Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.000
Filter
1.
Biomed Environ Sci ; 37(6): 581-593, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38988109

ABSTRACT

Objective: Triple-negative breast cancer (TNBC) poses a significant challenge for treatment efficacy. CD8+ T cells, which are pivotal immune cells, can be effectively analyzed for differential gene expression across diverse cell populations owing to rapid advancements in sequencing technology. By leveraging these genes, our objective was to develop a prognostic model that accurately predicts the prognosis of patients with TNBC and their responsiveness to immunotherapy. Methods: Sample information and clinical data of TNBC were sourced from The Cancer Genome Atlas and METABRIC databases. In the initial stage, we identified 67 differentially expressed genes associated with immune response in CD8+ T cells. Subsequently, we narrowed our focus to three key genes, namely CXCL13, GBP2, and GZMB, which were used to construct a prognostic model. The accuracy of the model was assessed using the validation set data and receiver operating characteristic (ROC) curves. Furthermore, we employed various methods, including Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, immune infiltration, and correlation analyses with CD274 (PD-L1) to explore the model's predictive efficacy in immunotherapeutic responses. Additionally, we investigated the potential underlying biological pathways that contribute to divergent treatment responses. Results: We successfully developed a model capable of predicting the prognosis of patients with TNBC. The areas under the curve (AUC) values for the 1-, 3-, and 5-year survival predictions were 0.618, 0.652, and 0.826, respectively. Employing this risk model, we stratified the samples into high- and low-risk groups. Through KEGG enrichment analysis, we observed that the high-risk group predominantly exhibited enrichment in metabolism-related pathways such as drug and chlorophyll metabolism, whereas the low-risk group demonstrated significant enrichment in cytokine pathways. Furthermore, immune landscape analysis revealed noteworthy variations between (PD-L1) expression and risk scores, indicating that our model effectively predicted the response of patients to immune-based treatments. Conclusion: Our study demonstrates the potential of CXCL13, GBP2, and GZMB as prognostic indicators of clinical outcomes and immunotherapy responses in patients with TNBC. These findings provide valuable insights and novel avenues for developing immunotherapeutic approaches targeting TNBC.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Humans , CD8-Positive T-Lymphocytes/immunology , Prognosis , Female
2.
Sci Rep ; 14(1): 16073, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992094

ABSTRACT

Triple-negative breast cancer (TNBC) is often treated with neoadjuvant systemic therapy (NAST). We investigated if radiomic models based on multiparametric Magnetic Resonance Imaging (MRI) obtained early during NAST predict pathologic complete response (pCR). We included 163 patients with stage I-III TNBC with multiparametric MRI at baseline and after 2 (C2) and 4 cycles of NAST. Seventy-eight patients (48%) had pCR, and 85 (52%) had non-pCR. Thirty-six multivariate models combining radiomic features from dynamic contrast-enhanced MRI and diffusion-weighted imaging had an area under the receiver operating characteristics curve (AUC) > 0.7. The top-performing model combined 35 radiomic features of relative difference between C2 and baseline; had an AUC = 0.905 in the training and AUC = 0.802 in the testing set. There was high inter-reader agreement and very similar AUC values of the pCR prediction models for the 2 readers. Our data supports multiparametric MRI-based radiomic models for early prediction of NAST response in TNBC.


Subject(s)
Multiparametric Magnetic Resonance Imaging , Neoadjuvant Therapy , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Female , Neoadjuvant Therapy/methods , Middle Aged , Multiparametric Magnetic Resonance Imaging/methods , Adult , Aged , Treatment Outcome , ROC Curve , Magnetic Resonance Imaging/methods , Radiomics
3.
Biomed Eng Online ; 23(1): 67, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003472

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is a subtype of breast cancer with the worst prognosis. Radiotherapy (RT) is one of the core modalities for the disease; however, the ionizing radiation of RT has severe side effects. The consistent development direction of RT is to achieve better therapeutic effect with lower radiation dose. Studies have demonstrated that synergistic effects can be achieved by combining RT with non-ionizing radiation therapies such as light and magnetic therapy, thereby achieving the goal of dose reduction and efficacy enhancement. METHODS: In this study, we applied FeCo NPs with magneto thermal function and phototherapeutic agent IR-780 to construct an ionizing and non-ionizing radiation synergistic nanoparticle (INS NPs). INS NPs are first subjected to morphology, size, colloidal stability, loading capacity, and photothermal conversion tests. Subsequently, the cell inhibitory and cellular internalization were evaluated using cell lines in vitro. Following comprehensive assessment of the NPs' in vivo biocompatibility, tumor-bearing mouse model was established to evaluate their distribution, targeted delivery, and anti-tumor effects in vivo. RESULTS: INS NPs have a saturation magnetization exceeding 72 emu/g, a hydrodynamic particle size of approximately 40 nm, a negatively charged surface, and good colloidal stability and encapsulation properties. INS NPs maintain the spectral characteristics of IR-780 at 808 nm. Under laser irradiation, the maximum temperature was 92 °C, INS NPs also achieved the effective heat temperature in vivo. Both in vivo and in vitro tests have proven that INS NPs have good biocompatibility. INS NPs remained effective for more than a week after one injection in vivo, and can also be guided and accumulated in tumors through permanent magnets. Later, the results exhibited that under low-dose RT and laser irradiation, the combined intervention group showed significant synergetic effects, and the ROS production rate was much higher than that of the RT and phototherapy-treated groups. In the mice model, 60% of the tumors were completely eradicated. CONCLUSIONS: INS NPs effectively overcome many shortcomings of RT for TNBC and provide experimental basis for the development of novel clinical treatment methods for TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/radiotherapy , Triple Negative Breast Neoplasms/therapy , Animals , Cell Line, Tumor , Mice , Humans , Female , Nanoparticles/chemistry , Radiation, Ionizing , Drug Carriers/chemistry , Combined Modality Therapy , Indoles
4.
PeerJ ; 12: e17667, 2024.
Article in English | MEDLINE | ID: mdl-39006029

ABSTRACT

Advances in understanding the pathological mechanisms of breast cancer have resulted in the emergence of novel therapeutic strategies. However, triple-negative breast cancer (TNBC), a molecular subtype of breast cancer with a poor prognosis, lacks classical and general therapeutic targets, hindering the clinical application of several therapies to breast cancer. As insights into the unique immunity and molecular mechanisms of TNBC have become more extensive, immunotherapy has gradually become a valuable complementary approach to classical radiotherapy and chemotherapy. CD8+ cells are significant actors in the tumor immunity cycle; thus, research on TNBC immunotherapy is increasingly focused in this direction. Recently, CD8+ tissue-resident memory (TRM) cells, a subpopulation of CD8+ cells, have been explored in relation to breast cancer and found to seemingly play an undeniably important role in tumor surveillance and lymphocytic infiltration. In this review, we summarize the recent advances in the mechanisms and relative targets of CD8+ T cells, and discuss the features and potential applications of CD8+ TRM cells in non-luminal breast cancer immunotherapy.


Subject(s)
Immunologic Memory , T-Lymphocytes, Cytotoxic , Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Immunotherapy/methods , CD8-Positive T-Lymphocytes/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Memory T Cells/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism
5.
BMC Med Imaging ; 24(1): 136, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844842

ABSTRACT

BACKGROUND: To develop and validate a peritumoral vascular and intratumoral radiomics model to improve pretreatment predictions for pathologic complete responses (pCRs) to neoadjuvant chemoradiotherapy (NAC) in patients with triple-negative breast cancer (TNBC). METHODS: A total of 282 TNBC patients (93 in the primary cohort, 113 in the validation cohort, and 76 in The Cancer Imaging Archive [TCIA] cohort) were retrospectively included. The peritumoral vasculature on the maximum intensity projection (MIP) from pretreatment DCE-MRI was segmented by a Hessian matrix-based filter and then edited by a radiologist. Radiomics features were extracted from the tumor and peritumoral vasculature of the MIP images. The LASSO method was used for feature selection, and the k-nearest neighbor (k-NN) classifier was trained and validated to build a predictive model. The diagnostic performance was assessed using the ROC analysis. RESULTS: One hundred of the 282 patient (35.5%) with TNBC achieved pCRs after NAC. In predicting pCRs, the combined peritumoral vascular and intratumoral model (fusion model) yields a maximum AUC of 0.82 (95% confidence interval [CI]: 0.75, 0.88) in the primary cohort, a maximum AUC of 0.67 (95% CI: 0.57, 0.76) in the internal validation cohort, and a maximum AUC of 0.65 (95% CI: 0.52, 0.78) in TCIA cohort. The fusion model showed improved performance over the intratumoral model and the peritumoral vascular model, but not significantly (p > 0.05). CONCLUSION: This study suggested that combined peritumoral vascular and intratumoral radiomics model could provide a non-invasive tool to enable prediction of pCR in TNBC patients treated with NAC.


Subject(s)
Magnetic Resonance Imaging , Neoadjuvant Therapy , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Female , Middle Aged , Retrospective Studies , Magnetic Resonance Imaging/methods , Adult , Aged , Treatment Outcome , Pathologic Complete Response , Radiomics
6.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928364

ABSTRACT

Triple-negative breast cancer (TNBC) remains one of the most challenging subtypes since it is initially characterized by the absence of specific biomarkers and corresponding targeted therapies. Advances in methodology, translational informatics, genomics, and proteomics have significantly contributed to the identification of therapeutic targets. The development of innovative treatments, such as antibody-drug conjugates and immune checkpoint inhibitors, alongside chemotherapy, has now become the standard of care. However, the quest for biomarkers defining therapy outcomes is still ongoing. Peroxiporins, which comprise a subgroup of aquaporins, which are membrane pores facilitating the transport of water, glycerol, and hydrogen peroxide, have emerged as potential biomarkers for therapy response. Research on peroxiporins reveals their involvement beyond traditional channeling activities, which is also reflected in their cellular localization and roles in cellular signaling pathways. This research on peroxiporins provides fresh insights into the mechanisms of therapy resistance in tumors, offering potential avenues for predicting treatment outcomes and tailoring successful TNBC therapies.


Subject(s)
Biomarkers, Tumor , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/therapy , Biomarkers, Tumor/metabolism , Female , Aquaporins/metabolism , Signal Transduction , Animals
7.
Int J Pharm ; 660: 124346, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38889853

ABSTRACT

Breast cancer, the second leading global cause of death, affects 2.1 million women annually, with an alarming 15 percent mortality rate. Among its diverse forms, Triple-negative breast cancer (TNBC) emerges as the deadliest, characterized by the absence of hormone receptors. This article underscores the urgent need for innovative treatment approaches in tackling TNBC, emphasizing the transformative potential of polymeric nanomaterials (PNMs). Evolved through nanotechnology, PNMs offer versatile biomedical applications, particularly in addressing the intricate challenges of TNBC. The synthesis methods of PNMs, explored within the tumor microenvironment using cellular models, showcase their dynamic nature in cancer treatment. The article anticipates the future of TNBC therapeutics through the optimization of PNMs-based strategies, integrating them into photothermal (PT), photodynamic (PT), and hyperthermia therapy (HTT), drug delivery, and active tumor targeting strategies. Advancements in synthetic methods, coupled with a nuanced understanding of the tumor microenvironment, hold promise for personalized interventions. Comparative investigations of therapeutic models and a thorough exploration of polymeric nanoplatforms toxicological perspectives become imperative for ensuring efficacy and safety. We have explored the interdisciplinary collaboration between nanotechnology, oncology, and molecular biology as pivotal in translating PNMs innovations into tangible benefits for TNBC patients.


Subject(s)
Nanostructures , Polymers , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/therapy , Humans , Nanostructures/chemistry , Nanostructures/administration & dosage , Polymers/chemistry , Female , Animals , Theranostic Nanomedicine/methods , Drug Delivery Systems/methods , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Tumor Microenvironment/drug effects , Hyperthermia, Induced/methods
8.
Front Immunol ; 15: 1420107, 2024.
Article in English | MEDLINE | ID: mdl-38933280

ABSTRACT

Triple-negative breast cancer (TNBC) is a subtype of breast cancer that presents significant therapeutic challenges due to the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. As a result, conventional hormonal and targeted therapies are largely ineffective, underscoring the urgent need for novel treatment strategies. γδT cells, known for their robust anti-tumor properties, show considerable potential in TNBC treatment as they can identify and eliminate tumor cells without reliance on MHC restrictions. These cells demonstrate extensive proliferation both in vitro and in vivo, and can directly target tumors through cytotoxic effects or indirectly by promoting other immune responses. Studies suggest that expansion and adoptive transfer strategies targeting Vδ2 and Vδ1 γδT cell subtypes have shown promise in preclinical TNBC models. This review compiles and discusses the existing literature on the primary subgroups of γδT cells, their roles in cancer therapy, their contributions to tumor cell cytotoxicity and immune modulation, and proposes potential strategies for future γδT cell-based immunotherapies in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Animals , Female , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Immunotherapy, Adoptive/methods , Immunotherapy/methods
10.
Breast ; 76: 103757, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38843710

ABSTRACT

INTRODUCTION: Breast cancer stands as the second most common solid tumors with a propensity for brain metastasis. Among metastatic breast cancer cases, the brain metastasis incidence ranges from 10 % to 30 %, with triple-negative breast cancer (TNBC) displaying a heightened risk and poorer prognosis. SRS has emerged as an effective local treatment modality for brain metastases; however, data on its outcomes specifically in pure triple-negative subtype remain scarce. METHOD: We retrospectively reviewed the electronic medical records of all brain metastasis (BM) TNBC patients treated with SRS. Patient, tumour characteristics and treatment details data were collected. This retrospective cohort study aimed to evaluate local control (LC), distant brain metastasis free survival (DBMFS), and overall survival (OS) outcomes in TNBC patients undergoing SRS for brain metastases while identifying potential prognostic factors. RESULT: Forty-three patients with TNBC and brain metastases treated with SRS between January 2017 and 2023 were included. The study found rates of LC (99 % at 1 year) and DBMFS (76 % at 1 year) after SRS, with brain metastasis count (p = 0,003) and systemic treatment modality (p = 0,001) being significant predictors of DBMFS. The median OS following SRS was 19.5 months, with neurological deficit (p = 0.003) and systemic treatment modality (p = 0.019) identified as significant predictors of OS. CONCLUSION: SRS demonstrates favourable outcomes in terms of local control and distant brain metastasis-free survival in TNBC. Neurological deficit and systemic treatment significantly influence overall survival, emphasizing the importance of personalized treatment approaches and (magnetic resonance imaging) MRI surveillance based on these factors.


Subject(s)
Brain Neoplasms , Radiosurgery , Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy , Middle Aged , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Retrospective Studies , Radiosurgery/methods , Prognosis , Adult , Aged , Treatment Outcome , Immunotherapy/methods
11.
Nano Lett ; 24(25): 7764-7773, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38864366

ABSTRACT

Inducing immunogenic cell death (ICD) during photothermal therapy (PTT) has the potential to effectively trigger photothermal immunotherapy (PTI). However, ICD induced by PTT alone is often limited by inefficient PTT, low immunogenicity of tumor cells, and a dysregulated redox microenvironment. Herein, we develop MoSe2 nanosheets with high-percentage metallic 1T phase and rich exposed active Mo centers through phase and defect engineering of MoSe2 as an effective nanoagent for PTI. The metallic 1T phase in MoSe2 nanosheets endows them with strong PTT performance, and the abundant exposed active Mo centers endow them with high activity for glutathione (GSH) depletion. The MoSe2-mediated high-performance PTT synergizing with efficient GSH depletion facilitates the release of tumor-associated antigens to induce robust ICD, thus significantly enhancing checkpoint blockade immunotherapy and activating systemic immune response in mouse models of colorectal cancer and triple-negative metastatic breast cancer.


Subject(s)
Immunotherapy , Molybdenum , Photothermal Therapy , Animals , Mice , Immunotherapy/methods , Humans , Molybdenum/chemistry , Female , Cell Line, Tumor , Nanostructures/chemistry , Nanostructures/therapeutic use , Glutathione/chemistry , Glutathione/metabolism , Colorectal Neoplasms/therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Immunogenic Cell Death/drug effects , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Infrared Rays , Selenium/chemistry , Selenium/therapeutic use , Phototherapy/methods
12.
ACS Biomater Sci Eng ; 10(6): 3568-3598, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38815129

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype with the worst prognosis and highest recurrence rates. The treatment choices are limited due to the scarcity of endocrine and HER2 targets, except for chemotherapy. However, the side effects of chemotherapy restrict its long-term usage. Immunotherapy shows potential as a promising therapeutic strategy, such as inducing immunogenic cell death, immune checkpoint therapy, and immune adjuvant therapy. Nanotechnology offers unique advantages in the field of immunotherapy, such as improved delivery and targeted release of immunotherapeutic agents and enhanced bioavailability of immunomodulators. As well as the potential for combination therapy synergistically enhanced by nanocarriers. Nanoparticles-based combined application of multiple immunotherapies is designed to take the tactics of enhancing immunogenicity and reversing immunosuppression. Moreover, the increasing abundance of biomedical materials holds more promise for the development of this field. This review summarizes the advances in the field of nanoparticle-mediated immunotherapy in terms of both immune strategies for treatment and the development of biomaterials and presents challenges and hopes for the future.


Subject(s)
Immunotherapy , Nanoparticles , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/drug therapy , Immunotherapy/methods , Nanoparticles/therapeutic use , Nanoparticles/chemistry , Female , Animals
13.
Comput Biol Med ; 176: 108537, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744008

ABSTRACT

BACKGROUND: Anti-PD-1/PD-L1 treatment has achieved durable responses in TNBC patients, whereas a fraction of them showed non-sensitivity to the treatment and the mechanism is still unclear. METHODS: Pre- and post-treatment plasma samples from triple negative breast cancer (TNBC) patients treated with immunotherapy were measured by tandem mass tag (TMT) mass spectrometry. Public proteome data of lung cancer and melanoma treated with immunotherapy were employed to validate the findings. Blood and tissue single-cell RNA sequencing (scRNA-seq) data of TNBC patients treated with or without immunotherapy were analyzed to identify the derivations of plasma proteins. RNA-seq data from IMvigor210 and other cancer types were used to validate plasma proteins in predicting response to immunotherapy. RESULTS: A random forest model constructed by FAP, LRG1, LBP and COMP could well predict the response to immunotherapy. The activation of complement cascade was observed in responders, whereas FAP and COMP showed a higher abundance in non-responders and negative correlated with the activation of complements. scRNA-seq and bulk RNA-seq analysis suggested that FAP, COMP and complements were derived from fibroblasts of tumor tissues. CONCLUSIONS: We constructe an effective plasma proteomic model in predicting response to immunotherapy, and find that FAP+ and COMP+ fibroblasts are potential targets for reversing immunotherapy resistance.


Subject(s)
Immunotherapy , Proteomics , Single-Cell Analysis , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Female , Immunotherapy/methods , Single-Cell Analysis/methods , Proteomics/methods , B7-H1 Antigen/blood , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Transcriptome , Immune Checkpoint Inhibitors/therapeutic use , Gene Expression Profiling , Proteome
14.
Front Immunol ; 15: 1355130, 2024.
Article in English | MEDLINE | ID: mdl-38742103

ABSTRACT

Pre-operative radiation therapy is not currently integrated into the treatment protocols for breast cancer. However, transforming immunological "cold" breast cancers by neoadjuvant irradiation into their "hot" variants is supposed to elicit an endogenous tumor immune defense and, thus, enhance immunotherapy efficiency. We investigated cellular and immunological effects of sub-lethal, neoadjuvant irradiation of ER pos., HER2 pos., and triple-negative breast cancer subtypes in-vitro and in-vivo in humanized tumor mice (HTM). This mouse model is characterized by a human-like immune system and therefore facilitates detailed analysis of the mechanisms and efficiency of neoadjuvant, irradiation-induced "in-situ vaccination", especially in the context of concurrently applied checkpoint therapy. Similar to clinical appearances, we observed a gradually increased immunogenicity from the luminal over the HER2-pos. to the triple negative subtype in HTM indicated by an increasing immune cell infiltration into the tumor tissue. Anti-PD-L1 therapy divided the HER2-pos. and triple negative HTM groups into responder and non-responder, while the luminal HTMs were basically irresponsive. Irradiation alone was effective in the HER2-pos. and luminal subtype-specific HTM and was supportive for overcoming irresponsiveness to single anti-PD-L1 treatment. The treatment success correlated with a significantly increased T cell proportion and PD-1 expression in the spleen. In all subtype-specific HTM combination therapy proved most effective in diminishing tumor growth, enhancing the immune response, and converted non-responder into responder during anti-PD-L1 therapy. In HTM, neoadjuvant irradiation reinforced anti-PD-L1 checkpoint treatment of breast cancer in a subtype -specific manner. According to the "bench to bedside" principle, this study offers a vital foundation for clinical translating the use of neoadjuvant irradiation in the context of checkpoint therapy.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Neoadjuvant Therapy , Receptor, ErbB-2 , Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , Cell Line, Tumor , Disease Models, Animal , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Neoadjuvant Therapy/methods , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/radiotherapy , Triple Negative Breast Neoplasms/therapy , Xenograft Model Antitumor Assays
15.
Ann Surg Oncol ; 31(8): 5180-5188, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38767803

ABSTRACT

BACKGROUND: We examined the association between immunotherapy-containing and standard chemotherapy regimens with treatment delays and postoperative complications in stage II-III triple-negative breast cancer. The effect of immune-related adverse events (irAEs) was compared. PATIENTS AND METHODS: We compared 139 women treated with neoadjuvant pembrolizumab plus chemotherapy (KEYNOTE-522 regimen) from August 2021 to September 2022 with 287 consecutive patients who received neoadjuvant chemotherapy alone prior to July 2021 and underwent surgery. Baseline characteristics, time to treatments, and surgical complications were compared using two-sample non-parametric tests. Linear regression evaluated association of irAEs with time to surgery and radiation. Logistic regression identified factors associated with surgical complications. RESULTS: Age, body mass index, race, American Society of Anesthesiologists (ASA) class, and mastectomy rates were similar among cohorts. No clinically relevant difference in time from end of neoadjuvant treatment to surgery was observed [KEYNOTE-522: median 32 (IQR 27, 43) days; non-KEYNOTE-522: median 31 (IQR 26, 37) days; P = 0.048]. Time to radiation did not differ (P = 0.7). A total of 26 patients (9%; non-KEYNOTE-522) versus 11 (8%; KEYNOTE-522) experienced postoperative complications (P = 0.6). In the KEYNOTE-522 cohort, 59 (43%) of 137 patients experienced 82 irAEs; 40 (68%) required treatment. Older age (P = 0.018) and ASA class 4 (P = 0.007) were associated with delays to surgery after adjusting for clinical factors. Experiencing ≥ 1 irAE was associated with delay to radiation (P = 0.029). IrAEs were not associated with surgical complications (P = 0.4). CONCLUSIONS: We observed no clinically meaningful difference between times to surgery/adjuvant radiation or postoperative complications and type of preoperative chemotherapy. IrAEs were associated with delay to adjuvant radiation but not with postoperative complications or delay to surgery.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Mastectomy , Neoadjuvant Therapy , Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Follow-Up Studies , Time-to-Treatment , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Postoperative Complications , Prognosis , Survival Rate , Immunotherapy , Adult , Retrospective Studies , Radiotherapy, Adjuvant
17.
J Control Release ; 370: 707-720, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744346

ABSTRACT

Immunotherapy is gaining prominence as a promising strategy for treating triple-negative breast cancer (TNBC). Neoantigens (neoAgs) and cancer-testis antigens (CTAs) are tumor-specific targets originating from somatic mutations and epigenetic changes in cancer cells. These antigens hold great promise for personalized cancer vaccines, as supported by preclinical and early clinical evidence in TNBC. This review delves into the potential of neoAgs and CTAs as vaccine candidates, emphasizing diverse strategies and delivery approaches. It also highlights the current status of vaccination modalities undergoing clinical trials in TNBC therapy. A comprehensive understanding of neoAgs, CTAs, vaccination strategies, and innovative delivery methods is crucial for optimizing neoAg-based immunotherapies in clinical practice.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Immunotherapy , Triple Negative Breast Neoplasms , Humans , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Antigens, Neoplasm/immunology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Female , Animals , Immunotherapy/methods , Clinical Trials as Topic , Drug Delivery Systems/methods
18.
Cancer Treat Rev ; 128: 102762, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776613

ABSTRACT

Immunotherapy has revolutionized cancer therapy and now represents a standard of care for many tumor types, including triple-negative breast cancer. Despite the positive results that have led to the approval of immunotherapy in both early- and advanced-stage triple-negative breast cancer, pivotal clinical trials cannot address the myriad questions arising in everyday clinical practice, often falling short in delivering all the information that clinicians require. In this manuscript, we aim to address some of these practical questions, with the purpose of providing clinicians with a guide for optimizing the use of immune checkpoint inhibitors in the management of breast cancer patients and identifying opportunities for future research to clarify unresolved questions.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology , Immunotherapy/methods , Female , Immune Checkpoint Inhibitors/therapeutic use
19.
Pharmacol Res ; 204: 107205, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719195

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype lacking estrogen receptors, progesterone receptors and lacks HER2 overexpression. This absence of critical molecular targets poses significant challenges for conventional therapies. Immunotherapy, remarkably immune checkpoint blockade, offers promise for TNBC treatment, but its efficacy remains limited. Epigenetic dysregulation, including altered DNA methylation, histone modifications, and imbalances in regulators such as BET proteins, plays a crucial role in TNBC development and resistance to treatment. Hypermethylation of tumor suppressor gene promoters and the imbalance of histone methyltransferases such as EZH2 and histone deacetylases (HDACs) profoundly influence tumor cell proliferation, survival, and metastasis. In addition, epigenetic alterations critically shape the tumor microenvironment (TME), including immune cell composition, cytokine signaling, and immune checkpoint expression, ultimately contributing to immune evasion. Targeting these epigenetic mechanisms with specific inhibitors such as EZH2 and HDAC inhibitors in combination with immunotherapy represents a compelling strategy to remodel the TME, potentially overcoming immune evasion and enhancing therapeutic outcomes in TNBC. This review aims to comprehensively elucidate the current understanding of epigenetic modulation in TNBC, its influence on the TME, and the potential of combining epigenetic therapies with immunotherapy to overcome the challenges posed by this aggressive breast cancer subtype.


Subject(s)
Epigenesis, Genetic , Triple Negative Breast Neoplasms , Tumor Microenvironment , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/drug therapy , Tumor Microenvironment/genetics , Animals , Female , Immunotherapy , DNA Methylation , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylase Inhibitors/pharmacology
20.
J Hematol Oncol ; 17(1): 36, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783389

ABSTRACT

Oncolytic viruses (OVs) offer a novel approach to treat solid tumors; however, their efficacy is frequently suboptimal due to various limiting factors. To address this challenge, we engineered an OV containing targets for neuron-specific microRNA-124 and Granulocyte-macrophage colony-stimulating factor (GM-CSF), significantly enhancing its neuronal safety while minimally compromising its replication capacity. Moreover, we identified PARP1 as an HSV-1 replication restriction factor using genome-wide CRISPR screening. In models of glioblastoma (GBM) and triple-negative breast cancer (TNBC), we showed that the combination of OV and a PARP inhibitor (PARPi) exhibited superior efficacy compared to either monotherapy. Additionally, single-cell RNA sequencing (scRNA-seq) revealed that this combination therapy sensitized TNBC to immune checkpoint blockade, and the incorporation of an immune checkpoint inhibitor (ICI) further increased the survival rate of tumor-bearing mice. The combination of PARPi and ICI synergistically enhanced the ability of OV to establish durable tumor-specific immune responses. Our study effectively overcomes the inherent limitations of OV therapy, providing valuable insights for the clinical treatment of TNBC, GBM, and other malignancies.


Subject(s)
Oncolytic Virotherapy , Oncolytic Virotherapy/methods , Animals , Humans , Mice , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Glioblastoma/therapy , Glioblastoma/genetics , Oncolytic Viruses/genetics , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/genetics , Female , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Herpesvirus 1, Human/genetics , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , MicroRNAs/genetics , Xenograft Model Antitumor Assays , CRISPR-Cas Systems
SELECTION OF CITATIONS
SEARCH DETAIL