1
|
Huang X, Hou S, Li Y, Xu G, Xia N, Duan Z, Luo K, Tian B. Targeting lipid metabolism via nanomedicine: A prospective strategy for cancer therapy. Biomaterials 2025; 317:123022. [PMID: 39754967 DOI: 10.1016/j.biomaterials.2024.123022] [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: 09/09/2024] [Revised: 11/20/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025]
Abstract
Lipid metabolism has been increasingly recognized to play an influencing role in tumor initiation, progression, metastasis, and therapeutic drug resistance. Targeting lipid metabolic reprogramming represents a promising therapeutic strategy. Despite their structural complexity and poor targeting efficacy, lipid-metabolizing drugs, either used alone or in combination with chemotherapeutic agents, have been employed in clinical practice. The advent of nanotechnology offers new approaches to enhancing therapeutic effects, includingthe targeted delivery and integration of lipid metabolic reprogramming with chemotherapy, photodynamic therapy (PDT), and immunotherapy. The integrated nanoformulation, nanomedicine, could significantly advance the field of lipid metabolism therapy. In this review, we will briefly introduce the concept of cancer lipid metabolism reprogramming, then elaborate the latest advances in engineered nanomedicine for targeting lipid metabolism during cancer treatment, and finally provide our insights into future perspectives of nanomedicine for interference with lipid metabolism in the tumor microenvironment.
Collapse
Affiliation(s)
- Xing Huang
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shengzhong Hou
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinggang Li
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Xu
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, 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
| | - Ning Xia
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, 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.
| | - Kui Luo
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, 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.
| | - Bole Tian
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Mirkin CA, Langer R, Mrksich M, Margolin AA, Petrosko SH, Artzi N. Blueprints for Better Drugs: The Structural Revolution in Nanomedicine. ACS NANO 2025; 19:18889-18901. [PMID: 40359339 DOI: 10.1021/acsnano.5c06380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Structural nanomedicines are engineered constructs that arrange therapeutic components into well-defined architectures to maximize efficacy. Their multivalent, multifunctional design offers key advantages over unstructured formulations, including targeted delivery, expanded therapeutic windows, and enhanced target engagement. The mRNA COVID-19 vaccines exemplify their transformative potential. However, structural precision varies, and more well-defined architectures will streamline optimization, manufacturing, and regulation. Unlike small molecule drugs, nanomedicines within a batch are not identical. Identifying the most effective, least toxic structures will advance our understanding of structure-function relationships and therapeutic mechanisms. This work highlights structural nanomedicines─small molecules, nucleic acids, and biologics─to galvanize the field and drive innovation toward even safer, more effective treatments that benefit patients.
Collapse
Affiliation(s)
- Chad A Mirkin
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
- CZ Biohub Chicago, LLC, Chicago, Illinois 60642, United States
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Milan Mrksich
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Cellular and Developmental Biology, Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Adam A Margolin
- Flashpoint Therapeutics, Evanston, Illinois 60201, United States
| | - Sarah Hurst Petrosko
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Natalie Artzi
- Brigham and Women's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts 02215, United States
- Institute for Biomedical Engineering and Science, Biomedical Engineering Division, Massachusetts Institute of Technology, Cambridge, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| |
Collapse
|
3
|
Chen LE, Nittayacharn P, Exner AA. Progress and potential of nanobubbles for ultrasound-mediated drug delivery. Expert Opin Drug Deliv 2025:1-24. [PMID: 40353846 DOI: 10.1080/17425247.2025.2505044] [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: 02/13/2025] [Revised: 04/18/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Despite much progress, nanomedicine-based drug therapies in oncology remain limited by systemic toxicity and insufficient particle accumulation in the tumor. To address these barriers, formulations responsive to external physical stimuli have emerged. One most promising system is the ultrasound stimulation of drug-loaded, gas-core particles (bubbles). Ultrasound induces bubble cavitation for cell and tissue permeabilization, triggers on-demand drug release, and provides opportunities for real-time imaging of delivery. AREAS COVERED Here, we focus on shell-stabilized, gas-core nanoparticles (also termed nanobubbles or ultrafine bubbles) and their role in ultrasound-mediated therapeutic delivery to tumors. This review frames the advantages of nanobubbles within the ongoing deficits in nanomedicine, describes mechanisms of ultrasound-mediated therapy, and details formulation techniques for nanobubble delivery systems. It then highlights the past decade of research in nanobubble-facilitated drug delivery for cancer therapy and anticipates new directions in the field. EXPERT OPINION Nanobubble ultrasound contrast agents offer a spatiotemporally triggerable therapeutic coupled with a safe, accessible imaging modality. Nanobubbles can be loaded with diverse therapeutic cargoes to treat disease and overcome numerous barriers limiting delivery to solid tumors. Close attention to formulation, characterization methods, acoustic testing parameters, and the biological mechanisms of nanobubble delivery will facilitate preclinical research toward clinical adoption.
Collapse
Affiliation(s)
- Laura E Chen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pinunta Nittayacharn
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
4
|
Boix-Montesinos P, Carrascosa-Marco P, Armiñán A, Vicent MJ. Identification of functional biomarkers for personalized nanomedicine in advanced breast cancer in vitro models. J Control Release 2025; 381:113584. [PMID: 40086758 DOI: 10.1016/j.jconrel.2025.113584] [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: 11/10/2024] [Revised: 01/30/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Nanomedicines represent promising advanced therapeutics for the enhanced treatment of breast cancer, the primary cause of cancer-related deaths in women; however, the clinical translation of nanomedicines remains challenging. Advanced in vitro models of breast cancer may improve preclinical evaluations and the identification of biomarkers that aid the stratification of patients who would benefit from a given nanomedicine. In this study, we first developed a matrix-embedded breast cancer cell spheroid model representing the extracellular matrix and confirmed the faithful recapitulation of disease aggressiveness in vitro. We then characterized factors influencing nanomedicine drug release (i.e., cathepsin B levels/activity, reactive oxygen species levels, glutathione levels, and cytoplasmic pH values) and evaluated nanomedicine internalization and cytotoxicity evaluation in our spheroid model. We confirmed the reduced-to-oxidized glutathione ratio as a functional biomarker of disulfide linker-containing polypeptide-drug conjugate effectiveness. We then established a biobank of patient-derived breast cancer organoids that recapitulate clinical intra-tumor and inter-tumor heterogeneity as a more advanced model. Analysis in organoids revealed that patient-specific responses to a polypeptide-based nanomedicine correlated with cathepsin B levels, supporting the potential of the functional biomarker for patient-tailored nanomedicine selection. Our findings highlight that exhaustively characterized advanced in vitro models support the evaluation of nanomedicines and the identification of functional biomarkers.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Paula Carrascosa-Marco
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Ana Armiñán
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCCIII, Madrid, Spain.
| | - María J Vicent
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCCIII, Madrid, Spain.
| |
Collapse
|
5
|
Song B, Shuang L, Zhang S, Tong C, Chen Q, Li Y, Hao M, Niu W, Jin CH. Research progress of nano drug delivery systems in the anti-tumor treatment of traditional Chinese medicine monomers. PeerJ 2025; 13:e19332. [PMID: 40292112 PMCID: PMC12034246 DOI: 10.7717/peerj.19332] [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: 12/04/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Tumors pose a serious threat to global public health and are usually treated from two aspects: tumor cells and tumor microenvironment. Compared with traditional chemotherapy drugs, traditional Chinese medicine (TCM) monomers have advantages in tumor treatment, such as multiple targets, multiple levels and synergistic intervention. However, most TCM active ingredients have disadvantages such as poor water solubility and stability, which restrict their clinical application. Nano drug delivery systems have the functions of improving the bioavailability of TCM anti-tumor active ingredients, enhancing tissue targeting, achieving controlled drug release, and inhibiting tumor multidrug resistance. Compared with free monomers, they have higher therapeutic effects and fewer side effects. This article summarizes five commonly used anti-tumor TCM monomer nanocarriers, including lipid nanomaterials, exosomes, polymer micelles, carbon nanotubes, and dendrimers, and explains their anti-tumor mechanisms after combining with TCM, such as inhibiting tumor cell proliferation and metastasis, regulating tumor microenvironment, etc. At the same time, the potential of nano drug delivery systems combined with radiotherapy and immunotherapy is discussed, as well as the current problems of potential toxicity, long-term stability, and complex amplification process, as well as future development directions, aiming to provide a reference for promoting the clinical application of nano drug delivery systems for TCM anti-tumor active ingredients.
Collapse
Affiliation(s)
- Bocui Song
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Li Shuang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Shuang Zhang
- Comprehensive Service Center, Yongji Economic Development Zone, Jilin, Jilin, China
| | - Chunyu Tong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Qian Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yuqi Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Meihan Hao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Wenqi Niu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Cheng-Hao Jin
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
- College of Life Science and Technology, College of Life Science and Technology, Daqing, Heilongjiang, China
| |
Collapse
|
6
|
Saqlain M, Muhammad Zohaib H, Ahmad Khan M, Qamar S, Masood S, Lauqman M, Ilyas M, Irfan M, Li H. Evaluating the Drug Delivery Capacity of 3D Coordination Polymer for Anticancer Drugs. Chem Asian J 2025; 20:e202401475. [PMID: 39903798 DOI: 10.1002/asia.202401475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/16/2025] [Indexed: 02/06/2025]
Abstract
We synthesized {[Cd2(dTMP)2(4,4'-azpy)2(H2O)2] ⋅ 3(O)}n a novel three-dimensional metal nucleotide coordination polymer (CP-1). An assessment of the CP-1 binding affinity for anticancer drugs was conducted using molecular dynamic simulations. The virtual screening results depict that CP-1 has a lot of potential for encapsulating the anthracycline anticancer drug doxorubicin (DOX). It hasn't yet been investigated how to accomplish high loading capacity, efficiency, and controlled release of DOX in dTMP-based 3D metal coordination polymers. Utilizing DOX as a drug model and our system as a drug-loading vehicle, we used UV-visible and circular dichroism titrations to examine the effects of its encapsulation and release. The mechanism of drug loading and release was investigated through pH-responsive behavior by adjusting the pH value to 8, 7, 6, and 5. The results indicate the CP-1 has a robust affinity for DOX at pH 7, which facilitates its loading on 3D porous coordination polymer. However, the maximum cumulative drug release of 87.11 % was observed at pH 5. The higher correlation coefficient (R2) was obtained at pH 5 with the Higuchi equation. It indicated that the drug released was primarily controlled with the diffusion mechanism. The CP-1 polymer's ability to encapsulate DOX while also permitting a possible controlled-release mechanism is confirmed by the combined insights from the experimental findings, energy graphs, RMSD analysis, and radius of gyration (Rg) data from MD simulations.
Collapse
Affiliation(s)
- Madiha Saqlain
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Hafiz Muhammad Zohaib
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Maroof Ahmad Khan
- State Key Laboratory of Marine Resource Utilization in South China Sea, Collaborative Innovation Center of Hainan University, 570228, Haikou, P. R. China
| | - Samina Qamar
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Sara Masood
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Lauqman
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Mubashar Ilyas
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Muhammad Irfan
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Hui Li
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| |
Collapse
|
7
|
Liu H, Zou J, Li X, Ge Y, He W. Drug delivery for platinum therapeutics. J Control Release 2025; 380:503-523. [PMID: 39923853 DOI: 10.1016/j.jconrel.2025.02.006] [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: 07/02/2024] [Revised: 01/18/2025] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
Cancer remains a severe threat to human health. Platinum drugs, such as cisplatin (CDDP), oxaliplatin, and carboplatin, are extensively utilized for treating various cancers and have become the primary drugs in first-line treatments for numerous solid tumors due to their effective anticancer properties. However, their side effects, including drug resistance, nephrotoxicity and ototoxicity, limit the clinical application. Therefore, there is an urgent need to develop targeted delivery and controlled release systems for platinum drugs to address the disadvantages, enhancing tumor accumulation and improving therapeutic effects. In this review, we first review the progress of platinum drugs, their anticancer mechanism, clinical applications and limitations. Then, we comprehensively summarize the platinum-based delivery using drug carriers and responsive strategies. We especially highlight the platinum-delivery formulations in ongoing clinical trials. Finally, we provide perspectives for this field. The review could provide an increasingly in-depth understanding of platinum therapeutics and motivate increasing delivery tactics to overcome the limitations of platinum application.
Collapse
Affiliation(s)
- Hui Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Yizhi Ge
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, PR China.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, PR China.
| |
Collapse
|
8
|
Su B, Chen Q, Li X, Fang M, Wang Y, Song H, You H, Zhou Z, Wu Y, Zhao Z, Chen Y, Fan H, Li C, Jiang C, Sun T. A Methionine Allocation Nanoregulator for the Suppression of Cancer Stem Cells and Support to the Immune Cells by Epigenetic Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415207. [PMID: 39985256 PMCID: PMC12005795 DOI: 10.1002/advs.202415207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Epigenetic dysregulation is prevalent in human cancers, affecting gene expression and metabolic patterns to meet the demands of malignant evolution and abnormal epigenetic processes, and resulting in a protumor immune microenvironment. Tumors require a steady supply of methionine for maintaining epigenetic flexibility, which is the only exogenous precursor of methyl donor S-adenosylmethionine for methylation, crucial for their resistance to therapies and survival in a nutrient-deficient microenvironment. Thus, tumor cells upregulate the Lat4 transporter to compete and deprive methionine in the microenvironment, sustaining their malignant phenotypes and also impairing immune cell functions. Addressing this methionine addiction is the key to overcoming drug resistance and improving immune response. Despite the challenge of lacking specific Lat4 inhibitors, an oxaliplatin prodrug crosslinked fluorinated polycation/anti-Lat4 small interfering RNA complex nanoregulator (AS-F-NP) has been designed and developed here. This nanoregulator restricted the greedy methionine uptake of tumor cells by knocking down Lat4, which in turn inhibited the malignant evolution of the tumor while restoring the viability and function of tumor-infiltrating immune cells.
Collapse
Affiliation(s)
- Boyu Su
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Qinjun Chen
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Xuwen Li
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Mingzhu Fang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yu Wang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Haolin Song
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Haoyu You
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Zheng Zhou
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yuxing Wu
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Zhenhao Zhao
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yun Chen
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Hongrui Fan
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Chufeng Li
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Chen Jiang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
- Department of Digestive DiseasesNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350212China
| | - Tao Sun
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
- Quzhou Fudan InstituteQuzhou324003China
| |
Collapse
|
9
|
Fang M, Su B, Zhang S, Li F, Guo Y, Chen Q, Wu Y, Liu H, Jiang C, Sun T. Engineered Intelligent Microenvironment Responsive Prodrug Conjugates Navigated by Bioinspired Lipoproteins for Reversing Liver Fibrosis. SMALL METHODS 2025:e2402247. [PMID: 40103435 DOI: 10.1002/smtd.202402247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/23/2025] [Indexed: 03/20/2025]
Abstract
Liver fibrosis (LF) is characterized by excessive production of reactive oxygen species (ROS), abnormal activation of hepatic stellate cells (HSCs), and subsequent extracellular matrix (ECM) deposition. The complexity of multiple interrelated pathways involved in this process makes it challenging for monotherapy to achieve the desired therapeutic effects. To address this issue, this study designs a ROS-activated heterodimer conjugate (VTO) to collaboratively alleviate LF. Additionally, a biomimetic high-density lipoprotein is utilized for encapsulation, resulting in the formation of PL-VTO, which enables natural liver targeting. Once PL-VTO is delivered to the fibrotic liver, it can respond and release both parent drugs upon encountering the high ROS microenvironment, effectively scavenge ROS, induce quiescence of activated HSCs, and reduce collagen deposition, ultimately reversing LF. Overall, this study presents a feasible and versatile nanotherapeutic approach to enhance the prodrug-driven treatment of LF.
Collapse
Affiliation(s)
- Mingzhu Fang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Boyu Su
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shilin Zhang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Fangxin Li
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yun Guo
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qinjun Chen
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yuxing Wu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Huiyi Liu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Quzhou Fudan Institute, Quzhou, 324003, China
| |
Collapse
|
10
|
Yuan J, Chen Q, Zuo M, Li X, Ou C, Chen Q, Yu D, Li H, Hao C, Yang J, Liu S, Cheng D. Enhanced combination therapy through tumor microenvironment-activated cellular uptake and ROS-sensitive drug release using a dual-sensitive nanogel. Biomater Sci 2025; 13:1554-1567. [PMID: 39957553 DOI: 10.1039/d4bm01377h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
Although the co-delivery of chemotherapeutic and photodynamic agents has been studied for years, developing a simple and efficient nanoplatform for high co-delivery efficiency remains a challenge for clinical applications. In this study, we prepared a reactive oxygen species (ROS) and pH dual-sensitive nanogel for the co-encapsulation of doxorubicin (DOX) and indocyanine green (ICG)-conjugated bovine serum albumin (BSA) via a simple inverse miniemulsion polymerization process. This was followed by modification with pegylated cell-penetrating peptides (CPPs) containing citraconic anhydride (CDM) linkers, which are sensitive to weakly acidic microenvironments (pH 6.5). Pegylation endowed the nanogel with extended blood circulation, while the de-shielding of polyethylene glycol (PEG) exposed the CPPs, significantly enhancing cellular uptake. Upon near-infrared (NIR) irradiation, ROS generated by ICG not only killed tumor cells but also triggered the release of DOX through nanogel disintegration. Serial experiments verified the nanogel's high co-delivery efficiency, tumor tissue matrix microenvironment-triggered cellular uptake, controlled drug release, and synergistic antitumor effects. Therefore, this dual-sensitive nanogel, prepared via inverse miniemulsion polymerization, offers a facile approach to improving co-delivery efficiency for combination therapy.
Collapse
Affiliation(s)
- Jianming Yuan
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Qinfeng Chen
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Mingxiang Zuo
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Xiaoxia Li
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - ChiYi Ou
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Qinghua Chen
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Dongsheng Yu
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Haowen Li
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Chenhui Hao
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Jing Yang
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Shuang Liu
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| | - Du Cheng
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, PR China.
| |
Collapse
|
11
|
Xiao W, Jiang W, Chen Z, Huang Y, Mao J, Zheng W, Hu Y, Shi J. Advance in peptide-based drug development: delivery platforms, therapeutics and vaccines. Signal Transduct Target Ther 2025; 10:74. [PMID: 40038239 PMCID: PMC11880366 DOI: 10.1038/s41392-024-02107-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 11/01/2024] [Accepted: 12/13/2024] [Indexed: 03/06/2025] Open
Abstract
The successful approval of peptide-based drugs can be attributed to a collaborative effort across multiple disciplines. The integration of novel drug design and synthesis techniques, display library technology, delivery systems, bioengineering advancements, and artificial intelligence have significantly expedited the development of groundbreaking peptide-based drugs, effectively addressing the obstacles associated with their character, such as the rapid clearance and degradation, necessitating subcutaneous injection leading to increasing patient discomfort, and ultimately advancing translational research efforts. Peptides are presently employed in the management and diagnosis of a diverse array of medical conditions, such as diabetes mellitus, weight loss, oncology, and rare diseases, and are additionally garnering interest in facilitating targeted drug delivery platforms and the advancement of peptide-based vaccines. This paper provides an overview of the present market and clinical trial progress of peptide-based therapeutics, delivery platforms, and vaccines. It examines the key areas of research in peptide-based drug development through a literature analysis and emphasizes the structural modification principles of peptide-based drugs, as well as the recent advancements in screening, design, and delivery technologies. The accelerated advancement in the development of novel peptide-based therapeutics, including peptide-drug complexes, new peptide-based vaccines, and innovative peptide-based diagnostic reagents, has the potential to promote the era of precise customization of disease therapeutic schedule.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Wenjie Jiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yu Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Junyi Mao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wei Zheng
- Department of Integrative Medicine, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yonghe Hu
- School of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
12
|
Khurana S, Sharma S, Goyal PK. Tumor microenvironment as a target for developing anticancer hydrogels. Drug Dev Ind Pharm 2025; 51:157-168. [PMID: 39829011 DOI: 10.1080/03639045.2025.2455424] [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: 11/04/2024] [Revised: 12/28/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE It has been reported that cancer cells get protected by a complex and rich multicellular environment i.e. the tumor microenvironment (TME) consisting of varying immune cells, endothelial cells, dendritic cells, fibroblasts, etc. This manuscript is aimed at the characteristic features of TME considered as potential target(s) for developing smart anticancer hydrogels. SIGNIFICANCE The stimuli-specific drug delivery systems especially hydrogels that can respond to the characteristic features of TME are fabricated for treating cancer. For developing anticancer formulations, TME targeting can be considered an alternative way as it enhances the cytotoxic potential and reduces the unwanted effects. This manuscript shall be of quite interest to academicians, researchers, and clinicians engaged in oncology. METHODS The manuscript was prepared by using the data available in the public domain in online resources such as Google Scholar, PubMed, Science Direct, Scopus, Web of Science, Research Gate, etc. RESULTS Smart hydrogels, sensitive to some specific features of TME such as low pH, high concentration of glutathione, specific enzymes, etc., are promising anticancer formulations as these improve the efficacy and lower the side effects of chemotherapy. CONCLUSION The stimuli-responsive hydrogels have been gaining more attention for delivering cytotoxic drugs to the TME in response to specific stimuli. The stimuli-responsive hydrogels, comprising of cytotoxic drug(s) and specific polymers have some special features such as similarity with biological matrix, ability to respond to various internal as well as external stimuli, improved permeability, porosity, biocompatibility, resemblance with soft living tissues, etc.; and are considered as the promising anticancer candidates.
Collapse
Affiliation(s)
- Suman Khurana
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram, India
- Department of Pharmacy, Panipat Institute of Engineering and Technology, Panipat, India
| | - Shrestha Sharma
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram, India
| | - Parveen Kumar Goyal
- Department of Pharmacy, Panipat Institute of Engineering and Technology, Panipat, India
| |
Collapse
|
13
|
Li A, Qi F, Zeng Y, Liu R, Cai H, He M, Li D, Gu Y, Liu J. Cryoshocked Adipocytes Mediated Dual-Modal Strategy Combining Photodynamic Therapy and Triptolide Palmitate for Pulmonary Metastatic Melanoma Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414307. [PMID: 39804941 PMCID: PMC11884613 DOI: 10.1002/advs.202414307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/19/2024] [Indexed: 01/16/2025]
Abstract
Pulmonary metastasis represents one of the most prevalent forms of metastasis in advanced melanoma, with mortality rates reaching 70%. Current treatments including chemotherapy, targeted therapy, and immunotherapy frequently exhibit limited efficacy or present high costs. To address these clinical needs, this study presents a biomimetic drug delivery system (Ce6-pTP-CsA) utilizing cryoshocked adipocytes (CsA) encapsulating the prodrug triptolide palmitate (pTP) and the photosensitizer Ce6, exploiting the characteristic of tumor cells to recruit and lipolyze adipocytes for energy. CsA substantially enhances the drug-loading capacity of adipocytes, with its particle size characteristics enabling targeted delivery of pTP to the lungs. The combination of photodynamic therapy (PDT) and pTP activates the caspase cascade, promoting apoptosis in tumor cells. Notably, the cleavage of disulfide bonds in pTP depletes glutathione (GSH), reducing its scavenging effect on reactive oxygen species (ROS) and enhancing the efficacy of PDT. Results demonstrate that Ce6-pTP-CsA effectively inhibits the proliferation and invasion of pulmonary metastatic melanoma cells in vitro and induces apoptosis, while significantly suppressing lung metastasis of SCID mice models in vivo. In conclusion, this novel biomimetic drug delivery system based on adipocytes provides a promising strategy for targeted therapy in pulmonary metastatic melanoma.
Collapse
Affiliation(s)
- Aixue Li
- Department of PharmacyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- College of PharmacyShandong University of Traditional Chinese MedicineJinanShandong250355China
| | - Fu Qi
- Department of PharmacyTongji HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Yuanye Zeng
- Department of PharmacyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Rongmei Liu
- Department of PharmacyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- College of PharmacyShandong University of Traditional Chinese MedicineJinanShandong250355China
| | - Huanhuan Cai
- Department of PharmacyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- College of PharmacyShandong University of Traditional Chinese MedicineJinanShandong250355China
| | - Mengyuan He
- Department of PharmacyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- College of PharmacyShandong University of Traditional Chinese MedicineJinanShandong250355China
| | - Dan Li
- Department of PharmacyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yongwei Gu
- Department of PharmacyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jiyong Liu
- Department of PharmacyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- College of PharmacyShandong University of Traditional Chinese MedicineJinanShandong250355China
| |
Collapse
|
14
|
Pan Y, Zhao H, Huang W, Liu S, Qi Y, Huang Y. Metal-Protein Hybrid Materials: Unlocking New Frontiers in Biomedical Applications. Adv Healthc Mater 2025; 14:e2404405. [PMID: 39778029 DOI: 10.1002/adhm.202404405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Metal-protein hybrid materials represent a novel class of functional materials that exhibit exceptional physicochemical properties and tunable structures, rendering them remarkable applications in diverse fields, including materials engineering, biocatalysis, biosensing, and biomedicine. The design and development of multifunctional and biocompatible metal-protein hybrid materials have been the subject of extensive research and a key aspiration for practical applications in clinical settings. This review provides a comprehensive analysis of the design strategies, intrinsic properties, and biomedical applications of these hybrid materials, with a specific emphasis on their potential in cancer therapy, drug and vaccine delivery, antibacterial treatments, and tissue regeneration. Through rational design, stable metal-protein hybrid materials can be synthesized using straightforward methods, enabling them with therapeutic, delivery, immunomodulatory, and other desired functionalities. Finally, the review outlines the existing limitations and challenges associated with metal-protein hybrid materials and evaluates their potential for clinical translation, providing insights into their practical implementation within biomedical applications.
Collapse
Affiliation(s)
- Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Han Zhao
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Wenyong Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Siyang Liu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| |
Collapse
|
15
|
Hu D, Huang Z, Li W, Shan L, Wu MY, Feng S, Wan Y. Macrophage Membrane-Cloaked ROS-Responsive Albumin Nanoplatforms for Targeted Delivery of Curcumin to Alleviate Acute Liver Injury. Mol Pharm 2025; 22:771-786. [PMID: 39783460 DOI: 10.1021/acs.molpharmaceut.4c00808] [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] [Indexed: 01/12/2025]
Abstract
Developing low-toxicity, high-efficacy, and fast-acting strategies to manage acute liver injury (ALI) is critical due to its rapid progression and potential for severe outcomes. Curcumin (CUR) has shown promise in ALI therapy due to its ability to modulate the inflammatory microenvironment by scavenging reactive oxygen species (ROS). Nevertheless, CUR is highly hydrophobic limiting its bioavailability and effective in vivo transport, which hinders its further application. In this study, we developed an inflammatory microenvironment-targeted drug delivery system by covalently coupling human serum albumin (HSA) with ROS-sensitive thioketal linkers and loading it with CUR to form nanoparticles (HSA-TK/CUR). These nanoparticles were then coated with a macrophage membrane (CM@HSA-TK/CUR), resulting in negatively charged spherical particles (≈ -23.26 mV) with an average particle size of around 165 nm. ROS responsiveness was confirmed through drug release assays and enhanced ROS depletion was further demonstrated by Diacetyldichlorofluorescein (DCFH-DA) ROS detection experiments. CM@HSA-TK/CUR treatment resulted in a 94.7% reduction in ROS levels in inflammatory cells. In addition, cellular uptake and in vivo distribution experiments demonstrated that camouflaging HSA-TK/CUR with macrophage membranes significantly enhanced its targeting of the inflammatory microenvironment. The findings revealed that CM@HSA-TK/CUR rapidly accumulated in the injured liver within 6 h, inhibited the production of pro-inflammatory factors (IL-1β, IL-6, and TNF-α), shifted macrophage polarization from M1 to M2 in vivo, and protected hepatocytes from oxidative stress-associated cell death, significantly attenuating the inflammatory response in ALI mice. In conclusion, CM@HSA-TK/CUR has excellent potential in treating mice with ALI.
Collapse
Affiliation(s)
- Dandan Hu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zhenqiu Huang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenlong Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lianhai Shan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Ming-Yu Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yu Wan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
16
|
Dai X, Yin Y, Wang C, Xu H. Hyaluronic acid regulated facile synthesis of size-tunable multifunctional nanomedicine for effective cancer therapy. Int J Biol Macromol 2025; 288:138668. [PMID: 39667478 DOI: 10.1016/j.ijbiomac.2024.138668] [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: 10/06/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
The complex and heterogeneous nature of cancer necessitates the development of innovative multifunctional nanomedicines (MN). Hyaluronic acid (HA) is a functional carbohydrate polysaccharide that is widely used in various biomedical fields. In this study, we employed HA as a stabilizer and regulator for the synthesis of a size-tunable nanomedicine comprising ferric ions, doxorubicin, and epigallocatechin gallate (EGCG), referred to as HDE-MN, for cancer therapy. A change in the HA ratio can yield HDE-MNs with sizes varying from ~20 nm to over 100 nm. Modified HA can respond to hyaluronidase (HAase) to provide controllable pH/HAase dual-responsive drug release for improved cancer therapy. Moreover, HA can mediate the targeted delivery of HDE-MNs both in vitro and in vivo to cancer cells. In addition, HDE-MNs reversed multidrug resistance owing to the incorporation of EGCG, inducing ferroptosis due to the involvement of ferric ions. More importantly, HDE-MNs have a photothermal conversion effect, enabling photothermal therapy, photothermally enhanced drug release, and ferroptosis, which collectively contribute to significantly improved cancer therapy. Therefore, the HDE-MNs with laser irradiation achieved full ablation of the in vivo tumors. Together with its good biocompatibility, HDE-MNs may be promising for effective cancer therapy.
Collapse
Affiliation(s)
- Xiuliang Dai
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, China
| | - Yina Yin
- Obstetrics and Gynecology Department, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Hongbin Xu
- Obstetrics and Gynecology Department, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China.
| |
Collapse
|
17
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
18
|
Liu L, Fu S, Gu H, Li Y, Zhu G, Ai H, Li W. Platinum(IV)-Backboned Polymer Prodrug-Functionalized Manganese Oxide Nanoparticles for Enhanced Lung Cancer Chemoimmunotherapy via Amplifying Stimulator of Interferon Genes Activation. ACS NANO 2025; 19:2726-2741. [PMID: 39772457 DOI: 10.1021/acsnano.4c15115] [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: 01/11/2025]
Abstract
The stimulator of interferon genes (STING) pathway exhibits great potential in remodeling the immunosuppressive tumor microenvironment and initiating antitumor immunity. However, how to effectively activate STING and avoid undesired toxicity after systemic administration remains challenging. Herein, platinum(IV)-backboned polymer prodrug-coated manganese oxide nanoparticles (DHP/MnO2NP) with pH/redox dual responsive properties are developed to precisely release cisplatin and Mn2+ in the tumor microenvironment and synergistically amplify STING activation. In vitro, we demonstrate that DHP/MnO2NP can effectively induce tumor cell DNA damage and leak into the cytoplasm, cooperating with Mn2+ to promote STING activation and significantly upregulate the expression of proinflammatory cytokines. Additionally, DHP/MnO2NP can selectively release cisplatin and Mn2+ to mediate tumor killing while reducing toxicity to normal cells. In vivo, DHP/MnO2NP exerted increased therapeutic efficacy by inducing STING activation and initiating robust antitumor immunity. Specifically, DHP/MnO2NP effectively skewed tumor-associated macrophages toward a proinflammatory phenotype and upregulated the expression of proinflammatory cytokines in tumors by up to 99-fold relative to the control. And the infiltration of CD8+ T cells was also significantly increased. When STING signaling was blocked, the antitumor effects and immunostimulatory efficacy of DHP/MnO2NP were significantly inhibited. Moreover, DHP/MnO2NP possess the advantage of enhanced tumor homing and retention, resulting in stronger and longer-lasting anticancer effects. Overall, DHP/MnO2NP provide a potential platform for potentiating cancer chemoimmunotherapy and hold promise for precision treatment.
Collapse
Affiliation(s)
- Li Liu
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, P. R. China
| | - Shengxiang Fu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, P. R. China
| | - Haojie Gu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, P. R. China
| | - Yangqian Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, P. R. China
| | - Guonian Zhu
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, P. R. China
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, P. R. China
| | - Weimin Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, P. R. China
| |
Collapse
|
19
|
Sun H, Zhong Z. Bioresponsive Polymeric Nanoparticles: From Design, Targeted Therapy to Cancer Immunotherapy. Biomacromolecules 2025; 26:33-42. [PMID: 39667037 DOI: 10.1021/acs.biomac.4c01257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Bioresponsive polymeric nanoparticles (NPs) that are capable of delivering and releasing therapeutics and biotherapeutics to target sites have attracted vivid interest in cancer therapy and immunotherapy. In contrast to enthusiastic evolution in the academic world, the clinical translation of these smart systems is scarce, partly due to concerns about safety, stability, complexity, and scalability. The moderate targetability, responsivity, and benefits are other concerns. In the past 17 years, we have devoted ourselves to exploring elegant strategies to address the above basic and translational problems by introducing diverse functional groups and/or targeting ligands to safe biomedical materials, such as biodegradable polymers and water-soluble polymers. This minimal modification is critical for further clinical translation. We have tailor-made various bioresponsive NPs including shell-sheddable and/or acid-sensitive biodegradable NPs, disulfide-cross-linked biodegradable micelles and polymersomes, and blood-brain barrier (BBB)-permeable NPs, to target different tumors. This perspective provides an overview of our work path toward targeted nanomedicines and personalized vaccines, which might inspire clinical translation and future research on cancer therapy.
Collapse
Affiliation(s)
- Huanli Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, 215222, PR China
| |
Collapse
|
20
|
Zhu A, Jiang Y, Pan L, Li J, Huang Y, Shi M, Di L, Wang L, Wang R. Cell inspired delivery system equipped with natural membrane structures in applications for rescuing ischemic stroke. J Control Release 2025; 377:54-80. [PMID: 39547421 DOI: 10.1016/j.jconrel.2024.11.013] [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: 07/04/2024] [Revised: 10/10/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Ischemic stroke (IS), accounting for 87 % of stroke incidences, constitutes a paramount health challenge owing to neurological impairments and irreversible tissue damage arising from cerebral ischemia. Chief among therapeutic obstacles are the restrictive penetration of the blood-brain barrier (BBB) and insufficient targeting precision, hindering the accumulation of drugs in ischemic brain areas. Motivated by the remarkable capabilities of natural membrane-based delivery vehicles in achieving targeted delivery and traversing the BBB, thanks to their biocompatible architecture and bioactive components, numerous membrane-engineered systems such as cells, cell membranes and extracellular vesicles have emerged as promising platforms to augment IS treatment efficacy with the help of nanotechnology. This review consolidates the primary pathological manifestations following IS, elucidates the unique functionalities of natural membrane drug delivery systems (DDSs) with nanotechnology, as well as delineates the structural characteristics of various natural membranes alongside rational design strategies employed. The review illuminates both the potential and challenges encountered when employing natural membrane DDSs in IS drug therapy, offering fresh perspectives and insights for devising efficacious and practical delivery systems tailored to IS intervention.
Collapse
Affiliation(s)
- Anran Zhu
- School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yingyu Jiang
- School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Longxiang Pan
- School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiale Li
- School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yao Huang
- School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Minghui Shi
- School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Ruoning Wang
- School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
21
|
Chen Q, Diao J. Editorial: Super-resolution imaging of sub-cellular dynamics of drug molecules. Adv Drug Deliv Rev 2025; 216:115455. [PMID: 39278312 DOI: 10.1016/j.addr.2024.115455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Affiliation(s)
- Qixin Chen
- State Key Laboratory of Advanced Drug Delivery and Release Systems, School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, PR China.
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
22
|
Rath G, Mazzali D, Zarbakhsh A, Resmini M. NIPAm Microgels Synthesised in Water: Tailored Control of Particles' Size and Thermoresponsive Properties. Polymers (Basel) 2024; 16:3532. [PMID: 39771384 PMCID: PMC11679721 DOI: 10.3390/polym16243532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Microgels, combining the properties of hydrogels and microparticles, are emerging as versatile materials for varied applications such as drug delivery and sensing, although the precise control of particle size remains a challenge. Advances in synthetic methodologies have provided new tools for tailoring of properties, however costs and scalability of the processes remains a limitation. We report here the water-based synthesis of a library of N-isopropylacrylamide-based microgels covalently crosslinked with varying contents of N,N'-methylenebisacrylamide. The results highlight the versatility of water as a synthetic medium, which yields large and monodisperse microgels, with excellent control over size. Dynamic light scattering data demonstrate that by increasing the total monomer concentration from 1 to 3 wt%, the particle size is increased by up to 4.9-fold. Crosslinker content allows fine-tuning of microgel size, with greater relevance for functionalised microgels. Functional co-monomers such as N-(3-aminopropyl)methacrylamide hydrochloride and N-(hydroxymethyl)acrylamide are shown to influence size and thermoresponsive behaviour, with hydrogen-bonding monomers reducing particle size and increasing the volume phase transition temperature by 2 °C. Positively charged monomers show a size reduction upon heating but provide colloidal stability at temperatures up to 60 °C. These findings emphasize the importance of tailoring synthetic conditions and formulations to optimize microgel properties for specific applications.
Collapse
Affiliation(s)
| | | | | | - Marina Resmini
- School of Physical & Chemical Sciences, Queen Mary University of London, Joseph Priestley Building, Mile End Road, London E1 4NS, UK; (G.R.); (D.M.); (A.Z.)
| |
Collapse
|
23
|
Hu D, Li Y, Li R, Wang M, Zhou K, He C, Wei Q, Qian Z. Recent advances in reactive oxygen species (ROS)-responsive drug delivery systems for photodynamic therapy of cancer. Acta Pharm Sin B 2024; 14:5106-5131. [PMID: 39807318 PMCID: PMC11725102 DOI: 10.1016/j.apsb.2024.10.015] [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: 06/20/2024] [Revised: 09/21/2024] [Accepted: 09/28/2024] [Indexed: 01/16/2025] Open
Abstract
Reactive oxygen species (ROS)-responsive drug delivery systems (DDSs) have garnered significant attention in cancer research because of their potential for precise spatiotemporal drug release tailored to high ROS levels within tumors. Despite the challenges posed by ROS distribution heterogeneity and endogenous supply constraints, this review highlights the strategic alliance of ROS-responsive DDSs with photodynamic therapy (PDT), enabling selective drug delivery and leveraging PDT-induced ROS for enhanced therapeutic efficacy. This review delves into the biological importance of ROS in cancer progression and treatment. We elucidate in detail the operational mechanisms of ROS-responsive linkers, including thioether, thioketal, selenide, diselencide, telluride and aryl boronic acids/esters, as well as the latest developments in ROS-responsive nanomedicines that integrate with PDT strategies. These insights are intended to inspire the design of innovative ROS-responsive nanocarriers for enhanced cancer PDT.
Collapse
Affiliation(s)
- Danrong Hu
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yicong Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ran Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kai Zhou
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyong Qian
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
24
|
Yang H, Zhang Y, Tan Z, Liu Z, Yan Y, Li Q, Saw PE, Liufu N, Ji F. Nucleus-targeted Silencer nanoplatform regulating ZEB1-AS1 in head and neck squamous cell carcinoma therapy. DISCOVER NANO 2024; 19:192. [PMID: 39579302 PMCID: PMC11585530 DOI: 10.1186/s11671-024-04148-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/04/2024] [Indexed: 11/25/2024]
Abstract
Long noncoding RNAs have emerged as key players in the progression of head and neck squamous cell carcinoma (HNSC). Among them, ZEB1-AS1 was identified as an upregulated candidate in HNSC through comprehensive analysis of RNA-sequencing datasets. Here, elevated ZEB1-AS1 expression was correlated with poor prognosis in HNSC patients. Further investigations demonstrated that downregulation of ZEB1-AS1 induced epithelial-mesenchymal transition and increased sensitivity to cisplatin in Cal27 cells, while its upregulation reversed these effects, underscoring its pivotal role in tumor metastasis and cisplatin resistance in Cal27 cells. Mechanistically, ZEB1-AS1, located in cytoplasm and nucleus, directly regulated the expression of ZEB1, thereby influencing the expression of μ opioid receptor (MOR) and implicating in cancer progression. To advance clinical translation, we employed a nucleus-targeting nanoparticle platform for efficient delivery of a mixture of antisense oligonucleotides and siRNA (Silencer), effectively manipulating ZEB1-AS1 expression in vitro and in vivo. Besides, a predictive model for HNSC patients was developed by analyzing the expression levels of ZEB1-AS1, ZEB1, and MOR in the HNSC datasets. Our study underscored the critical role of ZEB1-AS1 in HNSC and its potential as a therapeutic target. By elucidating its functional mechanisms and utilizing a nucleus-targeting nanoparticle platform for efficient delivery, we proved the potential of ZEB1-AS1-targeted therapies in HNSC.
Collapse
Affiliation(s)
- Haojie Yang
- Department of Anesthesia, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yangfan Zhang
- Department of Anesthesia, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zicong Tan
- Department of Anesthesia, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zihao Liu
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Beijing University Cancer Hospital Yunnan Hospital, Kunming, China
| | - Yingzhe Yan
- Department of Anesthesia, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Qin Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, China.
| | - Ning Liufu
- Department of Anesthesia, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Fengtao Ji
- Department of Anesthesia, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
25
|
Tortella Fuentes G, Fincheira P, Rubilar O, Leiva S, Fernandez I, Schoebitz M, Pelegrino MT, Paganotti A, dos Reis RA, Seabra AB. Nanoparticle-Based Nitric Oxide Donors: Exploring Their Antimicrobial and Anti-Biofilm Capabilities. Antibiotics (Basel) 2024; 13:1047. [PMID: 39596741 PMCID: PMC11591520 DOI: 10.3390/antibiotics13111047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Nitric oxide (NO) is an antimicrobial and anti-biofilm agent with significant potential for combating biofilm-associated infections and antibiotic resistance. However, owing to its high reactivity due to the possession of a free radical and short half-life (1-5 s), the practical application of NO in clinical settings is challenging. Objectives: This review explores the development of NO-releasing nanoparticles that provide a controlled, targeted delivery system for NO, enhancing its antimicrobial efficacy while minimizing toxicity. The review discusses various NO donors, nanoparticle platforms, and how NO disrupts biofilm formation and eradicates pathogens. Additionally, we examine the highly encouraging and inspiring results of NO-releasing nanoparticles against multidrug-resistant strains and their applications in medical and environmental contexts. This review highlights the promising role of NO-based nanotechnologies in overcoming the challenges posed by increasing antibiotic resistance and biofilm-associated infections. Conclusions: Although NO donors and nanoparticle delivery systems show great potential for antimicrobial and anti-biofilm uses, addressing challenges related to controlled release, toxicity, biofilm penetration, resistance, and clinical application is crucial.
Collapse
Affiliation(s)
- Gonzalo Tortella Fuentes
- Centro de Excelencia en Investigación Biotecnológica Aplicada al Medio Ambiente-CIBAMA, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile; (O.R.); (S.L.); (I.F.)
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile
| | - Paola Fincheira
- Centro de Excelencia en Investigación Biotecnológica Aplicada al Medio Ambiente-CIBAMA, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile; (O.R.); (S.L.); (I.F.)
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile
| | - Olga Rubilar
- Centro de Excelencia en Investigación Biotecnológica Aplicada al Medio Ambiente-CIBAMA, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile; (O.R.); (S.L.); (I.F.)
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile
| | - Sebastian Leiva
- Centro de Excelencia en Investigación Biotecnológica Aplicada al Medio Ambiente-CIBAMA, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile; (O.R.); (S.L.); (I.F.)
| | - Ivette Fernandez
- Centro de Excelencia en Investigación Biotecnológica Aplicada al Medio Ambiente-CIBAMA, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile; (O.R.); (S.L.); (I.F.)
- Programa de Doctorado en Ciencias de Recursos Naturales, Universidad de La Frontera, Temuco 4811230, Chile
| | - Mauricio Schoebitz
- Departamento de Suelos y Recursos Naturales, Facultad de Agronomía, Campus Concepción, Universidad de Concepción, Casilla 160-C, Concepción 4030000, Chile;
- Center of Biotechnology, Universidad de Concepción, Barrio Universitario s/n, Concepción 4030000, Chile
| | | | - André Paganotti
- Departamento de Farmácia, Universidade Federal de São Paulo, Diadema 09972-270, SP, Brazil
| | - Roberta Albino dos Reis
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André 09606-045, SP, Brazil; (R.A.d.R.); (A.B.S.)
| | - Amedea B. Seabra
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André 09606-045, SP, Brazil; (R.A.d.R.); (A.B.S.)
| |
Collapse
|
26
|
Boix-Montesinos P, Medel M, Malfanti A, Đorđević S, Masiá E, Charbonnier D, Carrascosa-Marco P, Armiñán A, Vicent MJ. Rational design of a poly-L-glutamic acid-based combination conjugate for hormone-responsive breast cancer treatment. J Control Release 2024; 375:193-208. [PMID: 39242032 DOI: 10.1016/j.jconrel.2024.09.002] [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: 06/17/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
Breast cancer represents the most prevalent tumor type worldwide, with hormone-responsive breast cancer the most common subtype. Despite the effectiveness of endocrine therapy, advanced disease forms represent an unmet clinical need. While drug combination therapies remain promising, differences in pharmacokinetic profiles result in suboptimal ratios of free drugs reaching tumors. We identified a synergistic combination of bisdemethoxycurcumin and exemestane through drug screening and rationally designed star-shaped poly-L-glutamic acid-based combination conjugates carrying these drugs conjugated through pH-responsive linkers for hormone-responsive breast cancer treatment. We synthesized/characterized single and combination conjugates with synergistic drug ratios/loadings. Physicochemical characterization/drug release kinetics studies suggested that lower drug loading prompted a less compact conjugate conformation that supported optimal release. Screening in monolayer and spheroid breast cancer cell cultures revealed that combination conjugates possessed enhanced cytotoxicity/synergism compared to physical mixtures of single-drug conjugates/free drugs; moreover, a combination conjugate with the lowest drug loading outperformed remaining conjugates. This candidate inhibited proliferation-associated signaling, reduced inflammatory chemokine/exosome levels, and promoted autophagy in spheroids; furthermore, it outperformed a physical mixture of single-drug conjugates/free drugs regarding cytotoxicity in patient-derived breast cancer organoids. Our findings highlight the importance of rational design and advanced in vitro models for the selection of polypeptide-based combination conjugates.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - María Medel
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain
| | - Alessio Malfanti
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Snežana Đorđević
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain
| | - Esther Masiá
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - David Charbonnier
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), IISCIII and CIEMAT, Madrid, Spain
| | - Paula Carrascosa-Marco
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - Ana Armiñán
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain.
| | - María J Vicent
- Polymer Therapeutics Lab., Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Biomédica en Red en Cancer, (CIBERONC), Instituto de Salud Carlos III, Spain; Screening Platform, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| |
Collapse
|
27
|
Yao Y, Chen Y, Zhou C, Zhang Q, He X, Dong K, Yang C, Chu B, Qian Z. Bioorthogonal chemistry-based prodrug strategies for enhanced biosafety in tumor treatments: current progress and challenges. J Mater Chem B 2024; 12:10818-10834. [PMID: 39352785 DOI: 10.1039/d4tb01413h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Cancer is a significant global health challenge, and while chemotherapy remains a widely used treatment, its non-specific toxicity and broad distribution can lead to systemic side effects and limit its effectiveness against tumors. Therefore, the development of safer chemotherapy alternatives is crucial. Prodrugs hold great promise, as they remain inactive until they reach the cancer site, where they are selectively activated by enzymes or specific factors, thereby reducing side effects and improving targeting. However, subtle differences in the microenvironments between tumors and normal tissue may still result in unintended cytotoxicity. Bioorthogonal reactions, known for their selectivity and precision without interfering with natural biochemical processes, are gaining attention. When combined with prodrug strategies, these reactions offer the potential to create highly effective chemotherapy drugs. This review examines the safety and efficacy of prodrug strategies utilizing various bioorthogonal reactions in cancer treatment.
Collapse
Affiliation(s)
- Yongchao Yao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ying Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Chang Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Quanzhi Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xun He
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Kai Dong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Chengli Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Bingyang Chu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
28
|
Guo S, Cao Y, Cheng B, Zhou Y, Li X, Zhang M, Huang Y, Wei S, Luo K, Dai R, Wang R. A nanoprodrug derived from branched poly (ethylene glycol) recognizes prostate-specific membrane antigen to precisely suppress prostate cancer progression. Int J Biol Macromol 2024; 282:136831. [PMID: 39454922 DOI: 10.1016/j.ijbiomac.2024.136831] [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: 07/17/2023] [Revised: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Prostate-specific membrane antigen (PSMA) is overexpressed in 80-90 % of prostate cancers (PCa) and is widely used as a diagnostic and therapeutic biomarker. Docetaxel (DTX), an FDA-approved anti-microtubule drug, is commonly employed to manage metastatic castration-resistant PCa; however, DTX therapy is often associated with severe side effects. One promising strategy to mitigate these side effects is the development of nanomedicine by loading small molecules into biocompatible vectors. Poly (ethylene glycol) (PEG) has been extensively used in clinical settings for this purpose, with PEGylated drugs demonstrating significant success. Compared to linear PEG, branched PEG (multi-arm PEG) provides enhanced stability for nanomedicines. In this study, we developed a novel nanoprodrug 4armPEG-Docetaxel DCL (4armPEG-DD) by conjugating a 4-arm PEG with DTX via a reduction-sensitive disulfide bond and further modifying it with 2-[3-[5-amino-1-carboxypentyl]-ureido]-pentanedioic acid (DCL), a PSMA-targeting ligand. Both in vitro and in vivo results demonstrated that the designed nanoprodrug specifically recognized PSMA-positive PCa cells and effectively released DTX in response to the intracellular reducing environment, leading to potent cytotoxic effects on PSMA-positive prostate tumors. Importantly, 4armPEG-DD exhibited improved in vivo safety compared to small-molecule DTX. Thus, we propose that 4armPEG-DD represents a promising candidate for the clinical treatment of PSMA-positive PCa.
Collapse
Affiliation(s)
- Shiwei Guo
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Yu Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Bo Cheng
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Yong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Meng Zhang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui Province 230032, China
| | - Yilan Huang
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Siping Wei
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, Guangxi Province 541004, China
| | - Kui Luo
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Huaxi MR Research Center (HMRRC), Department of Radiology, Animal Experimental Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Rongyang Dai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China; Department of Biochemistry and Molecular Biology, School of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan Province 637000, China.
| | - Ronghao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
29
|
Ismail M, Wang Y, Li Y, Liu J, Zheng M, Zou Y. Stimuli-Responsive Polymeric Nanocarriers Accelerate On-Demand Drug Release to Combat Glioblastoma. Biomacromolecules 2024; 25:6250-6282. [PMID: 39259212 DOI: 10.1021/acs.biomac.4c00722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor with a poor prognosis and limited treatment options. Drug delivery by stimuli-responsive nanocarriers holds great promise for improving the treatment modalities of GBM. At the beginning of the review, we highlighted the stimuli-active polymeric nanocarriers carrying therapies that potentially boost anti-GBM responses by employing endogenous (pH, redox, hypoxia, enzyme) or exogenous stimuli (light, ultrasonic, magnetic, temperature, radiation) as triggers for controlled drug release mainly via hydrophobic/hydrophilic transition, degradability, ionizability, etc. Modifying these nanocarriers with target ligands further enhanced their capacity to traverse the blood-brain barrier (BBB) and preferentially accumulate in glioma cells. These unique features potentially lead to more effective brain cancer treatment with minimal adverse reactions and superior therapeutic outcomes. Finally, the review summarizes the existing difficulties and future prospects in stimuli-responsive nanocarriers for treating GBM. Overall, this review offers theoretical guidelines for developing intelligent and versatile stimuli-responsive nanocarriers to facilitate precise drug delivery and treatment of GBM in clinical settings.
Collapse
Affiliation(s)
- Muhammad Ismail
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yibin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yundong Li
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan Zou
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
30
|
Ding H, Zhou C, Li T. Nanomedicines with Versatile GSH-Responsive Linkers for Cancer Theranostics. ACS Biomater Sci Eng 2024; 10:5977-5994. [PMID: 39298132 DOI: 10.1021/acsbiomaterials.4c00897] [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] [Indexed: 10/15/2024]
Abstract
Glutathione (GSH)-responsive nanomedicines have generated significant interest in biochemistry, oncology, and material sciences due to their diverse applications, including chemical and biological sensors, diagnostics, and drug delivery systems. The effectiveness of these smart GSH-responsive nanomedicines depends critically on the choice of GSH-responsive linkers. Despite their crucial role, comprehensive reviews of GSH-responsive linkers are scarce, revealing a gap in the current literature. This review addresses this gap by systematically summarizing various GSH-responsive linkers and exploring their potential applications in cancer treatment. We provide an overview of the mechanisms of action of these linkers and their bioapplications, evaluating their advantages and limitations. The insights presented aim to guide the development of advanced GSH-responsive agents for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Huamin Ding
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai 200125, China
| | - Can Zhou
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai 200125, China
| |
Collapse
|
31
|
Ray P, Sedigh A, Confeld M, Alhalhooly L, Iduoku K, Casanola-Martin GM, Pham-The H, Rasulev B, Choi Y, Yang Z, Mallik S, Quadir M. Design and evaluation of nanoscale materials with programmed responsivity towards epigenetic enzymes. J Mater Chem B 2024; 12:9905-9920. [PMID: 39021201 DOI: 10.1039/d4tb00514g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Self-assembled materials capable of modulating their assembly properties in response to specific enzymes play a pivotal role in advancing 'intelligent' encapsulation platforms for biotechnological applications. Here, we introduce a previously unreported class of synthetic nanomaterials that programmatically interact with histone deacetylase (HDAC) as the triggering stimulus for disassembly. These nanomaterials consist of co-polypeptides comprising poly(acetyl L-lysine) and poly(ethylene glycol) blocks. Under neutral pH conditions, they self-assemble into particles. The hydrodynamic diameters of particles were typically withing the range of 108-190 nm, depending on degree of acetylation of the hydrophobic block. However, their stability is compromised upon exposure to HDACs, depending on enzyme concentration and exposure time. Our investigation, utilizing HDAC8 as the model enzyme, revealed that the primary mechanism behind disassembly involves a decrease in amphiphilicity within the block copolymer due to the deacetylation of lysine residues within the particles' hydrophobic domains. To elucidate the response mechanism, we encapsulated a fluorescent dye within these nanoparticles. Upon incubation with HDAC, the nanoparticle structure collapsed, leading to controlled release of the dye over time. Notably, this release was not triggered by denatured HDAC8, other proteolytic enzymes like trypsin, or the co-presence of HDAC8 and its inhibitor. We also demonstrated the biocompatibility and cellular effects of these materials in the context of drug delivery in different types of anticancer cell lines, such as MIA PaCa-2, PANC-1, cancer like stem cells (CSCs), and non-cancerous HPNE cells. We observed that the release of a model drug (such as a STAT3 pathway inhibitor, Napabucasin) can be loaded into these nanoparticles, with >90% of the dosage can be released over 3 h under the influence of HDAC8 enzyme in a controlled fashion. Further, we conducted a comprehensive computational study to unveil the possible interaction mechanism between enzymes and particles. By drawing parallels to the mechanism of naturally occurring histone proteins, this research represents a pioneering step toward developing functional materials capable of harnessing the activity of epigenetic enzymes such as HDACs.
Collapse
Affiliation(s)
- Priyanka Ray
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| | - Abbas Sedigh
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Matthew Confeld
- Deapartment of Physics, North Dakota State University, Fargo, ND 58102, USA
| | - Lina Alhalhooly
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND 58102, USA
| | - Kweeni Iduoku
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| | - Gerardo M Casanola-Martin
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| | - Hai Pham-The
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi 10000, Vietnam
| | - Bakhtiyor Rasulev
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| | - Yongki Choi
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND 58102, USA
| | - Zhongyu Yang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| |
Collapse
|
32
|
Kong F, Liu H, Zhao C, Qin J. Targeted codelivery of doxorubicin and oleanolic acid by reduction responsive hyaluronic acid-based prodrug nano-micelles for enhanced antitumor activity and reduced toxicity. Int J Biol Macromol 2024; 277:134135. [PMID: 39069033 DOI: 10.1016/j.ijbiomac.2024.134135] [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: 04/06/2024] [Revised: 06/29/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Chemotherapy remains one of the most commonly used strategies in cancer treatment but suffers from damages to healthy tissues and organs. How to precisely co-deliver two or more drugs with different mechanisms of action to the tumors for synergistic function is a challenge for chemotherapy. Herein, Oleanolic acid (OA)-conjugated Hyaluronic acid self-assembled nano-micelles loaded with Doxorubicin (DOX) (HSO NPs/DOX) were constructed for CD44 positive cancer targeted codelivery of DOX and OA. HSO NPs/DOX exhibited reduction triggered drug release under high concentration of glutathione, more efficient uptake by 4T1 breast cancer cells than free DOX leading to higher cytotoxicity, pro-apoptotic, and migration inhibitory activities against 4T1 cells. The ex vivo biodistribution experiment demonstrated more HSO NPs/DOX were accumulated in the tumor tissues than free DOX and less in the non-tumor tissues after injections in 4T1 tumor bearing mice. More importantly, synergistic anti-tumor effects of DOX and OA were obtained using HSO NPs/DOX in 4T1 breast tumor-bearing mice and toxicity of DOX to liver and heart were circumvented through regulating the Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) and Silent Information Regulator 1 (Sirt1) expressions. Taken together, HSO NPs/DOX may become a promising codelivery system for chemotherapeutics in cancer therapy.
Collapse
Affiliation(s)
- Fei Kong
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Hengqing Liu
- School of Life Science, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Changhong Zhao
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Jingcan Qin
- Department of Radiology, Changhai Hospital, Naval Medical University, Changhai Road 168, Shanghai 200433, China.
| |
Collapse
|
33
|
Wei J, Qian Y, Bao L, Song W, Bi Y. Disulfide bonds as a molecular switch of enzyme-activatable anticancer drug precise release for fluorescence imaging and enhancing tumor therapy. Talanta 2024; 278:126394. [PMID: 38924984 DOI: 10.1016/j.talanta.2024.126394] [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: 03/21/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Enzyme-activatable drug delivery systems have been developed for cancer diagnosis and therapy. However, targeted intracellular drug delivery is a challenge for precisely tumor imaging and therapy due to the increased stability of copolymer nanoparticles (NPs) is accompanied by a notable decrease in enzyme degradation. Herein, disulfide bond was designed as an enzyme-activatable molecular switch of SS-P(G2)2/DOX NPs. The copolymer NPs consists of polyvinylpyrrolidone (PVP) with disulfide bonds in the center and enzyme-degradable peptide dendrites (Phe-Lys) to form dendritic-linear-dendritic triblock copolymers (TBCs). The amphiphilic TBCs could be split into two identical amphiphilic diblock copolymers (DBCs) by glutathione (GSH) in cancer cells specifically while maintaining the same hydrophilic-lipophilic equilibrium. This structural transformation significantly reduced the stability of copolymer NPs and enhanced sensitivity of DOX release by cathepsin B-activated. Subsequently, the released DOX acted as an indicator of fluorescence imaging and chemotherapy drug for cancer cells. The polymeric NPs achieved excellent drug-loaded stability and prolonged blood circulation in vivo, and realized fluorescence imaging and specific cancer cell killing capabilities by responding to the overexpression of GSH and cathepsin B in tumor cells. Furthermore, the copolymer NPs demonstrated excellent blood compatibility and biosafety. Therefore, a novel strategy based on one tumor marker acting as the switch for another tumor microenvironment responsive drug delivery system could be designed for tumor intracellular imaging and chemotherapy.
Collapse
Affiliation(s)
- Junwu Wei
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, PR China
| | - Yangyang Qian
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, PR China; College of Tea (Pu'er), West Yunnan University of Applied Sciences, Pu'er, 665000, PR China
| | - Lijun Bao
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, PR China
| | - Wenjie Song
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, PR China
| | - Yunmei Bi
- College of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming, 650500, PR China.
| |
Collapse
|
34
|
Badparvar F, Poursattar Marjani A, Salehi R, Ramezani F, Beyrampour Basmenj H, Talebi M. Dual pH/redox-responsive size-switchable polymeric nano-carrier system for tumor microenvironment DTX release. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:2220-2249. [PMID: 38944817 DOI: 10.1080/09205063.2024.2371203] [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: 01/23/2024] [Accepted: 06/18/2024] [Indexed: 07/01/2024]
Abstract
Innovation chemotherapeutic nano drug delivery systems (NDDSs) with various pharmacological achievement have become one of the hopeful therapeutic strategies in cancer therapy. This study focused on low pH, and high levels of glutathione (GSH) as two prominent characteristics of the tumor microenvironment (TME) to design a novel TME-targeted pH/redox dual-responsive P (AMA-co-DMAEMA)-b-PCL-SS-PCL-b-P (AMA-co-DMAEMA) nanoparticles (NPs) for deep tumor penetration and targeted anti-tumor therapy. The positively charged NPs exhibit strong electrostatic interactions with negatively charged cell membranes, significantly enhancing cellular uptake. Moreover, these NPs possess the unique size-shrinkable property, transitioning from 98.24 ± 27.78 to 45.56 ± 20.62 nm within the TME. This remarkable size change fosters an impressive uptake of approximately 100% by MDA-MB-231 cells within just 30 min, thereby greatly improving drug delivery efficiency. This size switchability enables passive targeting through the enhanced permeability and retention (EPR) effect, facilitating deep penetration into tumors. The NPs also demonstrate improved pH/redox-triggered drug release (∼70% at 24 h) within the TME and exhibit no toxicity in cell viability test. The cell cycle results of treated cells with docetaxel (DTX)-loaded NPs revealed G2/M (84.6 ± 1.16%) arrest. The DTX-loaded NPs showed more apoptosis (62.6 ± 3.7%) than the free DTX (51.8 ± 3.2%) in treated cells. The western blot and RT-PCR assays revealed that apoptotic genes and proteins expression of treated cells were significantly upregulated with the DTX-loaded NPs vs. the free DTX (Pvalue<.001). In conclusion, these findings suggest that this novel-engineered NPs holds promise as a TME-targeted NDDS.
Collapse
Affiliation(s)
- Fahimeh Badparvar
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia, Iran
| | | | - Roya Salehi
- Clinical Research Development Unite of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ramezani
- Department of Molecular Mediciene, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Beyrampour Basmenj
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Department of Applied Cell Science, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Meng X, Shen Y, Zhao H, Lu X, Wang Z, Zhao Y. Redox-manipulating nanocarriers for anticancer drug delivery: a systematic review. J Nanobiotechnology 2024; 22:587. [PMID: 39342211 PMCID: PMC11438196 DOI: 10.1186/s12951-024-02859-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024] Open
Abstract
Spatiotemporally controlled cargo release is a key advantage of nanocarriers in anti-tumor therapy. Various external or internal stimuli-responsive nanomedicines have been reported for their ability to increase drug levels at the diseased site and enhance therapeutic efficacy through a triggered release mechanism. Redox-manipulating nanocarriers, by exploiting the redox imbalances in tumor tissues, can achieve precise drug release, enhancing therapeutic efficacy while minimizing damage to healthy cells. As a typical redox-sensitive bond, the disulfide bond is considered a promising tool for designing tumor-specific, stimulus-responsive drug delivery systems (DDS). The intracellular redox imbalance caused by tumor microenvironment (TME) regulation has emerged as an appealing therapeutic target for cancer treatment. Sustained glutathione (GSH) depletion in the TME by redox-manipulating nanocarriers can exacerbate oxidative stress through the exchange of disulfide-thiol bonds, thereby enhancing the efficacy of ROS-based cancer therapy. Intriguingly, GSH depletion is simultaneously associated with glutathione peroxidase 4 (GPX4) inhibition and dihydrolipoamide S-acetyltransferase (DLAT) oligomerization, triggering mechanisms such as ferroptosis and cuproptosis, which increase the sensitivity of tumor cells. Hence, in this review, we present a comprehensive summary of the advances in disulfide based redox-manipulating nanocarriers for anticancer drug delivery and provide an overview of some representative achievements for combinational therapy and theragnostic. The high concentration of GSH in the TME enables the engineering of redox-responsive nanocarriers for GSH-triggered on-demand drug delivery, which relies on the thiol-disulfide exchange reaction between GSH and disulfide-containing vehicles. Conversely, redox-manipulating nanocarriers can deplete GSH, thereby enhancing the efficacy of ROS-based treatment nanoplatforms. In brief, we summarize the up-to-date developments of the redox-manipulating nanocarriers for cancer therapy based on DDS and provide viewpoints for the establishment of more stringent anti-tumor nanoplatform.
Collapse
Affiliation(s)
- Xuan Meng
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, P.R. China.
| | - Yongli Shen
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, P.R. China
| | - Huanyu Zhao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, P.R. China
| | - Xinlei Lu
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, P.R. China
| | - Zheng Wang
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Yanjun Zhao
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China.
| |
Collapse
|
36
|
Yao L, Chu C, Li Y, Cao L, Yang J, Mu W. Sonodynamic effect based on vancomycin-loaded microbubbles or meropenem-loaded microbubbles enhances elimination of different biofilms and bactericidal efficacy. Bone Joint Res 2024; 13:441-451. [PMID: 39222931 PMCID: PMC11368542 DOI: 10.1302/2046-3758.139.bjr-2023-0319.r3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Aims This study investigated vancomycin-microbubbles (Vm-MBs) and meropenem (Mp)-MBs with ultrasound-targeted microbubble destruction (UTMD) to disrupt biofilms and improve bactericidal efficiency, providing a new and promising strategy for the treatment of device-related infections (DRIs). Methods A film hydration method was used to prepare Vm-MBs and Mp-MBs and examine their characterization. Biofilms of methicillin-resistant Staphylococcus aureus (MRSA) and Escherichia coli were treated with different groups. Biofilm biomass differences were determined by staining. Thickness and bacterial viability were observed with confocal laser scanning microscope (CLSM). Colony counts were determined by plate-counting. Scanning electron microscopy (SEM) observed bacterial morphology. Results The Vm-MBs and Mp-MBs met the experimental requirements. The biofilm biomass in the Vm, Vm-MBs, UTMD, and Vm-MBs + UTMD groups was significantly lower than in the control group. MRSA and E. coli biofilms were most notably damaged in the Vm-MBs + UTMD group and Mp-MBs + UTMD group, respectively, with mean 21.55% (SD 0.08) and 19.73% (SD 1.25) remaining in the biofilm biomass. Vm-MBs + UTMD significantly reduced biofilm thickness and bacterial viability (p = 0.005 and p < 0.0001, respectively). Mp-MBs + UTMD could significantly decrease biofilm thickness and bacterial viability (allp < 0.001). Plate-counting method showed that the numbers of MRSA and E. coli bacterial colonies were significantly lower in the Vm-MBs + UTMD group and the Mp, Mp-MBs, UTMD, Mp-MBs + UTMD groups compared to the control group (p = 0.031). SEM showed that the morphology and structure of MRSA and E. coli were significantly damaged in the Vm-MBs + UTMD and Mp-MBs + UTMD groups. Conclusion Vm-MBs or Mp-MBs combined with UTMD can effectively disrupt biofilms and protectively release antibiotics under ultrasound mediation, significantly reducing bacterial viability and improving the bactericidal effect of antibiotics.
Collapse
Affiliation(s)
- Liqin Yao
- Department of Sports Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chenghan Chu
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yicheng Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Li Cao
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Key Laboratory of High Incidence Disease Research in Xinjiang (Xinjiang Medical University), Ministry of Education, Urumqi, China
| | - Jianhua Yang
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Wenbo Mu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
37
|
Liu J, Liu J, Mu W, Ma Q, Zhai X, Jin B, Liu Y, Zhang N. Delivery Strategy to Enhance the Therapeutic Efficacy of Liver Fibrosis via Nanoparticle Drug Delivery Systems. ACS NANO 2024; 18:20861-20885. [PMID: 39082637 DOI: 10.1021/acsnano.4c02380] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Liver fibrosis (LF) is a pathological repair reaction caused by a chronic liver injury that affects the health of millions of people worldwide, progressing to life-threatening cirrhosis and liver cancer without timely intervention. Due to the complexity of LF pathology, multiple etiological characteristics, and the deposited extracellular matrix, traditional drugs cannot reach appropriate targets in a time-space matching way, thus decreasing the therapeutic effect. Nanoparticle drug delivery systems (NDDS) enable multidrug co-therapy and develop multifactor delivery strategies targeting pathological processes, showing great potential in LF therapy. Based on the pathogenesis and the current clinical treatment status of LF, we systematically elucidate the targeting mechanism of NDDS used in the treatment of LF. Subsequently, we focus on the progress of drug delivery applications for LF, including combined delivery for the liver fibrotic pathological environment, overcoming biological barriers, precise intracellular regulation, and intelligent responsive delivery for the liver fibrotic microenvironment. We hope that this review will inspire the rational design of NDDS for LF in the future in order to provide ideas and methods for promoting LF regression and cure.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jinhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiangyu Zhai
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Bin Jin
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
38
|
Yang Y, Li P, Feng H, Zeng R, Li S, Zhang Q. Macrocycle-Based Supramolecular Drug Delivery Systems: A Concise Review. Molecules 2024; 29:3828. [PMID: 39202907 PMCID: PMC11357536 DOI: 10.3390/molecules29163828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Efficient delivery of therapeutic agents to the lesion site or specific cells is an important way to achieve "toxicity reduction and efficacy enhancement". Macrocycles have always provided many novel ideas for drug or gene loading and delivery processes. Specifically, macrocycles represented by crown ethers, cyclodextrins, cucurbit[n]urils, calix[n]arenes, and pillar[n]arenes have unique properties, which are different cavity structures, good biocompatibility, and good stability. Benefited from these diverse properties, a variety of supramolecular drug delivery systems can be designed and constructed to effectively improve the physical and chemical properties of guest molecules as needed. This review provides an outlook on the current application status and main limitations of macrocycles in supramolecular drug delivery systems.
Collapse
Affiliation(s)
- Yanrui Yang
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Pengcheng Li
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Haibo Feng
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Rui Zeng
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Shanshan Li
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Qixiong Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Department of Pharmacy, Sichuan Provincial People’s Hospital Chuandong Hospital & Dazhou First People’s Hospital, Dazhou 635000, China
| |
Collapse
|
39
|
Cho H, Huh KM, Shim MS, Cho YY, Lee JY, Lee HS, Kang HC. Beyond Nanoparticle-Based Intracellular Drug Delivery: Cytosol/Organelle-Targeted Drug Release and Therapeutic Synergism. Macromol Biosci 2024; 24:e2300590. [PMID: 38488862 DOI: 10.1002/mabi.202300590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/07/2024] [Indexed: 07/16/2024]
Abstract
Nanoparticle (NP)-based drug delivery systems are conceived to solve poor water-solubility and chemical/physical instability, and their purpose expanded to target specific sites for maximizing therapeutic effects and minimizing unwanted events of payloads. Targeted sites are also narrowed from organs/tissues and cells to cytosol/organelles. Beyond specific site targeting, the particular release of payloads at the target sites is growing in importance. This review overviews various issues and their general strategies during multiple steps, from the preparation of drug-loaded NPs to their drug release at the target cytosol/organelles. In particular, this review focuses on current strategies for "first" delivery and "later" release of drugs to the cytosol or organelles of interest using specific stimuli in the target sites. Recognizing or distinguishing the presence/absence of stimuli or their differences in concentration/level/activity in one place from those in another is applied to stimuli-triggered release via bond cleavage or nanostructural transition. In addition, future directions on understanding the intracellular balance of stimuli and their counter-stimuli are demonstrated to synergize the therapeutic effects of payloads released from stimuli-sensitive NPs.
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| |
Collapse
|
40
|
Wu H, Zhang Z, Cao Y, Hu Y, Li Y, Zhang L, Cao X, Wen H, Zhang Y, Lv H, Jin X. A Self-Amplifying ROS-Responsive Nanoplatform for Simultaneous Cuproptosis and Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401047. [PMID: 38569217 PMCID: PMC11187900 DOI: 10.1002/advs.202401047] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/21/2024] [Indexed: 04/05/2024]
Abstract
Cuproptosis is an emerging cell death pathway that depends on the intracellular Cu ions. Elesclomol (ES) as an efficient Cu ionophore can specifically transport Cu into mitochondria and trigger cuproptosis. However, ES can be rapidly removed and metabolized during intravenous administration, leading to a short half-life and limited tumor accumulation, which hampers its clinical application. Here, the study develops a reactive oxygen species (ROS)-responsive polymer (PCP) based on cinnamaldehyde (CA) and polyethylene glycol (PEG) to encapsulate ES-Cu compound (EC), forming ECPCP. ECPCP significantly prolongs the systemic circulation of EC and enhances its tumor accumulation. After cellular internalization, the PCP coating stimulatingly dissociates exposing to the high-level ROS, and releases ES and Cu, thereby triggering cell death via cuproptosis. Meanwhile, Cu2+-stimulated Fenton-like reaction together with CA-stimulated ROS production simultaneously breaks the redox homeostasis, which compensates for the insufficient oxidative stress treated with ES alone, in turn inducing immunogenic cell death of tumor cells, achieving simultaneous cuproptosis and immunotherapy. Furthermore, the excessive ROS accelerates the stimuli-dissociation of ECPCP, forming a positive feedback therapy loop against tumor self-alleviation. Therefore, ECPCP as a nanoplatform for cuproptosis and immunotherapy improves the dual antitumor mechanism of ES and provides a potential optimization for ES clinical application.
Collapse
Affiliation(s)
- Hangyi Wu
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjing211198China
| | - Zhenhai Zhang
- Jiangsu Province Academy of Traditional Chinese MedicineNanjing210023China
| | - Yanni Cao
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjing211198China
| | - Yuhan Hu
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjing211198China
| | - Yi Li
- Department of PharmaceuticsThe Affiliated Suqian First People's Hospital of Nanjing Medical UniversitySuqianJiangsu223800China
| | - Lanyi Zhang
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjing211198China
| | - Xinyi Cao
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjing211198China
| | - Haitong Wen
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjing211198China
| | - Youwen Zhang
- Department of PharmaceuticsThe Affiliated Suqian First People's Hospital of Nanjing Medical UniversitySuqianJiangsu223800China
| | - Huixia Lv
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjing211198China
| | - Xin Jin
- Department of PharmaceuticsThe Affiliated Suqian First People's Hospital of Nanjing Medical UniversitySuqianJiangsu223800China
| |
Collapse
|
41
|
Zhang Y, Fang Z, Pan D, Li Y, Zhou J, Chen H, Li Z, Zhu M, Li C, Qin L, Ren X, Gong Q, Luo K. Dendritic Polymer-Based Nanomedicines Remodel the Tumor Stroma: Improve Drug Penetration and Enhance Antitumor Immune Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401304. [PMID: 38469918 DOI: 10.1002/adma.202401304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/09/2024] [Indexed: 03/13/2024]
Abstract
The dense extracellular matrix (ECM) in solid tumors, contributed by cancer-associated fibroblasts (CAFs), hinders penetration of drugs and diminishes their therapeutic outcomes. A sequential treatment strategy of remodeling the ECM via a CAF modifier (dasatinib, DAS) is proposed to promote penetration of an immunogenic cell death (ICD) inducer (epirubicin, Epi) via apoptotic vesicles, ultimately enhancing the treatment efficacy against breast cancer. Dendritic poly(oligo(ethylene glycol) methyl ether methacrylate) (POEGMA)-based nanomedicines (poly[OEGMA-Dendron(G2)-Gly-Phe-Leu-Gly-DAS] (P-DAS) and poly[OEGMA-Dendron(G2)-hydrazone-Epi] (P-Epi)) are developed for sequential delivery of DAS and Epi, respectively. P-DAS reprograms CAFs to reduce collagen by downregulating collagen anabolism and energy metabolism, thereby reducing the ECM deposition. The regulated ECM can enhance tumor penetration of P-Epi to strengthen its ICD effect, leading to an amplified antitumor immune response. In breast cancer-bearing mice, this approach alleviates the ECM barrier, resulting in reduced tumor burden and increased cytotoxic T lymphocyte infiltration, and more encouragingly, synergizes effectively with anti-programmed cell death 1 (PD-1) therapy, significantly inhibiting tumor growth and preventing lung metastasis. Furthermore, systemic toxicity is barely detectable after sequential treatment with P-DAS and P-Epi. This approach opens a new avenue for treating desmoplastic tumors by metabolically targeting CAFs to overcome the ECM barrier.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zaixiang Fang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Zhou
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongying Chen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiqian Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengli Zhu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cong Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liwen Qin
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiangyi Ren
- Department of Radiology, Huaxi MR Research Center (HMRRC), 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), 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, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), 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, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
42
|
Liu Y, Hamm T, Eichinger TR, Kamm W, Wieland HA, Loretz B, Hirsch AKH, Lee S, Lehr CM. Biodynamer Nano-Complexes and -Emulsions for Peptide and Protein Drug Delivery. Int J Nanomedicine 2024; 19:4429-4449. [PMID: 38784761 PMCID: PMC11114140 DOI: 10.2147/ijn.s448578] [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: 11/07/2023] [Accepted: 03/26/2024] [Indexed: 05/25/2024] Open
Abstract
Background Therapeutic proteins and peptides offer great advantages compared to traditional synthetic molecular drugs. However, stable protein loading and precise control of protein release pose significant challenges due to the extensive range of physicochemical properties inherent to proteins. The development of a comprehensive protein delivery strategy becomes imperative accounting for the diverse nature of therapeutic proteins. Methods Biodynamers are amphiphilic proteoid dynamic polymers consisting of amino acid derivatives connected through pH-responsive dynamic covalent chemistry. Taking advantage of the amphiphilic nature of the biodynamers, PNCs and DEs were possible to be prepared and investigated to compare the delivery efficiency in drug loading, stability, and cell uptake. Results As a result, the optimized PNCs showed 3-fold encapsulation (<90%) and 5-fold loading capacity (30%) compared to DE-NPs. PNCs enhanced the delivery efficiency into the cells but aggregated easily on the cell membrane due to the limited stability. Although DE-NPs were limited in loading capacity compared to PNCs, they exhibit superior adaptability in stability and capacity for delivering a wider range of proteins compared to PNCs. Conclusion Our study highlights the potential of formulating both PNCs and DE-NPs using the same biodynamers, providing a comparative view on protein delivery efficacy using formulation methods.
Collapse
Affiliation(s)
- Yun Liu
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Timo Hamm
- Department of Research and Development, Sanofi-Aventis Deutschland GmbH, Frankfurt Am Main, Germany
| | - Thomas Ralf Eichinger
- Department of Research and Development, Sanofi-Aventis Deutschland GmbH, Frankfurt Am Main, Germany
| | - Walter Kamm
- Department of Research and Development, Sanofi-Aventis Deutschland GmbH, Frankfurt Am Main, Germany
| | - Heike Andrea Wieland
- Department of Research and Development, Sanofi-Aventis Deutschland GmbH, Frankfurt Am Main, Germany
| | - Brigitta Loretz
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Anna K H Hirsch
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Department of Drug Design and Optimisation, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Sangeun Lee
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| |
Collapse
|
43
|
Wang G, Li Z, Wang G, Sun Q, Lin P, Wang Q, Zhang H, Wang Y, Zhang T, Cui F, Zhong Z. Advances in Engineered Nanoparticles for the Treatment of Ischemic Stroke by Enhancing Angiogenesis. Int J Nanomedicine 2024; 19:4377-4409. [PMID: 38774029 PMCID: PMC11108071 DOI: 10.2147/ijn.s463333] [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: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Gongchen Wang
- Department of Vascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People’s Hospital, Yantai, Shandong, 265600, People’s Republic of China
| | - Peng Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Feiyun Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
44
|
Zhao J, Li X, Ma T, Chang B, Zhang B, Fang J. Glutathione-triggered prodrugs: Design strategies, potential applications, and perspectives. Med Res Rev 2024; 44:1013-1054. [PMID: 38140851 DOI: 10.1002/med.22007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
The burgeoning prodrug strategy offers a promising avenue toward improving the efficacy and specificity of cytotoxic drugs. Elevated intracellular levels of glutathione (GSH) have been regarded as a hallmark of tumor cells and characteristic feature of the tumor microenvironment. Considering the pivotal involvement of elevated GSH in the tumorigenic process, a diverse repertoire of GSH-triggered prodrugs has been developed for cancer therapy, facilitating the attenuation of deleterious side effects associated with conventional chemotherapeutic agents and/or the attainment of more efficacious therapeutic outcomes. These prodrug formulations encompass a spectrum of architectures, spanning from small molecules to polymer-based and organic-inorganic nanomaterial constructs. Although the GSH-triggered prodrugs have been gaining increasing interests, a comprehensive review of the advancements made in the field is still lacking. To fill the existing lacuna, this review undertakes a retrospective analysis of noteworthy research endeavors, based on a categorization of these molecules by their diverse recognition units (i.e., disulfides, diselenides, Michael acceptors, and sulfonamides/sulfonates). This review also focuses on explaining the distinct benefits of employing various chemical architecture strategies in the design of these prodrug agents. Furthermore, we highlight the potential for synergistic functionality by incorporating multiple-targeting conjugates, theranostic entities, and combinational treatment modalities, all of which rely on the GSH-triggering. Overall, an extensive overview of the emerging field is presented in this review, highlighting the obstacles and opportunities that lie ahead. Our overarching goal is to furnish methodological guidance for the development of more efficacious GSH-triggered prodrugs in the future. By assessing the pros and cons of current GSH-triggered prodrugs, we expect that this review will be a handful reference for prodrug design, and would provide a guidance for improving the properties of prodrugs and discovering novel trigger scaffolds for constructing GSH-triggered prodrugs.
Collapse
Affiliation(s)
- Jintao Zhao
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Xinming Li
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Tao Ma
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Bingbing Chang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
45
|
Pang Y, Lv J, He C, Ju C, Lin Y, Zhang C, Li M. Covalent organic frameworks-derived carbon nanospheres based nanoplatform for tumor specific synergistic therapy via oxidative stress amplification and calcium overload. J Colloid Interface Sci 2024; 661:908-922. [PMID: 38330663 DOI: 10.1016/j.jcis.2024.01.217] [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: 10/30/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024]
Abstract
Combinational therapy in cancer treatment that integrates the merits of different therapies is an effective approach to improve therapeutic outcomes. Herein, a simple nanoplatform (N-CNS-CaO2-HA/Ce6 NCs) that synergized chemodynamic therapy (CDT), photodynamic therapy (PDT), photothermal therapy (PTT), and Ca2+ interference therapy (CIT) has been developed to combat hypoxic tumors. With high photothermal effect, excellent peroxidase-like activity, and inherent mesoporous structure, N-doped carbon nanospheres (N-CNSs) were prepared via in situ pyrolysis of an established nanoscale covalent organic frameworks (COFs) precursor. These N-CNSs acted as PTT/CDT agents and carriers for the photosensitizer chlorin e6 (Ce6), thereby yielding a minimally invasive PDT/PTT/CDT synergistic therapy. Hyaluronic acid (HA)-modified CaO2 nanoparticles (CaO2-HA NPs) coated on the surface of the nanoplatform endowed the nanoplatform with O2/H2O2 self-supply capability to respond to and modulate the tumor microenvironment (TME), which greatly facilitated the tumor-specific performance of CDT and PDT. Moreover, the reactive oxygen species (ROS) produced during PDT and CDT enhanced the Ca2+ overloading due to CaO2 decomposition, amplifying the intracellular oxidative stress and leading to mitochondrial dysfunction. Notably, the HA molecules not only increased the cancer-targeting efficiency but also prevented CaO2 degradation during blood circulation, providing double insurance of tumor-selective CIT. Such a nanotherapeutic system possessed boosted antitumor efficacy with minimized systemic toxicity and showed great potential for treating hypoxic tumors.
Collapse
Affiliation(s)
- Yu Pang
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Jie Lv
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China; Postdoctoral Mobile Station of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Chengcai He
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Chengda Ju
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Yulong Lin
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Cong Zhang
- Hebei Provincial Key Laboratory of Photoelectric Control on Surface and Interface, College of Science, Hebei University of Science and Technology, Shijiazhuang 050018, China.
| | - Meng Li
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
46
|
Yin B, Wang R, Guo Y, Li L, Hu X. Injectable Thermo-Responsive Peptide Hydrogels and Its Enzyme Triggered Dynamic Self-Assembly. Polymers (Basel) 2024; 16:1221. [PMID: 38732690 PMCID: PMC11085460 DOI: 10.3390/polym16091221] [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: 04/02/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Endogenous stimuli-responsive injectable hydrogels hold significant promise for practical applications due to their spatio-temporal controllable drug delivery. Herein, we report a facile strategy to construct a series of in situ formation polypeptide hydrogels with thermal responsiveness and enzyme-triggered dynamic self-assembly. The thermo-responsive hydrogels are from the diblock random copolymer mPEG-b-P(Glu-co-Tyr). The L-glutamic acid (Glu) segments with different γ-alkyl groups, including methyl, ethyl, and n-butyl, offer specific secondary structure, facilitating the formation of hydrogel. The L-tyrosine (Tyr) residues not only provide hydrogen-bond interactions and thus adjust the sol-gel transition temperatures, but also endow polypeptide enzyme-responsive properties. The PTyr segments could be phosphorylated, and the phosphotyrosine copolymers were amphiphilies, which could readily self-assemble into spherical aggregates and transform into sheet-like structures upon dephosphorylation by alkaline phosphatase (ALP). P(MGlu-co-Tyr/P) and P(MGlu-co-Tyr) copolymers showed good compatibility with both MC3T3-E1 and Hela cells, with cell viability above 80% at concentrations up to 1000 μg/mL. The prepared injectable polypeptide hydrogel and its enzyme-triggered self-assemblies show particular potential for biomedical applications.
Collapse
Affiliation(s)
| | | | | | | | - Xiuli Hu
- Institute of Polymer Science and Engineering, School of Chemical Engineering, Hebei University of Technology, Tianjin 300130, China; (B.Y.); (R.W.); (Y.G.); (L.L.)
| |
Collapse
|
47
|
Hu Y, Liu P. Diselenide-Bridged Doxorubicin Dimeric Prodrug: Synthesis and Redox-Triggered Drug Release. Molecules 2024; 29:1709. [PMID: 38675530 PMCID: PMC11052396 DOI: 10.3390/molecules29081709] [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: 02/14/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The diselenide bond has attracted intense interest in redox-responsive drug delivery systems (DDSs) in tumor chemotherapy, due to its higher sensitivity than the most investigated bond, namely the disulfide bond. Here, a diselenide-bridged doxorubicin dimeric prodrug (D-DOXSeSe) was designed by coupling two doxorubicin molecules with a diselenodiacetic acid (DSeDAA) molecule via α-amidation, as a redox-triggered drug self-delivery system (DSDS) for tumor-specific chemotherapy. The drug release profiles indicated that the D-DOXSeSe could be cleaved to release the derivatives selenol (DOX-SeH) and seleninic acid (DOX-SeOOH) with the triggering of high GSH and H2O2, respectively, indicating the double-edged sword effect of the lower electronegativity of the selenide atom. The resultant solubility-controlled slow drug release performance makes it a promising candidate as a long-acting DSDS in future tumor chemotherapy. Moreover, the interaction between the conjugations in the design of self-immolation traceless linkers was also proposed for the first time as another key factor for a desired precise tumor-specific chemotherapy, besides the conjugations themselves.
Collapse
Affiliation(s)
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
48
|
Pegoraro C, Domingo-Ortí I, Conejos-Sánchez I, Vicent MJ. Unlocking the Mitochondria for Nanomedicine-based Treatments: Overcoming Biological Barriers, Improving Designs, and Selecting Verification Techniques. Adv Drug Deliv Rev 2024; 207:115195. [PMID: 38325562 DOI: 10.1016/j.addr.2024.115195] [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: 10/24/2023] [Revised: 01/13/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Enhanced targeting approaches will support the treatment of diseases associated with dysfunctional mitochondria, which play critical roles in energy generation and cell survival. Obstacles to mitochondria-specific targeting include the presence of distinct biological barriers and the need to pass through (or avoid) various cell internalization mechanisms. A range of studies have reported the design of mitochondrially-targeted nanomedicines that navigate the complex routes required to influence mitochondrial function; nonetheless, a significant journey lies ahead before mitochondrially-targeted nanomedicines become suitable for clinical use. Moving swiftly forward will require safety studies, in vivo assays confirming effectiveness, and methodologies to validate mitochondria-targeted nanomedicines' subcellular location/activity. From a nanomedicine standpoint, we describe the biological routes involved (from administration to arrival within the mitochondria), the features influencing rational design, and the techniques used to identify/validate successful targeting. Overall, rationally-designed mitochondria-targeted-based nanomedicines hold great promise for precise subcellular therapeutic delivery.
Collapse
Affiliation(s)
- Camilla Pegoraro
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inés Domingo-Ortí
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
49
|
Ali I, Rizwan A, Vu TT, Jo SH, Oh CW, Kim YH, Park SH, Lim KT. NIR-responsive carboxymethyl-cellulose hydrogels containing thioketal-linkages for on-demand drug delivery system. Int J Biol Macromol 2024; 260:129549. [PMID: 38246444 DOI: 10.1016/j.ijbiomac.2024.129549] [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: 11/02/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Near-infrared (NIR) light-responsive hydrogels have emerged as a highly promising strategy for effective anticancer therapy owing to the remotely controlled release of chemotherapeutic molecules with minimal invasive manner. In this study, novel NIR-responsive hydrogels were developed from reactive oxygen species (ROS)-cleavable thioketal cross-linkers which possessed terminal tetrazine groups to undergo a bio-orthogonal inverse electron demand Diels Alder click reaction with norbornene modified carboxymethyl cellulose. The hydrogels were rapidly formed under physiological conditions and generated N2 gas as a by-product, which led to the formation of porous structures within the hydrogel networks. A NIR dye, indocyanine green (ICG) and chemotherapeutic doxorubicin (DOX) were co-encapsulated in the porous network of the hydrogels. Upon NIR-irradiation, the hydrogels showed spatiotemporal release of encapsulated DOX (>96 %) owing to the cleavage of thioketal bonds by interacting with ROS generated from ICG, whereas minimal release of encapsulated DOX (<25 %) was observed in the absence of NIR-light. The in vitro cytotoxicity results revealed that the hydrogels were highly cytocompatible and did not induce any toxic effect on the HEK-293 cells. In contrast, the DOX + ICG-encapsulated hydrogels enhanced the chemotherapeutic effect and effectively inhibited the proliferation of Hela cancer cells when irradiated with NIR-light.
Collapse
Affiliation(s)
- Israr Ali
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Ali Rizwan
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Trung Thang Vu
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Sung-Han Jo
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Chul-Woong Oh
- Department of Marine Biology, College of Fisheries Sciences, Pukyong National University, Busan, Republic of Korea
| | - Yong Hyun Kim
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Sang-Hyug Park
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Republic of Korea.
| | - Kwon Taek Lim
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, Republic of Korea; Institute of Display Semiconductor Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
50
|
Shi J, Wang Y, Wu Y, Li J, Fu C, Li Y, Xie X, Fan X, Hu Y, Hu C, Zhang J. Tumor Microenvironment ROS/pH Cascade-Responsive Supramolecular Nanoplatform with ROS Regeneration Property for Enhanced Hepatocellular Carcinoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:7576-7592. [PMID: 38316581 DOI: 10.1021/acsami.3c16022] [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: 02/07/2024]
Abstract
The low targeted drug delivery efficiency, including poor tumor accumulation and penetration and uncontrolled drug release, leads to the failure of cancer therapy. Herein, a multifunctional supramolecular nanoplatform loading triptolide (TPL/PBAETK@GA NPs) was fabricated via the host-guest interaction between glycyrrhetinic-acid-modified poly(ethylene glycol)-adamantanecarboxylic acid moiety and reactive oxygen species (ROS)/pH cascade-responsive copolymer poly(β-amino esters)-thioketal (TK)-β-cyclodextrin. TPL/PBAETK@GA NPs could accumulate in hepatocellular carcinoma (HCC) tissue effectively, mediated by nanoscale advantage and GA' recognition to specific receptors. The elevated concentration of ROS in tumor microenvironment (TME) quickly breaks the TK linkages, causing the detachment of shell (cyclodextrin) CD layer. Then, the accompanying negative-to-positive charge-reversal of NPs was realized via the PBAE moiety protonation under the slightly acidic TME, significantly enhancing the NPs' cellular internalization. Remarkably, the pH-responsive endo/lysosome escape of PBAE core triggered intracellular TPL burst release, promoting the cancer cell apoptosis, autophagy, and intracellular ROS generation, leading to the self-amplification of ROS in TME. Afterward, the ROS positive-feedback loop was generated to further promote size-shrinkage and charge-reversal of NPs. Both in vitro and in vivo tests verified that TPL/PBAETK@GA NPs produced a satisfactory anti-HCC therapy outcome. Collectively, this study offers a potential appealing paradigm to enhance TPL-based HCC therapy outcomes via multifunctionalized supramolecular nanodrugs.
Collapse
Affiliation(s)
- Jinfeng Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yehui Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Kowloon, Hong Kong SAR 999077, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi Li
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Xingliang Xie
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Xiaohong Fan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yichen Hu
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Chuan Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|