82
Views
0
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

Self-Amplified pH/ROS Dual-Responsive Co-Delivery Nano-System with Chemo-Photodynamic Combination Therapy in Hepatic Carcinoma Treatment

ORCID Icon, ORCID Icon, , , , ORCID Icon, , & ORCID Icon show all
Pages 3737-3751 | Received 19 Dec 2023, Accepted 12 Mar 2024, Published online: 24 Apr 2024
 

Abstract

Background

Chemo-photodynamic combination therapy has demonstrated significant potential in the treatment of cancer. Triptolide (TPL), a naturally derived anticancer agent, when combined with the photosensitizer Chlorin e6 (Ce6), has shown to provide enhanced anti-tumor benefits. However, the development of stimuli-responsive nanovehicles for the co-delivery of TPL and Ce6 could further enhance the efficacy of this combination therapy.

Methods

In this study, we synthesized a pH/ROS dual-responsive mPEG-TK-PBAE copolymer, which contains a pH-sensitive PBAE moiety and a ROS-sensitive thioketal (TK) linkage. Through a self-assembly process, TPL and Ce6 were successfully co-loaded into mPEG-TK-PBAE nanoparticles, hereafter referred to as TPL/Ce6 NPs. We evaluated the pH- and ROS-sensitive drug release and particle size changes. Furthermore, we investigated both the in vitro suppression of cellular proliferation and induction of apoptosis in HepG2 cells, as well as the in vivo anti-tumor efficacy of TPL/Ce6 NPs in H22 xenograft nude mice.

Results

The mPEG-TK-PBAE copolymer was synthesized through a one-pot Michael-addition reaction and successfully co-encapsulated both TPL and Ce6 by self-assembly. Upon exposure to acid pH values and high ROS levels, the payloads in TPL/Ce6 NPs were rapidly released. Notably, the abundant ROS generated by the released Ce6 under laser irradiation further accelerated the degradation of the nanosystem, thereby amplifying the tumor microenvironment-responsive drug release and enhancing anticancer efficacy. Consequently, TPL/Ce6 NPs significantly increased PDT-induced oxidative stress and augmented TPL-induced apoptosis in HepG2 cells, leading to synergistic anticancer effects in vitro. Moreover, administering TPL/Ce6 NPs (containing 0.3 mg/kg of TPL and 4 mg/kg of Ce6) seven times, accompanied by 650 nm laser irradiation, efficiently inhibited tumor growth in H22 tumor-bearing mice, while exhibiting lower systemic toxicity.

Conclusion

Overall, we have developed a tumor microenvironment-responsive nanosystem for the co-delivery of TPL and Ce6, demonstrating amplified synergistic effects of chemo-photodynamic therapy (chemo-PDT) for hepatocellular carcinoma (HCC) treatment.

Abbreviations

Ce6, chlorin e6; TPL, triptolide; mPEG-TK-PBAE, methoxy poly (ethylene glycol)-thioketals-poly (β-amino esters); PDT, photodynamic therapy; 1O2, cytotoxic oxygen; ROS, reactive oxygen species; HCC, hepatocellular Carcinoma; EPR, enhanced permeability and retention effect; GPC, gel permeation chromatography; HPLC, high-performance liquid chromatography; CLSM, confocal laser scanning microscopy; TEM, transmission electron microscopy; FTIR, Fourier transform infrared spectroscopy; H&E, hematoxylin and eosin; ALT, Alanine aminotransferase; AST, aspartate aminotransferase; BUN, urea nitrogen; Cr, creatinine; MMP, mitochondrial membrane potential.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (No.81973662) and Innovation Team and Talents Cultivation Program of National Chinese Medicine (No: ZYYCXTD-D-202209).

Disclosure

The authors report no conflicts of interest in this work.