101
|
Zhang Y, Shang H, Zhang J, Jiang Y, Li J, Xiong H, Chao T. Drug Treatment Direction Based on the Molecular Mechanism of Breast Cancer Brain Metastasis. Pharmaceuticals (Basel) 2025; 18:262. [PMID: 40006075 PMCID: PMC11859690 DOI: 10.3390/ph18020262] [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] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Today, breast cancer (BC) is the most frequently diagnosed malignancy and a leading cause of cancer-related deaths among women worldwide. Brain metastases (BMs) are a common complication among individuals with advanced breast cancer, significantly impacting both survival rates and the overall condition of life of patients. This review systematically analyzes the innovative approaches to drug treatment for breast cancer brain metastases (BCBMs), with particular emphasis placed on treatments targeting molecular mechanisms and signaling pathways and drug delivery strategies targeting the blood brain barrier (BBB). The article discusses various drugs that have demonstrated effectiveness against BCBM, featuring a mix of monoclonal antibodies, nimble small-molecule tyrosine kinase inhibitors (TKIs), and innovative antibody-drug conjugates (ADCs). This study of various drugs and techniques designed to boost the permeability of the BBB sheds light on how these innovations can improve the treatment of brain metastases. This review highlights the need to develop new therapies for BCBM and to optimize existing treatment strategies. With a deeper comprehension of the intricate molecular mechanisms and advances in drug delivery technology, it is expected that more effective personalized treatment options will become available in the future for patients with BCBM.
Collapse
Affiliation(s)
- Yumin Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Haotian Shang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Jiaxuan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yizhi Jiang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Jiahao Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (H.S.); (Y.J.); (J.L.)
| |
Collapse
|
102
|
Wang JF, Wang MC, Jiang LL, Lin NM. The neuroscience in breast cancer: Current insights and clinical opportunities. Heliyon 2025; 11:e42293. [PMID: 39975839 PMCID: PMC11835589 DOI: 10.1016/j.heliyon.2025.e42293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/21/2025] Open
Abstract
The involvement of nerves in the development of breast cancer has emerged as a significant factor. Interaction between the nervous system and breast cancer can influence tumor initiation, growth, invasion, metastasis, reverse resistance to drugs, promote inflammation in tumors, and impair the immune system's ability to combat cancer. This review examined the intricate relationship linking the nervous system with breast cancer, emphasizing both central and peripheral aspects of the nervous system. Moreover, we reviewed neural cell factors and their impact on breast cancer progression, alongside the interactions between nerves and immunology, microbiota in breast cancer. Furthermore, the study discussed the potential of nerves as biomarkers for diagnosing and prognosticating breast cancer, and evaluated prospects for improving chemotherapy and immunotherapy therapeutic outcomes in breast cancer treatment. We hope to provide a deeper understanding of the neurobiological underpinnings of breast cancer and pave the way for the discovery of innovative therapeutic targets and prognostic markers.
Collapse
Affiliation(s)
- Jia-feng Wang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Meng-chuan Wang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, China
| | - Lei-lei Jiang
- The First Affiliated Hospital of Anhui University of Chinese Medicine,Hefei, 230031, China
| | - Neng-ming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, 310024, China
| |
Collapse
|
103
|
Soosaipillai A, Nardi-Agmon I, Brinc D, Fabros A, Kavsak PA, Thavendiranathan P, Di Meo A. Macrotroponin interference and association with cardiotoxicity in patients receiving cardiotoxic breast cancer therapy: a pilot study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:18. [PMID: 39953627 PMCID: PMC11827144 DOI: 10.1186/s40959-025-00314-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Cancer therapy-related cardiac dysfunction (CTRCD) is an important adverse effect in patients receiving potential cardiotoxic cancer therapies. Interpretation of cardiac troponin results can be affected by presence of macrotroponin, which can complicate CTRCD assessment. We aimed to assess whether macrotroponin is detectable in women with ERBB2 + breast cancer receiving sequential therapy with anthracyclines and trastuzumab. METHODS A total of 20 serum samples from 12 ERBB2 + breast cancer patients (median age: 55 years, range: 30-69 years) who exhibited a significant increase in high-sensitivity cardiac troponin I (hs-cTnI) from baseline to post-anthracycline (~ 2 months after therapy initiation) and/or 3-months into trastuzumab therapy (~ 5 months after therapy initiation) and/or who had at least one hs-cTnI value above the female-specific 99th percentile (hs-cTnI > 16 ng/L) and had available banked blood for analysis were included in this pilot study. Samples were analyzed using the Abbott STAT High-Sensitive Troponin-I and Roche Elecsys Troponin T hs STAT assays. Macrotroponin was detected by treating the sample with protein G and re-measuring hs-cTn. Macrotroponin presence was defined as a hs-cTnI or hs-cTnT recovery of < 40% or 85%, respectively. RESULTS Macrotroponin was not identified after anthracycline treatment but was present in four patients 3-months into trastuzumab therapy, two of which had hs-cTnI concentrations above the 99th percentile. None of these patients exhibited a significant reduction in LVEF and/or GLS despite having significant elevations in hs-cTnI. CONCLUSIONS Clinicians should be cautious of benign hs-cTn elevations resulting from macrotroponin presence, as it can complicate CTRCD assessment.
Collapse
Affiliation(s)
- Andrea Soosaipillai
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Clinical Biochemistry, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Inbar Nardi-Agmon
- Department of Medicine, Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Davor Brinc
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Clinical Biochemistry, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Anselmo Fabros
- Division of Clinical Biochemistry, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Peter A Kavsak
- Deparment of Pathology and Molecular Medicine, Division of Clinical Pathology, McMaster University, Hamilton, ON, Canada
| | - Paaladinesh Thavendiranathan
- Department of Medicine, Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Ashley Di Meo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Division of Clinical Biochemistry, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
104
|
Xie H, Tan T, Li Q, Li T. Revolutionizing HER-2 assessment: multidimensional radiomics in breast cancer diagnosis. BMC Cancer 2025; 25:265. [PMID: 39953417 PMCID: PMC11829378 DOI: 10.1186/s12885-025-13549-7] [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: 11/06/2024] [Accepted: 01/17/2025] [Indexed: 02/17/2025] Open
Abstract
OBJECTIVE To explore the application value of multidimensional radiomics based on ultrasound imaging in assessing the HER-2 status of breast cancer. METHODS We retrospectively analyzed the ultrasound imaging, clinical, and laboratory data of 850 breast cancer patients from two centers. During the study, we first utilized automation technology to accurately delineate the tumor region of interest (ROI) in breast ultrasound imaging. Subsequently, the intra-tumoral ROI was automatically expanded by 1 cm and 2 cm to obtain larger areas including the peritumoral tissues, and further generated three-dimensional volumes of interest (VOI) within and around the tumor. Through the K-means clustering method, we identified the sub-regions of interest within the ROI and extracted corresponding radiomic features using the pyradiomics toolkit. Additionally, we employed an advanced Vision Transformer (VIT) model to perform deep radiomic feature extraction on the ROI. Based on feature selection, we utilized various machine learning algorithms for modeling and analysis to assess the HER-2 status of breast cancer. RESULTS After comprehensive comparison and evaluation of multiple models, we found that the diagnostic model based on multidimensional feature fusion exhibited excellent diagnostic performance in assessing the HER-2 status of breast cancer. In the training set, the model achieved an accuracy of 0.949 and an AUC value of 0.990 (95% CI: 0.986-0.995), with outstanding key performance indicators such as sensitivity, specificity, positive predictive value, negative predictive value, and F1 score. The model showed good generalization in the test set, with accuracy 0.747, AUC 0.848 (95% CI: 0.791-0.904), and sensitivity 0.911. Specificity was slightly lower, but other indicators remained high, and the F1 score was 0.703. Calibration and clinical decision curves further confirmed the model's effectiveness and reliability. CONCLUSION This study fully demonstrates that multidimensional breast ultrasonography-based radiomic features can effectively assess the HER-2 status of breast cancer. This finding not only provides new evidence for early diagnosis of breast cancer but also offers new ideas and methods for personalized treatment planning and prognosis assessment.
Collapse
Affiliation(s)
- Hui Xie
- Department of Radiation Oncology, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, P. R. China
- Faulty of Applied Sciences, Macao Polytechnic University, Macao, 999078, P. R. China
| | - Tao Tan
- Faulty of Applied Sciences, Macao Polytechnic University, Macao, 999078, P. R. China
| | - Qing Li
- Department of Radiation Oncology, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, P. R. China
- Key Experimental Project of Higher Education Institutes in Hunan Province (Key Laboratory of Tumor Precision Medicine), Chenzhou, 423000, P. R. China
- College of Medical Imaging, Laboratory Diagnostics, and Rehabilitation, Xiangnan University, Chenzhou, 423000, P. R. China
| | - Tao Li
- College of Medical Imaging, Laboratory Diagnostics, and Rehabilitation, Xiangnan University, Chenzhou, 423000, P. R. China.
- Department of Medical, Xiangnan University, Chenzhou, 423000, P. R. China.
| |
Collapse
|
105
|
Helal C, Djerroudi L, Ramtohul T, Laas E, Vincent-Salomon A, Jin M, Seban RD, Bieche I, Bello-Roufai D, Bidard FC, Cottu P, Loirat D, Carton M, Lerebours F, Kiavue N, Romano E, Bonneau C, Cabel L. Clinico-pathological factors predicting pathological response in early triple-negative breast cancer. NPJ Breast Cancer 2025; 11:15. [PMID: 39948122 PMCID: PMC11825670 DOI: 10.1038/s41523-025-00729-8] [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: 11/06/2024] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Pathological complete response (pCR) after neoadjuvant chemoimmunotherapy (NACi) is associated with improved patient outcomes in early triple-negative breast cancer (TNBC). This study aimed to identify factors associated with pCR after NACi. This cohort included all patients with stage II-III TNBC treated with NACi who underwent surgery at Institut Curie hospitals between 08/2021-06/2023. Among 208 patients, the overall pCR rate was 70% and was similar in ER < 1% (69%) and ER-low TNBC (73%, p = 0.6). In a multivariate model, Ki-67 ≥ 30% (OR 5.19 [1.73-17.3]), centralized TILs ≥ 30% (OR = 3.08 [1.42-7.04]), absence of DCIS at initial biopsy (OR = 2.56 [1.08-6.25]) and germline mutations in homologous recombination genes (OR = 9.50 [2.37-67.7]) remained strong independent predictors of pCR. These findings may guide treatment decisions in patients with TNBC undergoing NACi. Almost all patients with germline mutations in HR genes achieved pCR, supporting de-escalation trials. We suggest that ER-low tumors should be managed as TNBC tumors.
Collapse
Affiliation(s)
- Clara Helal
- Department of Medical Oncology, Institut Curie, Paris, France
| | | | | | - Enora Laas
- Department of Surgery, Institut Curie, Paris, France
| | - Anne Vincent-Salomon
- Department of Pathology, Institut Curie, Paris, France
- PSL University, Paris, France
| | - Maxime Jin
- Department of Radiology, Institut Curie, Paris, France
| | | | - Ivan Bieche
- Department of Genetic, Institut Curie, Paris, France
| | | | - Francois-Clement Bidard
- Department of Medical Oncology, Institut Curie, Paris, France
- Paris-Saclay University, UVSQ, Saint Cloud, France
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie, Paris, France
- Université Paris Cité, Paris, France
| | - Delphine Loirat
- Department of Medical Oncology, Institut Curie, Paris, France
| | | | | | - Nicolas Kiavue
- Department of Medical Oncology, Institut Curie, Paris, France
| | - Emanuela Romano
- Department of Medical Oncology, Institut Curie, Paris, France
- PSL University, Paris, France
- Department of Immunology, Institut Curie, Paris, France
| | - Claire Bonneau
- Department of Surgery, Institut Curie, Paris, France
- U900-STAMPM Team, Saint Cloud, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, Paris, France.
| |
Collapse
|
106
|
Bian K, Zhu Y, Wang Y, Ma Y, Ye Z. Cone-beam Breast CT Features Associated With Lymphovascular Invasion in Patients With Breast Cancer. Acad Radiol 2025:S1076-6332(25)00006-6. [PMID: 39952842 DOI: 10.1016/j.acra.2025.01.005] [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: 10/29/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 02/17/2025]
Abstract
RATIONALE AND OBJECTIVES To explore the association between contrast-enhanced cone-beam breast CT (CE-CBBCT) imaging features and lymphovascular invasion (LVI) status. MATERIALS AND METHODS Female patients with breast cancer by postoperative histopathology who underwent preoperative CE-CBBCT from August 2020 to December 2023 were retrospectively enrolled. Two radiologists reviewed the CBBCT image features independently. Clinicopathologic and CBBCT characteristics between LVI-positive and LVI-negative were compared by χ2 or Fisher's exact tests and Student's t or Mann-Whitney U test, as appropriate. Multivariate logistic regression analysis was performed to identify independent predictive factors of LVI. The receiver operating characteristic curve was used to evaluate predictive performance. RESULTS A total of 401 women were enrolled. LVI status of breast cancer was significantly associated with histologic type, Ki-67 index, adjacent vessel sign (AVS), increased ipsilateral whole-breast vascularity (IIV) number, and IIV degree (all p<0.05). In mass, calcification, AVS, IIV number, and IIV degree were significantly associated with LVI (all p<0.05). In non-mass enhancement (NME), AVS, IIV number, and IIV degree were associated with LVI (all p<0.05). Multivariate logistic regression showed AVS (OR=4.367, p<0.001) and IIV degree (OR of moderate and prominent IIV=4.732, 3.641, both p<0.005) as independent risk factors for LVI. Specifically, in mass, AVS (OR=4.397, p<0.001) and moderate-to-prominent IIV (OR=4.815, 3.563, both p<0.01) were independent predictors. For NME, moderate-to-prominent IIV (OR=13.695, 4.054, both p=0.001) was also an independent factor. The combined LVI prediction model which included AVS and IIV degree showed excellent performance (AUC=0.804). CONCLUSION CBBCT imaging features can help identify LVI status in breast cancer patients, which will guide the accurate planning of treatment management.
Collapse
Affiliation(s)
- Keyi Bian
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yueqiang Zhu
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Department of Medical Imaging, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, the Netherlands
| | - Yafei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yue Ma
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
107
|
Alves TM, Prata WM, Borin MC, de Carvalho AR, Dias CZ, Cherchiglia ML, de Figueiredo LO, de Assis Acurcio F, Alvares-Teodoro J, Júnior AAG. Seven-year overall survival of trastuzumabe versus alternative systemic therapies in a Brazilian breast cancer cohort. Sci Rep 2025; 15:5296. [PMID: 39939346 PMCID: PMC11821829 DOI: 10.1038/s41598-025-88575-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
This study analyzed breast cancer patients treated with trastuzumab in Brazil's unified health system (SUS) from 2008 to 2015. A non-concurrent cohort study using SUS data applied propensity score matching to reduce bias between trastuzumab and non-trastuzumab groups. Survival probabilities were estimated via Kaplan-Meier, with subgroup analysis using the log-rank test. Hazard ratios (HR) were calculated using Cox proportional hazards models. Among 20,852 patients, the overall survival rate was 92%, with 94% in the trastuzumab group and 90% in the non-trastuzumab group. Younger, black patients and those in the North region had poorer survival. Advanced disease stages and palliative treatments were linked to higher mortality, while adjuvant therapy and radiotherapy were protective. During follow-up, 8.1% of patients died, with better outcomes observed in the trastuzumab group (p < 0.0001). Late initiation of trastuzumab (after 16 months) improved survival, especially in early stages (I and II). Invasive tumors and stage IV disease were associated with worse prognoses. The study demonstrates trastuzumab's effectiveness in SUS, underscores survival disparities related to sociodemographic factors, and emphasizes the need for early detection, equitable access, and optimized treatment timelines to improve outcomes in public healthcare.
Collapse
Affiliation(s)
- Thaís Monteiro Alves
- Graduate Program in Medicines and Pharmaceutical Assistance, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| | - Wallace Mateus Prata
- Graduate Program in Medicines and Pharmaceutical Assistance, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Ezequiel Dias Foundation, Belo Horizonte, Brazil
| | - Marcus Carvalho Borin
- Graduate Program in Medicines and Pharmaceutical Assistance, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Carolina Zampirolli Dias
- Graduate Program in Public Health, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mariangela Leal Cherchiglia
- Graduate Program in Public Health, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Preventive and Social Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Francisco de Assis Acurcio
- Graduate Program in Medicines and Pharmaceutical Assistance, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Social Pharmacy, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Alvares-Teodoro
- Graduate Program in Medicines and Pharmaceutical Assistance, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Social Pharmacy, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Augusto Afonso Guerra Júnior
- Graduate Program in Medicines and Pharmaceutical Assistance, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil.
- Department of Social Pharmacy, School of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
108
|
Nie Y, Wei Y, Zhang Y, Liang Z, Lei Z, Chang M, Peng Y. Design and implication of a breast cancer-targeted drug delivery system utilizing the Kisspeptin/GPR54 system. Int J Pharm 2025; 670:125154. [PMID: 39755342 DOI: 10.1016/j.ijpharm.2024.125154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
Kisspeptins function as endogenous ligands for the G protein-coupled receptor GPR54. While the primary role of the Kisspeptin/GPR54 signaling pathway pertains to reproduction, several studies have shown that GPR54 is highly expressed in breast cancer, and we further confirmed this result that GPR54 expression is significantly upregulated in breast cancer cells. Based on this finding, we developed a liposomal drug delivery system utilizing the Kisspeptin/GPR54 system to treat breast cancer after confirming the safety of Kp-10-228. By surface-modifying liposomes with Kp-10-228 (228-K3-EG8-Liposome), we demonstrated enhanced accumulation of these liposomes in tumor cells, both in vitro and in vivo. Doxorubicin-loaded 228-K3-EG8-Liposome exhibited a remarkable inhibition of cancer cell proliferation, significantly extending the median survival time in mice with breast tumors compared to model mice treated with non-targeted liposomes or free doxorubicin. Our results suggest that the liposomal drug delivery system utilizing the Kisspeptin/GPR54 system is a promising novel strategy for the management of breast cancer.
Collapse
Affiliation(s)
- Yaoyan Nie
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, China; Institute of Biology, Gansu Academy of Sciences, Lanzhou, Gansu 730000, China
| | - Yanzhu Wei
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yuhuan Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zhuansheng Liang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zelin Lei
- The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Min Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Yali Peng
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, China.
| |
Collapse
|
109
|
Yang JS, Cao JM, Sun R, Zhou XJ, Chen ZH, Liu BW, Liu XF, Yu Y, Wang X. SMYD4 promotes MYH9 ubiquitination through lysine monomethylation modification to inhibit breast cancer progression. Breast Cancer Res 2025; 27:20. [PMID: 39930544 PMCID: PMC11812198 DOI: 10.1186/s13058-025-01973-3] [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: 10/22/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Breast cancer is the leading cause of female mortality worldwide. (SET And MYND Domain Containing 4) SMYD4 has been reported to be a tumour suppressor. However, the molecular mechanism of SMYD4 remains unclear. METHODS The expression level of SMYD4 in breast cancer cells was detected by qRT-PCR and western blot. The effect of SMYD4 was verified in vitro and in vivo. The interaction between SMYD4 and MYH9 was investigated by co‑IP assay. The regulation of SMYD4 on WNT signaling pathway was detected by luciferase reporter assay and ChIP analysis. RESULTS This study found that SMYD4 downregulation was associated with poor prognosis. SMYD4 was performed as a tumor suppressor both in vitro and in vivo. SMYD4 was found to interact with the downstream protein MYH9 and impede WNT signaling pathway. Further studies revealed that SMYD4 impeded the binding of MYH9 to the CTNNB1 promoter region by promoting lysine monomethylation and ubiquitination degradation of MYH9. CONCLUSIONS These findings reveal the emerging character of SMYD4 in Wnt/β‑catenin signaling and bring new sights of gene interaction. The discovery of this SMYD4/MYH9/CTNNB1/WNT/β-Catenin signalling pathway axis suggests that SMYD4 is a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Jin-Shuo Yang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jun-Ming Cao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Rui Sun
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xue-Jie Zhou
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Zhao-Hui Chen
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Bo-Wen Liu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xiao-Feng Liu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yue Yu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Xin Wang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Tianjin Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
110
|
Zhang J, Yuan C, Ma X. Efficacy and safety of different drugs in patients with HER2-positive gastric cancer: network meta-analysis. Syst Rev 2025; 14:40. [PMID: 39930467 PMCID: PMC11808970 DOI: 10.1186/s13643-025-02777-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/25/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND In the past decade, there has been a significant advancement in targeted therapy and immunotherapy, leading to the discovery of new drugs and changes in the treatment approach for patients with HER2-positive gastric cancer. Although several drugs are available for treating these patients, there is still no consensus on their selection, and there has been limited direct or indirect comparison among them. OBJECTIVE To address this gap, a network meta-analysis was conducted to assess the efficacy and safety of different drugs used in the treatment of HER2-positive gastric cancer. METHODS By searching through databases such as PubMed, Embase, Web of Science, and Cochrane Library, we identified 16 randomized controlled trials that involved a total of 4485 patients and utilized 9 different intervention measures. RESULTS Based on the current evidence, compared with chemotherapy alone, the hazard ratio (HR) of overall survival (OS) and progression-free survival (PFS) in gastric cancer patients treated with nivolumab were [hazard ratio (HR): 2.61 95%confidence interval (CI) (1.51, 4.51)] and [hazard ratio (HR): 2.01 95% confidence interval (CI) (1.18, 3.42)], respectively. Compared with chemotherapy alone, the hazard ratio (HR) of overall survival (OS) and progression-free survival (PFS) in gastric cancer patients treated with trastuzumab deruxtecan were [hazard ratio (HR): 1.7 95% confidence interval (CI) (1.13, 2.56)] and [hazard ratio (HR): 2.13 95% confidence interval (CI) (1.42, 3.22)], respectively. It is suggested that nivolumab and trastuzumab deruxtecan can effectively prolong overall survival (OS) and progression-free survival(PFS) in patients with HER2-positive gastric cancer, while also reducing the risk of adverse events to some extent. Therefore, these two regimens, nivolumab and trastuzumab deruxtecan, are considered to be effective and safe options for the treatment of patients with HER2-positive gastric cancer. CONCLUSIONS In previous studies, trastuzumab-based chemotherapy has been a common treatment for HER2-positive gastric cancer. To a certain extent, our study provides a reliable direction for future treatment options for HER2-positive gastric cancer. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42023420941.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, 222000, China
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, China
| | - Chunluan Yuan
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, 222000, China.
| | - Xiao Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
111
|
Lo C, Chang DY, Lu YS, Wang MY, Tsai LW, Huang CS, Tang CH, Lin CH. Benefit of adjuvant chemotherapy for T1cN0M0 and selected T1bN0M0 triple-negative breast cancer: a nationwide cancer registry-based study. Oncologist 2025; 30:oyae346. [PMID: 40042201 PMCID: PMC11880992 DOI: 10.1093/oncolo/oyae346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 11/13/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The efficacy of adjuvant chemotherapy for T1N0M0 triple-negative breast cancer (TNBC) has not been clearly elucidated. Thus, we aimed to evaluate the efficacy of adjuvant chemotherapy for patients with T1a-cN0M0 TNBC. PATIENTS AND METHODS Patients newly diagnosed with TNBC between 2011 and 2015 were identified and followed up until the end of 2020 using the Taiwan Cancer Registry. Univariate and multivariate Cox proportional hazards regression analyses were performed to compare the recurrence-free survival (RFS) and OS between patients who received and those who did not receive adjuvant chemotherapy. RESULTS Of the 62 483 patients registered during 2011-2015, 1074 patients with T1N0M0 TNBC (T1a, n = 103; T1b, n = 167; and T1c, n = 804) who underwent definitive breast surgery were included. Overall, 850 (79%) patients received adjuvant chemotherapy; these comprised 24.3%, 67.7%, and 88.6% of the patients with T1a, T1b, and T1c disease, respectively. Over a median follow-up of 7.18 years, a significant RFS and OS benefit from adjuvant chemotherapy was observed in the T1c subgroup but not in the T1a and T1b subgroups. However, subgroup analysis of T1b disease indicated that adjuvant chemotherapy yielded an OS benefit to patients with histological grade III disease (adjusted hazard ratio = 0.08, 95% CI, 0.01-0.77; P = .03). CONCLUSIONS Adjuvant chemotherapy improved the RFS and OS in patients with T1cN0M0 TNBC and improved the OS in patients with histological grade III T1bN0M0 disease. Our study advocates for the utilization of adjuvant chemotherapy in patients diagnosed with T1cN0M0 and histological grade III T1bN0M0 TNBC.
Collapse
Affiliation(s)
- Chiao Lo
- Department of Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100229, Taiwan
| | - Dwan-Ying Chang
- Department of Oncology, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Medical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| | - Yen-Shen Lu
- Department of Oncology, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Medical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| | - Ming-Yang Wang
- Department of Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Surgical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| | - Li-Wei Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100229, Taiwan
- Department of Surgical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Surgery, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| | - Chao-Hsiun Tang
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei 235603, Taiwan
| | - Ching-Hung Lin
- Department of Oncology, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Medical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| |
Collapse
|
112
|
Guo Z, Zhu Z, Luo M, Cao Y, Lin X, Wu Q, Wang S, Wang L, Zhou J. Efficacy of cyclin-dependent kinase inhibitors with concurrent proton pump inhibitors in patients with breast cancer: a systematic review and meta-analysis. Oncologist 2025; 30:oyae320. [PMID: 39963828 PMCID: PMC11833246 DOI: 10.1093/oncolo/oyae320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/09/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The impact of concurrent proton pump inhibitors (PPIs) use on the prognosis of patients with breast cancer undergoing cyclin-dependent kinase inhibitors (CDKIs) treatment is currently uncertain. Considerable divergence exists regarding the clinical studies. In this study, we aim to perform a comprehensive analysis to evaluate the influence of concomitant PPI use on the effectiveness and adverse effects of CDKIs in patients with breast cancer. METHODS This study encompassed all pertinent clinical studies published up to the present, following the PRISMA guidelines. The study used hazard ratio (HR) or odds ratio (OR) as a summary statistic and used fixed or random effects models for pooled estimation. RESULTS This study incorporated 10 research articles involving 2993 participants. Among patients with breast cancer undergoing treatment with CDKIs, the simultaneous administration of PPIs was associated with a notable reduction in overall survival (HR = 2.00; 95% CI, 1.35-2.96). Nevertheless, no substantial correlation was observed between the simultaneous utilization of PPIs and the progression-free survival (PFS) of patients (HR = 1.30; 95% CI, 0.98-1.74). PFS did not change significantly when considering different drugs, treatment lines, or regions alone. Furthermore, the simultaneous administration of PPIs was found to result in a notable decrease in the incidence of grades 3/4 risk factors (OR = 0.63, 95% CI, 0.46-0.85). CONCLUSION The concurrent administration of PPIs did not result in significant alterations in the risk of disease advancement among patients with breast cancer undergoing CDKIs treatment. The utilization of PPIs led to a decrease in the adverse effects linked to the administration of CDKIs.
Collapse
Affiliation(s)
- Zijie Guo
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, People’s Republic of China
| | - Ziyu Zhu
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, People’s Republic of China
| | - Mingpeng Luo
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, People’s Republic of China
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310014, People’s Republic of China
| | - Yijia Cao
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
| | - Xixi Lin
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, People’s Republic of China
| | - Qingliang Wu
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, People’s Republic of China
- The Ninth People’s Hospital of Hangzhou, Hangzhou, Zhejiang 310014, People’s Republic of China
| | - Shenkangle Wang
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, People’s Republic of China
| | - Linbo Wang
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, People’s Republic of China
| | - Jichun Zhou
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People’s Republic of China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, People’s Republic of China
| |
Collapse
|
113
|
Al-Balushi RA, Chaudhuri A, Kandimalla R, Haque A, Alenezi KM, Saeed M, Changez M, Al Harthy T, Al Hinaai M, Siddiqui S, Agrawal AK, Aqil F. In vitro anticancer effects of frankincense and its nanoemulsions for enhanced cancer cell targeting. Front Pharmacol 2025; 16:1403780. [PMID: 39981177 PMCID: PMC11839425 DOI: 10.3389/fphar.2025.1403780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 01/10/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Frankincense has demonstrated promising in vitro anticancer activity. However, its conventional delivery methods face significant challenges due to limited oral bioavailability. To address these limitations, this study focuses on developing optimized nanoemulsions (NEs) of Frankincense oil (FO) to enhance its therapeutic efficacy. Methods Frankincense resins were extracted and characterized using gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS), identifying key metabolites including isopinocarveol, α-thujene, p-cymene, carvone, germacrene A, and various methyl esters. FO-based nanoemulsions (FO-NEs) were prepared and optimized using a 3-factor, 3-level Box-Behnken Design (BBD), with 10% FO (v/v), 40% surfactant (cremophor EL), and co-surfactant (Transcutol P). The optimized FO-NEs were evaluated for particle size, polydispersity index (PDI), zeta potential, and morphology using scanning electron microscopy (SEM) and atomic force microscopy (AFM). Cytotoxicity, wound healing, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) assays were performed against breast cancer (MDA-MB-231, MDA-MB-231-TR) and lung cancer (A549, A549-TR, H1299) cell lines. Results The optimized FO-NEs exhibited an average particle size of 65.1 ± 4.21 nm, a PDI of 0.258 ± 0.04, and a zeta potential of -22.3 ± 1.2 mV. SEM and AFM confirmed the spherical morphology of the FO-NEs. In vitro cytotoxicity studies revealed enhanced anticancer activity of FO-NEs (IC50 = 13.2 μg/mL) compared to free FO (IC50 = 22.5 μg/mL) against resistant breast cancer MDA-MB-231-TR cells. FO-NEs significantly improved cancer cell internalization, disrupted mitochondrial membrane potential, and increased ROS generation, leading to enhanced cytotoxic effects. Discussion The results demonstrate that nanoemulsion-based delivery significantly enhances the bioactivity and cellular uptake of frankincense oil compared to its free form. FO-NEs exhibit potent anticancer activity, particularly against drug-resistant cancer cell lines, suggesting their potential as a viable strategy for improving the therapeutic efficacy of frankincense in cancer treatment.
Collapse
Affiliation(s)
- Rayya A. Al-Balushi
- Department of Basic and Applied Sciences, College of Applied and Health Sciences, A’Sharqiyah University, Ibra, Oman
| | - Aiswarya Chaudhuri
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, India
| | - Raghuram Kandimalla
- Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Ashanul Haque
- Department of Chemistry, College of Science, University of Hail, Hail, Saudi Arabia
| | - Khalaf M. Alenezi
- Department of Chemistry, College of Science, University of Hail, Hail, Saudi Arabia
| | - Mohd. Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Mohammad Changez
- College of Health Sciences, University of Buraimi, Al Buraimi, Oman
| | - Thuraya Al Harthy
- Department of Basic and Applied Sciences, College of Applied and Health Sciences, A’Sharqiyah University, Ibra, Oman
| | - Mohammed Al Hinaai
- Department of Basic and Applied Sciences, College of Applied and Health Sciences, A’Sharqiyah University, Ibra, Oman
| | - Samra Siddiqui
- Department Health Services Management, College of Public Health and Health Informatics, University of Hail, Hail, Saudi Arabia
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, India
| | - Farrukh Aqil
- Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Department of Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
114
|
Liu L, Meng Z, Yan J. Efficacy of acupuncture in treating chemotherapy-related cognitive impairment in breast cancer patients: A systematic review and meta-analysis protocol. PLoS One 2025; 20:e0318984. [PMID: 39913618 PMCID: PMC11801641 DOI: 10.1371/journal.pone.0318984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Chemotherapy-related cognitive impairment (CRCI) is a common adverse effect of chemotherapy in breast cancer patients, with a high incidence that significantly impacts treatment adherence and quality of life. Currently, there is no definitive and effective treatment for CRCI. Research indicates that acupuncture may serve as a promising intervention for CRCI. This study aims to assess the effectiveness of acupuncture in treating CRCI. METHODS This study is guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Protocols (PRISMA-P). From the time of database construction to October 2024, we will search seven literature databases, three clinical study registry databases, and three sources of gray literature. The restricted languages are English and Chinese. We will use the Participants-Interventions-Comparators-Outcome-Conclusion-Study Design (PICOS) criteria to identify included studies and the RoB2 tool to assess the quality of the included studies. Data synthesis and meta-analysis will be performed using RevMan 5.3. DISCUSSION The results of this study will clarify the potential benefits of acupuncture in the treatment of CRCI and address existing disagreements about its efficacy. Furthermore, the findings will assist clinicians in making informed decisions regarding the treatment of CRCI. TRAIL REGISTRATION PROSPERO registration number: CRD42024568093.
Collapse
Affiliation(s)
- Lishi Liu
- The Second Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zihan Meng
- The Second Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jun Yan
- The Second Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
115
|
Rampogu S, Al-Antari MA, Oh TH, Shaik B. A review of six bioactive compounds from preclinical studies as potential breast cancer inhibitors. Mol Biol Rep 2025; 52:203. [PMID: 39907697 DOI: 10.1007/s11033-025-10300-0] [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: 06/19/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Breast cancer is one of the predominant causes of mortality in women worldwide. Although therapeutics such as surgery, chemotherapy, hormonal therapy, and radiotherapy have been used, they are associated with adverse effects or multidrug resistance. The use of natural compounds is a promising strategy, owing to their abundance and medicinal value. This review focuses on six natural compounds, namely cinnamaldehyde, diosmin, taxifolin, phloretin, arctigenin, and eugenol, and details their mechanisms of breast cancer inhibition based on in vitro and in vivo studies. These compounds generally promote apoptosis and cell cycle arrest, hinder metastasis and invasion, and decrease tumor growth. This review reinforces the use of natural compounds as therapeutics for breast cancer from their preclinical studies. These compounds might be promising for drug development due to their abundance, high reliability, and safety.
Collapse
Affiliation(s)
| | - Mugahed A Al-Antari
- Department of Artificial Intelligence, College of Software & Convergence Technology, Daeyang AI Center, Sejong University, Seoul, 05006, Republic of Korea
| | - Tae Hwan Oh
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Baji Shaik
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
116
|
Nie HJ, Fu YJ, Long S, Wang JY, Zhao WS, Zhai LH, Yang YL, Tan MJ, Hu H, Chen XH. Chemoproteomics reveals proteome-wide covalent and non-covalent targets of withaferin A. Acta Pharmacol Sin 2025:10.1038/s41401-024-01468-5. [PMID: 39900821 DOI: 10.1038/s41401-024-01468-5] [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: 06/25/2024] [Accepted: 12/22/2024] [Indexed: 02/05/2025]
Abstract
Withaferin A (WA), a natural product used in traditional medicine, has recently garnered attention because of its diverse pharmacological effects. However, the direct targets responsible for these effects remain elusive. The discovery of targets is usually serendipitous and research has predominantly concentrated on covalent interactions, overlooking non-covalent targets. The unbiased and proteome-wide mapping of WA-interacting proteins in living cells remains largely unexplored. We have developed a chemical proteomics platform that enabled profiling of the covalent/non-covalent interactome and target occupancy in disease-related cells, which was used to reveal the landscape of the targets of WA in triple-negative breast cancer (TNBC) cells. Analysis of the discovered high-occupancy targets suggested that WA was substantially involved in the RNA metabolism pathway, in addition to other biological processes. Moreover, we biochemically validated a selection of previously unknown high-occupancy targets from various important biological pathways, including the non-covalent target MVK and covalent targets HNRNPF and CKAP4, which all play critical roles in TNBC. Collectively, these findings provided a target map for comprehensive understanding of the anti-TNBC activity of WA, and present WA-targetable proteins as new avenues for pharmacological intervention in TNBC. We anticipate that this platform will be applicable for the unbiased profiling of the targets of WA in various other disease-related cell models, as well as for other bioactive electrophilic natural products in different pathophysiological systems.
Collapse
Affiliation(s)
- Hui-Jun Nie
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ying-Jie Fu
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Shang Long
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia-Yu Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wen-Si Zhao
- School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lin-Hui Zhai
- School of Medicine, Tongji University, Shanghai, 200433, China
| | - Yin-Long Yang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Min-Jia Tan
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Hu
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Xiao-Hua Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
117
|
Wu Y, Liu Y, Wu H, Tong M, Du L, Ren S, Che Y. Advances in Ultrasound-Targeted Microbubble Destruction (UTMD) for Breast Cancer Therapy. Int J Nanomedicine 2025; 20:1425-1442. [PMID: 39925678 PMCID: PMC11804227 DOI: 10.2147/ijn.s504363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
Breast cancer is one of the most common types of cancer in women worldwide and is a leading cause of cancer deaths among women. As a result, various treatments have been developed to combat this disease. Breast cancer treatment varies based on its stage and type of pathology. Among the therapeutic options, ultrasound has been employed to assist in the treatment of breast cancer, including radiation therapy, chemotherapy, targeted immunotherapy, hormonal therapy, and, more recently, radiofrequency ablation for early-stage and inoperable patients. One notable advancement is ultrasound-targeted microbubble destruction (UTMD), which is gradually becoming a highly effective and non-invasive anti-tumor modality. This technique can enhance chemical, genetic, immune, and anti-vascular therapies through its physical and biological effects. Specifically, UTMD improves drug transfer efficiency and destroys tumor neovascularization while reducing toxic side effects on the body during tumor treatment. Given these developments, the application of ultrasound-assisted therapy to breast cancer has gained significant attention from research scholars. In this review, we will discuss the development of various therapeutic modalities for breast cancer and, importantly, highlight the application of ultrasound microbubble-targeted disruption techniques in breast cancer treatment.
Collapse
Affiliation(s)
- Yunfeng Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Yuxi Liu
- Department of Ultrasound, Shandong Second Medical University Affiliated Hospital, Shan Dong, Weifang, People’s Republic of China
| | - Han Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Mengying Tong
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Linyao Du
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Shuangsong Ren
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Ying Che
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| |
Collapse
|
118
|
Hsu SW, Chiang SC, Hsu JC, Ko Y. Validation of Risk Models for Predicting Febrile Neutropenia Among Breast Cancer Patients Receiving Chemotherapy: A Real-World Study. Clin Ther 2025; 47:e1-e4. [PMID: 39643453 DOI: 10.1016/j.clinthera.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 09/19/2024] [Accepted: 11/07/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Breast cancer patients receiving chemotherapy may develop a serious complication called febrile neutropenia (FN). We aimed to validate and compare three existing FN prediction models for breast cancer patients receiving chemotherapy in Taiwan. PATIENTS AND METHODS This was a retrospective observational real-world study. Data were acquired from the clinical research databases of three study hospitals. Breast cancer patients who have received at least one antineoplastic chemotherapy drug were chosen for the analysis. For evaluating the occurrence of FN, we used both broad (a body temperature above 38°C with an absolute neutrophil count (ANC) below 0.5 × 109/L or a body temperature above 38°C with a diagnosis of neutropenia) and narrow definitions (having both fever and neutropenia diagnoses or having both neutropenia and infection diagnoses). Sensitivity, specificity, accuracy, positive predictive value (PPV), and negative predictive value (NPV) were calculated for each selected FN model. RESULTS Among the 1903 patients identified, when the broad and narrow definitions of FN were applied, 70 (3.7%) and 60 (3.2%) patients developed FN in the first cycle, respectively. Using the broad FN definition, Aagaard's model was the highest in sensitivity (90.0%), followed by Chantharakhit's (40.0%) and Chen's (7.2%); in specificity, Chen's (93.6%) was the highest. In addition, the accuracy was highest with the Chen model (90.4%). All three models' PPVs were low, ranging from 0.5% to 4.2%, but all three models' NPVs were over 96.3%. When the narrow FN definition was used, Chantharakhit's model showed a relatively high improvement in sensitivity (53.3%) and PPV (3.9%) while negligible increases or even slight decreases were seen in the other two models and in the other performance indicators of Chantharakhit's model. CONCLUSION The results of this study provide important information for clinicians when selecting models to identify patients at high-risk of FN. As the model performance observed was less than satisfactory, improving the prediction ability of the models is needed.
Collapse
Affiliation(s)
- Shu-Wei Hsu
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Department of Pharmacy, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Shao-Chin Chiang
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Pharmacy, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Jason C Hsu
- International Ph.D. Program in Biotech and Healthcare Management, College of Management, Taipei Medical University, Taipei, Taiwan; Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu Ko
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Research Center of Pharmacoeconomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
119
|
Buono G, Capozzi M, Caputo R, Lauro VD, Cianniello D, Piezzo M, Cocco S, Martinelli C, Verrazzo A, Tafuro M, Calderaio C, Calabrese A, Nuzzo F, Pagliuca M, Laurentiis MD. CAR-T cell therapy for breast cancer: Current status and future perspective. Cancer Treat Rev 2025; 133:102868. [PMID: 39798230 DOI: 10.1016/j.ctrv.2024.102868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025]
Abstract
Within the expanding therapeutic landscape for breast cancer (BC), metastatic breast cancer (MBC) remains virtually incurable and tend to develop resistance to conventional treatments ultimately leading to metastatic progression and death. Cellular immunotherapy (CI), particularly chimeric antigen receptor-engineered T (CAR-T) cells, has emerged as a promising approach for addressing this challenge. In the wake of their striking efficacy against hematological cancers, CAR-T cells have also been used where the clinical need is greatest - in patients with aggressive BCs. Unfortunately, current outcomes fall considerably short of replicating that success, primarily owing to the scarcity of tumor-specific antigens and the immunosuppressive microenvironment within BC. Herein, we provide an up-to-date overview of both preclinical and clinical data concerning the application of CAR-T cell therapy in BC. By surveying the existing literature, we discuss the prevailing constrains of this therapeutic approach and overview possible strategies to advance it in the context of breast malignancies. Possible approaches include employing synthetic biology to refine antigen targeting and mitigate off-target toxicity, utilizing logic-gated CAR constructs to enhance specificity, and leveraging armored CARs to remodel the tumor micro-environment. Temporal and spatial regulation of CAR-T cells using inducible gene switches and external triggers further improves safety and functionality. In addition, promoting T cell homing through chemokine receptor engineering and enhancing manufacturing processes with universal CAR platforms expand therapeutic applicability. These innovations not only address antigen escape and T cell exhaustion but also optimize the efficacy and safety profile of CAR-T cell therapy. We, therefore, outline a trajectory wherein CAR-T cells may evolve from a promising experimental approach to a standard modality in BC therapy.
Collapse
Affiliation(s)
- Giuseppe Buono
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Monica Capozzi
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Roberta Caputo
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Vincenzo Di Lauro
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | | | - Michela Piezzo
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Stefania Cocco
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Claudia Martinelli
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy; Clinical and Translational Oncology, Scuola Superiore Meridionale (SSM), Napoli, Italy
| | - Annarita Verrazzo
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy; Clinical and Translational Oncology, Scuola Superiore Meridionale (SSM), Napoli, Italy
| | - Margherita Tafuro
- Clinical and Translational Oncology, Scuola Superiore Meridionale (SSM), Napoli, Italy; Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Claudia Calderaio
- Clinical and Translational Oncology, Scuola Superiore Meridionale (SSM), Napoli, Italy; Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | | | - Francesco Nuzzo
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Martina Pagliuca
- Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli, Italy; Clinical and Translational Oncology, Scuola Superiore Meridionale (SSM), Napoli, Italy; Université Paris-Saclay, Gustave Roussy, INSERM, Molecular Predictors and New Targets in Oncology, Villejuif, France.
| | | |
Collapse
|
120
|
Nash SH, Verhage E, McDowell BD, Neuner J, Chrischilles E, Lizarraga IM, Schroeder M. Body Weight and Breast Cancer Treatment Experiences: Results From the Share Thoughts on a Breast Cancer Study. Cancer Med 2025; 14:e70628. [PMID: 39905674 PMCID: PMC11794236 DOI: 10.1002/cam4.70628] [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: 07/02/2024] [Revised: 01/03/2025] [Accepted: 01/17/2025] [Indexed: 02/06/2025] Open
Abstract
PURPOSE Differences in breast cancer recurrence and survival occur by body size; the role of treatment differences in these disparities has been underexplored. Our objective was to evaluate differences in treatments received, patient experiences of care, and treatment decision-making processes among breast cancer survivors by body size. METHODS We used data from the Share Thoughts on Breast Cancer study. Participants (n = 1198) completed a survey that included information on demographics, treatments received, quality of care, and decision-making. We used descriptive statistics to evaluate differences in survey response by BMI category, and multivariable-adjusted multinomial and logistic regression to examine associations of BMI with treatments received. RESULTS Those with higher BMI were more likely to be older, report fair/poor health, not have a college-level education, be non-white, not be insured, have an income under $50,000, be unemployed, and report a history of several chronic diseases. Although there were unadjusted associations, after adjustment, women with obesity were not significantly less likely to receive mastectomy [OR 0.79 (0.50, 1.26) and OR 0.66 (0.38, 1.16), for BMI 30-35 and 35+ kg/m2 respectively] or contralateral prophylactic mastectomy [OR 0.92 (0.59, 1.44) and OR 0.80 (0.46, 1.39)] than those without obesity. Similarly, we found no association of BMI with reconstructive surgery [OR 0.97 (0.58, 1.60) and OR 0.58 (0.30, 1.11)] after adjustment. Women with obesity were less likely to report that their breast cancer care was excellent or very good (p = 0.026). CONCLUSIONS We observed no differences in breast cancer treatments received by BMI after adjustment for key covariates in this study sample. Further research is necessary to determine why quality of care may be perceived as lower among women with obesity.
Collapse
Affiliation(s)
- Sarah H. Nash
- Department of Epidemiology, College of Public HealthUniversity of IowaIowa CityIowaUSA
- Holden Comprehensive Cancer CenterUniversity of IowaIowa CityIowaUSA
| | - Elizabeth Verhage
- Department of Epidemiology, College of Public HealthUniversity of IowaIowa CityIowaUSA
| | | | - Joan Neuner
- Department of Internal MedicineMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Elizabeth Chrischilles
- Department of Epidemiology, College of Public HealthUniversity of IowaIowa CityIowaUSA
- Holden Comprehensive Cancer CenterUniversity of IowaIowa CityIowaUSA
| | - Ingrid M. Lizarraga
- Holden Comprehensive Cancer CenterUniversity of IowaIowa CityIowaUSA
- Department of Surgical Oncology, College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Mary Schroeder
- Division of Health Services Research, College of PharmacyUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
121
|
Zarei A, Moradi S, Hosseinzadeh L, Salavati MB, Jalilian F, Shahlaei M, Sadrjavadi K, Adibi H. Synthesis, characterization, cytotoxic investigation of curcumin-based chromene derivatives and study of DNA interaction via experimental and computational methods. J Mol Struct 2025; 1322:140331. [DOI: 10.1016/j.molstruc.2024.140331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
|
122
|
Zhang W, Li Z, Huang Y, Zhao J, Guo S, Wang Q, Guo S, Li Q. Complex Role of Circulating Triglycerides in Breast Cancer Onset and Survival: Insights From Two-Sample Mendelian Randomization Study. Cancer Med 2025; 14:e70698. [PMID: 39960141 PMCID: PMC11831496 DOI: 10.1002/cam4.70698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/20/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Reducing the incidence of breast cancer and improving its prognosis have become significant challenges for the global public health sector. We aimed to investigate the role of circulating triglycerides in the occurrence and survival of patients with breast cancer, while focusing on the possible differential effects by molecular subtypes of breast cancer. METHODS We used a Mendelian randomization approach to analyze publicly accessible genome-wide association study data, including triglyceride levels, breast cancer risk, and survival prognosis. We performed a two-sample causality inference analysis using the inverse-variance weighted method. We used both Mendelian randomization-Egger regression and weighted median methods for model verification. Heterogeneity was evaluated using Cochran's Q test, and sensitivity analyses were performed using the leave-one-out method, Mendelian randomization-Egger intercept test, and Mendelian Randomization Pleiotropy RESidual Sum and Outlier test. RESULTS The results revealed a negative causal relationship between triglyceride levels and overall breast cancer risk (odds ratio [OR] = 0.94, confidence interval [CI] = 0.89-0.99, p = 0.011), luminal A breast cancer risk (OR = 0.93, CI = 0.87-0.99, p = 0.014), and human epidermal growth factor receptor 2 (HER2)-enriched breast cancer risk (OR = 0.84, CI = 0.73-0.96, p = 0.010). However, no statistically significant correlations were observed for the luminal B, luminal B HER2-negative, and triple-negative subtypes. Furthermore, triglyceride levels showed a positive causal relationship with the risk of survival prognosis in patients with estrogen receptor-negative breast cancer (OR = 1.33, CI = 1.00-1.76, p = 0.047). However, no statistically significant impact was observed on the survival of patients with overall breast cancer or patients with estrogen receptor-positive, HER2-positive, and HER2-negative breast cancer. CONCLUSIONS The potentially complex role of circulating triglycerides in the incidence and survival of patients with breast cancer provides a new perspective on the heterogeneity of the effects of triglycerides on breast cancer, thereby promoting the development of precise medical strategies. Moreover, our findings contribute to an increased understanding of overall health among patients and clinicians alike.
Collapse
Affiliation(s)
- Wu Zhang
- Fourth Department of OncologyHebei General HospitalShijiazhuangChina
- Graduate SchoolNorth China University of Science and TechnologyTangshanHebeiChina
| | - Zhiru Li
- Fourth Department of OncologyHebei General HospitalShijiazhuangChina
- Graduate SchoolNorth China University of Science and TechnologyTangshanHebeiChina
| | - Yuquan Huang
- Department of PathologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Jing Zhao
- Sixth Department of OncologyHebei General HospitalShijiazhuangHebeiChina
| | - Shaowei Guo
- Fourth Department of OncologyHebei General HospitalShijiazhuangChina
| | - Qian Wang
- Fourth Department of OncologyHebei General HospitalShijiazhuangChina
| | - Sihan Guo
- Department of Computer ScienceDurham UniversityDurhamUK
| | - Qingxia Li
- Fourth Department of OncologyHebei General HospitalShijiazhuangChina
- Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
123
|
Earla JR, Kurian AW, Kponee-Shovein K, Mahendran M, Song Y, Hua Q, Hilts A, Sun Y, Hirshfield KM, Robson M, Mejia JA. Correlation Between Disease-Free Survival Endpoints and Overall Survival in Elderly Patients with Early-Stage HER2-Negative Breast Cancer: A SEER-Medicare Analysis. Adv Ther 2025; 42:886-903. [PMID: 39680314 PMCID: PMC11787175 DOI: 10.1007/s12325-024-03074-7] [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: 09/20/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION Recent trial-level meta-analyses have established disease-free survival (DFS) as a valid surrogate for overall survival (OS) in human epidermal growth factor receptor 2-negative (HER2-) breast cancer (BC), irrespective of disease stage, and in early-stage hormone receptor-positive (HR+)/HER2- BC. To advance the understanding of the association between additional DFS endpoints and OS, this study assessed the patient-level correlations between DFS and OS, invasive DFS (IDFS) and OS, and distant DFS (DDFS) and OS in Medicare beneficiaries with early-stage HER2- BC, overall and in subgroups of patients with HR+/HER2- BC and triple-negative BC (TNBC). METHODS Patients with stages I-III HER2- BC aged ≥ 66 years were identified from SEER-Medicare data (2010-2019). DFS, IDFS, DDFS, and OS were assessed using Kaplan-Meier analyses. Normal scores rank correlation was estimated between each DFS endpoint and OS, overall and separately in patients with HR+/HER2- BC and TNBC. RESULTS Of 28,655 patients, 90.4% had HR+/HER2- BC and 9.6% had TNBC (median follow-up 4 years). Median DFS, IDFS, and DDFS were 4.5, 5.9, and 6.3 years, respectively, in HR+/HER2- BC and 3.0, 3.8, and 4.4 years, respectively, in TNBC. Median OS was not reached (5-year OS, HR+/HER2- BC 83.7%; TNBC 67.7%). A significant positive correlation was observed between each DFS endpoint and OS across cohorts, with the strongest correlation observed between DDFS and OS in HR+/HER2- BC (correlation coefficient 0.60; 95% confidence interval 0.57-0.62; p < 0.001) and in TNBC (0.69; 0.65-0.71; p < 0.001). CONCLUSION We observed significant positive patient-level correlations between DFS and OS, IDFS and OS, and DDFS and OS in early-stage HER2- BC. Our IDFS and DDFS findings advance the understanding of the role of these DFS endpoints as predictors of OS, and their potential utility as surrogate endpoints in clinical trials of early-stage HER2- BC, given additional validation in trial-level meta-analyses.
Collapse
Affiliation(s)
| | | | | | | | - Yan Song
- Analysis Group, Inc, Boston, MA, USA
| | - Qi Hua
- Analysis Group, Inc, Boston, MA, USA
| | | | - Yezhou Sun
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| | | | - Mark Robson
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jaime A Mejia
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| |
Collapse
|
124
|
Kim J, Munster PN. Estrogens and breast cancer. Ann Oncol 2025; 36:134-148. [PMID: 39522613 DOI: 10.1016/j.annonc.2024.10.824] [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: 06/03/2024] [Revised: 09/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Estrogens have been associated with an increase in breast cancer risk. Yet emerging clinical and experimental evidence points to progestogens [endogenous progesterone or synthetic progesterone (progestin)] as the primary hormonal driver underlying seemingly estrogen-associated breast cancer risk. Estrogens may contribute to breast cancer risk indirectly by induction of the progesterone receptor and thus amplifying progesterone signaling. Large studies of hormonal contraceptives suggest that the small increase in breast cancer risk from hormonal contraceptives is mainly attributable to progestins, not estrogens. Estrogen-plus-progestin hormone replacement therapy (HRT) has consistently shown an increase in breast cancer risk among postmenopausal women, whereas estrogen-alone HRT has little impact on breast cancer risk in naturally or surgically menopausal women. In particular, the long-term follow-up of the Women's Health Initiative (WHI) randomized trials suggests a benefit of estrogen alone. Recent data further indicate that endogenously elevated estrogen during assisted reproductive technology (ART) exhibits little adverse effect on or potentially a reduction in breast cancer risk and recurrence. Also, accumulating evidence suggests that inhibition of progesterone signaling is a critical mechanism underlying the risk-reducing and therapeutic effects of antiestrogens. Estrogen HRT has shown an array of proven benefits, including ameliorating menopausal symptoms and improving bone health. Collective evidence thus suggests that estrogen HRT is likely to offer health benefits to perimenopausal or postmenopausal women, including breast cancer survivors, as well as young BRCA1/2 carriers with prophylactic oophorectomy for ovarian cancer prevention.
Collapse
Affiliation(s)
- J Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, USA.
| | - P N Munster
- Department of Medicine, Center for BRCA Research, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, USA
| |
Collapse
|
125
|
Sun R, Wang M, Zeng T, Chen H, Yoshitomi T, Takeguchi M, Kawazoe N, Yang Y, Chen G. Scaffolds functionalized with matrix metalloproteinase-responsive release of miRNA for synergistic magnetic hyperthermia and sensitizing chemotherapy of drug-tolerant breast cancer. Bioact Mater 2025; 44:205-219. [PMID: 39502841 PMCID: PMC11535879 DOI: 10.1016/j.bioactmat.2024.10.011] [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: 07/24/2024] [Revised: 09/19/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
Combining hyperthermia and chemotherapy for maximum anticancer efficacy remains a challenge because drug-tolerant cancer cells often evade this synergistic treatment due to drug resistance and asynchronous drug release. In this study, multifunctional scaffolds were designed to efficiently treat drug-tolerant breast cancer by improving the sensitization of breast cancer cells and synchronizing anticancer drug release with magnetic hyperthermia. The scaffolds contained microRNA-encapsulated matrix metalloproteinase-cleavable liposomes, doxorubicin-encapsulated thermoresponsive liposomes and Fe3O4 nanoparticles. The scaffolds could release microRNA specifically to improve the sensitization of breast cancer cells to anticancer drugs. The scaffolds also showed excellent hyperthermia effects under alternating magnetic field irradiation. Moreover, doxorubicin release was synchronized with magnetic hyperthermia. In vitro and in vivo studies demonstrated that the scaffolds effectively reduced drug resistance and eliminated doxorubicin-tolerant MDA-MB-231 cells through the synergistic effect of magnetic hyperthermia and sensitizing chemotherapy. Additionally, the scaffolds could support the proliferation and adipogenic differentiation of stem cells for adipose tissue regeneration after killing cancer cells at a late therapeutic stage. These composite scaffolds offer an innovative strategy for treating breast cancer, with synergistic anticancer effects and regenerative functions.
Collapse
Affiliation(s)
- Rui Sun
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Man Wang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Tianjiao Zeng
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Huajian Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
| | - Toru Yoshitomi
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
| | - Masaki Takeguchi
- Research Center for Energy and Environmental Materials, National Institute for Materials Science, Ibaraki, 305-0047, Japan
| | - Naoki Kawazoe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, Ibaraki, 305-8572, Japan
| | - Guoping Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| |
Collapse
|
126
|
Sinha P, Yadav AK. Unraveling the anti-breast cancer activity of Cimicifugae rhizoma using biological network pathways and molecular dynamics simulation. Mol Divers 2025; 29:241-254. [PMID: 38615110 DOI: 10.1007/s11030-024-10847-3] [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: 02/06/2024] [Accepted: 03/12/2024] [Indexed: 04/15/2024]
Abstract
Cimicifugae is a commonly used treatment for breast cancer, but the specific molecular mechanisms underlying its effectiveness remain unclear. In this research, we employ a combination of network pharmacology, molecular docking, and molecular dynamics simulations to uncover the most potent phytochemical within Cimicifugae rhizoma in order to delve into its interaction with the target protein in breast cancer treatment. We identified 18 active compounds and 89 associated targets, primarily associated to various biological processes such as lipid metabolism, the signaling pathway in diabetes, viral infections, and cancer-related pathways. Molecular docking analysis revealed that the two most active compounds, Formononetin and Cimigenol, exhibit strong binding to the target protein AKT1. Through molecular dynamics simulations, we found that the Cimigenol-AKT1 complex exhibits greater structural stability and lower interaction energy compared to the stigmasterol-AKT1 complex. Our study demonstrates that Cimicifugae rhizoma exerts its effects in breast cancer treatment through a multi-component, multi-target synergistic approach. Furthermore, we propose that Cimigenol, targeting AKT-1, represents the most effective compound, offering valuable insights into the molecular mechanisms underpinning its role in breast cancer therapy.
Collapse
Affiliation(s)
- Prashasti Sinha
- Department of Physics, School of Physical & Decision Science, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Anil Kumar Yadav
- Department of Physics, School of Physical & Decision Science, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| |
Collapse
|
127
|
Dhiman P, Patil CN, Konimeni S, Meenu M. Triple-Negative, HER2/Neu, and Ki67 Markers in Breast Cancer Patients Undergoing Standard of Care Treatment in India: Real-World Evidence on Tumor Aggressiveness and Survival Outcomes. Cureus 2025; 17:e78798. [PMID: 40078245 PMCID: PMC11897781 DOI: 10.7759/cureus.78798] [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] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
INTRODUCTION Breast cancer is among the most prevalent cancers in women globally, with patients' survival adversely impacted by Ki67 expression and triple-negative phenotypes. In this study, we examined the relationship between HER2/neu, triple-negative, and Ki67 phenotypes and tumor aggressiveness along with the survival of breast cancer patients from India. MATERIALS AND METHODS A retrospective cohort study was performed using hospital-based data from a tertiary care center spanning January 2013 to August 2023. The study included breast cancer patients who received neoadjuvant chemotherapy based on preoperative assessment and/or adjuvant chemotherapy following postoperative evaluation. Patients with other primary cancers or those treated with investigational drugs were excluded. Data on variables such as age, parity, menopause, cancer stage and grade, estrogen receptor (ER), progesterone receptor (PR), HER2/neu, Ki67 score, and the use of biologicals, hormones, and chemoradiotherapy were analyzed using correlation and regression tests to identify factors associated with aggressive tumor behavior. Kaplan-Meier survival model and Cox-proportional hazard test were applied. RESULTS A total of 389 breast cancer patients with a mean age of 54.3 years met the eligibility criteria and were included in the analysis. A higher prevalence of hormone receptor positivity was observed among Indian patients than in Western countries. Younger, premenopausal women were more likely to present with high-grade tumors and high Ki67 scores, poorer overall survival, and the need for chemoradiotherapy. On the other hand, nulliparous patients mostly had triple-negative tumors with high Ki67 scores. Aggressive tumor behavior was linked to HER2/neu positivity, ER and PR negativity, and the requirement for both neoadjuvant and adjuvant chemotherapy. At the same time, premenopausal patients more frequently were candidates for radiation therapy. HER2/neu expression demonstrated a moderate negative correlation with co-expression of hormone receptors (ER and PR). Cancer grade was associated with Ki67 levels, nulliparity, triple-negative status, and hormone receptor expressions (ER and PR). Median survival was not reached for the study cohort. CONCLUSION Tumor aggressiveness was associated with a high Ki67 score, HER2/neu positivity, and the absence of hormone receptor expression in patients.
Collapse
Affiliation(s)
- Pravesh Dhiman
- Medical Oncology, All India Institute of Medical Sciences Bilaspur, Bilaspur, IND
| | - C N Patil
- Medical Oncology, Aster Cauvery Medical Institute (CMI) Hospital, Bengaluru, IND
| | - Sneha Konimeni
- Medical Oncology, Aster Cauvery Medical Institute (CMI) Hospital, Bengaluru, IND
| | - Meenakshi Meenu
- Pharmacology, All India Institute of Medical Sciences Bilaspur, Bilaspur, IND
| |
Collapse
|
128
|
Ni G, Sun Y, Jia H, Xiahou Z, Li Y, Zhao F, Zang H. MAZ-mediated tumor progression and immune evasion in hormone receptor-positive breast cancer: Targeting tumor microenvironment and PCLAF+ subtype-specific therapy. Transl Oncol 2025; 52:102280. [PMID: 39805182 PMCID: PMC11780959 DOI: 10.1016/j.tranon.2025.102280] [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: 11/03/2024] [Revised: 12/19/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Breast cancer had been the most frequently diagnosed cancer among women, making up nearly one-third of all female cancers. Hormone receptor-positive breast cancer (HR+BC) was the most prevalent subtype of breast cancer and exhibited significant heterogeneity. Despite advancements in endocrine therapies, patients with advanced HR+BC often faced poor outcomes due to the development of resistance to treatment. Understanding the molecular mechanisms behind this resistance, including tumor heterogeneity and changes in the tumor microenvironment, was crucial for overcoming resistance, identifying new therapeutic targets, and developing more effective personalized treatments. METHODS The study utilized single-cell RNA sequencing (scRNA-seq) data sourced from the Gene Expression Omnibus database and The Cancer Genome Atlas to analyze HR+BC and identify key cellular characteristics. Cell type identification was achieved through Seurat's analytical tools, and subtype differentiation trajectories were inferred using Slingshot. Cellular communication dynamics between tumor cell subtypes and other cells were analyzed with the CellChat. The pySCENIC package was utilized to analyze transcription factors regulatory networks in the identified tumor cell subtypes. The results were verified by in vitro experiments. A risk scoring model was developed to assess patient outcomes. RESULTS This study employed scRNA-seq to conduct a comprehensive analysis of HR+BC tumor subtypes, identifying the C3 PCLAF+ tumor cells subtype, which demonstrated high proliferation and differentiation potential. C3 PCLAF+ tumor cells subtype was found to be closely associated with cancer-associated fibroblasts through the MK signaling pathway, facilitating tumor progression. Additionally, we discovered that MAZ was significantly expressed in C3 PCLAF+ tumor cells subtype, and in vitro experiments confirmed that MAZ knockdown inhibited tumor growth, accentuating its underlying ability as a therapeutic target. Furthermore, we developed a novel prognostic model based on the expression profile of key prognostic genes within the PCLAF+/MAZ regulatory network. This model linked high PCLAF+ tumor risk scores with poor survival outcomes and specific immune microenvironment characteristics. CONCLUSION This study utilized scRNA-seq to reveal the role of the C3 PCLAF+ tumor cells subtype in HR+BC, emphasizing its association with poor prognosis and resistance to endocrine therapies. MAZ, identified as a key regulator, contributed to tumor progression, while the tumor microenvironment had a pivotal identity in immune evasion. The findings underscored the importance of overcoming drug resistance, recognizing novel treatment targets, and crafting tailored diagnosis regimens.
Collapse
Affiliation(s)
- Gaofeng Ni
- Department of Breast Surgery, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai 264003, China
| | - Yuwei Sun
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Hongling Jia
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing 100084, China
| | - Yumeng Li
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Fu Zhao
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Hongyan Zang
- Department of Breast Surgery, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
129
|
Chen E, Chen C, Chen Y, You J, Chen N, Xu S, Wang Q, Cai Y, Hu X, Li Q. Investigating HER2-Low in Early Breast Cancer: Prognostic Implications and Age-Related Prognostic Stratification. Cancer Med 2025; 14:e70637. [PMID: 39945279 PMCID: PMC11822451 DOI: 10.1002/cam4.70637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 01/07/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Recent studies about human epidermal growth factor receptor 2 (HER2)-low values have garnered great interest among oncologists. We aimed to investigate whether HER2-low impacts the prognosis of early-stage breast cancer overall and in specific subgroups, explore differences in clinicopathologic markers, and examine the role of age in HER2-low prognostic stratification. MATERIALS & METHODS We conducted a retrospective analysis of 6920 HER2-negative breast cancer patients from the First Affiliated Hospital of Wenzhou Medical University (2010-2022). The study focused on the impact of HER2-low status (immunohistochemistry +1 or +2, in situ hybridization not amplified) on overall survival (OS), considering the age at diagnosis. RESULTS Generally, HER2-low status correlated with less aggressive cancer indicators. No significant prognostic differences were observed between HER2-low and HER2-0 in the entire cohort, HR-positive, and HR-negative groups. However, in TNBC patients aged ≥ 65, HER2-low correlated with significantly better OS (HR = 0.45, 95% CI 0.24-0.83, p = 0.011), a finding consistent after multivariable adjustment (HR = 0.34, 95% CI 0.14-0.80, p = 0.014). In other subgroups, prognosis did not significantly correlate with HER2 status. The combination of HER2-low status and age plays a key role in prognostic stratification in TNBC. Patients aged ≥ 65 with HER2-0 had considerably poorer prognoses compared to other subgroups. CONCLUSION This extensive retrospective study demonstrates that HER2-low status cannot serve as an independent prognostic factor in the entire cohort, nor in the HR-positive and HR-negative groups individually. However, the combined factors of HER2-low status and age may indicate a potential contribution to the prognostic stratification of TNBC.
Collapse
Affiliation(s)
- Endong Chen
- Department of Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Chen Chen
- The First School of Medicine, School of Information and EngineeringWenzhou Medical UniversityZhejiangChina
| | - Yingying Chen
- Department of Colorectal and Anal SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Jie You
- Department of Thyroid SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Nan Chen
- The First School of Medicine, School of Information and EngineeringWenzhou Medical UniversityZhejiangChina
| | - Shenlin Xu
- The First School of Medicine, School of Information and EngineeringWenzhou Medical UniversityZhejiangChina
| | - Qingxuan Wang
- Department of Thyroid SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Yefeng Cai
- Department of Thyroid SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Xiaoqu Hu
- Department of Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Quan Li
- Department of Breast SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityZhejiangChina
| |
Collapse
|
130
|
Du J, Wang J, Ge F, Ma H, Zhu H, Du J, Yan F, He Q, Yang B, Yuan T, Zhu H. JOSD2 promotes breast cancer metastasis by deubiquitinating and stabilizing SMAD4. Biochem Pharmacol 2025; 232:116748. [PMID: 39793716 DOI: 10.1016/j.bcp.2025.116748] [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: 09/30/2024] [Revised: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Breast cancer is one of the most common malignant tumors among women worldwide, and its high degree of metastasis significantly impacts treatment effectiveness leading to poor prognosis. The potential molecular mechanisms underlying breast cancer metastasis remain to be further elucidated. In this study, via database analysis, we revealed that the deubiquitinase josephin domain containing 2 (JOSD2) was abnormally amplified in patients with metastatic breast cancer, and was significantly negatively correlated with patient prognosis. By integrating data from the Gene Expression Omnibus (GEO) database and Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway enrichment analysis, we found that the transforming growth factor beta (TGF-β) signaling pathway was significantly activated in breast cancer patients with increased JOSD2 expression. Further studies revealed that JOSD2 interacted with and stabilized SMAD family member 4 (SMAD4) by removing polyubiquitin chains. Inhibition of JOSD2 by RNA interference effectively inhibited the metastasis of breast cancer cells both in vitro and in vivo. In conclusion, our study not only reveals the role of JOSD2 in promoting breast cancer metastasis for the first time, but also indicates promising directions for the future development of deubiquitinase inhibitors, which could yield significant therapeutic benefits. Nevertheless, extensive research and development are required to fully realize this potential.
Collapse
Affiliation(s)
- Jiamin Du
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiao Wang
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fujing Ge
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongrui Ma
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongdao Zhu
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiangxia Du
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, Zhejiang, China
| | - Fangjie Yan
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Tao Yuan
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Hong Zhu
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
131
|
Lou Y, Fu B, Liu L, Song J, Zhu M, Xu C. The tRF-33/IGF1 axis dysregulates mitochondrial homeostasis in HER2-negative breast cancer. Am J Physiol Cell Physiol 2025; 328:C627-C638. [PMID: 39792150 DOI: 10.1152/ajpcell.00588.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/29/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025]
Abstract
Transfer RNA-derived small RNAs (tsRNAs), a recently identified noncoding RNA subset, are mainly classified into transfer RNA (tRNA)-derived small RNA fragments (tRFs) and tRNA-derived stress-induced RNAs (tiRNAs). tsRNAs dysregulation is frequently observed in numerous cancer types, suggesting involvement in tumorigenesis. However, their functions in breast cancer (BC) remain to be fully understood. Here, it was discovered that tRF-33-MEF91SS2PMFI0Q (tRF-33), derived from mature tRNA-LysTTT, was markedly upregulated in human epidermal receptor 2 (HER2)-negative BC cells and tissue samples. tRF-33 stimulated the proliferation, migration, and invasiveness of BC cells in vitro and facilitated tumor progression in vivo. Mechanistically, tRF-33 was found for the first time to bind directly to the 3'-UTR of IGF1, resulting in downregulation of both its mRNA and protein and thus affecting mitochondrial homeostasis and progression of BC. These results demonstrate a novel tsRNA modulatory mechanism and a potential direction for treating HER2-negative BC.NEW & NOTEWORTHY In this study, we identified differential expression of tRNA fragments in HER2-negative BC tissues compared with adjacent normal tissues, observing significant upregulation of an i-tRF type tRF-33-MEF91SS2PMFI0Q (tRF-33) in the tumor tissue. We also found that tRF-33 promoted tumorigenesis in BC cells. We demonstrated for the first time that IGF1 was a target gene of tRF-33 and also showed that the tRF-33/IGF1 axis impaired mitochondrial dynamics, thus affecting mitochondrial homeostasis and promoting HER2-negative BC progression.
Collapse
Affiliation(s)
- Yuming Lou
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| | - Bifei Fu
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| | - Lutong Liu
- Department of School of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Jialu Song
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| | - Mengying Zhu
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| | - Chaoyang Xu
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| |
Collapse
|
132
|
Li J, Lee ARYB, Tariq A, Lau G, Yau CE, Tan LL, Tyebally SM, Lee MX, Koo CY, Sia CH. Comparing Renin-Angiotensin-Aldosterone Blockade Regimens for Long-Term Chemotherapy-Related Cardiac Dysfunction: A Network Meta-Analysis. Cardiovasc Drugs Ther 2025; 39:171-186. [PMID: 37314568 DOI: 10.1007/s10557-023-07457-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 06/15/2023]
Abstract
PURPOSE Cancer therapies including trastuzumab and anthracyclines are cardiotoxic and cause cardiac dysfunction. To prevent cardiotoxicity, pharmacological agents used in heart failure have been administered concomitantly with cardiotoxic cancer therapy, but few studies to date have performed a head-to-head comparison of these different agents. This systematic review and network meta-analysis of randomized-controlled trials aims to evaluate the efficacy of renin-angiotensin-aldosterone system (RAAS) blockers, namely angiotensin-converting enzyme inhibitors (ACE-Is), aldosterone receptor blockers (ARBs), and mineralocorticoid receptor antagonists (MRAs), in primary prevention against chemotherapy-related cardiac dysfunction in patients receiving anthracyclines and/or trastuzumab. METHODS A systematic search was performed in major web databases for studies from inception to 15 September 2022. A Bayesian network meta-analysis model was used to assess the relative effects of competing treatments on the primary outcomes of risk of significant decline in left ventricular ejection fraction (LVEF) and mean LVEF decline. Secondary outcomes included left ventricular diastolic function, global longitudinal strain, and cardiac biomarkers. This study is registered with PROSPERO, CRD42022357980. RESULTS AND CONCLUSION Nineteen studies reported the effects of 13 interventions (N = 1905 patients). Only enalapril (RR 0.05, 95% CI 0.00-0.20) was associated with reduced risk of patients developing significant decline in LVEF relative to placebo. Subgroup analysis showed that the beneficial effect of enalapril was driven by protection against anthracycline-associated toxicity. In addition, no RAAS-inhibiting agents showed efficacy in protection against treatment with both anthracycline and trastuzumab. The use of RAAS inhibition therapy did not conclusively impact on other markers of cardiac function, including left ventricular diastolic function and cardiac biomarkers.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | - Areeba Tariq
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Grace Lau
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chun En Yau
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Ling Tan
- Department of Cardiology, National University Heart Center Singapore, Singapore, Singapore
| | - Sara Moiz Tyebally
- Department of Cardiology, Ng Teng Fong General Hospital, Singapore, Singapore
| | - Matilda Xinwei Lee
- Department of Hematology-Oncology, National University Cancer Institute Singapore, Singapore, Singapore
| | - Chieh Yang Koo
- Department of Cardiology, National University Heart Center Singapore, Singapore, Singapore
| | - Ching-Hui Sia
- Department of Cardiology, National University Heart Center Singapore, Singapore, Singapore.
| |
Collapse
|
133
|
Liu X, Ma R, Wei F, Wang M, Jiang Y, Zheng P, Cao Z. Tumor-derived exosomal lncRNA-MIR193BHG promotes bone metastasis of breast cancer by targeting the miR-489-3p/DNMT3A signaling axis in osteoclasts. J Transl Med 2025; 23:142. [PMID: 39891171 PMCID: PMC11786480 DOI: 10.1186/s12967-025-06156-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/19/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Breast cancer exhibits high incidence and mortality among women, with distant metastasis, especially bone metastasis, being the leading cause of death. Despite advances in adjuvant therapies, bone metastasis remains a challenge for patient survival and quality of life. Exosomes, small vesicles capable of mediating intercellular communication, play a crucial role in tumor metastasis. RESULTS This study investigated the role of tumor-derived exosomal long noncoding RNA (lncRNA)-MIR193BHG in breast cancer bone metastasis. LncRNA-MIR193BHG was delivered to osteoclasts via exosomes and promoted osteoclast formation and activity by targeting the miR-489-3p/DNA methyltransferase 3A (DNMT3A) signaling axis, thereby accelerating breast cancer-induced osteolysis. Knockdown experiments demonstrated that reducing the levels of exosomal lncRNA-MIR193BHG significantly inhibited osteoclast differentiation and bone resorption, which was confirmed both in vitro and in vivo. Additionally, mechanistic studies revealed that lncRNA-MIR193BHG acted as a competitive endogenous RNA (ceRNA) interacting with miR-489-3p, regulating DNMT3A expression and subsequently affecting osteoclast differentiation. CONCLUSIONS These findings suggest that lncRNA-MIR193BHG plays a critical regulatory role in breast cancer bone metastasis, and the lncRNA-MIR193BHG/miR-489-3p/DNMT3A signaling axis could be a potential target for the treatment of breast cancer bone metastasis. Future studies should further explore the broader applicability of this mechanism and its clinical feasibility.
Collapse
Affiliation(s)
- Xiaoya Liu
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Rui Ma
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Feng Wei
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Maihuan Wang
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yiwei Jiang
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Peng Zheng
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China.
| | - Zhen Cao
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China.
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
134
|
Bartkowiak K, Mohammadi PM, Nissen P, Werner S, Agorku D, Andreas A, Geffken M, Peine S, Verpoort K, Deutsch TM, Michel LL, Schneeweiss A, Thewes V, Trumpp A, Hardt O, Müller V, Riethdorf S, Schlüter H, Pantel K. Discovery of a sushi domain-containing protein 2-positive phenotype in circulating tumor cells of metastatic breast cancer patients. Sci Rep 2025; 15:3913. [PMID: 39890941 PMCID: PMC11785953 DOI: 10.1038/s41598-025-87122-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Cell lines derived from circulating tumor cells (CTCs) in the blood provide important biological information on cancer metastasis. CTC-ITB-01 is a CTC cell line derived from a patient with metastatic estrogen receptor-alpha (ER-alpha) positive breast cancer two months before the death of the patient. After a LC-MC/MS based proteomics analysis of CTC-ITB-01, we found extraordinary high levels of the poorly characterized protein SUSD2 (sushi domain-containing protein 2) in CTC-ITB-01. Expression of SUSD2 on subsets of CTCs was validated on clinical blood samples of patients with metastatic breast cancer. SUSD2-positive CTCs could be captured specifically by a MACS-based approach. We overexpressed SUSD2 in the poorly-metastatic cell line MCF-7. This resulted in upregulation of ER-alpha, the tumor progression protein GRP78 (78-kDa glucose-regulated protein) and downregulation of the tumor suppressor protein PDCD4 (programmed cell death protein 4). We observed downregulation of SUSD2 and PDCD4 after hypoxia and simulation of re-oxygenation in the blood in MCF-7 and MDA-MB-468, while in CTC-ITB-01 SUSD2 levels remained unchanged, and only PDCD4 was downregulated under hypoxia. In conclusion, we show, for the first time, that SUSD2 is expressed in CTCs and appears to affect key proteins in tumor progression and survival.
Collapse
Affiliation(s)
- Kai Bartkowiak
- Department for Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Parinaz Mossahebi Mohammadi
- Department for Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Paula Nissen
- Section Mass Spectrometry and Proteomics, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Stefan Werner
- Department for Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg- Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - David Agorku
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429, Bergisch Gladbach, Germany
| | - Antje Andreas
- Department for Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Maria Geffken
- Department of Transfusion Medicine, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Sven Peine
- Department of Transfusion Medicine, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Karl Verpoort
- Superregional group practice for hematology and oncology, Hohe Weide 17 b, 20295, Hamburg, Germany
| | - Thomas M Deutsch
- Department of Obstetrics and Gynecology, University of Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany
| | - Laura L Michel
- National Center for Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Verena Thewes
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ- ZMBH Alliance, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Olaf Hardt
- Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429, Bergisch Gladbach, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Sabine Riethdorf
- Department for Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometry and Proteomics, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Klaus Pantel
- Department for Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
135
|
Zhang M, Parker J, An L, Liu Y, Sun X. Flexible analysis of spatial transcriptomics data (FAST): a deconvolution approach. BMC Bioinformatics 2025; 26:35. [PMID: 39891065 PMCID: PMC11786350 DOI: 10.1186/s12859-025-06054-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
MOTIVATION Spatial transcriptomics is a state-of-art technique that allows researchers to study gene expression patterns in tissues over the spatial domain. As a result of technical limitations, the majority of spatial transcriptomics techniques provide bulk data for each sequencing spot. Consequently, in order to obtain high-resolution spatial transcriptomics data, performing deconvolution becomes essential. Most existing deconvolution methods rely on reference data (e.g., single-cell data), which may not be available in real applications. Current reference-free methods encounter limitations due to their dependence on distribution assumptions, reliance on marker genes, or the absence of leveraging histology and spatial information. Consequently, there is a critical need for the development of highly flexible, robust, and user-friendly reference-free deconvolution methods capable of unifying or leveraging case-specific information in the analysis of spatial transcriptomics data. RESULTS We propose a novel reference-free method based on regularized non-negative matrix factorization (NMF), named Flexible Analysis of Spatial Transcriptomics (FAST), that can effectively incorporate gene expression data, spatial, and histology information into a unified deconvolution framework. Compared to existing methods, FAST imposes fewer distribution assumptions, utilizes the spatial structure information of tissues, and encourages interpretable factorization results. These features enable greater flexibility and accuracy, making FAST an effective tool for deciphering the complex cell-type composition of tissues and advancing our understanding of various biological processes and diseases. Extensive simulation studies have shown that FAST outperforms other existing reference-free methods. In real data applications, FAST is able to uncover the underlying tissue structures and identify the corresponding marker genes.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Mathematics, University of Arizona, 617 N. Santa Rita Ave., Tucson, AZ, 85721, USA
| | - Joel Parker
- Department of Epidemiology and Biostatistics, University of Arizona, 1295 N. Martin Ave., Tucson, AZ, 85721, USA
| | - Lingling An
- Department of Agricultural and Biosystems Engineering, University of Arizona, 1177 East Fourth Street, Tucson, AZ, 85721, USA
| | - Yiwen Liu
- Department of Epidemiology and Biostatistics, University of Arizona, 1295 N. Martin Ave., Tucson, AZ, 85721, USA.
| | - Xiaoxiao Sun
- Department of Epidemiology and Biostatistics, University of Arizona, 1295 N. Martin Ave., Tucson, AZ, 85721, USA.
| |
Collapse
|
136
|
Wang F, Wang Y, Xiong B, Yang Z, Wang J, Yao Y, Yu L, Fu Q, Li L, Zhang Q, Zheng C, Huang S, Liu L, Liu C, Sun H, Mao B, Liu DX, Yu Z. Neoadjuvant pyrotinib and trastuzumab in HER2-positive breast cancer with no early response (NeoPaTHer): efficacy, safety and biomarker analysis of a prospective, multicentre, response-adapted study. Signal Transduct Target Ther 2025; 10:45. [PMID: 39875376 PMCID: PMC11775149 DOI: 10.1038/s41392-025-02138-6] [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: 08/12/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
The potential benefits of pyrotinib for patients with trastuzumab-insensitive, HER2-positive early-stage breast cancer remain unclear. This prospective, multicentre, response-adapted study evaluated the efficacy and safety of adding pyrotinib to the neoadjuvant treatment of HER2-positive breast cancer patients with a poor response to initial docetaxel plus carboplatin and trastuzumab (TCbH). Early response was assessed using magnetic resonance imaging (MRI) after two cycles of treatment. Patients showing poor response, as defined by RECIST 1.1, could opt to receive additional pyrotinib or continue standard therapy. The primary endpoint was the total pathological complete response (tpCR; ypT0/isN0) rate. Of the 129 patients enroled, 62 (48.1%) were identified as MRI-responders (cohort A), 26 non-responders continued with four more cycles of TCbH (cohort B), and 41 non-responders received additional pyrotinib (cohort C). The tpCR rate was 30.6% (95% CI: 20.6-43.0%) in cohort A, 15.4% (95% CI: 6.2-33.5%) in cohort B, and 29.3% (95% CI: 17.6-44.5%) in cohort C. Multivariable logistic regression analyses demonstrated comparable odds of achieving tpCR between cohorts A and C (odds ratio = 1.04, 95% CI: 0.40-2.70). No new adverse events were observed with the addition of pyrotinib. Patients with co-mutations of TP53 and PIK3CA exhibited lower rates of early partial response compared to those without or with a single gene mutation (36.0% vs. 60.0%, P = 0.08). These findings suggest that adding pyrotinib may benefit patients who do not respond to neoadjuvant trastuzumab plus chemotherapy. Further investigation is warranted to identify biomarkers predicting patients' benefit from the addition of pyrotinib.
Collapse
Affiliation(s)
- Fei Wang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Yongjiu Wang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Bin Xiong
- Department of Breast Surgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Zhenlin Yang
- Department of Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Jingfen Wang
- Department of Breast Surgery, Linyi Cancer Hospital, Linyi, China
| | - Yumin Yao
- Department of Breast Surgery, Liaocheng People's Hospital, Liaocheng, China
| | - Lixiang Yu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Qinye Fu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Liang Li
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Qiang Zhang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Chao Zheng
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Shuya Huang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Liyuan Liu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Chun Liu
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Huaibo Sun
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Beibei Mao
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Dong-Xu Liu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Zhigang Yu
- Breast Center, The Second Hospital of Shandong University, Jinan, China.
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China.
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China.
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China.
| |
Collapse
|
137
|
Luo L, Wang X, Lin Y, Ma X, Tan A, Chan R, Vardhanabhuti V, Chu WC, Cheng KT, Chen H. Deep Learning in Breast Cancer Imaging: A Decade of Progress and Future Directions. IEEE Rev Biomed Eng 2025; 18:130-151. [PMID: 38265911 DOI: 10.1109/rbme.2024.3357877] [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: 01/26/2024]
Abstract
Breast cancer has reached the highest incidence rate worldwide among all malignancies since 2020. Breast imaging plays a significant role in early diagnosis and intervention to improve the outcome of breast cancer patients. In the past decade, deep learning has shown remarkable progress in breast cancer imaging analysis, holding great promise in interpreting the rich information and complex context of breast imaging modalities. Considering the rapid improvement in deep learning technology and the increasing severity of breast cancer, it is critical to summarize past progress and identify future challenges to be addressed. This paper provides an extensive review of deep learning-based breast cancer imaging research, covering studies on mammograms, ultrasound, magnetic resonance imaging, and digital pathology images over the past decade. The major deep learning methods and applications on imaging-based screening, diagnosis, treatment response prediction, and prognosis are elaborated and discussed. Drawn from the findings of this survey, we present a comprehensive discussion of the challenges and potential avenues for future research in deep learning-based breast cancer imaging.
Collapse
|
138
|
Marchio V, Augimeri G, Morelli C, Vivacqua A, Giordano C, Catalano S, Sisci D, Barone I, Bonofiglio D. Omega-3 fatty acids: molecular weapons against chemoresistance in breast cancer. Cell Mol Biol Lett 2025; 30:11. [PMID: 39863855 PMCID: PMC11762563 DOI: 10.1186/s11658-025-00694-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Breast cancer is the most commonly diagnosed type of cancer and the leading cause of cancer-related death in women worldwide. Highly targeted therapies have been developed for different subtypes of breast cancer, including hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, triple-negative breast cancer (TNBC) and metastatic breast cancer disease are primarily treated with chemotherapy, which improves disease-free and overall survival, but does not offer a curative solution for these aggressive forms of breast cancer. Moreover, the development of chemoresistance is a major cause of therapeutic failure in this neoplasia, leading to disease relapse and patient death. In addition, chemotherapy's adverse side effects may substantially worsen health-related quality of life. Therefore, to improve the outcome of patients with breast cancer who are undergoing chemotherapy, several therapeutic options are under investigation, including the combination of chemotherapeutic drugs with natural compounds. Omega-3 (ω-3) polyunsaturated fatty acids (PUFAs), including docosahexaenoic and eicosapentaenoic acids, have drawn attention for their antitumoral properties and their preventive activities against chemotherapy-induced toxicities in breast cancer. A literature review was conducted on PubMed using keywords related to breast cancer, omega-3, chemoresistance, and chemotherapy. This review aims to provide an overview of the molecular mechanisms driving breast cancer chemoresistance, focusing on the role of ω-3 PUFAs in these recognized cellular paths and presenting current findings on the effects of ω-3 PUFAs combined with chemotherapeutic drugs in breast cancer management.
Collapse
Affiliation(s)
- Vittoria Marchio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
| | - Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
| | - Catia Morelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Diego Sisci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy.
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| |
Collapse
|
139
|
Tan Y, Jiang H, Tian X, Ma F, Wang J, Zhang P, Xu B, Fan Y, Zhao W. Non-Luminal Disease Score for Everolimus in Patients with Hormone Receptor‑positive and Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer: A Multicenter and Retrospective Study. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:67-78. [PMID: 39877556 PMCID: PMC11774108 DOI: 10.2147/bctt.s493053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/23/2024] [Indexed: 01/31/2025]
Abstract
Purpose This study aims to explore the role of the non-luminal disease score (NOLUS) for everolimus in patients with hormone receptor-positive and human epidermal growth factor receptor 2-negative (HR+/HER2-) advanced breast cancer (ABC). Methods NOLUS has previously been established as an algorithm: NOLUS (0-100) = - 0.45 × ER(%) - 0.28 × PR(%) + 0.27 × Ki67(%) + 73. Information of cancer patients was retrospectively collected from three cancer centers in China. Results Totally, 198 HR+/HER2- ABC patients with complete records in expression rates (%) of ER, PR and Ki67 were enrolled in the study. The expression rates (%) of ER, PR, and Ki67 were 38.8 ± 27.9 versus 80.9 ± 14.2 (p < 0.001), 13.9 ± 14.3 versus 50.2 ± 30.4 (p < 0.001), and 37.8 ± 23.6 versus 28.7 ± 19.9 (p = 0.04), respectively, for NOLUS-positive patients and NOLUS-negative patients. For the overall population, the median PFS was 5.8 months versus 5.1 months in NOLUS-positive and NOLUS-negative patients (p = 0.16, HR = 0.75, 95% CI = 0.50, 1.12). The median 1L-, 2L, and 3L-PFS was 13.9 months versus 11.8 months (p = 0.22, HR = 1.63, 95% CI = 0.74, 3.62), 6.7 months versus 3.6 months (p = 0.08, HR = 0.34, 95% CI = 0.10, 1.18), and 4.6 months versus 4.0 months (p = 0.81, HR = 1.07, 95% CI = 0.63, 1.79) respectively, for NOLUS-positive patients and NOLUS-negative patients. Conclusion NOLUS-positive patients have a lower percentage of ER and PR, but a higher percentage of Ki67 index. The correlation between the benefits of everolimus and NOLUS failed to develop significance, suggesting that NOLUS may not be applicable in predicting everolimus efficacy in patients with HR+/HER2- ABC. Further research is expected.
Collapse
Affiliation(s)
- Yujing Tan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Hanfang Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, People’s Republic of China
| | - Xinzhu Tian
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Jiayu Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Pin Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Ying Fan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Weihong Zhao
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| |
Collapse
|
140
|
Shenasa E, He Y, Wang Z, Tu D, Gao D, Kos Z, Thornton S, Nielsen TO. Digital Profiling of Immune Biomarkers in Breast Cancer: Relation to Anthracycline Benefit. Mod Pathol 2025; 38:100718. [PMID: 39863112 DOI: 10.1016/j.modpat.2025.100718] [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: 08/13/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Assessment of the tumor-immune microenvironment can be used as a prognostic tool for improved survival and as a predictive biomarker for treatment benefit, particularly from immune-modulating treatments including cytotoxic chemotherapy. Using digital spatial profiling (DSP), we studied the tumor-immune microenvironment of 522 breast cancer cases by quantifying 35 immune biomarkers on tissue microarrays from the MA.5 phase III clinical trial. In this trial, node-positive breast cancer patients were randomized to receive either non-anthracycline chemotherapy (cyclophosphamide, methotrexate, 5'-fluorouracil [CMF]) or anthracycline-containing cytotoxic chemotherapy (CEF). Donor block hematoxylin and eosin (H&E)-stained sections were scored for the level of stromal tumor-infiltrating lymphocytes (sTILs), according to the international guidelines. We hypothesized that patients with higher levels of tumor-immune infiltration, assessed by either DSP or H&E staining, would benefit from CEF (relative to CMF) more than patients with lower immune infiltration. Unsupervised hierarchical clustering of digitally scored biomarkers revealed 2 patient clusters: immune infiltrated versus ignored. Following a prespecified statistical plan crafted to meet REMARK (REporting recommendations for tumor MARKer prognostic studies) guidelines, we found that the DSP-derived Immune Cluster assignment did not predict an improved 10-year relapse-free survival for patients receiving CEF compared with CMF. However, a secondary hypothesis revealed a significant predictive value for H&E sTILs assessed on full-faced sections for CEF benefit over CMF in the entire cohort and the human epidermal growth factor receptor 2-enriched subset. As exploratory analyses, supervised clustering of DSP-scored biomarkers suggested that low levels of T-cell immunoglobulin and mucin domain 3 TIM-3 and high levels of human leukocyte antigen HLA-DR and programmed cell death protein ligand PD-L-1 are associated with sensitivity to CEF. Although novel high-plex techniques provide a detailed insight into the tumor microenvironment, conventional H&E staining remains a powerful tool that can be applied to full-faced sections to assess the value of the immune microenvironment, particularly sTILs, in predicting benefits from immunogenic chemotherapies.
Collapse
Affiliation(s)
- Elahe Shenasa
- Interdisciplinary Oncology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ye He
- Visual Computing and Virtual Reality Key Laboratory of Sichuan Province, Sichuan Normal University, Sichuan, China
| | - Zehui Wang
- Mathematics and Statistics, Queen's University, Kingston, Ontario, Canada
| | - Dongsheng Tu
- Community Health & Epidemiology, Queen's University, Kingston, Ontario, Canada
| | - Dongxia Gao
- MAPcore, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zuzana Kos
- Pathology, BC Cancer Vancouver Centre, Vancouver, British Columbia, Canada
| | - Shelby Thornton
- MAPcore, University of British Columbia, Vancouver, British Columbia, Canada
| | - Torsten O Nielsen
- Interdisciplinary Oncology, University of British Columbia, Vancouver, British Columbia, Canada; MAPcore, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
141
|
Shadmani FK, Abedi Gheshlaghi L, Jafari-Khounigh A, Rezakhani L, Khazaei M. Burden and mortality of breast cancer attributed to diabetes and smoking in women: a secondary analysis based on Global Burden of Disease 2019 in North Africa and the Middle East. BMJ Open 2025; 15:e087178. [PMID: 39855664 PMCID: PMC11784419 DOI: 10.1136/bmjopen-2024-087178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 12/12/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is the second most common cancer in the world. This study aimed to investigate the burden of BC due to some risk factors in the Middle East and North Africa (MENA) countries from 1990 to 2019. METHODS This study is a secondary analysis based on the Global Burden of Disease 2019 data. In this study, we used data for disability-adjusted life years (DALYs) and mortality of BC in MENA countries. Also, we presented data for DALYs and mortality attributed to diabetes, smoking and passive smoking among women with BC. RESULTS The age-standardised DALY due to BC in women increased slowly from 395 (95% CI: 357 to 458) per 100 000 in 1990 to 473 (95% CI: 409 to 544) per 100 000 in 2019. But Kuwait, Bahrain, Jordan and Turkey saw a decrease in age-standardised DALY trends between 1990 and 2019. In 1990, the age-standardised mortality rate was 12.3 (95% CI: 11.0 to 14.2) per 100 000 people, and by 2019, it had increased to 15.2 (95% CI: 13.3 to 17.3) per 100 000. The rate in 2019 varied between countries. Diabetes, smoking and passive smoking contribute significantly to the burden of disease and mortality from BC, with diabetes playing a more impactful role than the other risk factors. Across the MENA region, the burden and mortality attributed to diabetes vary among countries. CONCLUSION In the MENA region, the attributable fraction of risk factors such as diabetes, smoking and secondary smoking over BC deaths and DALY has increased in recent years.
Collapse
Affiliation(s)
- Fatemeh Khosravi Shadmani
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran (the Islamic Republic of)
| | - Leili Abedi Gheshlaghi
- Trauma Research Center, Bam University of Medical Sciences, Bam, Iran (the Islamic Republic of)
| | - Ali Jafari-Khounigh
- Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran (the Islamic Republic of)
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran (the Islamic Republic of)
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran (the Islamic Republic of)
| |
Collapse
|
142
|
Jeong SH, Chun SM, Kim M, Lee YS, Kim J, Leigh JH, Choi YH. Multimodal treatments and the risk of breast cancer-related lymphedema: insights from a nationally representative cohort in South Korea. BMC Cancer 2025; 25:114. [PMID: 39844110 PMCID: PMC11753110 DOI: 10.1186/s12885-025-13513-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND As the roles of chemotherapy (Chemo) and radiation therapy (Radio) in the definitive treatment of breast cancer have expanded, a broader understanding of the factors associated with breast cancer-related lymphedema (BCRL) has become increasingly essential. Therefore, we investigated the association between multimodality treatment and the risk of BCRL. METHODS In this retrospective study conducted using National Health Insurance data and the Korea National Cancer Incidence Database (2006-2017), 114,638 participants who underwent Surgery (Surg) or Chemo within 6 months after breast cancer diagnosis were enrolled, and the effect of multimodality treatment on the risk of BCRL was analyzed using the Cox proportional-hazards model. Multimodality treatment administered through six months of treatment was grouped as only Surg; Surg/Chemo; Surg/ Chemo/Radio; Surg/Radio; only Chemo; and Chemo/Radio. RESULTS The risk of BCRL was higher in the Surg/Chemo group (hazard ratio [HR]: 1.54, 95% confidence interval [CI]: 1.43-1.65), Surg/Chemo/Radio group (HR: 1.51, 95% CI: 1.43-1.65), only Chemo group (HR: 1.58, 95% CI: 1.45-1.71), and Chemo/Radio group (HR: 1.13, 95% CI: 1.00-1.29) in comparison with the only Surg group. CONCLUSION BCRL occurrence is an after-effect of complex breast cancer treatments, and the risk may vary depending on the treatment method, including Surg, chemo, and radio. Our findings suggest that multidisciplinary strategies are required to minimize the risk of BCRL development in patients with breast cancer.
Collapse
Affiliation(s)
- Sung Hoon Jeong
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
- National Traffic Injury Rehabilitation Research Institute, National Traffic Injury Rehabilitation Hospital, Yangpyeong, 12564, Republic of Korea
| | - Seong Min Chun
- Department of Physical Medicine and Rehabilitation, Soonchunhyang University Hospital Seoul, Soonchunhyang University College of Medicine, 59, Daesagwan-ro, Yongsan-gu, Seoul, 04401, Republic of Korea
| | - Miji Kim
- Department of Biostatistics and Computing, Yonsei University Graduate School, Seoul, Republic of Korea
| | - Ye Seol Lee
- Department of Long-term Care Benefits, National Health Insurance Service, Wonju, Republic of Korea
| | - Jisun Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Ja-Ho Leigh
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea.
- National Traffic Injury Rehabilitation Research Institute, National Traffic Injury Rehabilitation Hospital, Yangpyeong, 12564, Republic of Korea.
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Yoon-Hee Choi
- Department of Physical Medicine and Rehabilitation, Soonchunhyang University Hospital Seoul, Soonchunhyang University College of Medicine, 59, Daesagwan-ro, Yongsan-gu, Seoul, 04401, Republic of Korea.
| |
Collapse
|
143
|
Zhang H, Wang L, Lin Y, Ha X, Huang C, Han C. Classification of Molecular Subtypes of Breast Cancer Using Radiomic Features of Preoperative Ultrasound Images. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01388-8. [PMID: 39843718 DOI: 10.1007/s10278-025-01388-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Radiomics has been used as a non-invasive medical image analysis technique for diagnosis and prognosis prediction of breast cancer. This study intended to use radiomics based on preoperative Doppler ultrasound images to classify four molecular subtypes of breast cancer. A total of 565 female breast cancer patients diagnosed by postoperative pathology in a hospital between 2014 and 2022 were included in this study. Radiomic features extracted from preoperative ultrasound images and clinical features were used to construct models for the classification of molecular subtypes of breast cancer. The least absolute shrinkage and selection operator (LASSO) regression was applied for the final screening of radiomic features and clinical features. Three classifiers including Logistic regression, support vector machine (SVM), and XGBoost were utilized to construct model. Model performance was assessed primarily by the area under the receiver operating characteristic curve (AUC) and 95% confidence interval (CI). The mean age of these patients was 54.58 (± 11.27) years. Of these 565 patients, 130 (23.01%) were Luminal A subtype, 329 (58.23%) were Luminal B subtype, 65 (11.50%) were human epidermal growth factor receptor-2 (HER-2) subtype, and 41 (7.26%) were triple negative (TN) subtype. A total of 12 clinical features and 8 radiomic features were selected for model construction. The AUC of the SVM model [0.826 (95%CI 0.808-0.845)] was higher than that of the Logistic regression model [0.776 (95%CI 0.756-0.796)] and the XGB model [0.800 (95%CI 0.779-0.821)] in the multiple classification of breast cancer. For the single classification of breast cancer, the AUC of the SVM model was 0.710 (95%CI 0.660-0.760) for Luminal A subtype, 0.639 (95%CI 0.592-0.685) for Luminal B subtype, 0.754 (95%CI 0.695-0.813) for HER-2 subtype, and 0.832 (95%CI 0.771-0.892) for TN subtype. The SVM model with radiomic features combined with clinical features shows good performance in classifying four molecular subtypes of breast cancer.
Collapse
Affiliation(s)
- Hongxia Zhang
- Department of Ultrasound, Yantaishan Hospital, No. 10087 Keji Avenue, Laishan District, Yantai, 264003, Shandong, P.R. China
| | - Leilei Wang
- Department of Ultrasound, Yantaishan Hospital, No. 10087 Keji Avenue, Laishan District, Yantai, 264003, Shandong, P.R. China
| | - Yayun Lin
- Department of Ultrasound, Yantaishan Hospital, No. 10087 Keji Avenue, Laishan District, Yantai, 264003, Shandong, P.R. China
| | - Xiaoming Ha
- Department of Ultrasound, Yantaishan Hospital, No. 10087 Keji Avenue, Laishan District, Yantai, 264003, Shandong, P.R. China
| | - Chunyan Huang
- Department of Ultrasound, Yantaishan Hospital, No. 10087 Keji Avenue, Laishan District, Yantai, 264003, Shandong, P.R. China
| | - Chao Han
- Department of Ultrasound, Yantaishan Hospital, No. 10087 Keji Avenue, Laishan District, Yantai, 264003, Shandong, P.R. China.
| |
Collapse
|
144
|
Lin W, Huang Z, Zhang X, Zheng D, Yang Y, Shi M, Yang D, Chu T, Ma W. Tanshinlactone triggers methuosis in breast cancer cells via NRF2 activation. Front Pharmacol 2025; 15:1534217. [PMID: 39906392 PMCID: PMC11790599 DOI: 10.3389/fphar.2024.1534217] [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: 11/25/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025] Open
Abstract
Background Tanshinlactone is a compound derived from the herb Salvia miltiorrhiza. Breast cancer is the most prevalent malignancy among women globally. While significant strides have been made in breast cancer management, these interventions are often impeded by substantial adverse effects that undermine patients' quality of life and confront limitations due to the eventual development of multi-drug resistance. Catastrophic macropinocytosis, also called methuosis, as a nonapoptotic cell death associated with cytoplasmic vacuolization, has gained increasing attention, largely because of its potential importance in cancer therapy. Methods The effect of tanshinlactone on the growth of human cancer cells was evaluated using sulforhodamine B and colony formation assay. Fluorescent dyes are used to label macropinosomes and lysosomes. Phase contrast, confocal and transmission electron microscopy were employed to observe cell morphological changes. RT-PCR, western blot, lentiviral-mediated gene overexpression, and pharmacological inhibitor assays were comprehensively designed to regulate the identified signaling pathways and confirm the mechanism of tanshinlactone. Human breast cancer cell lines-derived xenograft tumor explants assay was used to evaluate the compound's efficacy and to assess the induction of methuosis via NRF2 activation by tanshinlactone. Results Tanshinlactone selectively inhibits the growth of ER+ and HER2+/EGFR + breast cancer cells while showing limited cytotoxicity against other cancer types and normal cells. The selective anti-breast cancer activity is associated with the induction of methuosis, characterized by cytoplasmic vacuolization due to dysfunctional macropinocytosis. This process is mediated by the activation of the transcription factor NRF2, leading to the formation of macropinosomes that fail to fuse with lysosomes or recycle to the plasma membrane, resulting in cell death. The in vitro induction of methuosis via NRF2 activation was replicated in a murine xenograft explants model. Additionally, tanshinlactone demonstrated effectiveness against lapatinib-resistant breast cancer cells, suggesting its potential as a therapeutic agent for overcoming drug resistance in cancer treatment. Conclusion Tanshinlactone as a novel therapeutic agent, is capable of selectively inhibiting ER+ and HER2+/EGFR + breast tumors through a unique mechanism of inducing catastrophic macropinocytosis. This regimen holds promise for targeted therapy with minimized side effects and offers a new therapeutic avenue for breast patients with drug-resistant diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
145
|
Al-Ghamdi AR, Ahmed WU, Al-Wabli RI, Al-Mutairi MS, Rahman AFMM. Synthesis and Anticancer Evaluation of O-Alkylated ( E)-Chalcone Derivatives: A Focus on Estrogen Receptor Inhibition. Int J Mol Sci 2025; 26:833. [PMID: 39859546 PMCID: PMC11766267 DOI: 10.3390/ijms26020833] [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: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer remains a leading cause of morbidity and mortality worldwide, highlighting the urgent need for novel therapeutic agents. This study investigated the synthesis and biological evaluation of O-alkyl (E)-chalcone derivatives (4a-4v) as potential anticancer agents. The compounds were synthesized via aldol condensation of substituted aldehydes and acetophenones, with structures confirmed by IR, NMR, and mass spectrometry. In vitro cytotoxicity assays revealed varying effectiveness, with compounds 4a, 4b, 4q, and 4v exhibiting potent activity against MDA-MB-231 and MCF-7, showing IC50 values between 2.08 and 13.58 µM, besides HCT-116 and HeLa cancer cell lines (IC50 values between 6.59 and 22.64 µM). Notably, compound 4b displayed remarkable selectivity, with an IC50 of 54.59 µM against the non-cancerous WI-38 cell line. Additionally, protein kinase inhibition assays indicated that compounds 4b and 4q effectively inhibited EGFR and VEGFR-2, with 4b outperforming the standard inhibitor erlotinib. Molecular docking studies of compound 4q showed strong binding affinities in the ATP-binding pockets of EGFR, HER2, VEGFR2, and CDK2. In silico analyses further highlighted the favorable pharmacokinetic properties of compound 4q, underscoring its potential as a selective tyrosine kinase inhibitor. These findings suggest the therapeutic promise of O-alkyl (E)-chalcone derivatives in cancer treatment.
Collapse
Affiliation(s)
- Alwah R. Al-Ghamdi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.R.A.-G.); (R.I.A.-W.)
| | - Wahid U. Ahmed
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China;
| | - Reem I. Al-Wabli
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.R.A.-G.); (R.I.A.-W.)
| | - Maha S. Al-Mutairi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.R.A.-G.); (R.I.A.-W.)
| | - A. F. M. Motiur Rahman
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.R.A.-G.); (R.I.A.-W.)
| |
Collapse
|
146
|
Zhu Y, Song Y, Zhou X, Zhang W, Luo H. Antibody-drug conjugates in breast cancer. Carcinogenesis 2025; 46:bgae082. [PMID: 39742416 DOI: 10.1093/carcin/bgae082] [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: 10/30/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/03/2025] Open
Abstract
Antibody-drug conjugates (ADCs) have garnered significant attention as an innovative therapeutic strategy in cancer treatment. The mechanism of action for ADCs involves the targeted delivery of antibodies to specific receptors, followed by the release of cytotoxic payloads directly into tumor cells. In recent years, ADCs have made substantial progress in the treatment of breast cancer (BC), particularly demonstrating significant efficacy in the human epidermal growth factor receptor-2 (HER-2)-positive subgroup. Clinical evidence indicates that ADCs have notably improved treatment efficacy and survival outcomes for BC patients. However, challenges such as drug toxicities and the emergence of drug resistance necessitate further research and discussion. In this paper, we will summarize the advances in ADCs targeting various receptors in BC patients and explore the challenges and future directions in this field. We anticipate that the increasing availability of ADCs will lead to more effective and personalized treatment options for BC patients.
Collapse
Affiliation(s)
- Yinxing Zhu
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China
| | - Yaqi Song
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China
| | - Xilei Zhou
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China
| | - Wenwen Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Honglei Luo
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China
| |
Collapse
|
147
|
Fakeri M, Haghi M, Jahanban Esfahlan R, Fathi M, Hosseinpour Feizi MA. Targeted apoptosis in breast cancer cells via niosome-mediated delivery of cyclophosphamide and sodium oxamate. Mol Biol Rep 2025; 52:139. [PMID: 39827334 DOI: 10.1007/s11033-025-10241-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Breast cancer is the most frequently diagnosed cancer among women, posing significant health risks. This study investigates niosome nanoparticles as a delivery system for Cyclophosphamide (CYC) and Sodium Oxamate (SO) to target apoptotic pathways in MDA-MB-231 breast cancer cells. METHODS Lipid-based niosomes were prepared using the thin-film hydration method and characterized for size, zeta potential, and Fourier-transform infrared spectroscopy (FTIR) profiles. MDA-MB-231 cells were treated with CYC and SO-loaded niosomes, and cell viability was assessed using the MTT assay. Flow cytometry with annexin V/PI labeling evaluated apoptosis, while real-time PCR and Western blotting analyzed levels of NRF2 and key apoptotic proteins. RESULTS Niosomes exhibited favorable properties with a mean particle size of 87.98 nm and a zeta potential of - 7.44 mV. Treatment significantly reduced cell viability compared to controls, indicating cytotoxic effects. Flow cytometric analysis revealed a substantial increase in apoptotic cells post-treatment. Western blot analysis showed elevated NRF2 protein levels and increased expression of caspase-3 and Bax, confirming activation of apoptotic pathways. CONCLUSIONS The co-delivery of CYC and SO via niosomes effectively targets apoptotic pathways in MDA-MB-231 breast cancer cells. The enhanced cytotoxicity and induction of apoptosis suggest that this drug delivery system may serve as an effective strategy for managing breast cancer.
Collapse
Affiliation(s)
- Mahsa Fakeri
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mehdi Haghi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Rana Jahanban Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
148
|
Valouzi A, Shahbazi M, Erfani-Moghadam V, Ramezani M, Shamsabadi FT. Cancer-Specific Activation of the Vesicular Stomatitis Virus Matrix by Survivin Promoter in Breast Cancer Cells. Mol Biotechnol 2025:10.1007/s12033-024-01359-4. [PMID: 39820852 DOI: 10.1007/s12033-024-01359-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Oncolytic viral-based therapy and specific gene expression by promoters are modern targeted oncotherapy approaches that have gained significant attention in recent years. In this study, both strategies were combined by designing cancer-specific activation of vesicular stomatitis virus matrix expression under the survivin promoter. The matrix sequence was cloned downstream of the survivin promoter (pM). After transfecting MCF-7 cells with pM, cell proliferation and apoptosis induction were assessed. Additionally, the transcript levels of matrix and apoptosis-related genes in response to pM was assessed. The proliferation of MCF-7 cells was significantly reduced by the constructed matrix-expressing plasmid at 48 and 72 h post-transfection (p < 0.05). Enhanced matrix expression resulted in the down-regulation of MMP-9, TP53, and NF-kB, while simultaneously up-regulating Bax transcripts. Evaluating the effect of pM vector on apoptosis induction revealed a significant increase in the MCF-7 cells compared to untreated cells (p < 0.05). The absence of significant matrix gene expression in HDF cells, relative to MCF-7 cells, further underscores the specific function of the Survivin promoter in cancer cells. These findings suggest that the matrix may have various biological functions in a diverse set of non-apoptotic pathways. Further research on the association of the matrix with other genes could provide insights into the biomedical significance and future perspectives of the matrix in cancer gene therapy.
Collapse
Affiliation(s)
- Atefeh Valouzi
- Department of Medical Biotechnology, Golestan University of Medical Sciences, Gorgan, Iran
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Department of Medical Biotechnology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Vahid Erfani-Moghadam
- Department of Medical Biotechnology, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahboobeh Ramezani
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh T Shamsabadi
- Department of Medical Biotechnology, Golestan University of Medical Sciences, Gorgan, Iran.
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
149
|
Lai Y, Zhang R. Green one-pot synthesis of quinoxaline derivatives using sulfo-anthranilic acid functionalized alginate-MCFe 2O 4 nanostructures: a novel superparamagnetic catalyst with antiproliferative potential. RSC Adv 2025; 15:1698-1712. [PMID: 39835215 PMCID: PMC11744772 DOI: 10.1039/d4ra07892f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
This study reports a green, multi-component synthesis of 2-aminoimidazole-linked quinoxaline Schiff bases using a novel superparamagnetic acid catalyst. The catalyst consists of sulfo-anthranilic acid (SAA) immobilized on MnCoFe2O4@alginate magnetic nanorods (MNRs), achieving high SAA loading (1.8 mmol g-1) and product yields (91-97%). Characterization of the MCFe2O4@Alginate@SAA MNR catalyst revealed an inverse spinel structure (XRD), a saturation magnetization of 31 emu g-1 (VSM), 17.5% organic content (TGA), and a rod-like morphology with diameters of 30-60 nm and lengths of 150-250 nm (SEM). Elemental composition confirmed by EDX analysis indicated successful SAA immobilization and high catalyst purity. The synthesized quinoxaline derivatives were evaluated for antiproliferative activity against SKOV3 and HCT-116 cancer cell lines using the MTT assay. Several compounds, notably 4a, 4s, 4t, 4w, and 4x, exhibited potent activity, inhibiting HCT-116 proliferation by >50% at 50 μg mL-1. Compound 4a demonstrated the most significant inhibition, with 82.3% against SKOV3 cells after 48 h and 69.0% against HCT-116 cells after 24 h, both at 50 μg mL-1. These results suggest the potential of 2-aminoimidazole-linked quinoxaline Schiff bases, particularly 4a, as promising multi-target chemotherapy agents.
Collapse
Affiliation(s)
- Ying Lai
- Department of Life Science and Agriculture, Zhoukou Normal University Zhoukou Henan 466001 China
| | - Ruoyu Zhang
- Department of Life Science and Agriculture, Zhoukou Normal University Zhoukou Henan 466001 China
| |
Collapse
|
150
|
Debnath A, Mazumder R, Singh AK, Singh RK. Identification of novel cyclin-dependent kinase 4/6 inhibitors from marine natural products. PLoS One 2025; 20:e0313830. [PMID: 39813224 PMCID: PMC11734976 DOI: 10.1371/journal.pone.0313830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/31/2024] [Indexed: 01/18/2025] Open
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) are crucial regulators of cell cycle progression and represent important therapeutic targets in breast cancer. This study employs a comprehensive computational approach to identify novel CDK4/6 inhibitors from marine natural products. We utilized structure-based virtual screening of the CMNPD database and MNP library, followed by rigorous filtering based on drug-likeness criteria, PAINS filter, ADME properties, and toxicity profiles. From an initial hit of 9,497 compounds, 2,344 passed drug-likeness and PAINS filters. Further ADME filtering yielded 50 compounds, of which 25 exhibited non-toxic profiles. These 25 candidates underwent consensus molecular docking using seven distinct algorithms: AutoDockTools 4.2, idock, LeDock, Qvina 2, Smina, AutoDock Vina 1.2.0, PLANTS, and rDock. Based on these results, six top-scoring compounds were selected for comprehensive 500 nanosecond all-atom molecular dynamics simulations to evaluate their structural stability and interactions with CDK4/6. Our analysis revealed that compounds CMNPD11585 and CMNPD2744 demonstrated superior stability in their interactions with CDK4/6, exhibiting lower RMSD and RMSF values, more favorable binding free energies, and persistent hydrogen bonding patterns. These compounds also showed lower Solvent Accessible Surface Area values, indicating better compatibility with the CDK4/6 active site. Subsequent in-vitro studies using MTT assays on MCF-7 breast cancer cells confirmed the cytotoxic effects of these compounds, with CMNPD11585 showing the highest potency, followed by CMNPD2744.
Collapse
Affiliation(s)
- Abhijit Debnath
- Noida Institute of Engineering and Technology [Pharmacy Institute], Institutional Area, Greater Noida, Uttar Pradesh, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology [Pharmacy Institute], Institutional Area, Greater Noida, Uttar Pradesh, India
| | - Anil Kumar Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajesh Kumar Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|