Breast Disease Branch of Shenzhen Medical Association, Tumor Branch of Shenzhen Medical Association, Breast Specialist Physicians Branch of Shenzhen Medical Doctor Association, Breast Health and Rehabilitation Management Committee of Shenzhen Health Management Association, Breast Cancer Committee of Shenzhen Anti-Cancer Association, Tumor Radiation Therapy Committee of Shenzhen Anti-Cancer Association, Thyroid and Breast Diseases Committee of Shenzhen Integrated Traditional Chinese and Western Medicine Association
Neoadjuvant therapy becomes increasingly important in comprehensive treatment of breast cancer, especially in locally advanced patients who have a high proportion in China. This consensus, developed by a multidisciplinary expert panel, based on existing evidence-based medical evidence and clinical practice, clarifies key concepts such as efficacy prediction and evaluation of breast cancer neoadjuvant therapy and their application in clinical decision-making through a systematic literature review combined with expert opinions. The consensus proposes a three-dimensional framework of efficacy prediction-dynamic evaluation-comprehensive evaluation, establishes a multidimensional assessment system that incorporates clinical, imaging, pathological and molecular biological information, and emphasizes the implementation of individualized monitoring strategies based on molecular subtypes of breast cancer. The contents include: (1) comprehensive baseline assessment and efficacy prediction methods before treatment; (2) principles of dynamic monitoring and treatment adjustment during treatment; (3) evaluation of therapeutic efficacy and assistance in decision-making; (4) molecular subtype-specific evaluation strategies and key points for long-term follow-up; (5) individualized evaluation protocols for special subtypes and special populations. This consensus aims to provide clinicians with standardized and normalized guidance to facilitate the precision management of breast cancer neoadjuvant therapy, improve efficacy, and enhance patient outcomes.
The tumor microenvironment of breast cancer is highly heterogeneous and dynamic, influencing disease progression and treatment response. Traditionally, breast cancer was considered to have low immunogenicity, but studies have found that triple negative breast cancer and HER-2 positive subtype have more tumor-infiltrating lymphocytes, such as CD8+ T cells and tertiary lymphoid structures, which may be associated with better prognosis and immune therapy response. In addition to tumor-infiltrating lymphocytes, the tumor microenvironment also includes macrophages, dendritic cells, mast cells and fibroblasts, which promote immune suppression or activation through various mechanisms. Metabolic reprogramming can further regulate the immune status of tumor microenvironment, influencing the occurrence, development, metastasis and treatment response of breast cancer. Although immune checkpoint inhibitors, especially anti-PD-1/PD-L1 drugs, have shown survival benefits in triple negative breast cancer patients, their efficacy is still limited to only a portion of patients. Therefore, in the future, it is necessary to further clarify the beneficiary population, reduce ineffective treatment, optimize combination treatment strategies, develop new immunotherapy drugs and explore predictive biomarkers, which may ultimately achieve precise treatment based on individual immune characteristics. This review systematically summarizes the composition of tumor microenvironment of breast cancer and its metabolic reprogramming characteristics, and discusses the current progress in breast cancer immunotherapy, which may provide insights for the optimization of treatment strategies and individualized immunotherapy.
To explore the heterogeneity of tumor-associated macrophage (TAM) in breast cancer and the functional differences among different TAM subgroups, and analyze the impact of TAM-related genes on the prognosis of triple negative breast cancer (TNBC) patients.
Methods
Thirty breast cancer tissue samples and 13 normal breast tissue samples from the GSE161529 dataset were collected. TAM subgroups were identified using the Seurat pipeline and consensus non-negative matrix factorization algorithm. The immune functions of different TAM subgroups were analyzed through GO and immune response enrichment analysis. The tissue samples of 5 TNBC patients from the GSE148673 dataset were collected. Univariate Cox regression and Pearson correlation analysis were used to screen for significant gene pairs related to TAM, and a co-expression network of TAM prognostic genes was constructed. K-means unsupervised clustering was used to get molecular subtypes of 247 TNBC patients in the TCGA and GEO databases based on the co-expression network genes, and survival differences and clinical characteristics were compared among different subtypes. Survival analysis was performed using the Kaplan-Meier method and log-rank test.
Results
Eight cell types (epithelial cells, T cells, fibroblasts, macrophages, endothelial cells, tissue stem cells, B cells and common myeloid progenitor cells) were annotated in the GSE161529 dataset. Among them, six functionally distinct TAM subgroups were identified, which played roles in immune inflammation, energy metabolism and cell adhesion. In the GSE148673 dataset, high expression of TAM-related genes CLEC4E, CTSC, CTSH, and CTSS was associated with a favorable prognosis in TNBC patients (P<0.01), while high expression of STAB1, RNASE1, SDS, SPP1 and TREM2 indicated a poor prognosis (P<0.01). Through univariate Cox analysis and Pearson correlation analysis, a co-expression network of 26 TAM genes related to patient prognosis was constructed. TNBC patients were classified into protective type (Group A, 96 cases) and risky type (Group B, 99 cases) based on the expression of 26 TAM genes. The binary clustering heatmap showed high consistency within groups; the gene expression heatmap indicated that protective TAM genes were highly expressed in Group A and lowly expressed in Group B. Survival analysis showed that the overall survival of Group A patients was significantly better than that of Group B (χ2=6.63,P=0.010); the clinical heatmap further revealed differences in age and clinical stage between two groups.
Conclusion
TAMs in breast cancer and its tumor microenvironment are heterogeneous, and six TAM subgroups have distinct roles in immune inflammation, energy metabolism and cell adhesion. TAM-related genes have clinical typing and prognostic prediction value in TNBC patients.
To compare the clinical efficacy between hormone therapy and ultrasound-guided microwave ablation in treating non-lactating mastitis patients.
Methods
According to the inclusion and exclusion critieria, a retrospective analysis was conducted on 39 non-lactating mastitis patients admitted to the Guangdong North People's Hospital from January 2020 to March 2024. Patients were divided into two groups based on treatment methods: the hormone group (25 cases) and the ablation group (14 cases). The hormone group received conventional glucocorticoids and oral compound scutellaria barbata, while the ablation group underwent ultrasound-guided microwave ablation and oral compound scutellaria barbata. The median follow-up time was 29 months, during which symptom relief rates, satisfaction rates and major adverse reactions were recorded. Inter-group comparisons of symptom relief rates and patient satisfaction rates were performed using Fisher's exact test, while degree of symptom relief, satisfaction levels and adverse reactions were compared using non-parametric tests.
Results
The ablation group achieved a symptom relief rate of 85.7% (12/14), significantly higher than 48.0% (12/25) in the hormone group (P=0.038). No statistically significant difference was observed in degree of symptom relief between the two groups (Z=-1.612, P=0.104). The ablation group demonstrated the patient satisfaction rate of 85.7% (12/14), significantly higher than 44.0% (11/25) in the hormone group (P=0.017). Statistical significance was found in the satisfaction levels between the two groups (Z=-2.208, P=0.043). In the hormone group, adverse reactions include weight gain (19 cases), facial acne (17 cases), generalized rash (12 cases), and gastrointestinal adverse reactions (16 cases). In the ablation group, adverse reactions included swelling in the ablation area (6 cases), breast lumps (10 cases), skin ecchymosis in the ablation area (9 cases), and non-healing puncture sites (2 cases).
Conclusion
The ultrasound-guided microwave ablation combined with compound scutellaria barbata for non-lactating mastitis demonstrates shows a symptom relief similar to hormone therapy, but with higher patient satisfaction, worthy of clinical application.