1
|
Qayyum MA, Mahmood S, Bahadur A, Iqbal S, Zidan A, Saad M, Mahmood MH, Farooq T, Batool M, Atif M, Jaber F, Rahman KM, Farooq Z, Asiri YA, Farouk AE, Aloufi S. Study of toxic and essential elemental imbalances at more advance stage of breast carcinoma patients. Pathol Res Pract 2025; 269:155866. [PMID: 40037052 DOI: 10.1016/j.prp.2025.155866] [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: 09/03/2024] [Revised: 01/18/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Delayed diagnosis and limited access to healthcare resources are leading to advanced stage breast carcinoma posing a formidable health challenge to modern medical science. Avoiding the occurrence of breast carcinoma is only possible by ascertaining the risk factors contributing to the progression of this fatal malignancy. Among other factors, exposure to trace elements plays a decisive role in progression of breast carcinoma development. The purpose of this investigation was to evaluate the harmful traces and vital components (Pb, Ca, Mn, Cd, Na, Cr, Cu, Mg, Sr, K, Fe, Zn, Co, Li, Se, Ag, Hg, As, Al and Ni) contents in the blood (n = 161), scalp hair (n = 149) and nails (n = 135) of stage IV breast carcinoma patients and female controls by atomic absorption spectrometry. The results revealed that mean Cr, Cu, Pb and Cd levels were elevated significantly (p < 0.05) in the nails and blood, while Ni, Co, Cr, Pb and Cd levels (p < 0.05) were elevated in the scalp hair of stage IV breast carcinoma patients than the female controls. Based on pathological breast cancer types, Pb and Cd were elevated in invasive ductal carcinoma, while Cu and Co were higher in invasive lobular carcinoma patients. Multivariate analysis was applied for the source of identification and apportionment of toxic trace elements in both donor groups. Most of the factors showed significant differences between the malignant and control groups' dietary, housing, and tobacco use/nonuse behaviors. The correlation analysis revealed significantly different association patterns for toxic trace elements in patients and controls and has an influential effect on cancer risk. Overall, the current study indicates a potential role of toxic trace elements in the development of breast carcinoma and these findings could be significant in the predicting diagnosis/prognosis of breast malignancy.
Collapse
Affiliation(s)
- Muhammad Abdul Qayyum
- Department of Chemistry, Division of Science & Technology, University of Education Lahore, Pakistan.
| | - Sajid Mahmood
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315100, China; Low Dimensional Materials Research Center at Khazar University, AZ1096, Baku, Azerbaijan
| | - Ali Bahadur
- Nanomaterials Research Center, Department of Chemistry, College of Science, Mathematics, and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang 325060, China; Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, 1000 Morris Ave, Union, NJ 07083, USA.
| | - Shahid Iqbal
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315100, China.
| | - Ammar Zidan
- Biomedical Engineering Department, College of Engineering and Technologies, Al-Mustaqbal University, Babylon 51001, Iraq
| | - Muhammad Saad
- Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Konarskiego 22B, Gliwice 44-100, Poland; Joint Doctoral School, Silesian University of Technology, Akademicka 2A, Gliwice 44-100, Poland
| | - Mian Hr Mahmood
- Department of Chemistry, Division of Science & Technology, University of Education Lahore, Pakistan
| | - Tahir Farooq
- Department of Applied Chemistry, Government College University Faisalabad, 38000, Pakistan
| | - Marrium Batool
- Department of Chemistry, Division of Science & Technology, University of Education Lahore, Pakistan
| | - Muhammad Atif
- Department of Chemistry, Division of Science & Technology, University of Education Lahore, Pakistan
| | - Fadi Jaber
- Department of Biomedical Engineering, Ajman University, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Kk Mujeeb Rahman
- Department of Biomedical Engineering, Ajman University, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Zahid Farooq
- Department of Physics, Division of Science & Technology, University of Education Lahore, Pakistan
| | - Yousif A Asiri
- Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Abd-ElAziem Farouk
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Salman Aloufi
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
2
|
Scherübl H. [Generation X and millennials are at greater cancer risk than previous generations: Possible causes and implications]. Dtsch Med Wochenschr 2025; 150:575-583. [PMID: 40262757 DOI: 10.1055/a-2531-9761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The incidence of early-onset malignancy (EOM) diagnosed under the age of 50 has been increasing worldwide. In the USA, Generation X and the millennials are up to 2 times more likely to develop 17 of the 34 most common cancers than the baby boomers at any given age. Globally, the incidence of EOM increased by 79.1% between 1990 and 2019. Early-life exposure may be linked with EOM development and may forecast a greater disease burden in the future. Possible causes and consequences are discussed with regards to cancer prevention.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Innere Medizin - Gastroenterologie, Gastrointestinale Onkologie und Infektiologie, Vivantes Klinikum Am Urban, Berlin, GERMANY
| |
Collapse
|
3
|
Aksoyalp ZS, Kayki-Mutlu G, Wojnowski L, Michel MC. A year in pharmacology: new drugs approved by the US Food and Drug Administration in 2024. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5077-5099. [PMID: 40163152 PMCID: PMC11985671 DOI: 10.1007/s00210-025-04020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
The US Food and Drug Administration approved 50 new drugs and nine new cellular and gene therapy products in 2024, i.e., a total of 59 new medical therapies. The latter group represented three treatments each for oncology and hematology/immunotherapy, and one each for neurology, genetic disorders, and cardiovascular disorders. Oncology, hematology/immunotherapy, and neurological disorders (14, six, and seven, respectively) also were highly prevalent among classic medications. Looking at trends over the past 5 years, we observe a greater share in first-in-class medications, more fast-track approvals, and mRNA/gene/cell-based therapies. While small molecules remain the largest fraction, their percentage has been declining substantially over the past 5 years. Taking together, these findings testify to the commitment of the pharmaceutical industry for innovative treatments, including conditions for which no approved therapies existed. On the other hand, there also is a trend for approvals for narrowly focused conditions such as tumors defined by genetic alterations.
Collapse
Affiliation(s)
- Zinnet Sevval Aksoyalp
- Department of Pharmacology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - Gizem Kayki-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Leszek Wojnowski
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Martin C Michel
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
4
|
Aroujo J, Parker H, Boari A, Mason E, Otakpor MU, Betancourt T, Kornienko A, Ciavatta ML, Carbone M, Evidente A, Taube JH, Romo D. Derivatization of ophiobolin A and cytotoxicity toward breast and glioblastoma cancer stem cells: Varying the ketone and unsaturated aldehyde moieties. Bioorg Med Chem Lett 2025; 120:130112. [PMID: 39880174 DOI: 10.1016/j.bmcl.2025.130112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
To gain further insights into the importance of the unsaturated 1,4-ketoaldehyde moiety of ophiobolin A (OpA) for the potency and selectivity observed toward cancer stem cells, several derivatives were synthesized through controlled reduction and oxidations of the unsaturated aldehyde and ketone moieties. Structure elucidation of these new OpA derivatives was achieved through detailed NMR studies and comparison to OpA and known isolated congeners possessing variations in these regions. The relative stereochemistry of the newly generated stereocenters was determined by coupling constants in conjunction with conformational analyses (DFT) of the synthetic derivatives. The cytotoxicity of these derivatives was studied against breast cancer and glioblastoma cell lines possessing stem-cell like properties. In addition, the comparative activity toward HMLE and HMLE-TWIST mammary epithelial cells was studied, with the latter cell line representing an epithelial mesenchymal transition positive (EMT+) cell line.
Collapse
Affiliation(s)
- Jaquelin Aroujo
- Department of Chemistry and Biochemistry, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States
| | - Haleigh Parker
- Department of Biology, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States
| | - Angela Boari
- Institute of Sciences of Food Production, National Research Council, Via Amendola 122/O, 70125 Bari, Italy
| | - Evan Mason
- Department of Chemistry and Biochemistry, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States
| | - Mackenzie U Otakpor
- Department of Chemistry and Biochemistry, Texas State University, 601 University Dr., TX 78666, United States
| | - Tania Betancourt
- Department of Chemistry and Biochemistry, Texas State University, 601 University Dr., TX 78666, United States
| | - Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, 601 University Dr., TX 78666, United States.
| | - Maria Letizia Ciavatta
- Institute of Biomolecular Chemistry, National Research Council (CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Marianna Carbone
- Institute of Biomolecular Chemistry, National Research Council (CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Antonio Evidente
- Institute of Biomolecular Chemistry, National Research Council (CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy.
| | - Joseph H Taube
- Department of Biology, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States.
| | - Daniel Romo
- Department of Chemistry and Biochemistry, Baylor University, 101 Bagby Ave., Waco, TX 76798, United States.
| |
Collapse
|
5
|
Gary M, Krueger E, Kisiel M, Demirhan K, Barajas AG, Ryan JC. Survey of Health Care Providers on Social Determinants of Health and Treatment Decision-Making for Patients With HR+/HER2- Metastatic Breast Cancer. Oncol Ther 2025:10.1007/s40487-025-00337-3. [PMID: 40266506 DOI: 10.1007/s40487-025-00337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/26/2025] [Indexed: 04/24/2025] Open
Abstract
INTRODUCTION Social determinants of health (SDOH) are a range of nonmedical factors that contribute to outcome disparities among certain groups of patients; however, little is known about how SDOH affect treatment decision-making for patients with cancer, particularly those with metastatic breast cancer (mBC). This study sought to gain insights from physicians and advanced practice providers on the impact of SDOH on practice- and patient-level cancer care decision-making with a nationwide online survey. METHODS The Social Determinants of Health in Metastatic Breast Cancer Survey was developed by the Association of Cancer Care Centers (ACCC) in partnership with Pfizer Inc, and in consultation with SDOH specialists. The 23-question survey captured experiences with SDOH-informed treatment decision-making for patients with cancer via multiple choice, Likert-scale, and free-response questions. ACCC-member physicians and advanced practice providers in the USA completed the survey between 23 January and 8 February 2024. RESULTS Respondents (n = 145), a majority of whom were medical oncologists (60%), represented clinics from diverse geographic regions of the USA; approximately 65% of respondents' clinics served patient populations with ≥ 10% Black, Indigenous, or People of Color. Common comprehensive cancer care services were provided by at least 59% of clinics, and SDOH factors were assessed by approximately 75% of clinics at diagnosis. Navigation services were available for patients with mBC at approximately 75% of respondents' clinics. Financial considerations (51%) and the presence of a caregiver (35%) were the most frequently cited SDOH-related factors that impacted mBC treatment decision-making. CONCLUSION The surveyed ACCC-member care providers displayed a high degree of awareness regarding SDOH impacts on their practice and patients, but resource limitations were identified as barriers to comprehensive, SDOH-informed cancer care. Harnessing existing resources from local and national advocacy groups, especially navigator training programs, is an actionable, real-world solution for improving mBC care for patients facing SDOH barriers.
Collapse
Affiliation(s)
- Monique Gary
- Cancer Program, Grand View Health, Sellersville, PA, USA
| | - Editha Krueger
- Dignity Health-Cancer Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Molly Kisiel
- Association of Cancer Care Centers, Rockville, MD, USA
| | | | | | | |
Collapse
|
6
|
Ma Y, Wang Y, Zhang Y, Bian K, Zhu Y, Liu A, Li H, Yin L, Lu H, Ye Z. Comparison of contrast-enhanced cone-beam breast CT, MRI, and mammography for breast cancer characterization. Eur Radiol 2025:10.1007/s00330-025-11568-3. [PMID: 40240554 DOI: 10.1007/s00330-025-11568-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/12/2025] [Accepted: 03/08/2025] [Indexed: 04/18/2025]
Abstract
OBJECTIVES To compare the consistency of contrast-enhanced cone-beam breast CT (CE-CBBCT), MRI and mammography regarding characterization of breast cancer. METHODS In this retrospective study, patients with breast cancer who underwent preoperative CE-CBBCT, MRI, and mammography between January 2017 and July 2022 were enrolled. Three experienced radiologists independently interpreted the characteristics of breast lesions on each imaging mode referring to BI-RADS, with a 4-week wash-out period. One of the three radiologists reviewed the CE-CBBCT images 4 weeks after the initial evaluation. Cross-modality consistency was calculated by Cohen's Kappa based on majority report. Inter-and intra-reader agreement were assessed using Fleiss and Cohen's Kappa, respectively. The association between imaging factors and consistency levels was analyzed using chi-square and Mann-Whitney U test. RESULTS A total of 214 malignant lesions identified in 207 patients were enrolled. CE-CBBCT showed almost perfect agreement with MRI on lesion type identification (Kappa = 0.865, 95% CI: 0.802-0.928), but fair agreement with mammography (Kappa = 0.287, 95% CI: 0.205-0.369). CE-CBBCT showed substantial agreement on characterization with MRI for both mass (Kappa = 0.752-0.824) and non-mass enhancement (NME) (Kappa = 0.702-0.729), and non-contrast-enhanced CBBCT (NCE-CBBCT) showed substantial agreement with mammography for calcification (Kappa = 0.717-0.777). Inter- (Kappa = 0.611-0.738) and intra-reader (Kappa = 0.757-0.887) agreement were substantial on CE-CBBCT interpretation. There was no statistically significant difference in imaging factors between different consistency levels (all p > 0.05). CONCLUSIONS CE-CBBCT showed high consistency on mass and NME characterization with MRI, and on calcification with mammography, indicating that CE-CBBCT could combine morphology, hemodynamic and calcification features, and the corresponding descriptors have the feasibility to describe CE-CBBCT characteristics of breast cancer. KEY POINTS Question Contrast-enhanced cone-beam breast CT (CE-CBBCT) is widely used in the diagnosis and assessment of breast cancer, but there is no standardized lexicon for image interpretation. Findings CE-CBBCT showed high consistency and comparable reproducibility with MRI and mammography for characterizing breast cancer lesions. Clinical relevance The findings prove that CE-CBBCT could combine morphology, hemodynamic, and calcification features and provide support for the feasibility of applying the BI-RADS descriptors of MRI and mammography to interpret contrast-enhanced cone-beam breast CT images in clinic.
Collapse
Affiliation(s)
- Yue Ma
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Yafei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Yuwei Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Keyi Bian
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Yueqiang Zhu
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Aidi Liu
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Haijie Li
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Lu Yin
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Hong Lu
- Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China.
| |
Collapse
|
7
|
Mohamed AF, Hamed AEM, Mohamed SSA, Othman AA, El-Tawab NAA. Effect of nursing application of emotion freedom technique on perceived stress, resilience and sexual satisfaction among women after mastectomy. BMC Nurs 2025; 24:428. [PMID: 40234881 PMCID: PMC12001692 DOI: 10.1186/s12912-025-02977-2] [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: 10/25/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND A breast cancer diagnosis extends beyond physical health concerns, profoundly impacting a woman's psychological well-being, resilience and sexual satisfaction. Mastectomy intensifies these challenges, potentially affecting overall quality of life and long-term well-being. Understanding the interrelationships between perceived stress, resilience and sexual satisfaction is crucial for designing targeted interventions that effectively support mastectomized women. This study evaluates the effectiveness of the Emotional Freedom Technique in mitigating stress, enhancing resilience and improving sexual well-being post-mastectomy. METHODS A quasi-experimental pretest-posttest design was employed with a convenient sample of 112 Egyptian women who had undergone mastectomy. Participants were recruited from the Outpatient Oncology Clinic at Beni-Suef University Hospital. Data were collected over six months (January-June 2024) and analyzed using SPSS version 26.0. The intervention consisted of six structured EFT sessions delivered over six weeks. Paired t-tests assessed pre- and post-intervention differences, while Pearson and Spearman correlation analyses examined relationships between variables to accommodate different data distributions. RESULTS Post-intervention assessments revealed statistically significant improvements. Perceived stress scores decreased from 32.42 ± 1.70 to 17.27 ± 2.96 (t = 49.130, p < 0.001, Cohen's d = 3.2), resilience scores increased from 11.53 ± 1.67 to 31.46 ± 5.48 (t = 36.454, p < 0.001, Cohen's d = 2.8) and sexual satisfaction scores improved from 17.03 ± 1.55 to 31.00 ± 4.31 (t = 13.245, p < 0.001, Cohen's d = 2.5). Strong negative correlations were found between perceived stress and both resilience (r = -0.692, p < 0.001) and sexual satisfaction (r = -0.835, p < 0.001), while resilience and sexual satisfaction were strongly positively correlated (r = 0.890, p < 0.001). CONCLUSION EFT is a cost-effective, non-invasive intervention that significantly reduces stress, enhances resilience and improves sexual satisfaction in women post-mastectomy. To optimize clinical integration, healthcare institutions should develop structured EFT training programs for nurses, incorporating theoretical foundations, hands-on practice and competency assessments. Standardized protocols should be established to guide EFT implementation in post-mastectomy care. Further research should explore long-term effects and broader applicability across diverse healthcare settings. CLINICAL TRIAL NUMBER NCT06583629 on 4/9/2024.
Collapse
Affiliation(s)
- Ashgan Fathy Mohamed
- Psychiatric and Mental Health Nursing Faculty of Nursing, Beni-Suef University, Beni-Suef, Egypt
| | | | | | | | | |
Collapse
|
8
|
Davis MB, Martini R. Precision oncology and genetic ancestry: The science behind population-based cancer disparities. Cancer Cell 2025; 43:619-622. [PMID: 40233717 DOI: 10.1016/j.ccell.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025]
Abstract
Precision oncology has transformed cancer care but has barely benefited patients of diverse ancestry. Historically, self-reported race/ethnicity has served as a surrogate for biological differences, but genetic ancestry provides a more precise framework for understanding genetic drivers of cancer disparities, including associations between ancestry and tumor subtypes, and genetic variants affecting drug metabolism and treatment response. To improve precision oncology for all patients and reduce cancer disparities, we propose expanding ancestry-inclusive genomic data, reevaluating disease-associated variants within ancestrally diverse cohorts, and standardizing data-sharing practices.
Collapse
Affiliation(s)
- Melissa Boneta Davis
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, Atlanta, GA, USA; Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA; Georgia Research Alliance, Atlanta, GA, USA.
| | - Rachel Martini
- Institute of Translational Genomic Medicine, Morehouse School of Medicine, Atlanta, GA, USA; Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
9
|
Neophytou C, Charalambous A, Voutouri C, Angeli S, Panagi M, Stylianopoulos T, Mpekris F. Sonopermeation combined with stroma normalization enables complete cure using nano-immunotherapy in murine breast tumors. J Control Release 2025; 382:113722. [PMID: 40233830 DOI: 10.1016/j.jconrel.2025.113722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
Nano-immunotherapy shows great promise in improving patient outcomes, as seen in advanced triple-negative breast cancer, but it does not cure the disease, with median survival under two years. Therefore, understanding resistance mechanisms and developing strategies to enhance its effectiveness in breast cancer is crucial. A key resistance mechanism is the pronounced desmoplasia in the tumor microenvironment, which leads to dysfunction of tumor blood vessels and thus, to hypoperfusion, limited drug delivery and hypoxia. Ultrasound sonopermeation and agents that normalize the tumor stroma have been employed separately to restore vascular abnormalities in tumors with some success. Here, we performed in vivo studies in two murine, orthotopic breast tumor models to explore if combination of ultrasound sonopermeation with a stroma normalization drug can synergistically improve tumor perfusion and enhance the efficacy of nano-immunotherapy. We found that the proposed combinatorial treatment can drastically reduce primary tumor growth and in many cases tumors were no longer measurable. Overall survival studies showed that all mice that received the combination treatment survived and rechallenge experiments revealed that the survivors obtained immunological memory. Employing ultrasound elastography and contrast enhanced ultrasound along with proteomics analysis, flow cytometry and immunofluorescene staining, we found the combinatorial treatment reduced tumor stiffness to normal levels, restoring tumor perfusion and oxygenation. Furthermore, it increased infiltration and activity of immune cells and altered the levels of immunosupportive chemokines. Finally, using machine learning analysis, we identified that tumor stiffness, CD8+ T cells and M2-type macrophages were strong predictors of treatment response.
Collapse
Affiliation(s)
- Constantina Neophytou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Antonia Charalambous
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Stella Angeli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 1678, Cyprus; Cancer Genetics, Therapeutics & Ultrastructural Pathology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.
| |
Collapse
|
10
|
Jiang Y, Qian Z, Wang C, Wu D, Liu L, Ning X, You Y, Mei J, Zhao X, Zhang Y. Targeting B7-H3 inhibition-induced activation of fatty acid synthesis boosts anti-B7-H3 immunotherapy in triple-negative breast cancer. J Immunother Cancer 2025; 13:e010924. [PMID: 40221152 PMCID: PMC11997833 DOI: 10.1136/jitc-2024-010924] [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: 10/29/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most malignant breast cancer, highlighting the need for effective immunotherapeutic targets. The immune checkpoint molecule B7-H3 has recently gained attention as a promising therapeutic target due to its pivotal role in promoting tumorigenesis and cancer progression. However, the therapeutic impact of B7-H3 inhibitors (B7-H3i) remains unclear. METHODS Transcriptomic and metabolomic analyses were conducted to explore the underlying mechanisms of B7-H3 inhibition in TNBC. The therapeutic efficacy of the combined treatment strategy was substantiated through comprehensive phenotypic assays conducted in vitro and validated in vivo using animal models. RESULTS B7-H3 blockade induces a "primed for death" stress state in cancer cells, leading to distinct alterations in metabolic pathways. Specifically, B7-H3 knockdown activated the AKT signaling pathway and upregulated sterol regulatory element-binding protein 1 (SREBP1), which in turn elevated FASN expression. The simultaneous inhibition of both B7-H3 and FASN more effectively attenuated the malignant progression of TNBC. CONCLUSIONS Our findings propose an "immune attack-metabolic compensation" dynamic model and suggest the feasibility of a dual-targeting strategy that concurrently inhibits both B7-H3 and FASN to enhance therapeutic efficacy in TNBC patients.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhiwen Qian
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cengzhu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Danping Wu
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Lu Liu
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xin Ning
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yilan You
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Mei
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoqian Zhao
- Department of Breast Surgery, Women's Hospital of Jiangnan University, Wuxi, China
| | - Yan Zhang
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Ding X, Zhu M, Kan H, Wang Q, Chen H, Xia X, Zhao F, Li Z. Relationship between psychological resilience and cognitive function on breast cancer chemotherapy patients: a person-centered method. Breast Cancer 2025:10.1007/s12282-025-01697-4. [PMID: 40208506 DOI: 10.1007/s12282-025-01697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Previous studies have indicated a connection between resilience and cognitive function, but critical gaps persist regarding limited data exploring the relationship between resilience profiles and cognition using person-centered methodologies, especially in the context of breast cancer patients. OBJECTIVES While the general correlation between resilience and cognitive function is known, how various resilience profiles impact cognitive function in breast cancer patients remains unclear. The study aimed to identify resilience profiles and explore the relationship with cognitive function in breast cancer (BC) patients. METHODS This study was a cross-sectional study in descriptive research. BC patients (n = 425) from a tertiary oncology hospital completed the Functional Assessment of Cancer Therapy-Cognitive Function (FACT-Cog) scale and Connor-Davidson Resilience Scale (CD-RISC- 25). Latent profile analysis was adopted to identify subgroups of patients with distinct resilience profiles according to model fit indices. An ANOVA analysis and Chi-square test were also employed. RESULTS Three profiles were identified, including Class 3, "High resilience-positive coping group" (n = 187, 44.7%), Class 2, "Medium resilience-effort regulation group" (n = 157, 37.6%), and Class 1, "Low resilience-negative coping" (n = 74, 17.7%). The results demonstrate that perceived cognitive impairment (CogPCI), other people's appraisal (CogOth), perceived cognitive ability (CogPCA), impact on quality of life (CogQOL), and FACT-Cog were significant differences in all three potential latent resilience profiles (P < 0.001). The CogPCI and FACT-Cog scores in Class 3 were the best, while Class 1 was the lowest. CONCLUSION The psychological resilience profile contains three classes, and the findings provide evidence that high resilience potentially serves as a crucial protective factor for cognitive function in BC patients. Healthcare providers should improve their ability to recognize and evaluate factors influencing resilience, including social support and physical activity, which will enable the development of precise nursing interventions to mitigate the long-term adverse effects of trauma and reduce the impact of cognitive impairment on BC patients.
Collapse
Affiliation(s)
- Xiaotong Ding
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, 33 Ba Dachu Road, Shijingshan District, Beijing, 100144, China
| | - Mingyue Zhu
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, 33 Ba Dachu Road, Shijingshan District, Beijing, 100144, China
| | - Houming Kan
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Qing Wang
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, 33 Ba Dachu Road, Shijingshan District, Beijing, 100144, China
- School of Nursing, Lanzhou University, 28 Yanxi Road, Chengguan District, Lanzhou, 730010, China
| | - Hongli Chen
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, 33 Ba Dachu Road, Shijingshan District, Beijing, 100144, China
| | - Xuan Xia
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, 33 Ba Dachu Road, Shijingshan District, Beijing, 100144, China
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Fang Zhao
- Department of Internal Medicine, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Zheng Li
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, 33 Ba Dachu Road, Shijingshan District, Beijing, 100144, China.
| |
Collapse
|
12
|
Jin Y, Zhao M, Su T, Fan Y, Ouyang Z, Lv F. Comparing Random Survival Forests and Cox Regression for Nonresponders to Neoadjuvant Chemotherapy Among Patients With Breast Cancer: Multicenter Retrospective Cohort Study. J Med Internet Res 2025; 27:e69864. [PMID: 40198909 DOI: 10.2196/69864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/19/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Breast cancer is one of the most common malignancies among women worldwide. Patients who do not achieve a pathological complete response (pCR) or a clinical complete response (cCR) post-neoadjuvant chemotherapy (NAC) typically have a worse prognosis compared to those who do achieve these responses. OBJECTIVE This study aimed to develop and validate a random survival forest (RSF) model to predict survival risk in patients with breast cancer who do not achieve a pCR or cCR post-NAC. METHODS We analyzed patients with no pCR/cCR post-NAC treated at the First Affiliated Hospital of Chongqing Medical University from January 2019 to 2023, with external validation in Duke University and Surveillance, Epidemiology, and End Results (SEER) cohorts. RSF and Cox regression models were compared using the time-dependent area under the curve (AUC), the concordance index (C-index), and risk stratification. RESULTS The study cohort included 306 patients with breast cancer, with most aged 40-60 years (204/306, 66.7%). The majority had invasive ductal carcinoma (290/306, 94.8%), with estrogen receptor (ER)+ (182/306, 59.5%), progesterone receptor (PR)- (179/306, 58.5%), and human epidermal growth factor receptor 2 (HER2)+ (94/306, 30.7%) profiles. Most patients presented with T2 (185/306, 60.5%), N1 (142/306, 46.4%), and M0 (295/306, 96.4%) staging (TNM meaning "tumor, node, metastasis"), with 17.6% (54/306) experiencing disease progression during a median follow-up of 25.9 months (IQR 17.2-36.3). External validation using Duke (N=94) and SEER (N=2760) cohorts confirmed consistent patterns in age (40-60 years: 59/94, 63%, vs 1480/2760, 53.6%), HER2+ rates (26/94, 28%, vs 935/2760, 33.9%), and invasive ductal carcinoma prevalence (89/94, 95%, vs 2506/2760, 90.8%). In the internal cohort, the RSF achieved significantly higher time-dependent AUCs compared to Cox regression at 1-year (0.811 vs 0.763), 3-year (0.834 vs 0.783), and 5-year (0.810 vs 0.771) intervals (overall C-index: 0.803, 95% CI 0.747-0.859, vs 0.736, 95% CI 0.673-0.799). External validation confirmed robust generalizability: the Duke cohort showed 1-, 3-, and 5-year AUCs of 0.912, 0.803, and 0.776, respectively, while the SEER cohort maintained consistent performance with AUCs of 0.771, 0.729, and 0.702, respectively. Risk stratification using the RSF identified 25.8% (79/306) high-risk patients and a significantly reduced survival time (P<.001). Notably, the RSF maintained improved net benefits across decision thresholds in decision curve analysis (DCA); similar results were observed in external studies. The RSF model also showed promising performance across different molecular subtypes in all datasets. Based on the RSF predicted scores, patients were stratified into high- and low-risk groups, with notably poorer survival outcomes observed in the high-risk group compared to the low-risk group. CONCLUSIONS The RSF model, based solely on clinicopathological variables, provides a promising tool for identifying high-risk patients with breast cancer post-NAC. This approach may facilitate personalized treatment strategies and improve patient management in clinical practice.
Collapse
Affiliation(s)
- Yudi Jin
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Zhao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tong Su
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanjia Fan
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zubin Ouyang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fajin Lv
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Ma Z, Zheng M, Liu P. Identification of fatty acid metabolism-related genes in the tumor microenvironment of breast cancer by a development and validation of prognostic index signature. Hereditas 2025; 162:55. [PMID: 40197314 PMCID: PMC11974137 DOI: 10.1186/s41065-025-00425-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Breast cancer (BRCA) is a malignancy originating in the breast cells, characterized by a poor overall survival rate. Post-resection, chemotherapy is commonly recommended as a primary therapeutic approach; however, its efficacy remains limited. Recent advancements in lipidomics and metabolomics have provided new insights into the intricate landscape of fatty acid metabolism (FAM) and the fatty acid lipidome in both health and disease. A growing body of evidence suggests that dysregulations in FAM and fatty acid levels play a significant role in cancer initiation and progression. Despite these advances, the precise mechanisms through which FAM mediates the anti-cancer effects of lobaplatin in BRCA remain poorly understood and warrant further investigation. METHODS GEO and TCGA data were classified into two types. We aimed to show how FAMGs influence immune function, immune checkpoints, and m6a in BRCA. A co-expression analysis discovered that gene expression is strongly connected to pyroptosis. The TCGA gathered information about mRNAsi, gene mutations, CNV, and clinical features. RESULTS In the low-risk group, overall survival (OS) is longer. GSEA was utilized to identify immune and tumor-related pathways. Most of the FAMG-derived prognostic signatures predominantly modulate immunological and oncogenic signaling pathways, including the Wnt, neurotrophin, chemokine, and calcium signaling cascades. Among the genes involved are CEL, WT1, and ULBP2. Expression levels varied as well. The prognostic model, CNVs, single nucleotide polymorphism (SNP), and drug sensitivity all pointed to the gene. CONCLUSIONS The primary objective of this study is to identify and validate BRCA-associated FAMGs that can serve as prognostic indicators and provide insights into immune system function, while also offering evidence to support the development of fatty acid metabolism-related molecularly targeted therapeutics. Consequently, FAMGs and their interactions with the immune system, as well as their role in BRCA, may emerge as promising therapeutic targets.
Collapse
Affiliation(s)
- Zhaofeng Ma
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Man Zheng
- Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong Province, 257091, China
| | - Pulin Liu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China.
| |
Collapse
|
14
|
Wu Q, Cao H, Jin J, Ma D, Niu Y, Yu Y, Wang X, Xia Y. Integrated multi-omics analysis reveals the functional and prognostic significance of lactylation-related gene PRDX1 in breast cancer. Front Mol Biosci 2025; 12:1580622. [PMID: 40256656 PMCID: PMC12006012 DOI: 10.3389/fmolb.2025.1580622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/20/2025] [Indexed: 04/22/2025] Open
Abstract
Background Breast cancer (BRCA) is a significant threat to women's health worldwide, and its progression is closely associated with the tumor microenvironment and gene regulation. Lactylation modification, as a key epigenetic mechanism in cancer biology, has not yet been fully elucidated in the context of BRCA. This study examines the regulatory mechanisms of lactylation-related genes (LRGs), specifically PRDX1, and their prognostic significance in BRCA. Methods We integrated data from multiple databases, including Genome-Wide Association Study (GWAS) summary statistics, single-cell RNA sequencing, spatial transcriptomics, and bulk RNA sequencing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Using Summary-based Mendelian Randomization (SMR) analysis, we identified LRGs associated with BRCA and comprehensively analysed the expression patterns of PRDX1, cell-cell communication networks, and spatial heterogeneity. Furthermore, we constructed and validated a prognostic model based on the gene expression profile of PRDX1-positive monocytes, evaluating it through Cox regression and LASSO regression analyses. Results PRDX1 was identified as a key LRG significantly associated with BRCA risk (p_SMR = 0.0026). Single-cell RNA sequencing analysis revealed a significant upregulation of PRDX1 expression in monocytes, with enhanced cell-cell communication between PRDX1-positive monocytes and fibroblasts. Spatial transcriptomics analysis uncovered heterogeneous expression of PRDX1 in the tumor nest regions, highlighting the spatial interaction between PRDX1-positive monocytes and fibroblasts. The prognostic model constructed based on the gene expression profile of PRDX1-positive monocytes demonstrated high accuracy in predicting patient survival in both the training and validation cohorts. High-risk patients exhibited immune-suppressive microenvironment characteristics, including reduced immune cell infiltration and upregulation of immune checkpoint gene expression. Conclusion This study reveals the key role of PRDX1 in BRCA progression, mainly through the regulation of the tumor microenvironment and immune escape mechanisms. The survival prediction model based on PRDX1 shows robust prognostic potential, and future research should focus on integrating PRDX1 with other biomarkers to enhance the precision of personalised medicine.
Collapse
Affiliation(s)
- Qinqing Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, China
- School of Public Health, Shantou University, Shantou, China
| | - Heng Cao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangdong Jin
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongxu Ma
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yixiao Niu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, China
- School of Public Health, Shantou University, Shantou, China
| | - Yanping Yu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, China
- School of Public Health, Shantou University, Shantou, China
| | - Xiang Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiqin Xia
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Ruan G, Ye L, Ke J, Lin H, Wu M, Liu Z, Fang Y, Zhang S, Wang H, Liu Y, Song H. All-In-One Gadolinium-Doxorubicin Nanoassemblies for Spatial Delivery and Chemoresistance Reversal in Tumor Microenvironments. ACS APPLIED MATERIALS & INTERFACES 2025; 17:19348-19366. [PMID: 40117447 DOI: 10.1021/acsami.4c21725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Efficiently overcoming chemoresistance in tumor microenvironments remains a critical hurdle in cancer therapy due to tumor heterogeneity, limited drug penetration, and adaptive resistance mechanisms. Herein, we report the design and application of all-in-one gadolinium-doxorubicin nanoassemblies (GDNAs) for spatially targeted delivery and chemoresistance reversal. These multifunctional nanoassemblies integrate a lanthanide-based component for real-time imaging and doxorubicin for chemotherapy, coupled with bioinformatics-guided small interfering RNAs (siRNAs) to silence key resistance-associated genes such as BCL2 and BIRC5. The GDNAs demonstrate enhanced tumor penetration and specificity for chemoresistant cells, achieving deep tissue delivery and synergistic effects in human-derived organoids and xenograft breast cancer models. Remarkably, GDNAs significantly reduce tumor viability and growth while attenuating invasive potential, showcasing superior therapeutic efficacy compared to conventional treatments. Comprehensive preclinical evaluations confirm their biocompatibility and low systemic toxicity, underscoring the translational potential of this platform. This work introduces a paradigm-shifting strategy by integrating imaging, targeted therapy, and gene silencing to address chemoresistance, offering a versatile approach for personalized cancer treatment.
Collapse
Affiliation(s)
- Guanyu Ruan
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
- Fujian Maternity and Child Health Hospital College of Clinical Medical College for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Lixiang Ye
- Fujian Center for Safety Evaluation of New Drugs, Fujian Medical University, Fuzhou, Fujian 351004, P. R. China
| | - Jianxi Ke
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, P. R. China
| | - Hongyu Lin
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Minxia Wu
- Electron Microscopy Laboratory of Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian 351004, P. R. China
| | - Zhihong Liu
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian 350025, P. R. China
| | - Yu Fang
- Fujian Center for Safety Evaluation of New Drugs, Fujian Medical University, Fuzhou, Fujian 351004, P. R. China
| | - Shuihua Zhang
- Fujian Center for Safety Evaluation of New Drugs, Fujian Medical University, Fuzhou, Fujian 351004, P. R. China
| | - Hongmei Wang
- Fujian Maternity and Child Health Hospital College of Clinical Medical College for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
| | - Yongsheng Liu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, P. R. China
| | - Hongtao Song
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350001, P. R. China
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, Fujian 350025, P. R. China
| |
Collapse
|
16
|
Santucci C, Mignozzi S, Levi F, Malvezzi M, Boffetta P, Negri E, La Vecchia C. European cancer mortality predictions for the year 2025 with focus on breast cancer. Ann Oncol 2025; 36:460-468. [PMID: 40074664 DOI: 10.1016/j.annonc.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND We predicted the number of cancer deaths and rates for 2025 in the European Union (EU), its five most populous countries, and the UK, focusing on breast cancer. MATERIALS AND METHODS We derived population data and death certificates for all cancers and major sites for the EU, France, Germany, Italy, Poland, Spain, and the UK since 1970, from the World Health Organization and United Nations databases. Estimates for 2025 were computed by linear regression on recent trends identified through Poisson joinpoint regression, considering the slope of the most recent trend segment. Deaths averted from 1989 to 2025 were calculated by applying the 1988 peak rate to subsequent population data. RESULTS We estimated 1 280 000 cancer deaths in the EU in 2025, corresponding to age-standardised rates (ASRs) of 120.9/100 000 males (-3.5% versus 2020) and 79.1/100 000 females (-1.2%). In the UK, we predicted 173 000 cancer deaths and ASRs of 101.2/100 000 males (-10.1%) and 82.1/100 000 females (-6.3%). In the EU, favourable trends are predicted for major neoplasms, except pancreatic cancer, in males (+2.0%) and females (+3.0%), and lung (+3.8%) and bladder (+1.9%) cancers among females. Breast cancer mortality showed favourable trends in all countries. Substantial decreases were predicted for EU females aged 50-69 years (-9.8%) and 70-79 years (-12.4%). Between 1989 and 2025, we estimated about 6.8 million averted cancer deaths in the EU, including over 373 000 breast cancer deaths. Corresponding numbers for the UK were 1 500 000 and 197 000. CONCLUSION EU breast cancer rates have fallen by 30% since 1990, due to advances in prevention, treatment, and early detection. Contrasting trends in lung cancer among males and females reflect differing tobacco smoking patterns. Female lung cancer mortality is still increasing in the EU, though less than in the previous decade. Persistent unfavourable pancreatic cancer trends can be related to the increasing prevalence of obesity and limited therapeutic advances, requiring continued attention.
Collapse
Affiliation(s)
- C Santucci
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - S Mignozzi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - F Levi
- Department of Epidemiology and Health Services Research, Centre for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - M Malvezzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - P Boffetta
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, USA; Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - E Negri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - C La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| |
Collapse
|
17
|
Jung W, Park SH, Park YMM, Song YM, Park JH, Yu J, Cho IY, Kim BS, Han K, Shin DW. Weight change and cardiovascular disease incidence in breast cancer survivors: a nationwide cohort study. Breast Cancer Res Treat 2025; 210:583-593. [PMID: 39762706 DOI: 10.1007/s10549-024-07594-2] [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: 09/08/2024] [Accepted: 12/17/2024] [Indexed: 03/04/2025]
Abstract
BACKGROUND Breast cancer survivors (BCS) face a higher risk of cardiovascular disease (CVD) due to treatment-related cardiotoxicity and pre-existing conditions. We investigated how post-diagnosis weight changes and obesity impact CVD risk in this population. METHOD Using the Korean National Health Insurance Service database (2010-2019), BCS without previous history of CVD were enrolled. Weight change was determined using standardized anthropometric protocols during biennial health examinations pre- and post-diagnosis. The primary outcome was incident CVD, a composite of myocardial infarction (MI) and ischemic stroke. Adjusted hazard ratios (aHRs) and confidence intervals (CIs) were estimated, accounting for cardiovascular risk factors, cancer treatments, and sociodemographic variables. RESULTS During a mean follow-up of 3.70 years among the 42,547 BCS (mean [SD] age 53.4 [9.4] years), substantial weight gain (> 10%) was associated with increased CVD risk (aHR 1.66, 95% CI 1.05-2.62) and MI risk (aHR 1.83, 95% CI 1.01-3.33) compared to those who maintained their weight. The association between change in obesity status and CVD risk was not significant. Among BCS with sustained obesity, CVD risk was more pronounced in younger survivors (< 50 years) (aHR 3.58, 95% CI 1.94-6.61), and in those using tamoxifen (aHR 1.74, 95% CI 1.11-2.75) (P-interactions < 0.05). CONCLUSIONS Our findings suggest that BCS who experience substantial weight gain post-diagnosis have an increased risk of CVD. Further intervention studies (e.g., GLP-1 agonist) are needed to ascertain the effects of weight changes on CVD risks in BCS.
Collapse
Affiliation(s)
- Wonyoung Jung
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang Hyun Park
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong-Moon Mark Park
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yun-Mi Song
- Department of Family Medicine and Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jae Hyun Park
- Department of Social Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jonghan Yu
- Division of Breast and Endocrine Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - In Young Cho
- Department of Family Medicine and Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Bong Sung Kim
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Dong Wook Shin
- Department of Family Medicine and Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea.
| |
Collapse
|
18
|
Leone JP, Leone J, Hassett MJ, Freedman RA, Avila J, Vallejo CT, Tayob N, Tolaney SM, Lin NU. Incidence, treatment patterns, and mortality for patients with breast cancer during the first year of the COVID-19 pandemic: a population-based study. Breast Cancer Res Treat 2025; 210:285-294. [PMID: 39621161 DOI: 10.1007/s10549-024-07562-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/13/2024] [Indexed: 03/25/2025]
Abstract
PURPOSE The COVID-19 pandemic created significant disruptions in the diagnosis and treatment of breast cancer (BC). Several public health measures were taken with limited evidence on their potential impact. In this observational study, we sought to compare the incidence of BC, treatment patterns, and mortality during 2020 versus 2018 and 2019. METHODS Using the Surveillance, Epidemiology, and End Results program, we identified 37,834 patients with ductal carcinoma in situ (DCIS) and 199,594 with invasive BC between 2018 and 2020. We assessed age-adjusted incidence rates of DCIS and invasive BC as cases per 100,000, treatment patterns, and mortality in 2020 versus 2018 and 2019. RESULTS From 2019 to 2020, the incidence of female DCIS decreased from 36.4 to 31.0, and the incidence of female invasive BC decreased from 184.2 to 166.6. Among females, the relative reductions in incidence from 2019 to 2020 were 14.8% for DCIS, 12.1% for stage I, 5.8% for stage II, 2.6% for stage III, and 1.9% for stage IV. Comparing 2020 to 2018-2019 in invasive BC, we observed significant changes in treatment patterns with decreased use of surgery or radiation and increased use of chemotherapy. The 12-month mortality rates were 4.49%, 4.37%, and 4.57% for 2018, 2019 and 2020, respectively. In the Cox model, there were no significant differences in mortality between patients diagnosed in 2020 versus 2018 or 2019. CONCLUSIONS During 2020, the incidence of BC decreased significantly. There were reductions in surgery and radiation use, but not in chemotherapy. Although vaccines were largely unavailable and COVID-19 treatments were in development, we saw no differences in 12-month mortality in 2020 versus prior years. The impact on BC-specific outcomes requires further follow-up.
Collapse
Affiliation(s)
- José Pablo Leone
- Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Julieta Leone
- Grupo Oncológico Cooperativo del Sur, Neuquén, Argentina
| | - Michael J Hassett
- Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rachel A Freedman
- Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jorge Avila
- St. Elizabeth Medical Center, Boston, MA, USA
| | | | - Nabihah Tayob
- Harvard Medical School, Boston, MA, USA
- Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nancy U Lin
- Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Baş O, Tokatlı M, Şavklıyıldız M, Yazarkan Y, Guduk N, Kılınç C, Karahan L, Şahin TK, Guven DC, Aksoy S. Modified cachexia index and survival in metastatic breast cancer patients treated with CDK 4-6 inhibitors. Expert Rev Anticancer Ther 2025; 25:405-409. [PMID: 39989252 DOI: 10.1080/14737140.2025.2471010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/01/2025] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND The objective of this study is to evaluate the correlation between survival outcomes and the modified cachexia index (mCXI) in patients with metastatic breast cancer who have been treated with cyclin-dependent kinase (CDK) 4/6 inhibitors. METHODS This study was conducted on patients with metastatic breast cancer who received CDK 4/6 inhibitors (either Palbociclib or Ribociclib) between January 2020 and November 2024. RESULTS 240 patients were included. A total of 236 patients (98.3%) were female. Median age was 57 (IQR: 48-66 years). The median follow-up period from the initiation of CDK 4/6 inhibitors to the last control was 20 months. One hundred eighty-four patients (76.7%) received ribociclib, while 56 (23.3%) received palbociclib. At diagnosis, 179 patients (74.6%) had metastatic disease. Patients are classified as modified cachexia index-low (mCXI-Low) and mCXI-High according to the receiver operating characteristic (ROC) analysis results for overall survival (OS) prediction [AUC: 0,654 p:<0,001 Cut-off value = 93.5]. Following multivariate analysis, both progression-free survival [HR: 1.50 (95% CI 1.07-2.12), p = 0.02] and overall survival [HR: 3.22 (95% CI 1.90-5.46), p < 0.001] were found to be significantly associated with mCXI. CONCLUSION mCXI is associated with overall survival in metastatic breast cancer patients who are treated with CDK 4-6 inhibitors.
Collapse
Affiliation(s)
- Onur Baş
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Mert Tokatlı
- Department of Internal Medicine, Hacettepe University, Ankara, Turkey
| | | | - Yiğit Yazarkan
- Department of Internal Medicine, Hacettepe University, Ankara, Turkey
| | - Naciye Guduk
- Department of Internal Medicine, Hacettepe University, Ankara, Turkey
| | - Can Kılınç
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Latif Karahan
- Department of Internal Medicine, Hacettepe University, Ankara, Turkey
| | - Taha Koray Şahin
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Deniz Can Guven
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Sercan Aksoy
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
20
|
Tang H, Zhang W, Weng Y, Zhang X, Shen H, Li X, Liu Y, Liu W, Xiao H, Jing H. Dietary self-management behavior and associated factors among breast cancer patients receiving chemotherapy: A latent profile analysis. Eur J Oncol Nurs 2025; 75:102825. [PMID: 39914133 DOI: 10.1016/j.ejon.2025.102825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 03/09/2025]
Abstract
PURPOSE Breast cancer patients experience more gastrointestinal symptoms during chemotherapy than during the survival period post-treatment. This can interfere with daily dietary intake and increase the risk of malnutrition. This study aimed to evaluate the level and classification of dietary self-management behavior and the influence of self-care self-efficacy, outcome expectations, treatment self-regulation and perceived social support guided by "Integrated theory of Health Behavior Change". METHODS A multicenter cross-sectional study was conducted. The assessment tools included self-designed demographic and clinical instruments, Dietary Management Behavior Questionnaire, Strategies Used by People to Promote Health, Consideration of Future Consequences Scale-Food, the Treatment Self-regulation Questionnaire and Perceived Social Support Scale. RESULTS A total of 760 samples were analysed. The best-fitting model of the 4 classes was determined, with the lowest-to-highest scores as follows: Class 1-poor behavior group (17.84%), Class 2-medium behavior group (28.26%), Class 3-good behavior group (22.67%) and Class 4-excellent behavior group (31.23%). In the control group, being underweight (OR = 0.131), having no spouse (OR = 0.235), having a lower educational level (OR = 0.147-0.384), being premenopausal (OR = 0.220-0.255), having immediate consequence considerations (OR = 0.866-0.909) and receiving other support (OR = 0.830) were risk factors (all P < 0.05). CONCLUSIONS The performance of dietary self-management behavior needs to be improved. This study may provide a reference for determining key intervention populations that are underweight during premenopause, have no spouse and have lower education. Specific intervention content, including future consequence consideration, positive attitudes, decision-making, autonomous motivation, introjected regulation and friend support, should be added to improve the dietary practices during chemotherapy precisely.
Collapse
Affiliation(s)
- Han Tang
- School of Nursing, Wenzhou Medical University, 325035, Wenzhou, China.
| | - Wei Zhang
- School of Nursing, Wenzhou Medical University, 325035, Wenzhou, China
| | - Ye Weng
- School of Nursing, Wenzhou Medical University, 325035, Wenzhou, China
| | - Xiaoyang Zhang
- Department of Oncology, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Haiyan Shen
- Department of Orthopedics 1, Sichuan Provincial People's Hospital, 610072, Chengdu, China
| | - Xiaochun Li
- Medical Department, The First Affiliated Hospital of Air Force Medical University, 710032, Xi'an, China
| | - Yue Liu
- Department of Health Statistics, School of Public Health, The Fourth Military Medical University, 710032, Xi'an, China
| | - Wei Liu
- Department of Breast Surgery 2 (Area 1), The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Han Xiao
- Department of Breast Surgery 2 (Area 1), The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Haihong Jing
- Department of Breast Surgery 2 (Area 1), The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| |
Collapse
|
21
|
Sahin TK, Rizzo A, Guven DC, Aksoy S. Post-progression treatment options after CDK4/6 inhibitors in hormone receptor-positive, HER2-negative metastatic breast cancer. Cancer Treat Rev 2025; 135:102924. [PMID: 40121890 DOI: 10.1016/j.ctrv.2025.102924] [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: 10/31/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
The combination of cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) and endocrine therapy (ET) is the standard first-line treatment for hormone receptor-positive (HR + ) and HER2-negative metastatic breast cancer (mBC). Despite their efficacy, resistance inevitably develops, necessitating alternative therapeutic strategies post-progression. This review explores current and emerging treatment options following progression on CDK4/6i, focusing on endocrine therapies, targeted therapies, combination approaches, and the continued use of CDK4/6i. Endocrine therapies, including fulvestrant and novel oral selective estrogen receptor degraders (SERDs) like elacestrant, show promise, especially in patients with ESR1 mutations. Targeted therapies such as PI3K/AKT/mTOR inhibitors, exemplified by alpelisib and capivasertib, offer potential by addressing downstream signaling pathways involved in resistance. Additionally, FGFR inhibitors like erdafitinib are under investigation for their role in overcoming specific resistance mechanisms. Combination strategies involving CDK4/6 inhibitors with immune checkpoint inhibitors or other targeted agents are also being explored, with early trials suggesting possible synergistic effects, although further validation is required. Continuation of CDK4/6 inhibitors beyond progression has shown potential benefits in selected patients, but the data are heterogeneous, and further studies are needed to clarify their role. While chemotherapy remains a standard option for patients who progress on these treatments, the goal is to delay its use through the effective utilization of endocrine and targeted therapies. Understanding resistance mechanisms and tailoring treatment to individual patient profiles is crucial for optimizing outcomes. Ongoing clinical trials are expected to provide deeper insights, guiding the development of more effective post-progression therapeutic strategies. This evolving landscape highlights the need for continuous research and individualized patient care to improve survival and quality of life in HR + mBC patients.
Collapse
Affiliation(s)
- Taha Koray Sahin
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Turkey.
| | | | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
22
|
Ransohoff JD, Lewinsohn RM, Dickerson J, Luo I, Satoyoshi M, Ritter V, Chavez R, Wheeler AJ, Liang SY, Kenkare P, Gomez SL, Schapira L, John EM, Han SS, Kurian AW. Endocrine Therapy Interruption, Resumption, and Outcomes Associated With Pregnancy After Breast Cancer. JAMA Oncol 2025; 11:423-426. [PMID: 39946107 PMCID: PMC11826432 DOI: 10.1001/jamaoncol.2024.6868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 02/16/2025]
Abstract
This cohort study examines interruption and resumption of endocrine therapy among women with breast cancer who became pregnant.
Collapse
Affiliation(s)
- Julia D. Ransohoff
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Rebecca M. Lewinsohn
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - James Dickerson
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Ingrid Luo
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California
| | - Mina Satoyoshi
- Stanford Hospital/Technology and Digital Solutions/Research Technologies/Data Services, Stanford, California
| | - Victor Ritter
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California
| | - Rachael Chavez
- Palo Alto Medical Foundation Research Institute, Sutter Health, Palo Alto, California
| | - Amanda J. Wheeler
- Palo Alto Medical Foundation Research Institute, Sutter Health, Palo Alto, California
| | - Su-Ying Liang
- Palo Alto Medical Foundation Research Institute, Sutter Health, Palo Alto, California
| | - Pragati Kenkare
- Palo Alto Medical Foundation Research Institute, Sutter Health, Palo Alto, California
| | - Scarlett L. Gomez
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Lidia Schapira
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California
| | - Esther M. John
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California
| | - Summer S. Han
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Allison W. Kurian
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
23
|
Ilovaisky AI, Scherbakov AM, Miciurov D, Chernoburova EI, Merkulova VM, Bogdanov FB, Salnikova DI, Sorokin DV, Krasil'nikov MA, Bozhenko EI, Zavarzin IV, Terent'ev AO. Secosteroid - 1,3,4-oxadiazole hybrids: Synthesis and evaluation of their activity against hormone-dependent breast cancer cells. J Steroid Biochem Mol Biol 2025; 251:106745. [PMID: 40164235 DOI: 10.1016/j.jsbmb.2025.106745] [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: 01/20/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
This study focused on the synthesis of secosteroids with good antiproliferative properties against hormone-dependent breast cancer. A straightforward and efficient method for synthesizing secosteroid - 1,3,4-oxadiazole hybrids was developed starting from 13α-hydroxy-3-methoxy-13,17-secoestra-1,3,5(10)-trien-17-oic acid hydrazide. The cyclization of hydrazide moiety with CS2 into 1,3,4-oxadiazole-2(3H)-thione fragment followed by sulfur alkylation resulted in the formation of various secosteroid - 2-mercapto-1,3,4-oxadiazole hybrids. These novel compounds were evaluated for their antiproliferative activity against the hormone-dependent human breast cancer cell line MCF-7. Among the synthesized hybrids, compounds 3i, 3o, and 3q displayed notable antiproliferative effects, with IC50 values ranging from 6.5 to 8.9 µM, comparable to the reference drug cisplatin. Furthermore, compound 3i showed minimal toxicity toward non-cancerous hFB-hTERT fibroblasts, indicating high selectivity. Compounds 3o and 3q exhibited antiestrogenic activity. Additionally, their effects on PARP and Bcl-2 suggest a pro-apoptotic mechanism of action. These findings highlight the potential of secosteroidal hybrids as promising candidates for the development of new anti-breast cancer agents targeting ERα and apoptosis pathways.
Collapse
Affiliation(s)
- Alexey I Ilovaisky
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexander M Scherbakov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia; Gause Institute of New Antibiotics, Bol'shaya Pirogovskaya ulitsa 11, Moscow 119021, Russia
| | - Dumitru Miciurov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Elena I Chernoburova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Valentina M Merkulova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Fedor B Bogdanov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Diana I Salnikova
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Danila V Sorokin
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Mikhail A Krasil'nikov
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24, Moscow 115522, Russia
| | - Eugene I Bozhenko
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Igor V Zavarzin
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexander O Terent'ev
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| |
Collapse
|
24
|
Zhang Y, Rodriguez J, Mao X, Grassmann F, Tapia J, Eriksson M, Hall P, Czene K. Incidence and Risk Factors of Interval and Screen-Detected Breast Cancer. JAMA Oncol 2025:2832064. [PMID: 40146116 PMCID: PMC11950978 DOI: 10.1001/jamaoncol.2025.0167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 03/28/2025]
Abstract
Importance Mammographic screening is the only proven method for early detection and mortality reduction of breast cancer (BC). However, many patients are missed at prior screening; thus, they receive their diagnosis between the interval of screening rounds, called interval cancer (IntCa). Some IntCas are fast growing between screening rounds. Objective To investigate the incidence and proportion of IntCa and screen-detected breast cancer (ScrCa) and identify factors associated with IntCa. Design, Setting, and Participants This population-based cohort study was conducted from January 1989 to March 2020, with follow-up until 2020 and a mean (SD) follow-up of 13 (8.3) years. The statistical analysis was performed from February 2023 to June 2024. It included cancer-free women (N = 527 144) residing in Stockholm, Sweden, who were invited to undergo mammography screening (aged 40-74 years) during 1989 to 2020. An additional cohort of women were included who were participating in the Karolinska Mammography Project for Risk Prediction of Breast Cancer study and had mammography data available. Exposures Family cancer history (defined from the Swedish Multi-Generation Register and Cancer Register), mammographic density, and various demographic, reproductive, and other factors (multiple Swedish registers). Main Outcomes and Measures Incidence of ScrCa and IntCa (defined from the Swedish Cancer Register in conjunction with individual screening histories). Results A total of 29 049 women (5.5%) received a diagnosis of BC, of whom 10 631 (2.0%) had ScrCa and 4369 (0.8%) IntCa. ScrCa and IntCa incidences increased during the period. The proportion of IntCa among screened patients with BC was around 30%, which decreased with older age. Factors associated with increased risk of IntCa included older age at first childbirth, higher education level, hormone replacement therapy, and higher mammographic density. Risk estimates of family cancer history on IntCa were family history of BC (hazard ratio [HR], 1.85; 95% CI, 1.72-1.99), family history of IntCa (HR, 2.92; 95% CI, 2.39-3.55), and hereditary breast and ovarian cancers (HR, 1.45; 95% CI, 1.36-1.54), with risk further elevated with the number of relatives who received a diagnosis when younger than the median age. Women with IntCa were more likely to have estrogen receptor (ER)-negative cancers than women with ScrCa (22% vs 11%), and having family history of ER-negative BC was associated with 3-fold risk for ER-negative IntCa. Conclusions and Relevance The results of this cohort study suggest that IntCa rates have not decreased with age-based screening, and implementing risk-based screening considering IntCa-specific risk factors is necessary for improving outcomes.
Collapse
Affiliation(s)
- Yuqi Zhang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Juan Rodriguez
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Xinhe Mao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Felix Grassmann
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jose Tapia
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Sharma G, Panwar R, Saini S, Tuli HS, Wadhwa K, Pahwa R. Emerging phytochemical-based nanocarriers: redefining the perspectives of breast cancer therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04003-3. [PMID: 40137964 DOI: 10.1007/s00210-025-04003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/28/2025] [Indexed: 03/29/2025]
Abstract
Breast cancer is recognized as the most prevalent condition impacting women globally, despite several advancements in diagnosis and treatment. Existing therapeutic interventions including surgical procedures, radiation therapy, and chemotherapy often produce harmful effects on healthy tissues, trigger chemo-resistance, and augment the risk of relapse. In response to several unmet challenges, substantial research has been conducted to explore the therapeutic potential of natural compounds for breast cancer therapy. Progress in phytochemistry and pharmacology has facilitated the identification of diverse herbal bioactives with favorable safety profiles and multi-target mechanisms of action against breast cancer cells. Several phytochemicals like flavonoids and tannins have shown significant anticancer potential against breast cancer in diverse preclinical models. However, challenges like limited cellular absorption, low water solubility, and high molecular weight hinder their effective translation into clinical applications. Therefore, the development of novel therapies is imperative for overcoming these hurdles in breast cancer treatment effectively. Nanotechnology has reflected considerable perspective in tackling diverse challenges by encapsulating phytoconstituents within various nanocarriers including polymeric nanoparticles, lipidic nanoparticles, nanoemulsions, nanogels, gold nanoparticles, and silver nanoparticles. This manuscript emphasizes the recent advancements in phytochemical-loaded nanocarriers efficiently tailored for breast cancer therapy along with patents, current challenges, and future perspectives in this avenue.
Collapse
Affiliation(s)
- Gulshan Sharma
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Rohil Panwar
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Sanskriti Saini
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Hardeep Singh Tuli
- Department of Bio-Science and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| | - Rakesh Pahwa
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India.
| |
Collapse
|
26
|
Hockaden N, Leriger G, Wang J, Ray H, Chakrabarti S, Downing N, Desmond J, Williams D, Hollenhorst PC, Longmore G, Carpenter RL. Amyloidogenesis promotes HSF1 activity enhancing cell survival during breast cancer metastatic colonization. Cell Stress Chaperones 2025; 30:143-159. [PMID: 40147541 PMCID: PMC12002613 DOI: 10.1016/j.cstres.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/10/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer among women and the second leading cause of cancer deaths in women. A majority of these breast cancer deaths are due to metastasis, which occurs when primary tumor cells invade into the blood stream to travel and colonize at distant organ sites. Metastatic colonization is the rate-limiting step of metastasis. Heat shock factor 1 (HSF1) is a transcription factor that has been shown to be involved in promoting malignancy with a function in metastatic dissemination due to its contribution to promoting epithelial-to-mesenchymal transition. The role of HSF1 in colonization is unclear. In this study, we observed that HSF1 was essential for metastatic colonization. Consistent with these findings, we also observed that HSF1 was more active in human metastatic tumors compared to primary tumors. HSF1 was also seen to be activated during in vitro colony formation, which was accompanied by increases in amyloid beta (Aβ) fibrils, which was also observed in human metastatic tumors. Aβ fibrils led to HSF1 activation and depletion or inhibition of HSF1 led to increases in Aβ fibrils. HSF1 inhibition with small molecule inhibitors suppressed in vitro colony formation and mammosphere growth of metastatic breast cancer cells. These results suggest that colonization increases Aβ fibril formation that subsequently activates HSF1 as a cell survival mechanism that is essential for metastatic initiation and outgrowth.
Collapse
Affiliation(s)
| | - Gabi Leriger
- Medical Sciences, Indiana University, Bloomington, IN 47405
| | - John Wang
- Medical Sciences, Indiana University, Bloomington, IN 47405
| | - Haimanti Ray
- Medical Sciences, Indiana University, Bloomington, IN 47405
| | | | | | - Jacob Desmond
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - David Williams
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University, Bloomington, IN 47405; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202; Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Gregory Longmore
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Richard L Carpenter
- Medical Sciences, Indiana University, Bloomington, IN 47405; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202; Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202.
| |
Collapse
|
27
|
Niture S, Ghosh S, Jaboin J, Seneviratne D. Tumor Microenvironment Dynamics of Triple-Negative Breast Cancer Under Radiation Therapy. Int J Mol Sci 2025; 26:2795. [PMID: 40141437 PMCID: PMC11943269 DOI: 10.3390/ijms26062795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by the absence of estrogen receptors (ER), progesterone receptors (PR), and HER2 expression. While TNBC is relatively less common, accounting for only 10-15% of initial breast cancer diagnosis, due to its aggressive nature, it carries a worse prognosis in comparison to its hormone receptor-positive counterparts. Despite significant advancements in the screening, diagnosis, and treatment of breast cancer, TNBC remains an important public health burden. Following treatment with chemotherapy, surgery, and radiation, over 40% of TNBC patients experience relapse within 3 years and achieve the least benefit from post-mastectomy radiation. The tumor microenvironment environment (TME) is pivotal in TNBC initiation, progression, immune evasion, treatment resistance, and tumor prognosis. TME is a complex network that consists of immune cells, non-immune cells, and soluble factors located in the region adjacent to the tumor that modulates the therapeutic response differentially between hormone receptor-positive breast cancer and TNBC. While the mechanisms underlying the radiation resistance of TNBC remain unclear, the immunosuppressive TME of TNBC has been implicated in chemotherapeutic resistance. Radiation therapy (RT) is known to alter the TME; however, immune changes elicited by radiation are poorly characterized to date, and whether these immune changes contribute to radiation resistance remains unknown. This review delves into the distinct characteristics of the TNBC TME, explores how RT influences TME dynamics, and examines mechanisms underlying tumor radiosensitization, radioresistance, and immune responses.
Collapse
Affiliation(s)
- Suryakant Niture
- Department of Radiation Oncology, Stephenson Cancer Center, Oklahoma University, Oklahoma City, OK 73104, USA
| | | | | | - Danushka Seneviratne
- Department of Radiation Oncology, Stephenson Cancer Center, Oklahoma University, Oklahoma City, OK 73104, USA
| |
Collapse
|
28
|
Bullock ZN, Wehrens XHT, Young DW. Editorial to "A Plant-Powered Remedy: Hyperoside's Potential Against Trastuzumab-Induced Cardiotoxicity" by Bullock et al. Cardiovasc Drugs Ther 2025:10.1007/s10557-025-07684-3. [PMID: 40106211 DOI: 10.1007/s10557-025-07684-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Affiliation(s)
- Zaniqua N Bullock
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Center for Drug, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX, 77030, USA
| | - Damian W Young
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.
- Center for Drug, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Loganathan T, George Priya Doss C. Computational molecular insights into ibrutinib as a potent inhibitor of HER2-L755S mutant in breast cancer: gene expression studies, virtual screening, docking, and molecular dynamics analysis. Front Mol Biosci 2025; 12:1510896. [PMID: 40177517 PMCID: PMC11962039 DOI: 10.3389/fmolb.2025.1510896] [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: 10/15/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background The proposed study integrates several advanced computational techniques to unravel the molecular mechanisms underlying breast cancer progression and drug resistance. Methods We investigated HER2-L755S mutation through a multi-step approach, including gene expression analysis, molecular docking, and molecular dynamics simulations. Results and Discussion By conducting a network-based analysis of gene expression data from breast cancer samples, key hub genes such as MYC, EGFR, CDKN2A, ERBB2, CDK1, E2F1, TOP2A, MDM2, TGFB1, and FOXM1 were identified, all of which are critical in tumor growth and metastasis. The study mainly focuses on the ERBB2 gene, which encodes the HER2 protein, and its common mutation HER2-L755S, associated with breast cancer and resistance to the drug lapatinib. The HER2-L755S mutation contributes to both tumorigenesis and therapeutic failure. To address this, alternative therapeutic strategies were investigated using combinatorial computational approaches. The stability and flexibility of the HER2-L755S mutation were evaluated through comparative molecular dynamics simulations over 1000 ns using Gromacs in the unbound (Apo) state. Virtual screening with Schrodinger Glide identified ibrutinib as a promising alternative to lapatinib for targeting the HER2-L755S mutant. Detailed docking and molecular dynamics simulations in the bound (Holo) state demonstrated that the HER2-L755S-ibrutinib complex exhibited higher binding affinity and lower binding energy, indicating more stable interactions compared to other complexes. MM-PBSA analysis revealed that the HER2-L755S-ibrutinib complex had more negative binding energy than the HER2-L755S-afatinib, HER2-L755S-lapatinib, and HER2-L755S-neratinib complexes, suggesting that ibrutinib forms the most stable complex with favorable binding interactions. Conclusion These results provide in-depth atomic-level insights into the binding mechanisms of these inhibitors, highlighting ibrutinib as a potentially effective inhibitor for the clinical treatment of breast cancer.
Collapse
Affiliation(s)
| | - C. George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| |
Collapse
|
30
|
Peng Y, Zhang X, Wu J, Wang H, Huang X. Development and validation of a Nomogram to predict postoperative flap necrosis risk in breast cancer patients undergoing modified radical mastectomy. Am J Cancer Res 2025; 15:1291-1306. [PMID: 40226471 PMCID: PMC11982728 DOI: 10.62347/dyff7059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Flap necrosis is a critical complication following modified radical mastectomy (MRM) for breast cancer (BC). It not only impairs wound healing but also delays postoperative treatment, adversely affecting patient survival rate and the overall quality of life. Thus, developing an accurate prediction model is crucial for early intervention and improving patient prognosis. OBJECTIVE To develop and validate a Nomogram model based on Logistic regression to assess the risk of postoperative flap necrosis in BC patients undergoing MRM. METHODS A retrospective study was conducted on 605 BC patients who underwent MRM. These patients were stratified into a training group (n=406) and a validation group (n=199) in a 33:67 ratio. Univariate and multivariate Logistic regression analyses were performed to identify risk factors for flap necrosis, and a Nomogram prediction model was subsequently constructed. The model's discriminatory power (assessed via the receiver operating characteristic [ROC] curve), calibration accuracy (evaluated by calibration curve), and clinical benefit (analyzed through decision curve analysis) were comprehensively evaluated. Moreover, essential performance metrics such as sensitivity, specificity, and accuracy were systematically recorded and analyzed. RESULTS Nine independent risk factors were identified, including age, body mass index (BMI), neutrophil count, hemoglobin level, drainage volume on the third postoperative day, axillary lymph node metastasis (ALNM), surgical duration, intraoperative bleeding volume, and drainage duration. The area under the curve (AUC) of the Nomogram model was 0.898 in the training group and 0.886 in the validation group, indicating good discriminatory capacity. Calibration curves demonstrated good agreement between predicted values and actual values, with P-values for goodness-of-fit of 0.1761 (training) and 0.0648 (validation), respectively. Decision curve analysis revealed significant clinical benefits, with maximum benefit rates of 76.84% (training) and 80.40% (validation), respectively. CONCLUSION The Nomogram model developed in this study accurately predicts flap necrosis risk in BC patients post-MRM, offering significant clinical utility for risk management and improved patient outcomes.
Collapse
Affiliation(s)
- Yi Peng
- Department of Breast Surgery, Fujian Maternity and Child Health Hospital Fuzhou 350001, Fujian, China
| | - Xin Zhang
- Department of Breast Surgery, Fujian Maternity and Child Health Hospital Fuzhou 350001, Fujian, China
| | - Jianbin Wu
- Department of Breast Surgery, Fujian Maternity and Child Health Hospital Fuzhou 350001, Fujian, China
| | - Hongmei Wang
- Department of Breast Surgery, Fujian Maternity and Child Health Hospital Fuzhou 350001, Fujian, China
| | - Xiaoxi Huang
- Department of Breast Surgery, Fujian Maternity and Child Health Hospital Fuzhou 350001, Fujian, China
| |
Collapse
|
31
|
Doddapaneni R, Tucker JD, Lu PJ, Lu QL. Synergistic Effect of Ribitol and Shikonin Promotes Apoptosis in Breast Cancer Cells. Int J Mol Sci 2025; 26:2661. [PMID: 40141303 PMCID: PMC11942206 DOI: 10.3390/ijms26062661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/18/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
The mortality rate of breast cancer remains high, despite remarkable advances in chemotherapy. Therefore, it is imperative to identify new treatment options. In the present study, we investigated whether the metabolite ribitol enhances the cytotoxic effect of shikonin against breast cancer in vitro. Here, we screened a panel of small molecules targeting energy metabolism against breast cancer. The results of the study revealed that ribitol enhances shikonin's growth-inhibitory effects, with significant synergy. A significant (p < 0.01) increase in the percentage (56%) of apoptotic cells was detected in the combined treatment group, compared to shikonin single-treatment group (38%), respectively. The combined ribitol and shikonin treatment led to significant arrest of cell proliferation (40%) (p < 0.01) compared to untreated cells, as well as the induction of apoptosis. This was associated with upregulation of p53 (p < 0.05) and downregulation of c-Myc (p < 0.01), Bcl-xL (p < 0.001), and Mcl-1 (p < 0.05). Metabolomic analysis supports the premise that inhibition of the Warburg effect is involved in shikonin-induced cell death, which is likely further enhanced by dysregulation of glycolysis and the tricarboxylic acid (TCA) cycle, afflicted by ribitol treatment. In conclusion, the present study demonstrates that the metabolite ribitol selectively enhances the cytotoxic effect mediated by shikonin against breast cancer in vitro.
Collapse
Affiliation(s)
- Ravi Doddapaneni
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA
| | | | | | - Qi L. Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA
| |
Collapse
|
32
|
Smolar M, Kudelova E, Danova I, Lucansky V, Dankova Z, Musova D, Grendar M, Nosakova L, Uhrik P, Samec M. Utilization of lymph node elastography in the axillary staging of breast cancer. Front Oncol 2025; 15:1478701. [PMID: 40182053 PMCID: PMC11966495 DOI: 10.3389/fonc.2025.1478701] [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: 08/10/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background The differential diagnosis of lymphadenopathy is an important determinant of prognosis in patients with breast cancer (BC). Invasive, fine needle aspiration (FNA) biopsy has been long considered as the gold standard for differentiating malignant lymph nodes (LN) from benign ones. Ultrasonography (USG) evaluation is a useful, rapid, and user-friendly imaging tool for LN assessment due to its high resolution. Compared to USG, ultrasound elastography is a relatively novel non-invasive method to differentiate benign and malignant lesions based on the stiffness heterogeneity of the tissue. The purpose of our study was to compare non-invasive imaging techniques, conventional USG, and strain elastography, to differentiate benign and malignant LNs lesions in a cohort of patients with early BC. Methods In total, 50 patients (48 women and 2 men) with histologically confirmed early BC were evaluated by conventional USG in B-mode followed by strain elastography (using parameters: pattern, strain ratio, hue histogram) for assessment of axillary LNs status. The surgical treatment included surgery of regional LNs (sentinel LN biopsy or axillary dissection), which served as the gold standard in statistical processing. Results The USG B-mode was found to have a sensitivity of 68.75% and a specificity of 61.54%. Among strain elastography parameters, the elastographic pattern showed the highest specificity (66.67%) while the sensitivity was 83.3%. The strain ratio showed 100% sensitivity and 55.6% specificity, followed by a hue histogram with a sensitivity of 72.2%, but specificity was only 25.9%. Conclusion Despite promising data, monitored parameters currently cannot reliably replace sentinel LN biopsy. However, the monitored parameters represent an appropriate additional tool that can be used to refine preoperative staging, better targeting of FNA biopsy, and more accurate assessment of LNs in follow-up patients within the dispensary.
Collapse
Affiliation(s)
- Marek Smolar
- Clinic of General, Visceral and Transplant Surgery, Jessenius Faculty of Medicine, Comenius University in Bratislava, University Hospital Martin, Martin, Slovakia
| | - Eva Kudelova
- Clinic of General, Visceral and Transplant Surgery, Jessenius Faculty of Medicine, Comenius University in Bratislava, University Hospital Martin, Martin, Slovakia
| | - Ivana Danova
- Clinic of General, Visceral and Transplant Surgery, Jessenius Faculty of Medicine, Comenius University in Bratislava, University Hospital Martin, Martin, Slovakia
| | - Vincent Lucansky
- Department of Pathological Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Zuzana Dankova
- Biobank for Cancer and Rare Diseases, Jessenius Faculty of Medicine in Martin (JFMED CU), Comenius University in Bratislava, Martin, Slovakia
| | - Diana Musova
- Clinic of General, Visceral and Transplant Surgery, Jessenius Faculty of Medicine, Comenius University in Bratislava, University Hospital Martin, Martin, Slovakia
| | - Marian Grendar
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin (JFMED CU), Comenius University in Bratislava, Martin, Slovakia
| | - Lenka Nosakova
- Clinic of Gastroenterological Internal Medicine, Jessenius Faculty of Medicine, Comenius University in Bratislava, University Hospital, Martin, Slovakia
| | - Peter Uhrik
- Clinic of Gastroenterological Internal Medicine, Jessenius Faculty of Medicine, Comenius University in Bratislava, University Hospital, Martin, Slovakia
| | - Marek Samec
- Department of Medical Biology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| |
Collapse
|
33
|
Tolaney SM, Jhaveri K, Helsten T, Puhalla SL, Conlin A, Dees EC, Beeram M, Chapman SC, Lithio A, Litchfield LM, Goetz MP. Abemaciclib in combination with therapies for patients with metastatic breast cancer: a phase 1b study. Front Oncol 2025; 15:1555921. [PMID: 40144206 PMCID: PMC11937066 DOI: 10.3389/fonc.2025.1555921] [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: 01/05/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Background The oral, selective, and potent small molecule cyclin-dependent kinases (CDK) 4/6 inhibitor (CDK4/6i) abemaciclib has demonstrated efficacy in advanced breast cancer and high-risk early breast cancer. This Phase 1b study evaluated the safety, tolerability, pharmacokinetics, and antitumor activity of abemaciclib in combination with endocrine therapies (Parts A-D), exemestane + everolimus (Part E), or fulvestrant + LY3023414 (a PI3K/mTOR inhibitor; Part G) in patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (MBC), or trastuzumab (Part F), or trastuzumab + pertuzumab (Part H) in patients with HER2-positive (HER2+) MBC. Patients and methods This study enrolled women aged ≥18 years old with either HR+, HER2- (Parts E and G), or HER2+ (Parts F and H) MBC. Additional requirements included measurable disease or non-measurable but evaluable bone disease (Parts E and F), or measurable disease (Parts G and H), an Eastern Cooperative Oncology Group performance status of 0-1, and no prior treatment with CDK4/6i (Parts E, F, and H). Adverse events were graded, and tumor response was assessed. Results Nineteen patients in Part E received abemaciclib (150 mg, n=15; 200 mg, n=4) with exemestane + everolimus, 24 patients in Part F received abemaciclib (150 mg, n=18; 200 mg, n=6) with trastuzumab, 12 patients in Part G received 150 mg abemaciclib with fulvestrant + LY3023414 (100 mg, n=7; 150 mg, n=5), and four patients in Part H received abemaciclib (100 mg) with trastuzumab + pertuzumab (with prophylactic loperamide). The most common treatment-emergent adverse events (TEAEs) were diarrhea, fatigue, neutropenia, and nausea. Grade ≥3 TEAEs were reported in 16, 18, 10, and 4 patients in Parts E-H, respectively. Abemaciclib had no effect on the pharmacokinetics of the combination study drugs. The objective response rates for patients with measurable disease were 46.2%, 10.0%, 66.7%, and 25.0% in Parts E-H, respectively. A recommended Phase 2 dose was not established for Parts E, G, and H at the dose levels evaluated, and was determined to be 150 mg Q12H in Part F. Conclusions Overall, our results demonstrate safety profiles consistent with those previously established for abemaciclib and provide preliminary data for these combination therapies in the treatment of HR+, HER2- or HER2+ MBC.
Collapse
Affiliation(s)
- Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Komal Jhaveri
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Teresa Helsten
- Moores Cancer Center, University of California San Diego, San Diego, CA, United States
| | - Shannon L. Puhalla
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Alison Conlin
- Providence Cancer Center, Portland, OR, United States
| | - E. Claire Dees
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Muralidhar Beeram
- South Texas Accelerated Research Therapeutics, San Antonio, TX, United States
| | | | - Andrew Lithio
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | - Matthew P. Goetz
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
34
|
Wu X, Jin L, Ren D, Huang S, Meng X, Wu Z, Lv C, Ru J, Zhang H, Zhang S, Bao J, Wang O, Xia E. α-Hederin causes ferroptosis in triple-negative breast cancer through modulating IRF1 to suppress GPX4. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156611. [PMID: 40153970 DOI: 10.1016/j.phymed.2025.156611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/22/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Breast cancer ranks first in the global incidence rate of cancer among women. Triple-negative breast cancer (TNBC) is considered to be the most dangerous type because of the lack of specific therapeutic targets and rapid progression. The emergence of ferroptosis provides a new therapeutic perspective for TNBC. α-Hederin is a triterpenoid saponin derived from the traditional Chinese medicine Ivy, which has been proven to have anti-cancer effects on various cancers, but its efficacy and mechanism of inducing ferroptosis in TNBC remain to be further clarified. OBJECT To investigate the effect and mechanism of α-Hederin induced ferroptosis in TNBC. METHOD Cell viability was measured by CCK-8 assay, and cell proliferation and migration were evaluated by clone assay and scratch assay. The effect of α-Hederin on TNBC cell apoptosis was assessed by flow cytometry. Transcriptomics searches for critical pathways. Intracellular and lipid reactive oxygen species and Fe2+and Fe were detected by DCFH-DA probe, FerroOrange fluorescent probe and C11-BODIPY fluorescent probe, and the contents of malondialdehyde and reduced glutathione were detected by MDA and GSH kits. Erastin was used as a positive control for ferroptosis and Ferrrostatin-1(Fer-1) as an inhibitor. The relationship between α-Hederin and GPX4, IRF was analyzed by western blot and si-RNA, and the association was further confirmed by molecular simulation docking, external SPR experiments, and luciferase experiments. Constructing xenograft mouse models and human derived organoid models to evaluate the anti-TNBC efficacy of α-Hederin, and verifying the efficacy and ferroptosis mechanism of the drug in vivo through HE staining and IHC. RESULT α-Hederin significantly inhibited the progression of TNBC. In vitro, α-Hederin decreased cancer cell viability through ferroptosis, increased glutathione degradation and MDA production, and promoted intracellular Fe2+ and ROS production, whereas Fer-1, an ferroptosis inhibitor, reversed this effect. Mechanistically, molecular docking and SPR experiments showed binding of α-Hederin to the key regulator IRF1, and knockdown/overexpression of IRF1 significantly affected the expression of GPX4, a downstream target of the ferroptosis pathway. In vivo, α-Hederin prevented tumor growth in xenograft and organoid models via the IRF1/GPX4 axis. CONCLUSION We proved for the first time in this research that α-Hederin exerts anti-TNBC effects through a novel IRF1/GPX4 ferroptosis pathway.
Collapse
Affiliation(s)
- Xue Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Lingli Jin
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Zhejiang Provincial People's Hospital, Zhejiang 325000, China
| | - Disuo Ren
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shaolong Huang
- Graduate School of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xinyu Meng
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhixuan Wu
- Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chaoyue Lv
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jiatong Ru
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Heyu Zhang
- Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shuwei Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jingxia Bao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Ouchen Wang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Erjie Xia
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
35
|
Narwade M, Haldar N, Samanta R, Pawar A, Gajbhiye V, Gajbhiye KR. α vβ 3 integrin aptamer functionalized pH-responsive lipid polymer hybrid nanoparticles for targeted co-delivery of paclitaxel and tamoxifen. Int J Biol Macromol 2025; 306:141754. [PMID: 40049497 DOI: 10.1016/j.ijbiomac.2025.141754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
Triple-negative breast cancer (TNBC) is the deadliest type due to its aggressive behavior, high recurrence, metastatic, and mortality rates. This study was aimed at the targeted co-delivery of paclitaxel (PTX) and tamoxifen (TMF) via lipid polymer hybrid nanoparticles (LPHNPs) for treating TNBC. Here, we conjugated αvβ3 integrin aptamer over LPHNPs for targeting TNBC cells. The aptamer-conjugated LPHNPs showed significantly higher uptake in 4 T1 cells than non-targeted LPHNPs. The PTX + TMX co-loaded targeted LPHNPs have cell viabilities of 5.9 ± 0.7 and 7.8 ± 0.6 % in 4 T1 and MDA-MB-231 cells, respectively, in 48 h. The cell viabilities of PTX + TMX co-loaded non-targeted LPHNPs and free PTX + TMX were 17.27 ± 1.56 and 24.31 ± 0.81 % in 4 T1 cells and 16.07 ± 0.14 and 20.15 ± 1.11 % in MDA-MB-231 cells, respectively, in 48 h. Flow cytometry indicated that targeted LPHNP-mediated PTX + TMF delivery was considerably more efficient (~31 %) in inducing apoptosis than PTX + TMF co-loaded non-targeted LPHNPs (~21 %) and free PTX + TMF (~13 %). The anti-cancer efficiency was better when PTX and TMF were delivered together rather than separately. The cytotoxicity assessment in the 3D cell culture demonstrated higher anti-cancer effectiveness of aptamer-conjugated co-loaded LPHNPs, confirmed by significantly inducing cell death. Thus, the results concluded that PTX and TMF-loaded αvβ3 integrin aptamer conjugated LPHNPs have tremendous potential for treating TNBC.
Collapse
Affiliation(s)
- Mahavir Narwade
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth, Pune, India
| | - Niladri Haldar
- Nanobioscience, Agharkar Research Institute, Pune 411004, India; Savitribai Phule Pune University, Pune 411007, India
| | - Rajkumar Samanta
- Nanobioscience, Agharkar Research Institute, Pune 411004, India; Savitribai Phule Pune University, Pune 411007, India
| | - Atmaram Pawar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth, Pune, India
| | - Virendra Gajbhiye
- Nanobioscience, Agharkar Research Institute, Pune 411004, India; Savitribai Phule Pune University, Pune 411007, India.
| | - Kavita R Gajbhiye
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth, Pune, India.
| |
Collapse
|
36
|
Zhang W, Wang R, Guo R, Yi Z, Wang Y, Wang H, Li Y, Li X, Song J. The multiple biological activities of hyperoside: from molecular mechanisms to therapeutic perspectives in neoplastic and non-neoplastic diseases. Front Pharmacol 2025; 16:1538601. [PMID: 40098612 PMCID: PMC11911483 DOI: 10.3389/fphar.2025.1538601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
In recent years, hyperoside (quercetin 3-O-β-D-galactopyranoside) has garnered significant attention due to its diverse biological effects, which include vasoprotective, antioxidant, anti-inflammatory, and anti-tumor properties. Notably, hyperoside has shown remarkable potential in cancer therapy by targeting multiple mechanisms; it induces apoptosis, inhibits proliferation, blocks angiogenesis, and reduces the metastatic potential of cancer cells. Furthermore, hyperoside enhances the sensitivity of cancer cells to chemotherapy by modulating key signaling pathways. Beyond neoplastic diseases, hyperoside also presents promising therapeutic applications in managing non-cancerous conditions such as diabetes, Alzheimer's disease, and pulmonary fibrosis. This review comprehensively examines the molecular mechanisms underlying hyperoside's anti-cancer effects and highlights its role in the treatment of cancers, including lung and colorectal cancers. Additionally, it explores the latest research on hyperoside's potential in addressing non-neoplastic conditions, such as pulmonary fibrosis, diabetes, and Parkinson's disease. By summarizing current findings, this review underscores the unique therapeutic value of hyperoside and its potential as a multifunctional treatment in both neoplastic and non-neoplastic contexts.
Collapse
Affiliation(s)
- Weisong Zhang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Rui Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Rongqi Guo
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Zhongquan Yi
- Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Yihao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Hao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Yangyang Li
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Xia Li
- Department of General Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Jianxiang Song
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| |
Collapse
|
37
|
Kakati RT, Whitman AA, Haase S, Szenasi AT, Thai CH, Brunk E, Okumu DO, East MP, Perou CM, Johnson GL, Spanheimer PM. Kinase Plasticity in Response to Vandetanib Enhances Sensitivity to Tamoxifen in Estrogen Receptor Positive Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.19.629395. [PMID: 39975402 PMCID: PMC11838206 DOI: 10.1101/2024.12.19.629395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Resistance to endocrine therapy (ET) is common in estrogen receptor (ER) positive breast cancer. Multiple studies have demonstrated that upregulation of MAPK signaling pathways contributes to ET resistance. Herein we show that vandetanib treatment enhances sensitivity to ET in ET-sensitive and -resistant ER+ breast cancer models. Vandetanib treatment alters the gene expression program of ER+ breast cancer cells resulting in a less proliferative and more estrogen responsive Luminal-A like character. Tyrosine kinase network reprogramming was assessed using multiplexed kinase inhibitor beads-mass spectrometry (MIB/MS) assay to identify adaptive resistance mechanisms to vandetanib treatment, including upregulation of HER2 activity. Co-treatment to inhibit HER2 with lapatinib enhanced sensitivity to vandetanib, demonstrating biologic activity of HER2 upregulation. Using a CRISPR knockout model, we demonstrate that vandetanib effects are partially mediated by RET receptor tyrosine kinase. Finally, we use our operating room-to-laboratory assay that measures drug response in individual primary tumor cells in short term cultures to demonstrate conserved gene expression changes, including increased HER2 activity signatures, in vandetanib treated cells, and identify features associated with vandetanib response. These results support future investigation of RET targeting strategies considering reprogrammed networks, such as activated HER2, in patients with ET resistant ER+ breast cancer.
Collapse
Affiliation(s)
- Rasha T Kakati
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Austin A Whitman
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Santiago Haase
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Attila T Szenasi
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Christine Hnc Thai
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Elizabeth Brunk
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC
| | - Denis O Okumu
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC
| | - Michael P East
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, University of North Carolina, Chapel Hill, NC
- Computational Medicine Program, University of North Carolina, Chapel Hill, NC
| | - Gary L Johnson
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC
| | - Philip M Spanheimer
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
- Department of Genetics, University of North Carolina, Chapel Hill, NC
- Department of Surgery, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
38
|
Smith NJ, Watchalotone S, Sandhu S. Drug-Induced Liver Injury Secondary to Endocrine Therapy With Aromatase Inhibitors: A Case Report. Cureus 2025; 17:e80795. [PMID: 40255731 PMCID: PMC12006492 DOI: 10.7759/cureus.80795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Aromatase inhibitors (AIs) are routinely used in treating estrogen receptor (ER)-positive breast cancer in postmenopausal women and are effective in reducing the recurrence of metastatic hormone-sensitive breast cancer. We present the case of a 76-year-old female with a past medical history of hypothyroidism and dyslipidemia who presented for the treatment of ER-positive pleomorphic lobular carcinoma in situ (LCIS). After initiating anastrozole, she developed weakness, loss of appetite, darkening of urine, and jaundice. Upon admission to the hospital, results from the patient's initial workup revealed abnormal liver function tests. Anastrozole was identified as the causative agent for hepatic injury and was discontinued. The patient's liver enzymes normalized two months off of the medication, and she decided to proceed with routine observation rather than initiating new endocrine therapy. This case emphasizes the importance of recognizing drug-induced liver injury (DILI) as a rare and serious complication of anastrozole use and highlights the need for consideration of possible risks with endocrine therapies.
Collapse
Affiliation(s)
- Nicholas J Smith
- Oncology, Midwestern University Arizona College of Osteopathic Medicine, Glendale, USA
| | - Sariah Watchalotone
- Oncology, Midwestern University Arizona College of Osteopathic Medicine, Glendale, USA
| | - Sonia Sandhu
- Oncology, Ironwood Cancer and Research Centers, Glendale, USA
| |
Collapse
|
39
|
Saka AH, Giaquinto AN, McCullough LE, Tossas KY, Star J, Jemal A, Siegel RL. Cancer statistics for African American and Black people, 2025. CA Cancer J Clin 2025; 75:111-140. [PMID: 39976243 PMCID: PMC11929131 DOI: 10.3322/caac.21874] [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: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 02/21/2025] Open
Abstract
African American and other Black individuals (referred to as Black people in this article) have a disproportionate cancer burden, including the lowest survival of any racial or ethnic group for most cancers. Every 3 years, the American Cancer Society estimates the number of new cancer cases and deaths for Black people in the United States and compiles the most recent data on cancer incidence (herein through 2021), mortality (through 2022), survival, screening, and risk factors using population-based data from the National Cancer Institute and the Centers for Disease Control and Prevention. In 2025, there will be approximately 248,470 new cancer cases and 73,240 cancer deaths among Black people in the United States. Black men have experienced the largest relative decline in cancer mortality from 1991 to 2022 overall (49%) and in almost every 10-year age group, by as much as 65%-67% in the group aged 40-59 years. This progress largely reflects historical reductions in smoking initiation among Black teens, advances in treatment, and earlier detections for some cancers. Nevertheless, during the most recent 5 years, Black men had 16% higher mortality than White men despite just 4% higher incidence, and Black women had 10% higher mortality than White women despite 9% lower incidence. Larger inequalities for mortality than for incidence reflect two-fold higher death rates for prostate, uterine corpus, and stomach cancers and for myeloma, and 40%-50% higher rates for colorectal, breast, cervical, and liver cancers. The causes of ongoing disparities are multifactorial, but largely stem from inequalities in the social determinants of health that trace back to structural racism. Increasing diversity in clinical trials, enhancing provider education, and implementing financial incentives to ensure equitable care across the cancer care continuum would help close these gaps.
Collapse
Affiliation(s)
- Anatu H Saka
- Cancer Surveillance Research, American Cancer Society, Atlanta, Georgia, USA
| | - Angela N Giaquinto
- Cancer Surveillance Research, American Cancer Society, Atlanta, Georgia, USA
| | | | - Katherine Y Tossas
- Department of Social and Behavioral Sciences, School of Public Health, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jessica Star
- Risk Factors and Screening Research, American Cancer Society, Atlanta, Georgia, USA
| | - Ahmedin Jemal
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| | - Rebecca L Siegel
- Cancer Surveillance Research, American Cancer Society, Atlanta, Georgia, USA
| |
Collapse
|
40
|
Bhardwaj PV, Abdou Y. Managing pregnancy-associated breast cancer: A practical approach. Semin Perinatol 2025; 49:152037. [PMID: 40089317 DOI: 10.1016/j.semperi.2025.152037] [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] [Indexed: 03/17/2025]
Abstract
Pregnancy-Associated Breast Cancer (PABC) is a rare but complex condition that presents both professional and ethical challenges. Diagnosis is often delayed due to breast changes associated with pregnancy and puerperium, which can mask malignant findings. Management requires a multidisciplinary approach that carefully balances maternal and fetal risks. Chemotherapy is generally reserved for the second and third trimesters to avoid teratogenicity, with anthracyclines being the most well-studied and safest agents in this setting. Surgical decisions are influenced by factors such as cancer stage, gestational age, and the timing of potential radiation therapy. Notably, radiation therapy, endocrine therapy, and most targeted therapies are contraindicated during pregnancy due to potential harm to the fetus. Comprehensive care should include robust social and mental health support for the mother and her family to help navigate the physical and emotional challenges during this period.
Collapse
Affiliation(s)
- Prarthna V Bhardwaj
- Assistant Professor of Medicine, Division of Hematology-Oncology, University of Massachusetts Chan School of Medicine - Baystate, Springfield, MA, USA.
| | - Yara Abdou
- Assistant Professor, Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
41
|
Xu X, Guo X, Chen J, Pan Y, Li J, Chen J, Lai W, Lin L. Abemaciclib-associated kidney injuries: A retrospective analysis of the United States Food and Drug Administration adverse events reporting system. J Int Med Res 2025; 53:3000605251325961. [PMID: 40116800 PMCID: PMC12011124 DOI: 10.1177/03000605251325961] [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: 11/29/2024] [Accepted: 02/19/2025] [Indexed: 03/23/2025] Open
Abstract
BackgroundAbemaciclib, an oral kinase inhibitor, is used to treat hormone receptor-positive and HER2-negative breast cancer patients. However, there has been a decrease in studies reporting adverse reactions to abemaciclib-related kidney injuries. Thus, this study was aimed at assessing its safety profile using a large-scale pharmacovigilance database.MethodsAbemaciclib-related adverse drug reaction reports from the Food and Drug Administration Adverse Event Reporting System were obtained and scrutinized, and adverse drug reactions were selected using reporting odds ratio, the proportional reporting ratio methods, empirical Bayes geometric mean and UK Medicines and Healthcare products Regulatory Agency methods.ResultsWe selected 10,757 matched reports associated with abemaciclib, among which we found eight adverse reactions about kidney injuries correlated with abeamciclib, such as increased blood creatinine, renal disorder, decreased glomerular filtration rate, increased blood urea, hydronephrosis, abnormal renal function test, increased creatinine renal clearance and increased cystatin C. A demographic analysis of reported cases of abemaciclib-associated renal injury revealed that the majority were female, aged ≥46 years and had taken the drug ≥30 days.ConclusionThis study highlights the characteristics of adverse reactions with abemaciclib and those associated with renal damage, which are crucial for safety studies on the clinical use of this drug.
Collapse
Affiliation(s)
- Xiangchun Xu
- Department of Pharmacy, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xuzheng Guo
- Department of Pharmacy, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jinhui Chen
- Department of Pharmacy, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuhua Pan
- Department of Pharmacy, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Li
- Department of Pharmacy, Xiangtan Traditional Chinese Medicine Hospital, Xiangtan, Hunan, China
| | - Jing Chen
- Medical Research Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Weihua Lai
- Department of Pharmacy, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lu Lin
- Department of Pharmacy, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Ruan H, Zou Y, Huang L, Zha W, Ouyang Q, Yang L. PD-1/PD-L1 inhibitors plus chemotherapy versus chemotherapy alone for Asian patients with advanced triple-negative breast cancer: a phase III RCTs based meta-analysis. Front Oncol 2025; 15:1540538. [PMID: 40094013 PMCID: PMC11906427 DOI: 10.3389/fonc.2025.1540538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Background Advanced triple-negative breast cancer (TNBC) presents significant therapeutic challenges, particularly in Asian populations, which exhibit distinct biological and genetic characteristics. Immunotherapy combined with chemotherapy has emerged as a promising approach; however, its efficacy compared to chemotherapy alone remains under investigation. This meta-analysis aims to evaluate the clinical outcomes of PD-1/PD-L1 inhibitors combined with chemotherapy (PIC) versus chemotherapy alone in the treatment of advanced TNBC in Asian patients. Methods A systematic literature search was performed across six databases for phase 3 randomized controlled trials (RCTs). Only studies comparing the outcomes of PIC versus chemotherapy alone in patients with advanced TNBC, including subgroup analyses of Asian populations, were included. Data were pooled to assess overall survival (OS), progression-free survival (PFS), responses, and safety profiles. Results A total of 1041 patients from five phase 3 RCTs were included in the final analysis. Compared to chemotherapy alone, PIC therapy significantly improved PFS (hazard ratio [HR]: 0.74 [0.62, 0.88], P = 0.0008). No significant difference was observed in OS (HR: 0.78 [0.55, 1.12], P = 0.18), although a slight trend favoring PIC therapy was noted. Among PD-L1-positive patients, both OS (HR: 0.62 [0.44, 0.86], P = 0.005) and PFS (HR: 0.66 [0.50, 0.86], P = 0.003) were significantly improved in the PIC group. The PIC group also exhibited a substantially higher OS rate at 12-36 months and a higher PFS rate at 6-30 months. However, the incidence of immune-related AEs (irAEs) (risk ratio [RR]: 1.69 [1.33, 2.15], P < 0.0001) and grade 3-5 irAEs (RR: 3.11 [1.59, 6.10], P = 0.001) was significantly higher in the PIC group. The most common irAEs in the PIC group were hypothyroidism (14.40%), dermatitis (10.00%), and infusion reactions (8.85%). Both treatment groups exhibited similar response rates and treatment-related AEs (TRAEs). Conclusions In Asian patients with advanced TNBC, PIC significantly improved survival compared to chemotherapy alone. Although the combination therapy was associated with a higher incidence of irAEs, its clinical benefits support its use as a viable treatment option for this population. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024622428.
Collapse
Affiliation(s)
- Hua Ruan
- Department of Oncology, Xinyu People’s Hospital, Xinyu, China
| | - Yubin Zou
- Department of Oncology, Xinyu People’s Hospital, Xinyu, China
| | - Lifeng Huang
- Department of Oncology, Fenyi People’s Hospital, Xinyu, China
| | - Wenjuan Zha
- Department of Oncology, Xinyu People’s Hospital, Xinyu, China
| | - Qingqing Ouyang
- Department of Oncology, Xinyu People’s Hospital, Xinyu, China
| | - Ling Yang
- Department of Oncology, Xinyu People’s Hospital, Xinyu, China
| |
Collapse
|
43
|
Blanco R, Quezada-Romegialli C, Muñoz JP. Bovine Leukemia Virus and Human Breast Cancer: A Review of Clinical and Molecular Evidence. Viruses 2025; 17:324. [PMID: 40143252 PMCID: PMC11946124 DOI: 10.3390/v17030324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Despite significant advancements in early diagnosis and treatment, breast cancer (BC) remains a major global health challenge. Ongoing research is essential to identify novel risk factors, implement innovative screening programs, and develop personalized treatment approaches. Among the various risk factors, infection with certain oncogenic viruses has emerged as a potential contributor to BC development. Increasing evidence suggests that bovine leukemia virus (BLV) may contribute to zoonotic infections in humans, with a potential role in BC initiation and progression. This review evaluates clinical and experimental data on BLV presence in both malignant and non-malignant breast tissues, exploring potential mechanisms through which BLV may access human breast tissue and contribute to carcinogenesis. Current data reveal a higher prevalence of BLV infection in BC tissues compared to non-tumor tissues, correlating with an increased risk of BC development. In this context, dairy and meat products from BLV-infected animals have been proposed as potential transmission sources. BLV-encoded proteins disrupt key oncogenic pathways, which support their possible role in breast carcinogenesis. However, the interpretation of these findings is limited by potential confounding factors such as genetic predisposition, environmental exposures, and dietary influences. Further research, including well-controlled epidemiological studies, longitudinal cohorts, and mechanistic investigations into BLV proteins in human breast cells, is necessary to determine its role in BC development.
Collapse
Affiliation(s)
- Rancés Blanco
- Independent Researcher, Av. Vicuña Mackenna Poniente 6315, La Florida 8240000, Chile;
| | - Claudio Quezada-Romegialli
- Plataforma de Monitoreo Genómico y Ambiental, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile;
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| |
Collapse
|
44
|
Yitik AY, Sabir N, Yılmaz S. Comparative Evaluation of Superb Microvascular Imaging and Dynamic Contrast-Enhanced Magnetic Resonance Imaging in Differentiating Benign and Malignant Breast Masses. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2025. [PMID: 39991881 DOI: 10.1002/jum.16664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/25/2025]
Abstract
OBJECTIVES Our study aims to compare the diagnostic performance of superb microvascular imaging (SMI) and dynamic contrast-enhanced magnetic resonance imaging (MRI) in differentiating benign from malignant breast masses, using histopathological findings as the reference standard. METHODS This prospective study was conducted from April 2022 to March 2024. A total of 112 breast lesions from 110 patients were evaluated using gray-scale ultrasonography, SMI, and dynamic contrast-enhanced MRI. The vascular index (VI) obtained during SMI examination and kinetic curve patterns from MRI were analyzed. RESULTS Histopathological analysis revealed 62 benign and 50 malignant lesions. The VI showed a statistically significant difference between benign and malignant lesions, with a mean VI of 5.12 ± 4.66 in benign masses and 10.13 ± 5.48 in malignant masses (P < .001). The ROC analysis demonstrated an AUC of 0.79 for SMI with a VI cut-off value of 4.15, yielding a sensitivity of 92%, specificity of 60%, and accuracy of 74%. A statistically significant correlation was found between VI values and MRI contrast enhancement kinetic curve types (P < .05). MRI demonstrated superior diagnostic performance, with an AUC of 0.89 and sensitivity, specificity, and accuracy of 98, 80.65, and 88.39%, respectively. CONCLUSIONS SMI, when used in conjunction with conventional ultrasonography and MRI, provides significant diagnostic value in differentiating benign from malignant breast masses. The study supports the potential integration of SMI into routine breast cancer diagnostic workflows, particularly in settings where MRI is less accessible.
Collapse
Affiliation(s)
- Ahmet Yasin Yitik
- Department of Radiology, Pamukkale University Medical Faculty Hospital, Denizli, Turkey
| | - Nuran Sabir
- Department of Radiology, Pamukkale University Medical Faculty Hospital, Denizli, Turkey
| | - Sevda Yılmaz
- Department of General Surgery, Pamukkale University Medical Faculty Hospital, Denizli, Turkey
| |
Collapse
|
45
|
Freeman JQ, Schechter K, Nguyen LC, Omoleye OJ, Hara JH. Real-word study of racial/ethnic disparities and socioeconomic determinants of overall survival in male breast cancer. RESEARCH SQUARE 2025:rs.3.rs-5808248. [PMID: 40034441 PMCID: PMC11875317 DOI: 10.21203/rs.3.rs-5808248/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
This study assessed racial/ethnic disparities and socioeconomic determinants of overall survival in male breast cancer. Using the 2010-2021 US National Cancer Database, we identified 20,470 patients: 78.2% White, 13.8% Black, 4.0% Hispanic, and 2.5% Asian or Pacific Islander. After adjusting for clinicopathologic characteristics, Black patients had higher mortality than White patients (adjusted hazard ratio [AHR] 1.22, 95% CI: 1.12-1.32); however, when further adjusting for socioeconomic factors, this difference was no longer significant (AHR 1.09, 95% CI: 0.99-1.21). Hispanic patients had better survival. In the TNBC cohort, Asian or Pacific Islander patients had higher mortality than White patients (AHR 2.35, 95% CI: 1.21-4.55), warranting further investigation. In this US male breast cancer cohort, Black patients and White patients had similar mortality risk after further adjusting for socioeconomic indicators. Higher median household income and private insurance were linked to better survival. Strategies addressing socioeconomic inequities may help improve male breast cancer outcomes.
Collapse
|
46
|
Park JH, Kwon LM, Lee HK, Koo T, Suh YJ, Kwon MJ, Kim HY. Radiomic Analysis of Magnetic Resonance Imaging for Breast Cancer with TP53 Mutation: A Single Center Study. Diagnostics (Basel) 2025; 15:428. [PMID: 40002579 PMCID: PMC11854707 DOI: 10.3390/diagnostics15040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Radiomics is a non-invasive and cost-effective method for predicting the biological characteristics of tumors. In this study, we explored the association between radiomic features derived from magnetic resonance imaging (MRI) and genetic alterations in patients with breast cancer. Methods: We reviewed electronic medical records of patients with breast cancer patients with available targeted next-generation sequencing data available between August 2018 and May 2021. Substraction imaging of T1-weighted sequences was utilized. The tumor area on MRI was segmented semi-automatically, based on a seeded region growing algorithm. Radiomic features were extracted using the open-source software 3D slicer (version 5.6.1) with PyRadiomics extension. The association between genetic alterations and radiomic features was examined. Results: In total, 166 patients were included in this study. Among the 50 panel genes analyzed, only TP53 mutations were significantly associated with radiomic features. Compared with TP53 wild-type tumors, TP53 mutations were associated with larger tumor size, advanced stage, negative hormonal receptor status, and HER2 positivity. Tumors with TP53 mutations exhibited higher values for Gray Level Non-Uniformity, Dependence Non-Uniformity, and Run Length Non-Uniformity, and lower values for Sphericity, Low Gray Level Emphasis, and Small Dependence Low Gray Level emphasis compared to TP53 wild-type tumors. Six radiomic features were selected to develop a composite radiomics score. Receiver operating characteristic curve analysis showed an area under the curve of 0.786 (95% confidence interval, 0.719-0.854; p < 0.001). Conclusions: TP53 mutations in breast cancer can be predicted using MRI-derived radiomic analysis. Further research is needed to assess whether radiomics can help guide treatment decisions in clinical practice.
Collapse
Affiliation(s)
- Jung Ho Park
- Division of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea (Y.J.S.)
| | - Lyo Min Kwon
- Department of Radiology, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea;
| | - Hong Kyu Lee
- Department of Thoracic and Cardiovascular Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Taeryool Koo
- Department of Radiation Oncology, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Yong Joon Suh
- Division of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea (Y.J.S.)
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Ho Young Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| |
Collapse
|
47
|
Li J. Exploring the Potential of Adjuvant CDK4/6 Inhibitors in Hormone Receptor-Positive Early Breast Cancer: A Consistent Approach for All. Cancers (Basel) 2025; 17:561. [PMID: 40002156 PMCID: PMC11852482 DOI: 10.3390/cancers17040561] [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: 12/05/2024] [Revised: 01/05/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hormone receptor-positive, HER2-negative breast cancer is the most common subtype, with endocrine therapy as the standard treatment. Despite the advancements in adjuvant endocrine therapy, recurrence remains a challenge, particularly in high-risk patients. Recent trials on cyclin D kinase 4/6 (CDK4/6) inhibitors in adjuvant therapy have shown promise in reducing early recurrence and improving survival. METHODS This review analyzes the clinical evidence supporting the use of CDK4/6 inhibitors, focusing on the NATALEE and monarchE trials, which demonstrate comparable efficacy and manageable safety profiles for ribociclib and abemaciclib. RESULTS AND CONCLUSIONS Ribociclib, with its broader applicability and impact on the decision making for axillary lymph node surgery, may be the preferred option in high-risk populations. The review also addresses unanswered clinical questions and highlights the need for ongoing research to optimize the adjuvant therapy strategies.
Collapse
Affiliation(s)
- Jianbin Li
- Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing 100071, China;
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
48
|
Xie D, Sun L, Wu M, Li Q. From detection to elimination: iron-based nanomaterials driving tumor imaging and advanced therapies. Front Oncol 2025; 15:1536779. [PMID: 39990682 PMCID: PMC11842268 DOI: 10.3389/fonc.2025.1536779] [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: 11/29/2024] [Accepted: 01/16/2025] [Indexed: 02/25/2025] Open
Abstract
Iron-based nanomaterials (INMs), due to their particular magnetic property, excellent biocompatibility, and functionality, have been developed into powerful tools in both tumor diagnosis and therapy. We give an overview here on how INMs such as iron oxide nanoparticles, element-doped nanocomposites, and iron-based organic frameworks (MOFs) display versatility for tumor imaging and therapy improvement. In terms of imaging, INMs improve the sensitivity and accuracy of techniques such as magnetic resonance imaging (MRI) and photoacoustic imaging (PAI) and support the development of multimodal imaging platforms. Regarding treatment, INMs play a key role in advanced strategies such as immunotherapy, magnetic hyperthermia, and synergistic combination therapy, which effectively overcome tumor-induced drug resistance and reduce systemic toxicity. The integration of INMs with artificial intelligence (AI) and radiomics further expands its capabilities for precise tumor identification, and treatment optimization, and amplifies treatment monitoring. INMs now link materials science with advanced computing and clinical innovations to enable next-generation cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Dong Xie
- Department of Radiology, The Affiliated People’s Hospital of Ningbo University, Ningbo, China
| | - Linglin Sun
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Manxiang Wu
- Department of Radiology, The Affiliated People’s Hospital of Ningbo University, Ningbo, China
| | - Qiang Li
- Department of Radiology, The Affiliated People’s Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
49
|
Sun Y, Liu J, Zhu L, Huang F, Dong Y, Liu S, Chen S, Ji W, Lu J, Liu L, Li S. Treatment Response to Oncolytic Virus in Patient-Derived Breast Cancer and Hypopharyngeal Cancer Organoids: Evaluation via a Microfluidics Organ-on-a-Chip System. Bioengineering (Basel) 2025; 12:146. [PMID: 40001666 PMCID: PMC11851931 DOI: 10.3390/bioengineering12020146] [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: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
In this study, we present an oncolytic virus (OV) evaluation system established using microfluidic organ-on-a-chip (OOC) systems and patient-derived organoids (PDOs), which was used in the development of a novel oncolytic virus, AD4-GHPE. An OV offers advantages such as good targeting ability and minimal side effects, and it has achieved significant breakthroughs when combined with immunotherapy in recent clinical trials. The development of OVs has become an emerging research focus. PDOs can preserve the heterogeneity of in situ tumor tissues, whereas microfluidic OOC systems can automate and standardize various experimental procedures. These systems have been applied in cutting-edge drug screening and cell therapy experiments; however, their use in functionally complex oncolytic viruses remains to be explored. In this study, we constructed a novel recombinant oncolytic adenovirus, AD4-GHPE, and evaluated OOC systems and PDOs through various functional validations in hypopharyngeal and breast cancer organoids. The results confirmed that AD4-GHPE exhibits three antitumor mechanisms, namely, tumor-specific cytotoxicity, a reduction in programmed death ligand 1 (PD-L1) expression in tumor cells to increase CD8+ T-cell activity, and granulocyte-macrophage colony-stimulating factor (GM-CSF) secretion. The evaluation system combining OOC systems and PDOs was efficient and reliable, providing personalized OV treatment recommendations for patients and offering industrialized and standardized research ideas for the development of OVs.
Collapse
Affiliation(s)
- Yu Sun
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Jiaqi Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Li Zhu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Yanbo Dong
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
| | - Shuang Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Siyi Chen
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Wei Ji
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Jingjing Lu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Liangfa Liu
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| |
Collapse
|
50
|
Atale N, Wells A. Statins as Secondary Preventive Agent to Limit Breast Cancer Metastatic Outgrowth. Int J Mol Sci 2025; 26:1300. [PMID: 39941069 PMCID: PMC11818786 DOI: 10.3390/ijms26031300] [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: 12/22/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Metastasis is a leading cause of mortality in breast cancer, as metastatic disease is often aggressive and resistant to conventional treatments. Cancer cells that spread to distant organs can enter a dormant phase for extended periods, sometimes years or decades. During this dormant phase, cancer cells avoid immune and pharmacological response. Thus, new approaches are needed to prevent these disseminated cells from becoming lethal cancers. Statins are known inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase that have been extensively used in patients with cardiovascular diseases to lower cholesterol. However, recent research has demonstrated their potential in anticancer therapies. Epidemiological evidence suggests that statins are associated with a reduction in breast cancer-specific mortality, although they do not appear to affect the incidence of primary tumors. In this review, we discuss the role of statins in metastasis and dormancy, their cytocidal and cytostatic effects and their interactions with different cell types in the tumor microenvironment. The exact mechanisms by which statins reduce mortality without influencing primary tumor growth remain unclear, also warranting further investigation into their potential role in metastasis and tumor dormancy, which could ultimately help patients to improve survival and quality of life.
Collapse
Affiliation(s)
- Neha Atale
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Alan Wells
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Research and Development Service, Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
- Cell Biology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|