1
|
Zhang J, Zhang Q, Li X, Wei Y, Qiu M, Yang H, Sun X. Prominent supramolecular systems for cancer Therapy: From structural design to tailored applications. Eur J Med Chem 2025; 294:117754. [PMID: 40378574 DOI: 10.1016/j.ejmech.2025.117754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/19/2025]
Abstract
Supramolecular materials represent a powerful class of platforms in cancer diagnosis and therapy, owing to their dynamic architectures, stimuli responsiveness, and high biocompatibility. This review focused on three representative categories-Pillarene-based systems, virus-mimetic nanoparticles (VMNs), and metal-organic frameworks (MOFs)-each offering unique structural and functional properties. Pillarene-based assemblies enable precise host-guest interactions, by being classified into amphiphilic, ionic, and chiral varieties, the robust drug loading and controlled release capabilities of the Pillarene family were emphasized. At the same time, the VMNs, including virus-like particles and virosomes, show power in cancer cell targeting and membrane penetration by emulating natural viral architectures. By discussing the fabrication and application of single-metallic, multi-metallic, and composite MOFs, their potential in multimodal diagnosis and therapy was revealed. In addition, other supramolecular categories, such as cyclodextrin and dendrimers, were introduced as well. We highlighted representative approaches and emerging methods, and comparative perspectives with traditional nanocarriers were included. A critical evaluation of pharmacokinetic behaviors, biosafety concerns, and translational limitations was also proposed, aiming to guide future research in supramolecular cancer nanomedicine. Through an integrative and forward-looking analysis, this review provided a comprehensive framework for understanding and designing supramolecular systems for precision oncology. These emerging nanotechnologies hold promise to reshape cancer medicine by enabling adaptive, targeted, and multifunctional therapeutic strategies.
Collapse
Affiliation(s)
- Jiawei Zhang
- The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, China; School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang, China
| | - Qingya Zhang
- The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, China; School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenyang, China
| | - Xiaojia Li
- Teaching Center for Basic Medical Experiment, China Medical University, No.77, Puhe Road, Shenyang, China
| | - Yixuan Wei
- Teaching Center for Basic Medical Experiment, China Medical University, No.77, Puhe Road, Shenyang, China
| | - Min Qiu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang, China.
| | - Xiaoting Sun
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenyang, China.
| |
Collapse
|
2
|
Lin C, Sun J, Yang Y, Pan X, Wang S, Li X, Zhang Y, Gao H, Gan C. Peptide-based nanoassembly enhances ferroptosis in cancer to overcome paclitaxel resistance. J Control Release 2025; 384:113895. [PMID: 40441495 DOI: 10.1016/j.jconrel.2025.113895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 05/05/2025] [Accepted: 05/25/2025] [Indexed: 06/02/2025]
Abstract
The development of chemotherapy resistance poses a major challenge in cancer therapy. Ferroptosis, a unique type of cell death, offers a promising strategy to combat this resistance. Herein, a peptide-based nanoassembly (PTX@CPG) consisting of paclitaxel (PTX), chlorin e6 (Ce6), and FFVLKPLGLAGK-(PEG8)3 was constructed to promote ferroptosis through reactive oxygen species (ROS) accumulation and overcome chemoresistance. Specifically, the small-sized PTX@CPG nanoparticles effectively penetrate tumors, where the microenvironment-responsive peptide is selectively cleaved by the high expression of matrix metalloproteinase 2. This process facilitates the targeted release of PTX and its reassembly into nanofibers, improving the tumor retention of Ce6 and enhancing its cellular uptake. The synergistic therapeutic effects of PTX in combination with photodynamic therapy on triple-negative breast cancer cells were validated through both in vitro and in vivo experiments. Impressively, upon laser irradiation, PTX@CPG significantly increased ROS production, thereby amplifying the ferroptosis-inducing effects of PTX. Moreover, ferroptosis triggered by PTX@CPG with laser reduced the levels of P-glycoprotein and glutathione peroxidase 4, contributing to the alleviation of chemoresistance. Overall, PTX@CPG with laser demonstrated effective spatial targeting and drug retention, enhancing ferroptosis through ROS accumulation and showcasing a promising approach for overcoming chemotherapy resistance in cancer therapy.
Collapse
Affiliation(s)
- Congcong Lin
- College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| | - Jiamin Sun
- College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yun Yang
- College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xinyao Pan
- College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Shiyu Wang
- College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xiaoyang Li
- College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yan Zhang
- College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China.
| | - Chunli Gan
- College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
3
|
Wu S, Xia X, Zhou R, Zhao H. Hydrogel-enabled ROS-GSH modulation for sustained copper-mediated chemodynamic therapy of oral squamous cell carcinoma. J Control Release 2025; 383:113772. [PMID: 40280240 DOI: 10.1016/j.jconrel.2025.113772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Copper ion (Cu2+) has been revealed to be involved in the occurrence and development of oral squamous cell carcinoma (OSCC), making copper-mediated chemodynamic therapy (Cu-CDT) a promising treatment strategy for OSCC by elevating Cu2+ levels to generating a large amount of reactive oxygen species (ROS). However, the excessive reduced glutathione (GSH) in the tumor microenvironment can scavenge the ROS generated by Cu-CDT. While the directional co-delivery of Cu2+ and GSH-depleting agents shows promise for Cu-CDT in OSCC therapy, their rapid metabolism and the superficial nature of OSCC lesions necessitate tailored drug formulations to ensure effective bioavailability. To counteract this challenge, this work proposed a practical hydrogel-supported ROS-GSH regulation strategy, which involves the on-demand design of a copper ion-crosslinked guanosine-based hydrogel (GCD) containing dimethyl fumarate (DMF, which conjugates with GSH for consumption). It can directionally and sustainably co-deliver Cu2+ and DMF to OSCC lesions under mildly acidic pH conditions, thereby enhancing Cu-CDT efficiency through improved Cu2+ utilization and DMF-driven GSH depletion. As anticipated, the strategy sustains the generation of hydroxyl radicals, effectively inducing apoptosis and suppressing cell proliferation in CAL-27 cells, which consequently inhibits the growth of OSCC tumors. Therefore, this work highlights the GCD hydrogel's great potential as a promising Cu-CDT therapeutic platform.
Collapse
Affiliation(s)
- Shihong Wu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xin Xia
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Guo C, Lin L, Wang Y, Jing J, Gong Q, Luo K. Nano drug delivery systems for advanced immune checkpoint blockade therapy. Theranostics 2025; 15:5440-5480. [PMID: 40303342 PMCID: PMC12036873 DOI: 10.7150/thno.112475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been widely utilized in the first-line therapy of various types of cancer. However, immune-related adverse events (irAEs) and resistance to ICIs remain intractable challenges for immune checkpoint blockade (ICB) therapy during clinic treatment. Nano drug delivery systems (NDDSs) have shown promising potential to improve anticancer efficacy and reduce side effects of small molecular drugs. The combination of nanotechnology and ICB provides new opportunities to overcome the challenges of immunotherapy. Nanoplatforms have been employed for direct delivery of ICIs, and they are preferred vehicles for combination therapy of ICIs and other therapeutic agents. In this review, the strategies of using NDDSs for advancing ICB therapy in recent years are surveyed, emphasizing the employment of NDDSs for combination treatment by ICIs and other agents to manipulate antitumor immunity. Analysis of current strategies for applying NDDSs for ICB leads to future research directions and development trends.
Collapse
Affiliation(s)
- Chenqi Guo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Lin
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yihan Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- West China School of Medicine, Chengdu 610041, China
| | - Jing Jing
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Rehabilitation Therapy, Breast Center, Institute of Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and molecular imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
5
|
Feng S, Xu Q, Liu B, He Y, Song L, Zhao Q, Wang S. Modulating Intracellular Autophagy and Macropinocytosis for Increased Neighboring Drug Delivery. ACS NANO 2025; 19:13175-13190. [PMID: 40162609 DOI: 10.1021/acsnano.4c18465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Neighboring effects provided a valuable direction for in-depth penetration of nanoparticles into tumors. However, the uncontrollable drug transcytosis and limited drug uptake hindered by viscous cancer-associated fibroblasts (CAFs) greatly limit their in-depth penetration. Here, we proposed and demonstrated that intracellular autophagosomes could carry the remaining drugs to neighboring cells, and the enhanced macropinocytosis played a major role in neighboring delivery. To enhance the autophagosome-based neighboring delivery, Ca2+-doped polydopamine was prepared to load GLS1 inhibitor CB-839 and modified glutamine (839/CG) for triggering macropinocytosis-based active cells uptake. After Ca2+-release caused lysosome damage, 839/CG escaped from lysosomes and hindered the autophagosome maturation. Then, Ca2+-induced endoplasmic reticulum oscillations and glutamine starvation both increased and blocked autophagy flow, causing 839/CG-contained autophagosome accumulation. Meanwhile, the tumor increased its macropinocytosis in response to mTOR downregulation-induced glutamine hunger, causing "the more you eat, the hungrier you get". After tumor death, the 839/CG-contained autophagosomes were released and actively ingested by neighboring hungry tumor cells through macropinocytosis. Combined with the photothermal effect triggered CAF decrease, neighboring cells repeated the above process for in-depth tumor delivery. Also, immunogenic death enhanced the antigen presentation of DCs and infiltration of T cells, thereby inhibiting tumor growth and lung metastasis.
Collapse
Affiliation(s)
- Shuaipeng Feng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Qingqing Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Bin Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Ye He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Luming Song
- Department of Microbial and Biochemical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| |
Collapse
|
6
|
Song YH, Gu YJ, Lei Z, Li NK, Zhang YM, Yu Q, Liu Y. Fluorinated Cyclodextrin Supramolecular Nanoassembly Enables Oxygen-Enriched and Targeted Photodynamic Therapy. NANO LETTERS 2025; 25:4476-4484. [PMID: 40056123 DOI: 10.1021/acs.nanolett.5c00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2025]
Abstract
Photodynamic therapy has become a promising treatment modality against many diseases, but its dilemma-the intrinsic hypoxia of solid tumors and the high oxygen dependence for generation of cytotoxic species-has seriously hampered its practical translation. Herein a binary supramolecular nanocarrier, which is composed of fluorocarbon chain-appended β-cyclodextrin as an oxygen carrier and adamantane-grafted hyaluronic acid as a cell-targeting agent, can deliver different types of photosensitizers by multiple noncovalent interactions. Superior to the alkylated counterpart, the fluorinated amphiphilic β-cyclodextrin can spontaneously form a nanoparticulate assembly and exhibit high oxygen-enrichment performance. The obtained nanoassembly can alleviate hypoxia in the tumor microenvironment and enhance the efficacy of photodynamic therapy. Remarkable phototoxicity and minimal dark toxicity are observed in the cancer cells, and meanwhile, preferential accumulation and significant cancer ablation are realized in the tumor-bearing mice. To be envisioned, this supramolecular assembly capable of efficiently carrying oxygen can be explored as a universal platform for precise phototherapeutics.
Collapse
Affiliation(s)
- Ya-Hui Song
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, P. R. China
| | - Yi-Jun Gu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhuo Lei
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, P. R. China
| | - Nan-Kun Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, P. R. China
| | - Ying-Ming Zhang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, P. R. China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yu Liu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
7
|
Xu S, Xie X, He P, Zhu S, Li X, Chen Q, Ma X, Liang X. Nitric Oxide-Producing Multiple Functional Nanoparticle Remodeling Tumor Microenvironment for Synergistic Photodynamic Immunotherapy against Hypoxic Tumor. ACS NANO 2025; 19:6371-6387. [PMID: 39913864 DOI: 10.1021/acsnano.4c16329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
The treatment of pancreatic cancer faces significant challenges due to connective tissue hyperplasia and severe hypoxia. Unlike oxygen-dependent Type II photosensitizers, Type I photosensitizers can produce a substantial amount of reactive oxygen species, even under hypoxic conditions, making them more suitable for photodynamic therapy of pancreatic cancer. However, the dense extracellular matrix of pancreatic cancer limits the penetration efficiency of photosensitizers, and the presence of immunosuppressive cells in the tumor microenvironment reduces the therapeutic effect. To address these challenges, we designed the photoimmunotherapeutic M1@PAP nanoparticles composed of Type I photosensitizer and anti-PD-L1 siRNA (siPD-L1), which was encapsulated into M1 macrophage membrane vesicles. In this system, pyropheophorbide-a (PPA) was covalently conjugated to poly-l-arginine (Arg9). Notably, it was capable of generating sufficient superoxide anions under hypoxic conditions, thereby functioning as a Type I photosensitizer. Furthermore, Arg9 acted as a nitric oxide (NO) donor, enhancing the penetration efficiency of the nanophotosensitizer by inhibiting cancer-associated fibroblast (CAF) activation and decomposing the tumor extracellular matrix. Additionally, M1 macrophage membrane vesicles provided active targeting capabilities and reeducated immunosuppressed M2 macrophages. The reversal of immunosuppressive microenvironment further promoted the efficacy of anti-PD-L1 siRNA immunotherapy, showing great potential in synergistic photodynamic immunotherapy against hypoxic pancreatic tumor.
Collapse
Affiliation(s)
- Shuyu Xu
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xinxin Xie
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Ping He
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Shiwei Zhu
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xiaoda Li
- Peking University Health Science Center, Beijing 100191, China
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Xiaotu Ma
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xiaolong Liang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
8
|
Jiang M, Wu S, Zhang H, Shen M, Zhang Y, Zheng Z. Twin-Tail Tadpole-Shaped Ce6-Peptide Conjugate for Enhanced Photodynamic Cancer Therapy. ACS APPLIED BIO MATERIALS 2025; 8:1475-1483. [PMID: 39909401 DOI: 10.1021/acsabm.4c01696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Despite its therapeutic potential, photodynamic therapy faces several key limitations in clinical applications, including poor drug delivery and insufficient tumor selectivity. We engineered RFYFYR-Ce6-RFYFYR (R-Ce6-R), a twin-tail peptide-photosensitizer conjugate that self-assembles into nanostructures for improved cancer treatment. By incorporating arginine-rich peptide sequences, this design not only enhances cellular internalization but also promotes peroxynitrite (ONOO-) formation, amplifying the therapeutic effect. Our studies revealed that R-Ce6-R achieves 33-fold higher potency than unmodified Ce6, with an IC50 of 0.18 μM. The conjugate demonstrated selective accumulation in tumor tissue, robust ROS generation, and complete tumor regression in animal models while maintaining a favorable safety profile. These results establish R-Ce6-R as an innovative approach for advancing photodynamic therapy in cancer treatment.
Collapse
Affiliation(s)
- Mengmeng Jiang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Shaowen Wu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Hongxia Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Menghuan Shen
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yirui Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhen Zheng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
9
|
Yu H, Huang Z, Wu J, Zhao Z, Hua Y, Yang Y. Chlorin e6: a promising photosensitizer of anti-tumor and anti-inflammatory effects in PDT. Nanomedicine (Lond) 2025; 20:389-400. [PMID: 39877963 PMCID: PMC11812356 DOI: 10.1080/17435889.2025.2456450] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Photodynamic therapy (PDT) involves the activation of photosensitizers (PSs) by visible laser light at the target site to catalyze the production of reactive oxygen species, resulting in tumor cell death and blood vessel closure. The efficacy of PDT depends on the PSs, the amount of oxygen, and the intensity of the excitation laser. PSs have been extensively researched, and great efforts have been made to develop an ideal photosensitizer. Chlorin-e6 is an FDA-approved second-generation PSs that has attracted widespread research interest in the medical field, especially with respect to antitumor and anti-inflammatory activity. Chlorin-e6 possesses the advantages of a large absorption coefficient, high strength, low residue in the body, and relatively high safety and thus has promising application prospects. Here we review the use of chlorin-e6 in PDT and discuss the prospects of further development of this technology.
Collapse
Affiliation(s)
- Hairong Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Ziling Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Jiale Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Ziming Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yabing Hua
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yihua Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
Zhang L, Que RB, Ke TT, Wang C, Xie W, Sun HJ, Zheng BY, Ke MR, Huang JD, Li X. A tumor-pH-responsive phthalocyanine as activatable type I photosensitizer for improved photodynamic immunotherapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 261:113067. [PMID: 39577037 DOI: 10.1016/j.jphotobiol.2024.113067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
The development of a simple drug formulation capable of achieving both activatable type I photoreaction and tumor-responsive release of immunomodulator is crucial for advancing photodynamic immunotherapy (PDIT). Herein, we present a nanostructured photosensitizer (NP5) that is activated by the acidic tumor microenvironment to produce type I reactive oxygen species (ROS) under light irradiation and release the immunomodulator demethylcantharidin (DMC) for PDIT. The NP5 is formed by self-assembly of a versatile phthalocyanine molecule which is composed of DMC and phthalocyanine linked via a pH-responsive amide bond. NP5 produces minimal ROS under light irradiation at the condition of pH 7.4. However, NP5 can release DMC at the condition of pH 6.5 and concurrently trigger type I photoreactions. The results of in vivo experiments indicate that NP5-mediated PDIT induce the increase of cytotoxic T lymphocytes and decrease of regulatory T lymphocytes, which can effectively inhibit the bilateral tumor growth. This work is anticipated to serve as a reference for the development of innovative agents for precise PDIT of hypoxic tumors.
Collapse
Affiliation(s)
- Ling Zhang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Rong-Bin Que
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Ting-Ting Ke
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Chao Wang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Wei Xie
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Hong-Jie Sun
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Bi-Yuan Zheng
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Mei-Rong Ke
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Jian-Dong Huang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China.
| | - Xingshu Li
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350108, China.
| |
Collapse
|
11
|
Hang L, Li H, Li M, Sun Y, Wu W, Fang L, Diao Y, Qu H, Zhang T, Li S, Jiang G. Lactoferrin docking NIR-II cyanine dye as a potentiated phototheranostic for synchronous multimodal bioimaging and tumor photo-immunotherapy. Theranostics 2024; 14:6671-6691. [PMID: 39479444 PMCID: PMC11519796 DOI: 10.7150/thno.102663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
Rationale: A promising dye for phototheranostics, IR-1048 is a near-infrared region II (NIR-II) cyanine dye that exhibits exceptional optical characteristics in NIR-II spectrum. Unfortunately, the biological applications of IR-1048 are challenged by its hydrophobic nature, the formation of face-to-face stacked dimeric aggregates (H-aggregates) that result in pronounced spectral blue shifts, and issues related to fluorescence quenching. Method: We present a novel docking strategy involving bovine serum albumin (BSA) and lactoferrin (Lf) to construct BSA@IR-1048 and Lf@IR-1048 nanoprobes. The NIR-II optical characteristics of these nanoprobes have been thoroughly investigated through both theoretical and experimental approaches. In addition, we conducted in vitro and in vivo evaluations of their NIR-II photothermal and photodynamic properties, multimodal imaging capabilities, and effectiveness in photoimmunotherapy. Results: Following the protein docking process, both BSA@IR-1048 and Lf@IR-1048 probes exhibited a red-shifted absorbance peak and an "ON" state in NIR-II fluorescence. Theoretical analyses alongside experimental results indicate that Lf@IR-1048, which has a higher docking binding energy of -10.83 kcal/mol, significantly enhances optical characteristics in the NIR-II region. Notably, when utilizing a single NIR-II light source, Lf@IR-1048 was effective in producing single-linear state oxygen and converting photons into heat energy, achieving a photo-thermal conversion efficiency of 41.9%. The overexpression of transferrin receptors in tumor cells also improved tumor-targeting and enrichment capabilities of Lf@IR-1048, as demonstrated vitro and in vivo studies. Comparatively, Lf@IR-1048 facilitated multimodal imaging-guided NIR-II phototherapy, showing an impressive tumor development inhibition rate of 94.8%. Furthermore, in bilateral CT26 tumor-bearing mice, the Lf@IR-1048-based photo-immunotherapy exhibited significant antitumor activity, attributed to enhanced dendritic cell maturation and infiltration of cytotoxic T lymphocytes. Conclusion: Lf@IR-1048 displays a powerful combination of photothermal therapy, photodynamic therapy, and tumor-targeting potential for effective multimodal imaging-guided NIR-II phototherapy, leading to substantial inhibition of tumor growth.
Collapse
Affiliation(s)
- Lifeng Hang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 518037, P. R. China
| | - Haijian Li
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 518037, P. R. China
| | - Meng Li
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 518037, P. R. China
| | - Yiqiang Sun
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Wenjiao Wu
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 518037, P. R. China
| | - Laiping Fang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 518037, P. R. China
| | - Yanzhao Diao
- School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
| | - Hong Qu
- School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
| | - Tao Zhang
- School of Physical and Mathematical sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Shumei Li
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 518037, P. R. China
| | - Guihua Jiang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 518037, P. R. China
- School of Medicine, Jinan University, Guangzhou, 510632, P. R. China
| |
Collapse
|
12
|
Wang J, Liu M, Wang J, Li Z, Feng Z, Xu M, Wang H, Li H, Li Z, Yu J, Liu J, Wei Q, Zhang S, Zhang X. Zinc oxide nanoparticles with catalase-like nanozyme activity and near-infrared light response: A combination of effective photodynamic therapy, autophagy, ferroptosis, and antitumor immunity. Acta Pharm Sin B 2024; 14:4493-4508. [PMID: 39525585 PMCID: PMC11544279 DOI: 10.1016/j.apsb.2024.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 11/16/2024] Open
Abstract
We prepared biocompatible and environment-friendly zinc oxide nanoparticles (ZnO NPs) with upconversion properties and catalase-like nanozyme activity. Photodynamic therapy (PDT) application is severely limited by the poor penetration of UV-Visible light and a hypoxic tumor environment. Here, we used ZnO NPs as a carrier for the photosensitizer chlorin e6 (Ce6) to construct zinc oxide-chlorin e6 nanoparticles (ZnO-Ce6 NPs), simultaneously addressing both problems. In terms of penetration, ZnO NPs convert 808 nm near-infrared light into 401 nm visible light to excite Ce6, achieving deep-penetrating photodynamic therapy under long-wavelength light. Interestingly, the ability to emit short-wavelength light under long-wavelength light is usually observed in upconversion nanoparticles. As nanozymes, ZnO NPs can catalyze the decomposition of hydrogen peroxide in tumors, providing oxygen for photodynamic action and relieving hypoxia. The enhanced photodynamic action produces a large amount of reactive oxygen species, which overactivate autophagy and trigger immunogenic cell death (ICD), leading to antitumor immunotherapy. In addition, even in the absence of light, ZnO and ZnO-Ce6 NPs can induce ferroptosis of tumor cells and exert antitumor effects.
Collapse
Affiliation(s)
- Jingru Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Man Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingwen Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhuoyue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhenhan Feng
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Meiqi Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hui Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hui Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhantao Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianming Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Junwei Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qingchao Wei
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Shuang Zhang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo 315832, China
| |
Collapse
|
13
|
Zhang H, Felthaus O, Eigenberger A, Klein S, Prantl L. Treg Cell Therapeutic Strategies for Breast Cancer: Holistic to Local Aspects. Cells 2024; 13:1526. [PMID: 39329710 PMCID: PMC11429654 DOI: 10.3390/cells13181526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Regulatory T cells (Tregs) play a key role in maintaining immune homeostasis and preventing autoimmunity through their immunosuppressive function. There have been numerous reports confirming that high levels of Tregs in the tumor microenvironment (TME) are associated with a poor prognosis, highlighting their role in promoting an immunosuppressive environment. In breast cancer (BC), Tregs interact with cancer cells, ultimately leading to the suppression of immune surveillance and promoting tumor progression. This review discusses the dual role of Tregs in breast cancer, and explores the controversies and therapeutic potential associated with targeting these cells. Researchers are investigating various strategies to deplete or inhibit Tregs, such as immune checkpoint inhibitors, cytokine antagonists, and metabolic inhibition. However, the heterogeneity of Tregs and the variable precision of treatments pose significant challenges. Understanding the functional diversity of Tregs and the latest advances in targeted therapies is critical for the development of effective therapies. This review highlights the latest approaches to Tregs for BC treatment that both attenuate Treg-mediated immunosuppression in tumors and maintain immune tolerance, and advocates precise combination therapy strategies to optimize breast cancer outcomes.
Collapse
Affiliation(s)
- Hanwen Zhang
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss Allee 11, 93053 Regensburg, Germany (L.P.)
| | | | | | | | | |
Collapse
|
14
|
Chen Y, Zhou Q, Jia Z, Cheng N, Zhang S, Chen W, Wang L. Enhancing cancer immunotherapy: Nanotechnology-mediated immunotherapy overcoming immunosuppression. Acta Pharm Sin B 2024; 14:3834-3854. [PMID: 39309502 PMCID: PMC11413684 DOI: 10.1016/j.apsb.2024.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/12/2024] [Accepted: 05/24/2024] [Indexed: 09/25/2024] Open
Abstract
Immunotherapy is an important cancer treatment method that offers hope for curing cancer patients. While immunotherapy has achieved initial success, a major obstacle to its widespread adoption is the inability to benefit the majority of patients. The success or failure of immunotherapy is closely linked to the tumor's immune microenvironment. Recently, there has been significant attention on strategies to regulate the tumor immune microenvironment in order to stimulate anti-tumor immune responses in cancer immunotherapy. The distinctive physical properties and design flexibility of nanomedicines have been extensively utilized to target immune cells (including tumor-associated macrophages (TAMs), T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated fibroblasts (TAFs)), offering promising advancements in cancer immunotherapy. In this article, we have reviewed treatment strategies aimed at targeting various immune cells to regulate the tumor immune microenvironment. The focus is on cancer immunotherapy models that are based on nanomedicines, with the goal of inducing or enhancing anti-tumor immune responses to improve immunotherapy. It is worth noting that combining cancer immunotherapy with other treatments, such as chemotherapy, radiotherapy, and photodynamic therapy, can maximize the therapeutic effects. Finally, we have identified the challenges that nanotechnology-mediated immunotherapy needs to overcome in order to design more effective nanosystems.
Collapse
Affiliation(s)
- Yunna Chen
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Qianqian Zhou
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Zongfang Jia
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Nuo Cheng
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Sheng Zhang
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Weidong Chen
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Lei Wang
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| |
Collapse
|
15
|
Ma W, Wang X, Zhang D, Mu X. Research Progress of Disulfide Bond Based Tumor Microenvironment Targeted Drug Delivery System. Int J Nanomedicine 2024; 19:7547-7566. [PMID: 39071505 PMCID: PMC11283832 DOI: 10.2147/ijn.s471734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer poses a significant threat to human life and health. Chemotherapy is currently one of the effective cancer treatments, but many chemotherapy drugs have cell toxicity, low solubility, poor stability, a narrow therapeutic window, and unfavorable pharmacokinetic properties. To solve the above problems, target drug delivery to tumor cells, and reduce the side effects of drugs, an anti-tumor drug delivery system based on tumor microenvironment has become a focus of research in recent years. The construction of a reduction-sensitive nanomedicine delivery system based on disulfide bonds has attracted much attention. Disulfide bonds have good reductive responsiveness and can effectively target the high glutathione (GSH) levels in the tumor environment, enabling precise drug delivery. To further enhance targeting and accelerate drug release, disulfide bonds are often combined with pH-responsive nanocarriers and highly expressed ligands in tumor cells to construct drug delivery systems. Disulfide bonds can connect drug molecules and polymer molecules in the drug delivery system, as well as between different drug molecules and carrier molecules. This article summarized the drug delivery systems (DDS) that researchers have constructed in recent years based on disulfide bond drug delivery systems targeting the tumor microenvironment, disulfide bond cleavage-triggering conditions, various drug loading strategies, and carrier design. In this review, we also discuss the controlled release mechanisms and effects of these DDS and further discuss the clinical applicability of delivery systems based on disulfide bonds and the challenges faced in clinical translation.
Collapse
Affiliation(s)
- Weiran Ma
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
- Jilin University School of Pharmaceutical Sciences, Changchun, 130021, People’s Republic of China
| | - Xiaoying Wang
- Jilin University School of Pharmaceutical Sciences, Changchun, 130021, People’s Republic of China
| | - Dongqi Zhang
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
16
|
Chaudhari R, Patel V, Kumar A. Cutting-edge approaches for targeted drug delivery in breast cancer: beyond conventional therapies. NANOSCALE ADVANCES 2024; 6:2270-2286. [PMID: 38694472 PMCID: PMC11059480 DOI: 10.1039/d4na00086b] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/07/2024] [Indexed: 05/04/2024]
Abstract
Breast cancer is a global health challenge with staggering statistics underscoring its pervasive impact. The burden of this disease is measured in terms of its prevalence and the challenges it poses to healthcare systems, necessitating a closer look at its epidemiology and impact. Current breast cancer treatments, including surgery, chemotherapy, radiation therapy, and targeted therapies, have made significant strides in improving patient outcomes. However, they are not without limitations, often leading to adverse effects and the development of drug resistance. This comprehensive review delves into the complex landscape of breast cancer, including its incidence, current treatment modalities, and the inherent limitations of existing therapeutic approaches. It also sheds light on the promising role of nanotechnology, encompassing both inorganic and organic nanoparticles equipped with the ability to selectively deliver therapeutic agents to tumor sites, in the battle against breast cancer. The review also addresses the emerging therapies, their associated challenges, and the future prospects of targeted drug delivery in breast cancer management.
Collapse
Affiliation(s)
- Ramesh Chaudhari
- Biological & Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| | - Vishva Patel
- Biological & Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| | - Ashutosh Kumar
- Biological & Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| |
Collapse
|
17
|
Qiu ZW, Zhong YT, Lu ZM, Yan N, Kong RJ, Huang JQ, Li ZF, Nie JM, Li R, Cheng H. Breaking Physical Barrier of Fibrotic Breast Cancer for Photodynamic Immunotherapy by Remodeling Tumor Extracellular Matrix and Reprogramming Cancer-Associated Fibroblasts. ACS NANO 2024; 18:9713-9735. [PMID: 38507590 DOI: 10.1021/acsnano.4c01499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Cancer-associated fibroblasts (CAFs) assist in breast cancer (BRCA) invasion and immune resistance by overproduction of extracellular matrix (ECM). Herein, we develop FPC@S, a photodynamic immunomodulator that targets the ECM, to improve the photodynamic immunotherapy for fibrotic BRCA. FPC@S combines a tumor ECM-targeting peptide, a photosensitizer (protoporphyrin IX) and an antifibrotic drug (SIS3). After anchoring to the ECM, FPC@S causes ECM remodeling and BRCA cell death by generating reactive oxygen species (ROS) in situ. Interestingly, the ROS-mediated ECM remodeling can normalize the tumor blood vessel to improve hypoxia and in turn facilitate more ROS production. Besides, upon the acidic tumor microenvironment, FPC@S will release SIS3 for reprograming CAFs to reduce their activity but not kill them, thus inhibiting fibrosis while preventing BRCA metastasis. The natural physical barrier formed by the dense ECM is consequently eliminated in fibrotic BRCA, allowing the drugs and immune cells to penetrate deep into tumors and have better efficacy. Furthermore, FPC@S can stimulate the immune system and effectively suppress primary, distant and metastatic tumors by combining with immune checkpoint blockade therapy. This study provides different insights for the development of fibrotic tumor targeted delivery systems and exploration of synergistic immunotherapeutic mechanisms against aggressive BRCA.
Collapse
Affiliation(s)
- Zi-Wen Qiu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Ying-Tao Zhong
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Zhen-Ming Lu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Ni Yan
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Ren-Jiang Kong
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jia-Qi Huang
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Zhuo-Feng Li
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jun-Mei Nie
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Runqing Li
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Hong Cheng
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|