Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Introduction: Triple-negative breast cancer (TNBC) demonstrates poor prognosis due to its heterogeneity-related biological barrier, immunosuppressive tumor microenvironment (TME) and escape of cancer cells on immune surveillance. Exosomes are membrane-encased vehicles with properties of inter- and intra-cellular communication and regulation for therapeutic use, even as drug vehicles. M1 macrophage-derived exosomes (M1-Exos) can communicate with adjacent M2 macrophages and reprogram them to M1 subtypes to reshape TME. Celastrol (CEL) is a highly promising natural antitumor drug and plays an important role in immunotherapy but with high toxicity and low water solubility.

Objectives: This work aims to provide reinforced immunotherapy to reshape tumor immunosuppressive microenvironment of TNBC by employing M1-Exos as a nanoscale co-delivery vehicle for CEL, offering increased tumor targeting and antitumor efficacy while reducing systemic toxicity to achieve improved treatment outcomes.

Methods: A versatile combinatorial delivery system, tLyP-1-M1-Exos/CEL, was engineered by modifying tLyP-1 peptide and loading CEL to M1-Exos, then the size distribution, morphological features were characterized by transmission electron microscope, nanoparticle tracking analysis, and biomarkers were characterized by western blot. The efficacy and mechanism were assessed using CCK-8, qPCR, immunofluorescence, in vivo fluorescence imaging system, flow cytometry, western blot, H&E staining and other methods at cellular level and in nude mice.

Results: The synthesized tLyP-1-M1-Exos/CEL exhibited specific tumor-targeting ability, greater tumor-suppressing properties and lower toxicity. The expression of M1 markers was upregulated and the expression of M2 markers was downregulated in M2-phenotype macrophages co-incubated with tLyP-1-M1-Exos/CEL. tLyP-1-M1-Exos/CEL decreased the expression of MHC I by inhibiting IRF1-CIITA signal pathway, making tumor cells more susceptible to be phagocytosed by macrophages.

Conclusion: Our study showed a reinforced immunotherapy that the synthesized tLyP-1-M1-Exos/CEL could not only reverse M2 macrophages into M1 to reshape immunosuppressive TME but also decrease the expression of MHC Ⅰ on tumor surface to enhance macrophage phagocytic autoimmunity.

Download full-text PDF

Source
http://dx.doi.org/10.1016/j.jare.2025.07.024DOI Listing

Publication Analysis

Top Keywords

reinforced immunotherapy
12
macrophage-derived exosomes
8
western blot
8
synthesized tlyp-1-m1-exos/cel
8
expression markers
8
expression mhc
8
tumor
5
tlyp-1-m1-exos/cel
5
immunotherapy macrophage-derived
4
cel
4

Similar Publications

Cellular Senescence and Immunosenescence in Melanoma: Insights From the Tumor Microenvironment.

Cancer Med

September 2025

Department of Chinese Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.

Background: Melanoma is one of the most immunogenic malignancies, yet resistance to immune checkpoint inhibitors (ICIs) remains a major obstacle to durable therapeutic success. Emerging evidence indicates that aging-related processes, including cellular senescence and immunosenescence, can reshape the tumor microenvironment (TME) to favor immune evasion and disease progression. Senescent melanoma and stromal cells secrete a senescence-associated secretory phenotype (SASP) that alters immune cell recruitment and function, while immunosenescence leads to diminished cytotoxic responses and the accumulation of dysfunctional or suppressive immune subsets.

View Article and Find Full Text PDF

Tumor microenvironment-responsive CA@ZIF-8/MnO nanoreactor for self-reinforcing cascade chemodynamic therapy and immunomodulation.

Colloids Surf B Biointerfaces

August 2025

State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.

Chemodynamic therapy (CDT), which utilizes endogenous hydrogen peroxide (HO) to generate hydroxyl radicals (OH) via Fenton-like reactions, faces critical limitations in clinical translation, including insufficient intratumoral HO levels and glutathione (GSH)-mediated ROS scavenging. To address these challenges, we developed a tumor microenvironment (TME)-responsive nanoreactor, CA@ZIF-8/MnO (CZM), integrating dual functionalities of GSH-depleting and HO self-supplying for cascade-amplified CDT. The ZIF-8 framework serves as a biodegradable carrier for chlorogenic acid (CA), which converts superoxide (O) into HO, while the MnO shell depletes GSH to yield Mn, a Fenton-like catalyst.

View Article and Find Full Text PDF

Although checkpoint immunotherapy has primarily focused on CD8⁺ T cells, emerging evidence highlights an important role for cytotoxic CD4⁺ T cells in mediating therapeutic responses. However, research on the functional properties of cytotoxic CD4⁺ T cells in the context of immunotherapy is still at an early stage and remains insufficiently defined. Utilizing single-cell RNA-sequencing datasets obtained from metastatic melanoma patients treated with checkpoint inhibitors targeting PD-1 and/or CTLA-4, we performed transcriptomic profiling of conventional CD4⁺ T cells, excluding proliferative and regulatory (FOXP3⁺) subsets, and compared responders and non-responders as distinct groups.

View Article and Find Full Text PDF

Inhalable Nanovaccine Based on Bioengineered Bacteria-Derived Membrane Vesicles Against Lung Metastasis.

Adv Mater

September 2025

Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China.

Lung metastases pose a challenge in cancer treatment due to the lung's vascular network and immunosuppressive microenvironment. Conventional subcutaneous vaccines typically fail to elicit localized immune responses at metastatic sites. To address this, an inhalable nanovaccine, BMVax (bacterial membrane-based vaccine), is developed using bacterial membrane vesicles from engineered E.

View Article and Find Full Text PDF

Objective: Pembrolizumab is a key drug in the immunotherapy of endometrial cancer (EC) and has improved the prognosis to some extent. However, adverse drug events (ADEs) have hindered the achievement of expected therapeutic outcomes in EC. This study, therefore, aims to investigate the ADEs of pembrolizumab using the FAERS database, offering new insights for clinical practice in EC treatment.

View Article and Find Full Text PDF