Targeting mesenchymal monocyte-derived macrophages to enhance the sensitivity of glioblastoma to temozolomide by inhibiting TNF/CELSR2/p65/Kla-HDAC1/EPAS1 axis.

J Adv Res

The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer

Published: May 2025


Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Introduction: Temozolomide (TMZ) resistance poses a significant challenge to the treatment of aggressive and highly lethal glioblastomas (GBM). Monocyte-derived Macrophages (MDM) within the tumor microenvironment are key factors contributing to TMZ resistance in GBM. Lactate-mediated histone lysine lactylation (Kla) plays a crucial role in the regulation of tumor progression. However, the mechanism through which MDM-induced Kla expression promotes TMZ resistance in GBM remains unclear.

Objectives: The objective of this study s to identify a subtype of MDM with therapeutic potential target and to elucidate the mechanisms through which this subtype of MDM contributes to tumor malignant progression and TMZ resistance.

Methods: We integrated single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics data to evaluate whether mesenchymal (MES) MDM is associated with poor prognosis. By establishing a subtype model of GBM cells for the first time, we validated the mechanism by which MES-MDM promotes subtype conversion of tumor cells. Using patient-derived GBM organoids and an intracranial orthotopic GBM model, we demonstrated that targeting MES-MDM increased GBM sensitivity to TMZ treatment.

Results: We identified a novel MDM subtype, MES-MDM, in the hypoxic niches of the perinecrotic region characterized by high TREM1 expression, which fueled GBM progression. Hypoxia drived MES-MDM signatures by activating ATF3 transcription. MES-MDM facilitated the transition from the NPC to the MES subtype in GBM cells, in which Histone Deacetylase 1 (HDAC1) Kla, induced by the TNF-CELSR2/p65 signaling pathway, promoted this conversion, thereby promoting TMZ resistance. Targeting MES-MDM with TREM1 inhibitory peptides amplified TMZ sensitivity, offering a potential strategy for overcoming resistance to therapy in GBM. Targeting TREM1 enhanced the effectiveness of anti-PD-1 immunotherapy.

Conclusion: This study provides a potential therapeutic strategy for patients with MES-subtype GBM by targeting MES-MDM in combination with TMZ or PD-1 antibody treatment.

Download full-text PDF

Source
http://dx.doi.org/10.1016/j.jare.2025.05.032DOI Listing

Publication Analysis

Top Keywords

tmz resistance
16
targeting mes-mdm
12
gbm
11
monocyte-derived macrophages
8
tmz
8
resistance gbm
8
subtype mdm
8
gbm cells
8
gbm targeting
8
mes-mdm
7

Similar Publications

More than a third of patients with glioblastoma experience tumor progression during adjuvant therapy. In this study, we performed a high-throughput drug repurposing screen of FDA-approved agents capable of crossing the blood-brain barrier in order to find agents to counteract acquired or inherent glioma cell resistance to temozolomide-associated cytotoxicity. We identified the cholesterol processing inhibitor, lomitapide, as a potential chemosensitizer in glioblastoma.

View Article and Find Full Text PDF

Background: Standard treatment for glioblastoma includes chemotherapy, alkylating agents such as temozolomide (TMZ); however, MGMT resistance leads to recurrence. Demethoxycurcumin (DMC) has been reported to inhibit cancer cell growth, induce apoptosis, and prevent metastasis in different cancer models. We investigated the DMC-induced apoptosis and autophagy via inhibition of the AKT/mTOR pathway in human glioma U87MG and T98G cell lines.

View Article and Find Full Text PDF

Dual-action nanotherapy: Temozolomide-loaded, anti-PD-L1 scFv-functionalized lipid nanocarriers for targeted glioblastoma therapy.

Eur 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 PDF

Novel Thioredoxin reductase 1 inhibitor BS1801 relieves treatment resistance and triggers endoplasmic reticulum stress by elevating reactive oxygen species in glioma.

Redox Biol

August 2025

Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, No.119 South 4th Ring Road West, Beijing, China; Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China; Beijing Engineering Research Center of Target

Glioma patients will inevitably develop resistance to temozolomide (TMZ) leading to tumor recurrence. By comparing genomic differences between primary and recurrent glioma patients, Thioredoxin reductase 1 (TrxR1) was identified as a crucial role in TMZ resistance. Glioma cells elevate the expression level of TXNRD1 to against TMZ-induced reactive oxygen species (ROS), thereby conferring TMZ resistance.

View Article and Find Full Text PDF

Temozolomide-Derived Therapeutic Strategies to Overcome Resistance in Glioblastoma.

J Med Chem

September 2025

Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas 79968, United States.

Glioblastoma multiforme (GBM) accounts for nearly half of malignant CNS tumors and has a dismal 5-year survival rate of 5.5%. The current standard of care comprises maximal surgical resection, followed by radiotherapy with concurrent temozolomide (TMZ) and subsequent adjuvant TMZ chemotherapy.

View Article and Find Full Text PDF