Improved Antitumor Efficacy of a Dextran-based Docetaxel-coupled Conjugate against Triple-Negative Breast Cancer.

Curr Drug Deliv

National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate Based Medicine, Shandong University, Qingdao, Shandong 266237, China.

Published: January 2024

Background: Most chemotherapeutic agents are characterized by poor water solubility and non-specific distribution. Polymer-based conjugates are promising strategies for overcoming these limitations.

Objective: This study aims to fabricate a polysaccharide, dextran-based, dual-drug conjugate by covalently grafting docetaxel (DTX) and docosahexaenoic acid (DHA) onto the bifunctionalized dextran through a long linker, and to investigate the antitumor efficacy of this conjugate against breast cancer.

Methods: DTX was firstly coupled with DHA and covalently bounded with the bifunctionalized dextran (100 kDa) through a long linker to produce a conjugate dextran-DHA-DTX (termed C-DDD). Cytotoxicity and cellular uptake of this conjugate were measured . Drug biodistribution and pharmacokinetics were investigated through liquid chromatography/mass spectrometry analysis. The inhibitory effects on tumor growth were evaluated in MCF-7- and 4T1-tumor-bearing mice.

Results: The loading capacity of the C-DDD for DTX was 15.90 (weight/weight). The C-DDD possessed good water solubility and was able to self-assemble into nanoparticles measuring 76.8 ± 5.5 nm. The maximum plasma concentration and area under the curve (0-∞) for the released DTX and total DTX from the C-DDD were significantly enhanced compared with the conventional DTX formulation. The C-DDD selectively accumulated in the tumor, with limited distribution was observed in normal tissues. The C-DDD exhibited greater antitumor activity than the conventional DTX in the triplenegative breast cancer model. Furthermore, the C-DDD nearly eliminated all MCF-7 tumors in nude mice without leading to systemic adverse effects.

Conclusion: This dual-drug C-DDD has the potential to become a candidate for clinical application through the optimization of the linker.

Download full-text PDF

Source
http://dx.doi.org/10.2174/1567201820666230622105503DOI Listing

Publication Analysis

Top Keywords

antitumor efficacy
8
breast cancer
8
water solubility
8
bifunctionalized dextran
8
long linker
8
c-ddd
8
conventional dtx
8
dtx
7
conjugate
5
improved antitumor
4

Similar Publications

A Integrated Molecule Based on Ferritin Nanoplatforms for Inducing Tumor Ferroptosis with the Synergistic Photo/Chemodynamic Treatment.

ACS Appl Mater Interfaces

January 2025

Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, Guangdong 523058, China.

Ferroptosis combined with photodynamic therapy (PDT) has emerged as a powerful approach to induce cancer cell death by producing and accumulating lethal reactive oxygen species (ROS) in the tumor microenvironment (TME). Despite its efficacy and safety, challenges persist in delivering multiple drugs to the tumor site for enhanced antitumor efficacy and improved tissue targeting. Hence, we designed a method of inducing ferroptosis through laser-mediated and human homologation-specific efficient activation, which is also a ferroptosis therapy with higher safety through ROS-mediated.

View Article and Find Full Text PDF

Treatment options for recurrent high-risk non-muscle-invasive bladder cancer (HR NMIBC) and muscle-invasive bladder cancer (MIBC) are limited, highlighting a need for clinically effective, accessible, and better-tolerated alternatives. In this review we examine the clinical development program of TAR-200, a novel targeted releasing system designed to provide sustained intravesical delivery of gemcitabine to address the needs of patients with NMIBC and of those with MIBC. We describe the concept and design of TAR-200 and the clinical development of this gemcitabine intravesical system in the SunRISe portfolio of studies.

View Article and Find Full Text PDF

PD1-TLR10 fusion protein enhances the antitumor efficacy of CAR-T cells in colon cancer.

Int Immunopharmacol

January 2025

TriArm Therapeutics, Niudun Road 200, 201203 Shanghai, China. Electronic address:

Background: The immunosuppressive microenvironment negatively affects the efficacy of chimeric antigen receptor T (CAR-T) cells in solid tumors. Fusion protein that combining extracellular domain of inhibitory checkpoint protein and the cytoplasmic domain of stimulatory molecule may improve the efficacy of CAR-T cells by reversing the suppressive signals.

Methods: To generate optimal PD1-TLR10 fusion proteins, PD1 extracellular domain and TLR10 intracellular domain were connected by transmembrane domain from PD1, CD28, or TLR10, respectively.

View Article and Find Full Text PDF

Tamoxifen (TAM) is employed to treat premenopausal ER-positive breast cancer patients, but TAM resistance is the main reason affecting its efficacy. Thus, addressing TAM resistance is crucial for improving therapeutic outcomes. This study explored the potential role of Tinagl1, a secreted extracellular matrix protein, whose expression is compromised in TAM-resistant MCF-7 breast cancer cells (MCF-7R).

View Article and Find Full Text PDF

BI 1703880, a novel STimulator of INterferon Genes (STING) agonist, has demonstrated preclinical antitumor activity. As STING activation can upregulate programmed death ligand 1 and human leukocyte antigen in tumor cells, a combination of BI 1703880 and an anti-programmed cell death protein 1-antibody, such as ezabenlimab, may improve efficacy. This first-in-human phase Ia study (NCT05471856) is evaluating BI 1703880 plus ezabenlimab in patients with advanced solid tumors.

View Article and Find Full Text PDF

Want AI Summaries of new PubMed Abstracts delivered to your In-box?

Enter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!