Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (7,727)

Search Parameters:
Keywords = breast tumor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 3550 KB  
Article
Three-Dimensional Tumor Spheroids Reveal B7-H3 CAR T Cell Infiltration Dynamics and Microenvironment-Induced Functional Reprogramming in Solid Tumors
by Feng Chen, Ke Ning, Yuanyuan Xie, Xiaoyan Yang, Ling Yu and Xinhui Wang
Cells 2026, 15(2), 169; https://doi.org/10.3390/cells15020169 (registering DOI) - 16 Jan 2026
Abstract
Chimeric antigen receptor (CAR) T cell therapy has demonstrated clinical success in hematologic malignancies but has limited efficacy in solid tumors due to tumor microenvironment (TME) barriers that impede CAR T cell recognition, infiltration, and sustained function. Traditional 2D assays inadequately recapitulate these [...] Read more.
Chimeric antigen receptor (CAR) T cell therapy has demonstrated clinical success in hematologic malignancies but has limited efficacy in solid tumors due to tumor microenvironment (TME) barriers that impede CAR T cell recognition, infiltration, and sustained function. Traditional 2D assays inadequately recapitulate these constraints, necessitating improved in vitro models. This study validated a 3D tumor spheroid platform using an agarose microwell system to generate uniform B7-H3-positive spheroids from multiple solid tumor cell lines, enabling the evaluation of CAR T cell activity. TME-relevant immune modulation under 3D conditions was analyzed by flow cytometry for B7-H3, MHC I/II, and antigen processing machinery (APM), followed by co-culture with B7-H3 CAR T cells to assess cytotoxicity, spheroid integrity, tumor viability, and CAR T cell activation, exhaustion, and cytokine production. Two human cancer-cell-line-derived spheroids, DU 145 (prostate cancer) and SUM159 (breast cancer), retained B7-H3 expression, while MC38 (mouse colon cancer)-derived spheroids served as a B7-H3 negative control. Under 3D culture conditions, DU 145 and SUM159 spheroids acquire TME-like immune evasion characteristics and specifically downregulated MHC-I and APM (TAP1, TAP2, LMP7) with concurrent upregulation of MHC-II and calreticulin. Co-culture showed effective spheroid infiltration, cytotoxicity, and structural disruption, with infiltrating CAR T cells displaying higher CD4+ fraction, activation, exhaustion, effector/terminal differentiation, and IFN-γ/TNF-α production. This 3D platform recapitulates critical TME constraints and provides a cost-effective, feasible preclinical tool to assess CAR T therapies beyond conventional 2D assays. Full article
(This article belongs to the Section Cell Methods)
18 pages, 4066 KB  
Article
Machine Learning Model Based on Multiparametric MRI for Distinguishing HER2 Expression Level in Breast Cancer
by Yongxin Chen, Weifeng Liu, Wenjie Tang, Qingcong Kong, Siyi Chen, Shuang Liu, Liwen Pan, Yuan Guo and Xinqing Jiang
Curr. Oncol. 2026, 33(1), 53; https://doi.org/10.3390/curroncol33010053 - 16 Jan 2026
Abstract
This study aimed to develop machine learning models based on conventional MRI features to classify HER2 expression levels in invasive breast cancer and explore their association with disease-free survival (DFS). A total of 678 patients from two centers were included, with Center 1 [...] Read more.
This study aimed to develop machine learning models based on conventional MRI features to classify HER2 expression levels in invasive breast cancer and explore their association with disease-free survival (DFS). A total of 678 patients from two centers were included, with Center 1 divided into training and internal test sets and Center 2 serving as an external test set. Random Forest models were trained to distinguish HER2-positive vs. HER2-negative (Task 1) and HER2-low vs. HER2-zero tumors (Task 2) using BI-RADS–based MRI features. SHapley Additive exPlanations were applied to rank feature importance, assist feature selection, and enhance model interpretability. DFS was analyzed using Kaplan–Meier curves and log-rank tests. In Task 1, key features included tumor size, axillary lymph nodes, fibroglandular tissue, peritumoral edema, and multifocal, achieving AUCs of 0.75 and 0.73 in the internal and external test sets, respectively. In Task 2, tumor size, peritumoral edema, and multifocal yielded AUCs of 0.73 and 0.72, respectively. Higher task-specific model scores were associated with shorter DFS in Task 1 (p = 0.037) and longer DFS in Task 2 (p = 0.046). MRI-based machine learning models can noninvasively stratify HER2 expression levels, with potential for prognostic stratification and clinical application. Full article
(This article belongs to the Section Breast Cancer)
Show Figures

Figure 1

13 pages, 1251 KB  
Article
Circulating Lipid Peroxides Associate with More Aggressive Tumor Phenotypes and Increased Risks of Recurrence and Mortality in Breast Cancer Patients
by Julia Fernandes Gois Orrutéa, Rafaela Oliveira Matos, João Paulo Araújo Pinto, André Cherubini Cechinel, Bruna Yukie Koizumi, Rafael Gomes Paz, Rafaella Frederico Almeida, Janaína Carla da Silva, Tatiane Renata Fagundes, Daniel Rech, Guilherme Welter Wendt and Carolina Panis
Med. Sci. 2026, 14(1), 43; https://doi.org/10.3390/medsci14010043 - 16 Jan 2026
Abstract
Background/Objectives: Breast cancer is the most common neoplasm among women and remains the leading cause of cancer-related mortality in the female population worldwide. Tumor cells exist within a highly oxidative microenvironment, which promotes the formation of substantial amounts of lipid peroxides. However, [...] Read more.
Background/Objectives: Breast cancer is the most common neoplasm among women and remains the leading cause of cancer-related mortality in the female population worldwide. Tumor cells exist within a highly oxidative microenvironment, which promotes the formation of substantial amounts of lipid peroxides. However, the clinical significance of circulating lipid peroxides in breast cancer is still not well understood. Methods: In this study, we quantified systemic lipid peroxide levels in plasma samples from 408 breast cancer patients and examined their associations with key clinicopathological parameters to evaluate their potential as disease biomarkers. Data are reported as relative light units (RLU). Results: Our findings revealed significantly higher lipid peroxide levels in HER2-amplified tumors compared with estrogen-receptor-positive tumors (1,133,494 ± 102,409 RLU vs. 951,883 ± 47,535 RLU; p = 0.0438). Elevated levels were also observed in patients with triple-negative breast cancer relative to those with Luminal A (1,163,323 ± 109,640 RLU vs. 875,633 ± 49,601 RLU; p = 0.0356) and Luminal B tumors (1,163,323 ± 109,640 RLU vs. 1,071,779 ± 98,329 RLU; p = 0.0254). In addition, increased lipid peroxidation was detected in patients with high-grade tumors (G3: 1,141,035 ± 101,045 RLU vs. G1–G2: 949,658 ± 46,119 RLU; p = 0.0346) and in those classified as at high risk of recurrence or death compared with low-risk patients (1,209,530 ± 95,396 RLU vs. 978,318 ± 229,526 RLU; p = 0.0054). Overweight patients also exhibited higher lipid peroxide levels than eutrophic individuals (1,131,233 ± 59,633 RLU vs. 820,772 ± 57,653 RLU; p = 0.0142). Conclusions: Collectively, these results suggest that circulating lipid peroxides may serve as potential biomarkers for recurrence and death risk in breast cancer, particularly among patients with more aggressive tumor phenotypes. Full article
(This article belongs to the Section Cancer and Cancer-Related Research)
Show Figures

Figure 1

31 pages, 1485 KB  
Article
Explainable Multi-Modal Medical Image Analysis Through Dual-Stream Multi-Feature Fusion and Class-Specific Selection
by Naeem Ullah, Ivanoe De Falco and Giovanna Sannino
AI 2026, 7(1), 30; https://doi.org/10.3390/ai7010030 - 16 Jan 2026
Abstract
Effective and transparent medical diagnosis relies on accurate and interpretable classification of medical images across multiple modalities. This paper introduces an explainable multi-modal image analysis framework based on a dual-stream architecture that fuses handcrafted descriptors with deep features extracted from a custom MobileNet. [...] Read more.
Effective and transparent medical diagnosis relies on accurate and interpretable classification of medical images across multiple modalities. This paper introduces an explainable multi-modal image analysis framework based on a dual-stream architecture that fuses handcrafted descriptors with deep features extracted from a custom MobileNet. Handcrafted descriptors include frequency-domain and texture features, while deep features are summarized using 26 statistical metrics to enhance interpretability. In the fusion stage, complementary features are combined at both the feature and decision levels. Decision-level integration combines calibrated soft voting, weighted voting, and stacking ensembles with optimized classifiers, including decision trees, random forests, gradient boosting, and logistic regression. To further refine performance, a hybrid class-specific feature selection strategy is proposed, combining mutual information, recursive elimination, and random forest importance to select the most discriminative features for each class. This hybrid selection approach eliminates redundancy, improves computational efficiency, and ensures robust classification. Explainability is provided through Local Interpretable Model-Agnostic Explanations, which offer transparent details about the ensemble model’s predictions and link influential handcrafted features to clinically meaningful image characteristics. The framework is validated on three benchmark datasets, i.e., BTTypes (brain MRI), Ultrasound Breast Images, and ACRIMA Retinal Fundus Images, demonstrating generalizability across modalities (MRI, ultrasound, retinal fundus) and disease categories (brain tumor, breast cancer, glaucoma). Full article
(This article belongs to the Special Issue Digital Health: AI-Driven Personalized Healthcare and Applications)
Show Figures

Figure 1

43 pages, 2779 KB  
Review
Molecular and Immune Mechanisms Governing Cancer Metastasis, Including Dormancy, Microenvironmental Niches, and Tumor-Specific Programs
by Dae Joong Kim
Int. J. Mol. Sci. 2026, 27(2), 875; https://doi.org/10.3390/ijms27020875 - 15 Jan 2026
Abstract
Metastasis is still the leading cause of cancer-related death. It happens when disseminated tumor cells (DTCs) successfully navigate a series of steps and adapt to the unique conditions of distant organs. In this review, key molecular and immune mechanisms that shape metastatic spread, [...] Read more.
Metastasis is still the leading cause of cancer-related death. It happens when disseminated tumor cells (DTCs) successfully navigate a series of steps and adapt to the unique conditions of distant organs. In this review, key molecular and immune mechanisms that shape metastatic spread, long-term survival, and eventual outgrowth are examined, with a focus on how tumor-intrinsic programs interact with extracellular matrix (ECM) remodeling, angiogenesis, and immune regulation. Gene networks that sustain tumor-cell plasticity and invasion are described, including EMT-linked transcription factors such as SNAIL and TWIST, as well as broader transcriptional regulators like SP1. Also, how epigenetic mechanisms, such as EZH2 activity, DNA methylation, chromatin remodeling, and noncoding RNAs, lock in pro-metastatic states and support adaptation under therapeutic pressure. Finally, proteases and matrix-modifying enzymes that physically and biochemically reshape tissues, including MMPs, uPA, cathepsins, LOX/LOXL2, and heparinase, are discussed for their roles in releasing stored growth signals and building permissive niches that enable seeding and colonization. In parallel, immune-evasion strategies that protect circulating and newly seeded tumor cells are discussed, including platelet-mediated shielding, suppressive myeloid populations, checkpoint signaling, and stromal barriers that exclude effector lymphocytes. A major focus is metastatic dormancy, cellular, angiogenic, and immune-mediated, framed as a reversible survival state regulated by stress signaling, adhesion cues, metabolic rewiring, and niche constraints, and as a key determinant of late relapse. Tumor-specific metastatic programs across mesenchymal malignancies (osteosarcoma, chondrosarcoma, and liposarcoma) and selected high-burden cancers (melanoma, hepatocellular carcinoma, glioblastoma, and breast cancer) are highlighted, emphasizing shared principles and divergent organotropisms. Emerging therapeutic strategies that target both the “seed” and the “soil” are also discussed, including immunotherapy combinations, stromal/ECM normalization, chemokine-axis inhibition, epigenetic reprogramming, and liquid-biopsy-enabled minimal residual disease monitoring, to prevent reactivation and improve durable control of metastatic disease. Full article
(This article belongs to the Special Issue Molecular Mechanism Involved in Cancer Metastasis)
Show Figures

Figure 1

25 pages, 3718 KB  
Article
The WISP1/Src/MIF Axis Promotes the Malignant Phenotype of Non-Invasive MCF7 Breast Cancer Cells
by Maria-Elpida Christopoulou, Panagiota Karamitsou, Alexios Aletras and Spyros S. Skandalis
Cells 2026, 15(2), 160; https://doi.org/10.3390/cells15020160 - 15 Jan 2026
Abstract
Breast cancer is a heterogeneous disease that exists in multiple subtypes, some of which still lack targeted and effective therapy. A major challenge is to unravel their underlying molecular mechanisms and bring to light novel therapeutic targets. In this study, we investigated the [...] Read more.
Breast cancer is a heterogeneous disease that exists in multiple subtypes, some of which still lack targeted and effective therapy. A major challenge is to unravel their underlying molecular mechanisms and bring to light novel therapeutic targets. In this study, we investigated the role of WNT-inducible signaling pathway protein 1 (WISP1) matricellular protein in the acquirement of an invasive phenotype by breast cancer cells. To this aim, we treated non-invasive MCF7 cells with WISP1 and assessed the expression levels of macrophage migration inhibitory factor (MIF) and its cellular receptor CD74. Next, we examined the expression of epithelial-to-mesenchymal transition (EMT) markers as well as molecular effectors of the tumor microenvironment, such as CD44, the main hyaluronan receptor that also acts as a co-receptor for MIF, the hyaluronan oncogenic network, and specific matrix metalloproteinases (MMPs) and their endogenous inhibitors, tissue inhibitors of metalloproteinases (TIMPs). The results showed that WISP1 potently induces the expression of MIF cytokine and affects the expression of specific extracellular matrix molecules with established roles in the promotion of malignant properties. Notably, Src kinases and MIF are critically involved in these processes. Collectively, the present study demonstrates for first time a WISP1/Src/MIF axis as well as its ability to induce an invasive phenotype in MCF7 cells and highlights novel cellular and molecular processes involved in the epithelial-to-mesenchymal transition and the development of invasive breast cancer. This suggests that specific cues from the tumor microenvironment can activate a migratory/invasive phenotype in a subpopulation of cells residing within the heterogeneous breast tumor. Full article
Show Figures

Figure 1

16 pages, 7824 KB  
Article
Tumor Growth Rate Predicts Pathological Outcomes in Breast Fibroepithelial Tumors: A Pilot Study and Review of Literature
by Hisham F. Bahmad, Adriana Falcon, Abdallah Araji, Karem Gharzeddine, Youley Tjendra, Elena F. Brachtel, Natalie Pula, Nicole Brofman, Merce Jorda and Carmen Gomez-Fernández
Cancers 2026, 18(2), 269; https://doi.org/10.3390/cancers18020269 - 15 Jan 2026
Abstract
Background/Objectives: Fibroepithelial tumors (FETs) of the breast, including fibroadenomas (FAs) and phyllodes tumors (PTs), are among the most common breast masses encountered by breast radiologists and pathologists. Differentiating FAs from benign or borderline PTs can be challenging, especially on core biopsy specimens where [...] Read more.
Background/Objectives: Fibroepithelial tumors (FETs) of the breast, including fibroadenomas (FAs) and phyllodes tumors (PTs), are among the most common breast masses encountered by breast radiologists and pathologists. Differentiating FAs from benign or borderline PTs can be challenging, especially on core biopsy specimens where sampling limitations obscure key histologic features. Although imaging techniques provide useful diagnostic context, their predictive accuracy for pathologic classification remains limited. Methods: We conducted a single-institution pilot study to assess whether tumor growth rate (TGR) derived from serial imaging could serve as a noninvasive correlate of histopathologic outcomes in FETs. Thirty-two patients with serial imaging and subsequent surgical excision (January 2020–May 2025) were analyzed. TGR, expressed as percentage volume increase per month, was calculated from diameter-based volumetrics. Results: The cohort included conventional FA (n = 10), cellular FA (n = 4), benign PT (n = 8), borderline PT (n = 6), and malignant PT (n = 4). Malignant PTs demonstrated significantly higher median TGRs (180.4%/month) and shorter imaging intervals (1.1 months) compared with other groups (p = 0.0357 and p = 0.005, respectively). These large effect-size differences suggest clinically meaningful growth dynamics. Conclusions: As a pilot, this study establishes foundational variance and effect-size estimates for powering a multicenter trial. If validated, TGR may provide an objective, noninvasive metric to enhance preoperative risk stratification and guide management of breast FETs. Full article
Show Figures

Figure 1

17 pages, 3282 KB  
Systematic Review
Effects of Nutritional Supplementation on Tumor Growth: A Systematic Review and Meta-Analysis of Studies Using Animal Models of Mammary Cancer
by Bruna Ribeiro-Silva, José Antônio Orellana Turri, Ricardo dos Santos Simões, José Cipolla-Neto, Edmund Chada Baracat and José Maria Soares-Jr
Biology 2026, 15(2), 150; https://doi.org/10.3390/biology15020150 - 14 Jan 2026
Viewed by 19
Abstract
Breast cancer remains the most prevalent malignancy among women worldwide, and experimental tumor models are essential for evaluating new therapeutic approaches before clinical application. The Walker-256 carcinoma model exhibits biological behavior comparable to human carcinoma and allows short-term assessment of tumor progression. This [...] Read more.
Breast cancer remains the most prevalent malignancy among women worldwide, and experimental tumor models are essential for evaluating new therapeutic approaches before clinical application. The Walker-256 carcinoma model exhibits biological behavior comparable to human carcinoma and allows short-term assessment of tumor progression. This study aimed to conduct a systematic review and meta-analysis of the effects of dietary supplementation on Walker-256 tumor growth. We followed the PRISMA guidelines and searched the PubMed, Embase, and Cochrane databases without time restrictions. The meta-analysis included clustering based on the type of supplementation received, comparing treatment groups with control groups. Twenty-one studies were included, analyzing 18 different supplements, and 12 studies were considered for quantitative synthesis. The overall effect indicated a significant reduction in tumor growth with supplementation (SMD = 2.83, 95% CI = 1.99–3.66, p < 0.001), despite high heterogeneity (I2 = 93.3%). Oils had the greatest impact, particularly fish oil (SMD = 6.99, 95% CI = 3.15–10.83, p < 0.001) and shark liver oil (SMD = 4.43, 95% CI = 2.19–6.67, p < 0.01). Full article
(This article belongs to the Section Cancer Biology)
Show Figures

Figure 1

25 pages, 18497 KB  
Article
Carvacrol Selectively Induces Mitochondria-Related Apoptotic Signaling in Primary Breast Cancer-Associated Fibroblasts
by Nail Besli, Nilufer Ercin, Merve Tokocin, Sümeyra Emine Boluk, Rabia Kalkan Cakmak, Kamil Ozdogan, Talar Vartanoglu Aktokmakyan, Mehtap Toprak, Gulcin Ercan, Merve Beker, Ulkan Celik, Emir Capkinoglu and Yusuf Tutar
Pharmaceuticals 2026, 19(1), 142; https://doi.org/10.3390/ph19010142 - 14 Jan 2026
Viewed by 52
Abstract
Background/Objectives: Cancer-associated fibroblasts (CAFs) are key stromal mediators of breast tumor progression and therapy resistance. Carvacrol, a dietary monoterpenic phenol, exhibits antiproliferative activity in cancer cells, but its effects on primary human breast CAFs remain unclear. This study aimed to determine whether [...] Read more.
Background/Objectives: Cancer-associated fibroblasts (CAFs) are key stromal mediators of breast tumor progression and therapy resistance. Carvacrol, a dietary monoterpenic phenol, exhibits antiproliferative activity in cancer cells, but its effects on primary human breast CAFs remain unclear. This study aimed to determine whether carvacrol selectively induces mitochondria-related apoptotic signaling in breast CAFs while sparing normal fibroblasts (NFs). Methods: Primary fibroblast cultures were established from invasive ductal carcinoma tissues (CAFs, n = 9) and nonmalignant breast tissues (NFs, n = 5) and validated by α-SMA and FAP immunofluorescence. Cells were exposed to 400 μM carvacrol. Apoptosis was assessed by TUNEL assay and BAX/BCL-XL Western blotting. Changes in signaling pathways were evaluated by analyzing PPARα/NF-κB, sirtuin (SIRT1, SIRT3), autophagy-related markers (LAMP2A, p62), and matrix metalloproteinases (MMP-2, MMP-3). In silico molecular docking and 100-ns molecular dynamics simulations were performed to examine interactions between carvacrol and caspase-3 and caspase-9. Results: Carvacrol induced a pronounced, time-dependent apoptotic response in CAFs, with TUNEL-based viability declining to approximately 10% of control levels by 12 h and a marked increase in the BAX/BCL-XL ratio. In contrast, NFs exhibited minimal TUNEL positivity and no significant change in BAX/BCL-XL. In CAFs, but not NFs, carvacrol reduced PPARα expression and NF-κB nuclear localization, increased SIRT1 and SIRT3 levels, selectively suppressed MMP-3 while partially normalizing MMP-2, and altered autophagy-related markers (decreased LAMP2A and accumulation of p62), consistent with autophagic stress and possible impairment of autophagic flux. Computational analyses revealed stable carvacrol binding to caspase-3 and caspase-9 with modest stabilization of active-site loops, supporting caspase-dependent, mitochondria-related apoptosis. Conclusions: Carvacrol selectively targets breast cancer-associated fibroblasts by inducing mitochondria-related apoptotic signaling while largely sparing normal fibroblasts. This effect is accompanied by coordinated modulation of PPARα/NF-κB, sirtuin, autophagy, and MMP pathways. These findings support further evaluation of carvacrol as a microenvironment-directed adjunct in breast cancer therapy. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

17 pages, 638 KB  
Review
Mast Cells Accumulate in the Stroma of Breast Adenocarcinoma and Secrete Pro-Inflammatory Cytokines and Tumor-Damaging Mediators: Could IL-37 and IL-38 Play an Anti-Tumor Role?
by Pio Conti, Carla E. Gallenga, Ciro Annicchiarico, Armando Coppola, Raffaello Pellegrino, Michelangelo J. Conti and Filiberto Mastrangelo
Int. J. Mol. Sci. 2026, 27(2), 824; https://doi.org/10.3390/ijms27020824 - 14 Jan 2026
Viewed by 35
Abstract
Tumor tissue is surrounded by mast cells (MCs), which participate in the inflammatory immune response by producing cytokines, proteases, and other molecules. MCs are involved in both innate and acquired immunity and are associated with the IgE response through the FcεRI receptor. MCs [...] Read more.
Tumor tissue is surrounded by mast cells (MCs), which participate in the inflammatory immune response by producing cytokines, proteases, and other molecules. MCs are involved in both innate and acquired immunity and are associated with the IgE response through the FcεRI receptor. MCs mediate inflammation in several immune reactions, including acute hyperreactivity, leukocyte recruitment, acute tissue swelling, anaphylaxis, and pro-inflammatory cytokine production. They not only function as pro-inflammatory effector cells but may also contribute to the regulation of the acquired immune response in tumor tissue. Therefore, MCs may mediate immunity in breast cancer by promoting remodelling and counteracting cancer growth. They also produce anti-inflammatory substances, such as histamine, transforming growth factor-β (TGF-β)1, IL-10, and IL-4, which inhibit the acquired immune response and reduce the inflammatory state. IL-37 and IL-38 are novel natural regulators of inflammation and are anti-inflammatory members of the IL-1 family. IL-1, generated by immune cells such as macrophages and lymphocytes, is released downstream of oncogenes in breast cancer, triggering an inflammatory response by stimulating other pro-inflammatory cytokines such as IL-6, tumor necrosis factor (TNF), and IL-33 (an early warning cytokine). Therefore, blocking IL-1 with IL-37 or IL-38 could represent a novel therapeutic strategy that, when combined with other treatments, could be beneficial in breast cancer. This review focuses on the new discoveries and insights into the role of MCs in breast cancer. We also analyzed molecules that can promote tumor growth and those that can inhibit cancer development and metastasis. This review aims to study the role of MCs accumulated in the stroma of breast adenocarcinoma in relation to secreted anti-inflammatory cytokines, such as IL-37 and IL-38. Full article
(This article belongs to the Special Issue Cytokines and Inflammatory Diseases)
Show Figures

Figure 1

19 pages, 2879 KB  
Article
Prevalence and Outcomes of HER2-Low Versus HER2-0 Status in Patients with Metastatic Breast Cancer
by Akshara Singareeka Raghavendra, Diane D. Liu, Senthil Damodaran, Sarah Pasyar, Yu Shen, Jason A. Mouabbi, Carlos H. Barcenas, Kelly K. Hunt and Debu Tripathy
Cancers 2026, 18(2), 253; https://doi.org/10.3390/cancers18020253 - 14 Jan 2026
Viewed by 33
Abstract
Background: HER2-low breast cancer (HER2 immunohistochemical [IHC] score 1+, or IHC 2+ without HER2 gene amplification) is distinct from HER2-positive and HER2-0 breast cancer (IHC 0), with a differing prognosis and specific therapeutic options. The DESTINY-Breast04 trial demonstrated notable efficacy of the HER2 [...] Read more.
Background: HER2-low breast cancer (HER2 immunohistochemical [IHC] score 1+, or IHC 2+ without HER2 gene amplification) is distinct from HER2-positive and HER2-0 breast cancer (IHC 0), with a differing prognosis and specific therapeutic options. The DESTINY-Breast04 trial demonstrated notable efficacy of the HER2 antibody–drug conjugate trastuzumab deruxtecan over standard chemotherapy in patients with metastatic breast cancer (MBC) defined as HER2-low. More recently, the DESTINY-Breast06 trial confirmed this benefit in hormone receptor-positive and HER2-ultralow (less than 1+, but with ≤10% of infiltrating cancer cells showing incomplete and faint/weak membrane staining) cases, prompting re-evaluation of HER2 diagnostic thresholds and treatment strategies. Methods: Eligible patients were women with HER2-low or HER2-0 MBC evaluated at MD Anderson between January 2006 and January 2019. HER2-low was defined as either (1) IHC 1+ or (2) IHC 2+ and negative on fluorescence in situ hybridization. Multivariate logistic regression was used to evaluate distinct clinicopathologic features of patients with HER2-low status. Overall survival (OS) was estimated by the Kaplan–Meier method. Multivariate Cox proportional hazards regression was applied to assess the effects of covariates of interest on OS across different HER2 groups. Results: We included 3834 women: 2637 (69%) with recurrent and 1197 (31%) with de novo MBC; HER2-low disease was present in 1575 (60%) and 712 (59%), respectively. In de novo cases, higher nuclear grade was associated with HER2-low status (grade 2 vs. 1, OR = 2.02, p = 0.007; grade 3 vs. 1, OR = 1.87, p = 0.015), while recurrent cases were associated with ER-positivity (OR = 1.96, p < 0.001) and prior adjuvant radiotherapy (OR = 0.79, p = 0.007). Median OS was 3.2 years (95% CI 3.0–3.5). In de novo disease, Black race (HR = 1.48), metaplastic (HR = 3.15) or other non-ductal/lobular histologies (HR = 2.36), and grade 3 (HR = 1.67) predicted worse OS, whereas Hispanic ethnicity (HR = 0.74) and Other races (HR = 0.57), higher ER (HR = 0.48–0.41) and PR (HR = 0.72–0.53), and HER2-low status (HR = 0.77) conferred improved outcomes. In recurrent disease, Black race predicted worse OS (HR = 1.21, 95% CI 1.05–1.39), while Other race (HR = 0.78, 95% CI 0.62–0.97), higher ER (HR = 0.69–0.44) and PR (HR = 0.73–0.73), and HER2-low (HR = 0.89) were protective. HER2 discordance between primary and metastatic sites occurred in 38.8% of recurrent and 13.1% of de novo cases. Conclusions: HER2-low status was significantly associated with longer OS compared to HER2-0 status in both recurrent and de novo MBC cases. These real-world data help establish the prevalence of HER2-low status and its distinct outcomes. The discrepancy in HER2-low status between the primary tumor and metastatic sites highlights the potential for changes in HER2 expression over time, exploring the interaction between HER2-low breast cancer and the tumor microenvironment and emphasizing the importance of monitoring and reassessing HER2 status at various stages to guide treatment decisions effectively and the need for more quantitative and reproducible HER assays. Full article
(This article belongs to the Special Issue Cancer Metastasis in 2025–2026)
Show Figures

Figure 1

33 pages, 4569 KB  
Article
Heterotypic 3D Model of Breast Cancer Based on Tumor, Stromal and Endothelial Cells: Cytokines Interaction in the Tumor Microenvironment
by Anastasia Leonteva, Alina Kazakova, Ekaterina Berezutskaya, Anna Ilyina, David Sergeevichev, Sergey Vladimirov, Maria Bogachek, Igor Vakhrushev, Pavel Makarevich, Vladimir Richter and Anna Nushtaeva
Cells 2026, 15(2), 145; https://doi.org/10.3390/cells15020145 - 14 Jan 2026
Viewed by 39
Abstract
The recreation of the tumor microenvironment remains a significant challenge in the development of experimental cancer models. The present study constitutes an investigation into the interconnection between tumor, endothelial and stromal cells in heterotypic breast cancer spheroids. The generation of models was achieved [...] Read more.
The recreation of the tumor microenvironment remains a significant challenge in the development of experimental cancer models. The present study constitutes an investigation into the interconnection between tumor, endothelial and stromal cells in heterotypic breast cancer spheroids. The generation of models was achieved through the utilization of MCF7, MDA-MB-231, and SK-BR-3 tumor cell lines, in conjunction with endothelial TIME-RFP cells and either cancer-associated (BrC4f) or normal (BN120f) fibroblasts, within ultra-low attachment plates. It was established that stromal cells, most notably fibroblasts, were conducive to the aggregation of tumor cells into spheroids and the formation of pseudovessels in close proximity to fibroblast bands. In contrast to the more aggressive tumor models MDA-MB-231 and SK-BR-3, microenvironment cells do not influence the migration ability of MCF7 tumor cells. Heterotypic spheroids incorporating CAFs demonstrated a more aggressive and immunosuppressive phenotype. Multiplex immunoassay analysis of cytokines, followed by STRING cluster analysis, was used to identify key processes including angiogenesis, invasion, stem cell maintenance, and immunosuppression. Furthermore, a cluster of cytokines (LIF, SDF-1, HGF, SCGFb) was identified as potentially involved in the regulation of PD-L1 expression by tumor cells. This finding reveals a potential mechanism of immune evasion and suggests new avenues for therapeutic investigation. Full article
(This article belongs to the Special Issue Cell-to-Cell Crosstalk as a Target of Therapies)
Show Figures

Figure 1

31 pages, 3388 KB  
Review
Molecular Insights into Dominant Pseudouridine RNA Modification: Implications for Women’s Health and Disease
by Qiwei Yang, Ayman Al-Hendy and Thomas G. Boyer
Biology 2026, 15(2), 142; https://doi.org/10.3390/biology15020142 - 14 Jan 2026
Viewed by 60
Abstract
Pseudouridine (Ψ), the most abundant RNA modification, plays essential roles in shaping RNA structure, stability, and translational output. Beyond cancer, Ψ is dynamically regulated across numerous physiological and pathological contexts—including immune activation, metabolic disorders, stress responses, and pregnancy-related conditions such as preeclampsia—where elevated [...] Read more.
Pseudouridine (Ψ), the most abundant RNA modification, plays essential roles in shaping RNA structure, stability, and translational output. Beyond cancer, Ψ is dynamically regulated across numerous physiological and pathological contexts—including immune activation, metabolic disorders, stress responses, and pregnancy-related conditions such as preeclampsia—where elevated Ψ levels reflect intensified RNA turnover and modification activity. These broad functional roles highlight pseudouridylation as a central regulator of cellular homeostasis. Emerging evidence demonstrates that Ψ dysregulation contributes directly to the development and progression of several women’s cancers, including breast, ovarian, endometrial, and cervical malignancies. Elevated Ψ levels in tissues, blood, and urine correlate with tumor burden, metastatic potential, and therapeutic responsiveness. Aberrant activity of Ψ synthases such as PUS1, PUS7, and the H/ACA ribonucleoprotein component dyskerin alters pseudouridylation patterns across multiple RNA substrates, including rRNA, tRNA, mRNA, snoRNAs, and ncRNAs. These widespread modifications reshape ribosome function, modify transcript stability and translational efficiency, reprogram RNA–protein interactions, and activate oncogenic signaling programs. Advances in high-resolution, site-specific Ψ mapping technologies have further revealed mechanistic links between pseudouridylation and malignant transformation, highlighting how modification of distinct RNA classes contributes to altered cellular identity and tumor progression. Collectively, Ψ and its modifying enzymes represent promising biomarkers and therapeutic targets across women’s cancers, while also serving as sensitive indicators of diverse non-cancer physiological and disease states. Full article
(This article belongs to the Section Developmental and Reproductive Biology)
Show Figures

Figure 1

36 pages, 2843 KB  
Review
Bone Metastasis in Estrogen Receptor-Positive Breast Cancer: Molecular Insights and Therapeutic Advances
by Zhuoran Huang, Yi Wu and Yanshu Li
Int. J. Mol. Sci. 2026, 27(2), 785; https://doi.org/10.3390/ijms27020785 - 13 Jan 2026
Viewed by 74
Abstract
Estrogen receptor-positive (ER+) breast cancer represents the most prevalent molecular subtype of breast cancer, characterized by hormone-dependent growth, relatively indolent progression, and a pronounced tendency to metastasize to bone. While endocrine therapies remain the cornerstone of treatment, a significant proportion of [...] Read more.
Estrogen receptor-positive (ER+) breast cancer represents the most prevalent molecular subtype of breast cancer, characterized by hormone-dependent growth, relatively indolent progression, and a pronounced tendency to metastasize to bone. While endocrine therapies remain the cornerstone of treatment, a significant proportion of ER+ tumors eventually develop resistance, culminating in distant metastases—most frequently to the bone. Bone metastasis substantially compromises patient survival and quality of life, highlighting the critical need to elucidate its molecular underpinnings. Recent multi-omics and mechanistic studies have shed light on the complex interplay between tumor-intrinsic signaling pathways, such as dysregulated ER signaling, PI3K/AKT/mTOR, TGF-β, and Hippo pathways, and the bone microenvironment, including osteoclast activation, immune suppression, and stromal remodeling. This review systematically summarizes the current understanding of the molecular mechanisms driving bone metastasis in ER+ breast cancer, with a particular focus on tumor–bone microenvironment crosstalk and key regulatory pathways. Additionally, we discuss recent advances in therapeutic strategies, encompassing next-generation endocrine therapies, CDK4/6 inhibitors, bone-targeted agents, and pathway-specific inhibitors. Together, these insights pave the way for more effective and personalized interventions against ER+ breast cancer with bone involvement. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

44 pages, 10841 KB  
Article
Study on Dual-Targeted Liposomes Containing Curcumin-Copper Chelate in the Treatment of Triple-Negative Breast Cancer
by Lina Wu, Xueli Guo and Pan Guo
Pharmaceuticals 2026, 19(1), 135; https://doi.org/10.3390/ph19010135 - 13 Jan 2026
Viewed by 76
Abstract
Background: Triple-negative breast cancer (TNBC) remains primarily treated with chemotherapy due to the lack of effective therapeutic targets, but this approach carries significant systemic toxicity and a high risk of drug resistance. Curcumin (Cur), despite its multifaceted antitumor activity, faces limitations in [...] Read more.
Background: Triple-negative breast cancer (TNBC) remains primarily treated with chemotherapy due to the lack of effective therapeutic targets, but this approach carries significant systemic toxicity and a high risk of drug resistance. Curcumin (Cur), despite its multifaceted antitumor activity, faces limitations in clinical application due to poor water solubility and weak targeting properties. This study aims to develop a folate/mitochondria dual-targeted curcumin–copper chelate liposome (Cu-Cur DTLPs) formulation that enables copper accumulation within tumor cells and induces copper-mediated cell death, thereby providing an effective and relatively low-toxicity therapeutic strategy for triple-negative breast cancer. Methods: Curcumin–copper chelates (Cu-Cur) were first synthesized and characterized using mass spectrometry, NMR, and infrared spectroscopy. Subsequently, dual-targeted liposomes (Cu-Cur DTLPs) were prepared via the thin-film dispersion method, with systematic evaluation of particle size, zeta potential, encapsulation efficiency, and in vitro release profiles. In vitro cytotoxicity was assessed against 4T-1 and MDA-MB-231 cells using the MTT assay. In a 4T-1 tumor-bearing BALB/c mouse model, comprehensive evaluation of targeting efficiency, antitumor efficacy, and mechanisms of action was conducted via in vivo imaging, tumor volume monitoring, immunohistochemistry (detecting FDX1 and DLAT proteins), and TUNEL staining. Results: Cu-Cur DTLPs with a uniform particle size of approximately 104.4 nm were successfully synthesized. In vitro and in vivo studies demonstrated that compared to free curcumin and conventional liposomes, Cu-Cur DTLPs significantly enhanced drug accumulation in tumor tissues and exhibited effective tumor growth inhibition. Mechanistic studies confirmed that this formulation specifically accumulates copper ions within tumor cells, upregulates FDX1, promotes DLAT oligomerization, and induces mitochondrial dysfunction, thereby driving copper death. TUNEL staining ruled out apoptosis as the primary mechanism. Safety evaluation revealed no significant toxicity in major organs. Conclusions: The Cu-Cur DTLPs developed in this study effectively induce copper-mediated death in TNBC through a dual-targeted delivery system, significantly enhancing antitumor activity with favorable safety profiles. This establishes a highly promising novel nanotherapeutic strategy for TNBC treatment. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

Back to TopTop