Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Artesunate (ART), an antimalarial drug, has been identified as a ferroptotic agent, inducing the generation of reactive oxygen species (ROS) and lipid peroxidation, which, in turn, activate endoplasmic reticulum (ER) stress responses and promote mitochondrial-dependent apoptosis. In our previous studies, we demonstrated that ART enhances tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through crosstalk between the ER stress-mediated signal pathway and the Bid-Bax mitochondrial apoptotic cascade. To further explore the mechanisms underlying ferroptotic-apoptotic crosstalk and evaluate the potential of intra-arterial drug-eluting microspheres for targeted tumor therapy, we developed artesunate-eluting microspheres (ART-EMs) and investigated the tumoricidal efficacy of ART-EMs combined with TRAIL. Our findings reveal that the combined ART-EMs with TRAIL (AT) treatment synergistically enhances cancer cell death. Specifically, we observed increased apoptosis in HCT116 and BxPC-3 cell lines, accompanied by notable morphological changes and enhanced cytotoxicity. Importantly, our results demonstrate that the pro-apoptotic proteins Bid and Bax play essential roles in driving synergistic apoptosis during AT treatment. Furthermore, the contrasting apoptotic responses between AT treatment and the chemotherapeutic agent mitomycin C's dependence on p53-Bak-associated pathways underscore the differential activation of intrinsic apoptosis pathways across cancer cell lines. This study provides deeper insight into the roles of Bak and Bax in orchestrating apoptosis, offering potential strategies for more effective cancer treatments.

Download full-text PDF

Source
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC12149531PMC
http://dx.doi.org/10.34133/bmr.0217DOI Listing

Publication Analysis

Top Keywords

tumoricidal efficacy
8
cell death
8
cancer cell
8
cell lines
8
apoptosis
6
efficacy artesunate-eluting
4
artesunate-eluting microsphere
4
microsphere differential
4
differential role
4
role bax/bak
4

Similar Publications

Enhance therapeutic efficacy of BiTE (HER2/CD3) for HER2- positive tumors through expression.

Int J Pharm X

December 2025

Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, China, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.

Bispecific T-cell engagers (BiTEs) are small-molecule antibodies that exhibits potent tumoricidal activity but suffer from a short plasma half-life. Mesenchymal stromal cells (MSCs) represent promising delivery vehicles for sustained therapeutic protein expression. In this study, we used human umbilical cord blood-MSCs (hUC-MSCs) as a delivery system to to secrete HER2/CD3 BiTE antibodies, thereby addressing the pharmacokinetic limitations of conventional BiTE therapies.

View Article and Find Full Text PDF

A Hybrid Protein-Oxygen Nanomedicine Overcomes Osimertinib Resistance in NSCLC via HIF-1α/VEGF/EGFR Inhibition.

Int J Nanomedicine

September 2025

Department of Thoracic Surgery, Tenth Affiliated Hospital, Southern Medical University (Dongguan People's hospital), Dongguan, Guangdong, 523059, People's Republic of China.

Purpose: Osimertinib, established as the frontline treatment for advanced non-small cell lung cancer (NSCLC), can effectively prolong progression-free survival. However, it faces the problem of reduced treatment persistence due to acquired drug resistance. Meanwhile, tumor hypoxia is also a key driver of drug resistance.

View Article and Find Full Text PDF

The Biophysics of Flash Radiotherapy: Tools for Measuring Tumor and Normal Tissues Microenvironment.

Antioxidants (Basel)

July 2025

Departments of Machine Learning and Radiation Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA.

Ultra-high dose rate radiotherapy known as Flash radiotherapy (FLASH-RT) offers tremendous opportunities to improve the therapeutic ratio of radiotherapy by sparing the normal tissue while maintaining similar tumoricidal efficacy. However, the underlying biophysical basis of the FLASH effect remains under active investigation with several proposed mechanisms involving oxygen depletion, altered free-radical chemistry, and differential biological responses. This article provides an overview of available experimental and computational tools that can be utilized to probe the tumor and normal tissue microenvironment.

View Article and Find Full Text PDF

IL-7-PD-L1 nano-antibody mediated "zipper" effect augments the tumoricidal activity of tumor-infiltrating lymphocytes.

Exp Hematol Oncol

August 2025

R&D, Qingdao Sino‑cell Biomedicine Co., Ltd., 1 Changcheng South Road, Chengyang, Qingdao, 266000, Shandong, China.

Cancer represents a pressing global health concern, characterized by a substantial number of unmet clinical needs. Cell therapy has emerged as a promising and efficacious approach for cancer treatment, particularly tumor-infiltrating lymphocytes (TILs), which have demonstrated remarkable improvements in patients' overall survival rates across various clinical studies. However, the tumor microenvironment exerts a adverse effect on TILs, leading to their rapid exhaustion and functional disorder.

View Article and Find Full Text PDF

Lifespan-Regulated CAR-Macrophages from Myeloid Progenitors for Enhanced Colorectal Cancer Therapy.

Adv Sci (Weinh)

August 2025

State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.

Adoptive cell therapies for solid tumors face persistent challenges from poor tumor infiltration and immunosuppressive microenvironment. To overcome these limitations, a clinically scalable platform is developed to generate chimeric antigen receptor macrophages (CAR-HMs) from tamoxifen-regulated immortalized Hoxb8-transduced myeloid progenitors, achieving >95% CAR transduction efficiency and 60-fold expansion within 10 days. Engineered with a colorectal cancer-specific anti-carcinoembryonic antigen (CEA) CAR, these FcγRI-CAR-HMs demonstrated potent tumoricidal activity (>80% CRC cell lysis in vitro), deep tissue penetration (>100 µm in 3D tumor spheroids), and significant therapeutic efficacy (≈89% tumor regression in vivo).

View Article and Find Full Text PDF