ORIGINAL RESEARCH article
Front. Immunol.
Sec. Cancer Immunity and Immunotherapy
Volume 16 - 2025 | doi: 10.3389/fimmu.2025.1684611
This article is part of the Research TopicDeciphering Membrane and Intracellular Protein Targets in Cancer: Subcellular Mechanisms, Drug Discovery, and Translational InterventionsView all 3 articles
Selenium Nanoparticle-Delivered MDM2 Inhibitor Reactivates p53 and Reprograms Tumor Immune Microenvironment in Colorectal Cancer
Provisionally accepted- 1The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- 2The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
Select one of your emails
You have multiple emails registered with Frontiers:
Notify me on publication
Please enter your email address:
If you already have an account, please login
You don't have a Frontiers account ? You can register here
Background: Colorectal cancer (CRC) remains a major global health challenge, with limited immunotherapy efficacy in microsatellite stable (MSS) tumors that comprise ~85% of cases. The p53 tumor suppressor pathway, frequently inactivated through the mouse double minute 2 homolog (MDM2) overexpression in wild-type tumor protein 53(TP53) tumors, represents a promising therapeutic target for both direct antitumor effects and immune modulation. Methods: We developed selenium nanoparticles loaded with the MDM2-targeting peptide inhibitor MI (Se@MI) using a one-pot synthesis approach. The nanoparticles were characterized by transmission electron microscopy, dynamic light scattering, and UV-Vis spectroscopy. In vitro assays included MTT cytotoxicity evaluation, cellular uptake by flow cytometry, RNA-seq, and Western blotting of p53-pathway/apoptosis markers. Antitumor efficacy was evaluated in CT26 murine colorectal cancer models, with mechanistic studies including transcriptomic analysis, immunohistochemistry, and flow cytometry. Safety profiles were assessed through body weight monitoring, hematological analysis, histopathological examination of major organs, and serum biomarker evaluation. Results: Se@MI nanoparticles demonstrated uniform spherical morphology (45-50nm diameter) and displayed a positive zeta potential (+24.69 mV), indicative of favorable colloidal dispersibility. Enhanced cellular uptake (74.3% positive cells) and potent cytotoxicity (IC₅₀ = 1.00 μM) were observed in CT26 cells. Transcriptomic analysis revealed significant activation of p53 signaling pathways (NES = 1.504, P = 0.029) and protein analyses confirmed induction of p21, PUMA, Bax, and cleaved caspase-3. In vivo, Se@MI treatment achieved 72.23% tumor growth inhibition, significantly outperforming controls. Mechanistically, Se@MI restored p53 function by disrupting MDM2-p53 interactions, inducing apoptosis and cell cycle arrest. Importantly, Se@MI reprogrammed the tumor immune milieu through increased infiltration of CD8+ T cell and cytotoxic function while suppressing regulatory T cells. Comprehensive safety evaluation revealed excellent biocompatibility with no adverse effects on body weight, hematological parameters, organ histology, inflammatory cytokines, or hepatic/renal function markers. Conclusions: Se@MI represents a novel nanomedicine strategy that combines direct p53 pathway reactivation with immune microenvironment modulation. This dual mechanism of action offers a potential strategy to boost immunotherapeutic outcomes in colorectal carcinoma and may help overcome resistance mechanisms to immune checkpoint blockade
Keywords: Selenium nanoparticles, p53 reactivation, antitumor, Tumor Microenvironment, peptide
Received: 12 Aug 2025; Accepted: 20 Oct 2025.
Copyright: © 2025 You, Feng, Guo, Yan and Yan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
* Correspondence:
Jin Yan, yanjin19920602@xjtu.edu.cn
Siqi Yan, yansiqi92@xjtu.edu.cn
Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.