98%
921
2 minutes
20
Background: PD-L1 blockade has been found to be effective in treating multiple malignancies. Combined therapy is proposed to provide better therapeutic effects. Cordycepin, a prominent bioactive compound found in cordyceps, can inhibit the development of various cancers.
Purpose: This study aimed to determine the efficacy of combined anti-PD-L1 antibody and cordycepin in tumor treatment.
Methods: A single-cell RNA sequencing was used to analyze the mechanism of combined treatment.
Results: Combination therapy of anti-PD-L1 and cordycepin significantly inhibited tumor growth by regulating the T cell ratio and improving the function of CD8T cells. Furthermore, cordycepin promoted the reprogramming of type-II macrophages into type-I macrophages, a process confirmed through flow cytometry analysis of the underlying mechanism.
Conclusion: Our findings demonstrate that the combination of anti-PD-L1 and cordycepin effectively suppressed tumor growth by regulating the proportion of T cells and reprograming type-II macrophages.
Download full-text PDF |
Source |
---|---|
http://dx.doi.org/10.1016/j.ejphar.2024.177089 | DOI Listing |
Oncol Res
September 2025
Department of Biliary-Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
Hepatocellular carcinoma (HCC) is a highly aggressive malignancy, largely driven by an immunosuppressive tumor microenvironment (TME) that facilitates tumor growth, immune escape, and resistance to therapy. Although immunotherapy-particularly immune checkpoint inhibitors (ICIs)-has transformed the therapeutic landscape by restoring T cell-mediated anti-tumor responses, their clinical benefit as monotherapy remains suboptimal. This limitation is primarily attributed to immunosuppressive components within the TME, including tumor-associated macrophages, regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSCs).
View Article and Find Full Text PDFEur J Pharm Sci
September 2025
Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA, 02129, USA. Electronic address:
Glioblastoma (GBM) is a highly malignant brain tumor with limited treatment options and poor prognosis. GBM exhibits resistance to conventional therapies, including temozolomide (TMZ), radiotherapy, and immunotherapy, partly due to immunosuppressive mechanisms such as programmed death-ligand 1 (PD-L1) overexpression. To address these challenges, we developed TMZ-loaded nanostructured lipid carriers (NLCs) conjugated with anti-PD-L1 single-chain variable fragments (scFv) for dual chemo-immunotherapy.
View Article and Find Full Text PDFBackground: Triggering receptor expressed on myeloid cells 1 (TREM-1), an inflammation amplifier, is an emerging target in inflammation and oncology.
Objective: To test my hypothesis that pan-TREM-1 and macrophage-restricted TREM-1 blockades may differ in their efficacy in cancer and other inflammatory diseases.
Methods: Ligand-independent TREM-1 inhibitory peptides GF9 and GA31 (the latter in a form of macrophage-targeted lipopeptide complexes, GA31-LPC) were used as pan-TREM-1 and macrophage-restricted TREM-1 inhibitors, respectively, to test the hypothesis in multiple animal models of cancer, sepsis, pulmonary inflammation and fibrosis.
Front Immunol
September 2025
Department of Experimental Pediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany.
Tumors frequently evade immune destruction by impairing cytotoxic CD8 T-cell responses, highlighting the need for strategies that restore T-cell functionality. Here, we identify SLAMF7 (CD319) as a key enhancer of human CD8 T-cell responses against tumors. SLAMF7 expression is induced by pro-inflammatory signals such as IL-12 and CD28 co-stimulation.
View Article and Find Full Text PDFCancer Med
September 2025
Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
Background: Although immune checkpoint inhibitors (ICIs) have markedly improved first-line management of non-small cell lung cancer (NSCLC), many tumors eventually escape control after anti-PD-(L)1 therapy, leaving a clear therapeutic gap. Preclinical studies and preliminary clinical data suggest that coupling ICIs with anti-angiogenesis therapy can yield complementary antitumor effects. Consequently, we launched this investigation to evaluate the therapeutic benefit and tolerability of sintilimab, a PD-(L)1-blocking monoclonal antibody, together with the oral multi-target anti-angiogenesis agent anlotinib in metastatic NSCLC individuals experiencing progression after first-line PD-(L)1 inhibition.
View Article and Find Full Text PDF