1
|
Tan K, Zhang H, Yang J, Wang H, Li Y, Ding G, Gu P, Yang S, Li J, Fan X. Organelle-oriented nanomedicines in tumor therapy: Targeting, escaping, or collaborating? Bioact Mater 2025; 49:291-339. [PMID: 40161442 PMCID: PMC11953998 DOI: 10.1016/j.bioactmat.2025.02.040] [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: 12/25/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Precise tumor therapy is essential for improving treatment specificity, enhancing efficacy, and minimizing side effects. Targeting organelles is a key strategy for achieving this goal and is a frontier research area attracting a considerable amount of attention. The concept of organelle targeting has a significant effect on the structural design of the nanodrugs employed. Most notably, the intricate interactions among different organelles in a tumor cell essentially create a unified system. Unfortunately, this aspect might have been somewhat overlooked when existing organelle-targeting nanodrugs were designed. In this review, we underscore the synergistic relationship among the various organelles and advocate for a holistic view of organelle-targeting design. Through the integration of biology and material science, recent advancements in organelle targeting, escaping, and collaborating are consolidated to offer fresh perspectives for the development of antitumor nanomedicines.
Collapse
Affiliation(s)
- Kexin Tan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Haiyang Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Jianyuan Yang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Hang Wang
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Yongqiang Li
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Guqiao Ding
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Siwei Yang
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jipeng Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| |
Collapse
|
2
|
Sabari S, Chinchankar S, Ambite I, Nazari A, Carneiro APN, Svenningsson A, Svanborg C, Chaudhuri A. Rapid ER remodeling induced by a peptide-lipid complex in dying tumor cells. Life Sci Alliance 2025; 8:e202403114. [PMID: 40132886 PMCID: PMC11938384 DOI: 10.26508/lsa.202403114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
The membranous ER spans the entire cell, creating a network for the biosynthesis of proteins and lipids, cell-wide communication, and nuclear delivery of molecules, including therapeutic agents. Here, we identify a novel ER response triggered by the tumoricidal complex alpha1-oleate, defined by a loss of peripheral ER structure, extensive ER vesiculation. Alpha1-oleate was present in the ER-derived vesicle membranes, also decorated by ER-resident and ER-interacting proteins, calnexin and ORP3, and in their lumen, also enriched for KDEL, confirming their ER origin distinct from lipid droplets. Rapid nuclear uptake of the complex constituents resulted in diffuse nuclear staining, and the asymmetrical perinuclear enrichment of the collapsing ER with its content of alpha1-oleate created large invaginations lined by the ER, inner nuclear membrane markers, and lamin nucleoskeleton. In parallel, a change in nuclear shape resulted in a volcano-like structure. This newly discovered, potent ER response to alpha1-oleate may have evolved to package ER-associated cellular contents in the nuclei of dying tumor cells, thus sequestering toxic cell debris associated with apoptotic cell death.
Collapse
Affiliation(s)
- Samudra Sabari
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Siddharth Chinchankar
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ines Ambite
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Atefeh Nazari
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - António Pedro Nbm Carneiro
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Axel Svenningsson
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Catharina Svanborg
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Arunima Chaudhuri
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
3
|
Kulkarni B, AlOtaiby S, Khashab NM, Hadjichristidis N. Redox-Responsive PEO- b-PCL-Based Block Copolymers for Synergistic Drug Delivery and Bioimaging in Cancer Cells. Biomacromolecules 2025; 26:3032-3043. [PMID: 40265982 DOI: 10.1021/acs.biomac.5c00102] [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: 04/24/2025]
Abstract
Stimuli-responsive polymer-based nanocarriers enhance the drug delivery efficiency by enabling targeted release at tumor sites. However, integrating therapeutic and diagnostic functions into a single nanoplatform while maintaining control over both remains a significant challenge. This study presents a stimuli-responsive, multifunctional poly(ethylene oxide)-b-poly(ε-caprolactone) (PEO-b-PCL) nanocarrier for combination cancer therapy and bioimaging. The system codelivers chlorambucil (CHL) and methotrexate (MTX) to enhance therapeutic efficacy and overcome multidrug resistance. A redox-responsive disulfide linker enables CHL release in the tumor's glutathione-rich environment, ensuring selective drug activation. Additionally, an aggregation-induced emission (AIE) fluorophore, tetraphenylethylene (TPE), facilitates the monitoring of cellular uptake and drug release. The resulting TPE-(PEO-b-PCL)-S-S-CHL (P3) micelles encapsulated with MTX (P3-MTX) exhibited favorable size, morphology, and enhanced cytotoxicity, demonstrating a synergistic effect in combination therapy. Confocal laser scanning microscopy (CLSM) confirmed intracellular uptake by using TPE-based fluorescence. Thus, these nanocarriers offer a promising theranostic platform for simultaneous cancer treatment and monitoring.
Collapse
Affiliation(s)
- Bhagyashree Kulkarni
- Polymer Synthesis Laboratory, KAUST Catalysis Center, Physical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Shahad AlOtaiby
- Smart Hybrid Materials (SHMs) Laboratory, Advanced Membranes and Porous Materials Center, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Research Center, King Fahad Medical City, Riyadh 11525, Saudi Arabia
| | - Niveen M Khashab
- Smart Hybrid Materials (SHMs) Laboratory, Advanced Membranes and Porous Materials Center, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Nikos Hadjichristidis
- Polymer Synthesis Laboratory, KAUST Catalysis Center, Physical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| |
Collapse
|
4
|
Shang P, Li G, Yang R, Wang L, Zhou H. Hydrogel microspheres based on aggregation induction to improve the solubility of insoluble drugs promoting bone repair. Colloids Surf B Biointerfaces 2025; 253:114686. [PMID: 40279815 DOI: 10.1016/j.colsurfb.2025.114686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/30/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025]
Abstract
Insoluble organic molecule drugs often accumulate in vivo, diminishing their efficacy. This study explores the utilization of cage oligosiloxane (POSS-SH) as a carrier for insoluble drugs. POSS-SH exhibits disaggregation properties and enhances cell phagocytosis. Using a thermodynamic approach, kartogenin (KGN) and diclofenac (DS) were covalently bonded via acrylyl chloride. Subsequently, a "one-pot method" was employed to graft double-bonded KGN, DS, and PEG onto POSS-SH, forming organic-inorganic POSS hybrid drugs. In the target molecule, water-soluble polyethylene glycol was introduced to enhance solubility and inhibit organic molecule aggregation within the target molecule. The resulting POSS nanoparticles were coated with methacrylate hyaluronic acid, generating hydrogel microspheres (MHS@PSD) for slow release in situ within the bone joint cavity using microfluidic technology. Slow release of PSD can ameliorate local joint inflammation and promote mesenchymal stem cell differentiation into chondrocytes. Compared to unmodified KGN and DS, our designed materials expedite and enhance cartilage regeneration. In summary, POSS-based hydrogel microspheres with deaggregation properties hold promise for cartilage regeneration, offering a framework for enhancing the efficacy of insoluble organic drugs.
Collapse
Affiliation(s)
- Peiyang Shang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China; Department of Orthopaedics, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Gen Li
- Department of Orthopaedics, Clinic Center for Sports Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Renhao Yang
- Department of Orthopaedics, Clinic Center for Sports Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Wang
- Department of Orthopaedics, Clinic Center for Sports Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Haibin Zhou
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China.
| |
Collapse
|
5
|
Redolfi-Bristol D, Yamamoto K, Zhu W, Mazda O, Riello P, Marin E, Pezzotti G. Mapping Selenium Nanoparticles Distribution Inside Cells through Confocal Raman Microspectroscopy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:18124-18133. [PMID: 40098475 PMCID: PMC11956006 DOI: 10.1021/acsami.5c00380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/19/2025]
Abstract
Selenium nanoparticles (SeNPs) exhibit significant potential in biomedical applications due to their antimicrobial, anticancer, and anti-inflammatory properties. In this study, we synthesized biocompatible SeNPs and employed confocal Raman microspectroscopy to map their distribution within human dermal fibroblast (HDF) cells. SeNPs possess a distinctive Raman band placed outside the cellular fingerprint region, which facilitates its detection and precise Raman imaging. Viability assays revealed that SeNPs exhibit cytotoxic effects only at the highest concentrations and for long exposure times while resulting in no harmful effects during all of the other treatments. For the first time, we achieved three-dimensional (3D) Raman mapping of SeNPs within cells, providing insights into their cellular penetration. Additionally, two-dimensional (2D) Raman mapping performed at different times and at sublethal concentrations demonstrated dynamic uptake and confirmed internalization. These findings highlight the effectiveness of SeNPs for biomedical imaging and therapeutic applications, offering an additional approach to studying nanoparticle-cell interactions.
Collapse
Affiliation(s)
- Davide Redolfi-Bristol
- Ceramic
Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
- Dipartimento
di Scienze Molecolari e Nanosistemi, Università
Ca’ Foscari di Venezia, Via Torino 155, 30172 Venezia, Italia
| | - Kenta Yamamoto
- Department
of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Wenliang Zhu
- Ceramic
Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
| | - Osam Mazda
- Department
of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Pietro Riello
- Dipartimento
di Scienze Molecolari e Nanosistemi, Università
Ca’ Foscari di Venezia, Via Torino 155, 30172 Venezia, Italia
| | - Elia Marin
- Biomaterials
Engineering Laboratory, Kyoto Institute
of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
- Department
Polytechnic of Engineering and Architecture, University of Udine, 33100 Udine, Italy
- Biomedical
Research Center, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
| | - Giuseppe Pezzotti
- Dipartimento
di Scienze Molecolari e Nanosistemi, Università
Ca’ Foscari di Venezia, Via Torino 155, 30172 Venezia, Italia
- Department
of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
- Biomedical
Engineering Center, Kansai Medical University, 1-9-11 Shinmachi, Hirakata, Osaka 573-1191, Japan
- Department
of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho,
Kamigyo-ku, Kyoto 602-8566, Japan
- Department
of Orthopedic Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
- Department
of Applied Science and Technology, Politecnico
di Torino, Corso Duca
degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
6
|
Chang YK, Hao SJ, Wu FG. Recent Biomedical Applications of Functional Materials Based on Polyhedral Oligomeric Silsesquioxane (POSS). SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401762. [PMID: 39279395 DOI: 10.1002/smll.202401762] [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: 03/05/2024] [Revised: 08/06/2024] [Indexed: 09/18/2024]
Abstract
Polyhedral oligomeric silsesquioxane (POSS) is a 3D, cage-like nanoparticle with an inorganic Si-O-Si core and eight tunable corner functional groups. Its well-defined structure grants it distinctive physical, chemical, and biological properties and has been widely used for preparing high-performance materials. Recently, click chemistry has enabled the synthesis of various functional POSS-based materials for diverse biomedical applications. This article reviews the recent applications of POSS-based materials in the biomedical field, including cancer treatment, tissue engineering, antibacterial use, and biomedical imaging. Representative examples are discussed in detail. Among the various POSS-based applications, cancer treatment and tissue engineering are the most important. Finally, this review presents the current limitations of POSS-based materials and provides guidance for future research.
Collapse
Affiliation(s)
- Yun-Kai Chang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Shi-Jie Hao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, P. R. China
| |
Collapse
|
7
|
Liu X, Li M, Woo S. Subcellular Drug Distribution: Exploring Organelle-Specific Characteristics for Enhanced Therapeutic Efficacy. Pharmaceutics 2024; 16:1167. [PMID: 39339204 PMCID: PMC11434838 DOI: 10.3390/pharmaceutics16091167] [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: 08/12/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
The efficacy and potential toxicity of drug treatments depends on the drug concentration at its site of action, intricately linked to its distribution within diverse organelles of mammalian cells. These organelles, including the nucleus, endosome, lysosome, mitochondria, endoplasmic reticulum, Golgi apparatus, lipid droplets, exosomes, and membrane-less structures, create distinct sub-compartments within the cell, each with unique biological features. Certain structures within these sub-compartments possess the ability to selectively accumulate or exclude drugs based on their physicochemical attributes, directly impacting drug efficacy. Under pathological conditions, such as cancer, many cells undergo dynamic alterations in subcellular organelles, leading to changes in the active concentration of drugs. A mechanistic and quantitative understanding of how organelle characteristics and abundance alter drug partition coefficients is crucial. This review explores biological factors and physicochemical properties influencing subcellular drug distribution, alongside strategies for modulation to enhance efficacy. Additionally, we discuss physiologically based computational models for subcellular drug distribution, providing a quantifiable means to simulate and predict drug distribution at the subcellular level, with the potential to optimize drug development strategies.
Collapse
Affiliation(s)
- Xin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY 14214-8033, USA;
| | - Miaomiao Li
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210-1267, USA;
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY 14214-8033, USA;
| |
Collapse
|
8
|
Xiao A, Yin L, Chen T, Qian H. Lipo/TK-CDN/TPP/Y6 nanoparticles inhibit cutaneous melanoma formation. J Drug Target 2024; 32:931-940. [PMID: 38838039 DOI: 10.1080/1061186x.2024.2365243] [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: 01/21/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Stimulation of the innate immune stimulator of interferon genes (STING) pathway has been shown to boost anti-tumour immunity. Nevertheless, the systemic delivery of STING agonists to the tumour presents challenges. Therefore, we designed a cyclic dinucleotide (CDN)-based drug delivery system (DDS) combined photothermal therapy (PTT)/photodynamic therapy (PDT)/immunotherapy for cutaneous melanoma. We coencapsulated a reactive oxygen species (ROS)-responsive prodrug thioketone-linked CDN (TK-CDN), and photoresponsive agents chlorin E6 (Y6) within mitochondria-targeting reagent triphenylphosphonium (TPP)-modified liposomes (Lipo/TK-CDN/TPP/Y6). Lipo/TK-CDN/TPP/Y6 exhibited a photothermal effect similar to Y6, along with a superior cellular uptake rate. Upon endocytosis by B16F10 cells, Lipo/TK-CDN/TPP/Y6 generated large amounts of ROS under laser irradiation for PDT. Mice bearing B16F10 tumours were intravenously injected with Lipo/TK-CDN/TPP/Y6 and exposed to irradiation, resulting in a substantial inhibition of tumour growth. Exploration of the mechanism of anti-tumour action showed that Lipo/TK-CDN/TPP/Y6 had a stronger stimulation of STING activation and anti-tumour immune cell infiltration compared to other groups. Hence, the Lipo/TK-CDN/TPP/Y6 nanoparticles offer great potential as a DDS for targeted and on-demand drug release at tumour sites. These nanoparticles exhibit promise as a candidate for precise and controllable combination therapy in the treatment of tumours.
Collapse
Affiliation(s)
- Anju Xiao
- Department of Dermatology and Venereology, Dejiang County People's Hospital, Affiliated to Zunyi Medical University, Dejiang, China
| | - Li Yin
- Department of Pathology, Dejiang County People's Hospital, Affiliated to Zunyi Medical University, Dejiang, China
| | - Ting Chen
- Department of Clinical Medicine, Dejiang County People's Hospital, Affiliated to Zunyi Medical University, Dejiang, China
| | - Huiling Qian
- Department of Endocrinology, Dejiang County People's Hospital, Affiliated to Zunyi Medical University, Dejiang, China
| |
Collapse
|
9
|
Gao F, Wu Y, Wang R, Yao Y, Liu Y, Fan L, Xu J, Zhang J, Han X, Guan X. Precise nano-system-based drug delivery and synergistic therapy against androgen receptor-positive triple-negative breast cancer. Acta Pharm Sin B 2024; 14:2685-2697. [PMID: 38828153 PMCID: PMC11143519 DOI: 10.1016/j.apsb.2024.03.012] [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: 12/25/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 06/05/2024] Open
Abstract
Targeting androgen receptor (AR) has shown great therapeutic potential in triple-negative breast cancer (TNBC), yet its efficacy remains unsatisfactory. Here, we aimed to identify promising targeted agents that synergize with enzalutamide, a second-generation AR inhibitor, in TNBC. By using a strategy for screening drug combinations based on the Sensitivity Index (SI), we found that MK-8776, a selective checkpoint kinase1 (CHK1) inhibitor, showed favorable synergism with enzalutamide in AR-positive TNBC. The combination of enzalutamide and MK-8776 was found to exert more significant anti-tumor effects in TNBC than the single application of enzalutamide or MK-8776, respectively. Furthermore, a nanoparticle-based on hyaluronic acid (HA)-modified hollow-manganese dioxide (HMnO2), named HMnE&M@H, was established to encapsulate and deliver enzalutamide and MK-8776. This HA-modified nanosystem managed targeted activation via pH/glutathione responsiveness. HMnE&M@H repressed tumor growth more obviously than the simple addition of enzalutamide and MK-8776 without a carrier. Collectively, our study elucidated the synergy of enzalutamide and MK-8776 in TNBC and developed a novel tumor-targeted nano drug delivery system HMnE&M@H, providing a potential therapeutic approach for the treatment of TNBC.
Collapse
Affiliation(s)
- Fangyan Gao
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yueyao Wu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Runtian Wang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuhui Yao
- Department of Oncology, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yiqiu Liu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lingling Fan
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jingtong Xu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xin Han
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoxiang Guan
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
10
|
Liu J, Cabral H, Mi P. Nanocarriers address intracellular barriers for efficient drug delivery, overcoming drug resistance, subcellular targeting and controlled release. Adv Drug Deliv Rev 2024; 207:115239. [PMID: 38437916 DOI: 10.1016/j.addr.2024.115239] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
11
|
Wu J, Zheng X, Lin W, Chen L, Wu ZS. Persistent Targeting DNA Nanocarrier Made of 3D Structural Unit Assembled from Only One Basic Multi-Palindromic Oligonucleotide for Precise Gene Cancer Therapy. Adv Healthc Mater 2024; 13:e2303865. [PMID: 38289018 DOI: 10.1002/adhm.202303865] [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: 11/06/2023] [Revised: 01/19/2024] [Indexed: 02/13/2024]
Abstract
Construction of a simple, reconfigurable, and stimuli-responsive DNA nanocarrier remains a technical challenge. In this contribution, by designing three palindromic fragments, a simplest four-sticky end-contained 3D structural unit (PS-unit) made of two same DNA components is proposed. Via regulating the rotation angle of central longitudinal axis of PS-unit, the oriented assembly of one-component spherical architecture is accomplished with high efficiency. Introduction of an aptamer and sticky tail warehouse into one component creates a size-change-reversible targeted siRNA delivery nanovehicle. Volume swelling of 20 nm allows one carrier to load 1987 siPLK1s. Once entering cancer cells and responding to glutathione (GSH) stimuli, siPLK1s are almost 100% released and original size of nanovehicle is restored, inhibiting the expression of PLK1 protein and substantially suppressing tumor growth (superior to commercial transfection agents) in tumor-bearing mice without systemic toxicity.
Collapse
Affiliation(s)
- Jingting Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiaoqi Zheng
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Wenqing Lin
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Linhuan Chen
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| |
Collapse
|
12
|
Huynh M, Vinck R, Gibert B, Gasser G. Strategies for the Nuclear Delivery of Metal Complexes to Cancer Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311437. [PMID: 38174785 DOI: 10.1002/adma.202311437] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/20/2023] [Indexed: 01/05/2024]
Abstract
The nucleus is an essential organelle for the function of cells. It holds most of the genetic material and plays a crucial role in the regulation of cell growth and proliferation. Since many antitumoral therapies target nucleic acids to induce cell death, tumor-specific nuclear drug delivery could potentiate therapeutic effects and prevent potential off-target side effects on healthy tissue. Due to their great structural variety, good biocompatibility, and unique physico-chemical properties, organometallic complexes and other metal-based compounds have sparked great interest as promising anticancer agents. In this review, strategies for specific nuclear delivery of metal complexes are summarized and discussed to highlight crucial parameters to consider for the design of new metal complexes as anticancer drug candidates. Moreover, the existing opportunities and challenges of tumor-specific, nucleus-targeting metal complexes are emphasized to outline some new perspectives and help in the design of new cancer treatments.
Collapse
Affiliation(s)
- Marie Huynh
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for Inorganic Chemistry, Paris, F-75005, France
- Gastroenterology and technologies for Health, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS5286, Université Lyon 1, Lyon, 69008, France
| | - Robin Vinck
- Orano, 125 avenue de Paris, Châtillon, 92320, France
| | - Benjamin Gibert
- Gastroenterology and technologies for Health, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS5286, Université Lyon 1, Lyon, 69008, France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for Inorganic Chemistry, Paris, F-75005, France
| |
Collapse
|
13
|
Zhu XY, Wang TY, Jia HR, Wu SY, Gao CZ, Li YH, Zhang X, Shan BH, Wu FG. A ferroptosis-reinforced nanocatalyst enhances chemodynamic therapy through dual H 2O 2 production and oxidative stress amplification. J Control Release 2024; 367:892-904. [PMID: 38278369 DOI: 10.1016/j.jconrel.2024.01.049] [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: 08/06/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
The existence of a delicate redox balance in tumors usually leads to cancer treatment failure. Breaking redox homeostasis by amplifying oxidative stress and reducing glutathione (GSH) can accelerate cancer cell death. Herein, we construct a ferroptosis-reinforced nanocatalyst (denoted as HBGL) to amplify intracellular oxidative stress via dual H2O2 production-assisted chemodynamic therapy (CDT). Specifically, a long-circulating liposome is employed to deliver hemin (a natural iron-containing substrate for Fenton reaction and ferroptosis), β-lapachone (a DNA topoisomerase inhibitor with H2O2 generation capacity for chemotherapy), and glucose oxidase (which can consume glucose for starvation therapy and generate H2O2). HBGL can achieve rapid, continuous, and massive H2O2 and •OH production and GSH depletion in cancer cells, resulting in increased intracellular oxidative stress. Additionally, hemin can reinforce the ferroptosis-inducing ability of HBGL, which is reflected in the downregulation of glutathione peroxidase-4 and the accumulation of lipid peroxide. Notably, HBGL can disrupt endo/lysosomes and impair mitochondrial function in cancer cells. HBGL exhibits effective tumor-killing ability without eliciting obvious side effects, indicating its clinical translation potential for synergistic starvation therapy, chemotherapy, ferroptosis therapy, and CDT. Overall, this nanocatalytic liposome may be a promising candidate for achieving potentiated cancer treatment.
Collapse
Affiliation(s)
- Xiao-Yu Zhu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China
| | - Tian-Yu Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China
| | - Hao-Ran Jia
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China
| | - Shun-Yu Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China
| | - Cheng-Zhe Gao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China
| | - Yan-Hong Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China
| | - Xinping Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China
| | - Bai-Hui Shan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, PR China.
| |
Collapse
|
14
|
Peng Y, Yang Z, Sun H, Li J, Lan X, Liu S. Nanomaterials in Medicine: Understanding Cellular Uptake, Localization, and Retention for Enhanced Disease Diagnosis and Therapy. Aging Dis 2024; 16:AD.2024.0206-1. [PMID: 38421835 PMCID: PMC11745437 DOI: 10.14336/ad.2024.0206-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Nanomaterials (NMs) have emerged as promising tools for disease diagnosis and therapy due to their unique physicochemical properties. To maximize the effectiveness and design of NMs-based medical applications, it is essential to comprehend the complex mechanisms of cellular uptake, subcellular localization, and cellular retention. This review illuminates the various pathways that NMs take to get from the extracellular environment to certain intracellular compartments by investigating the various mechanisms that underlie their interaction with cells. The cellular uptake of NMs involves complex interactions with cell membranes, encompassing endocytosis, phagocytosis, and other active transport mechanisms. Unique uptake patterns across cell types highlight the necessity for customized NMs designs. After internalization, NMs move through a variety of intracellular routes that affect where they are located subcellularly. Understanding these pathways is pivotal for enhancing the targeted delivery of therapeutic agents and imaging probes. Furthermore, the cellular retention of NMs plays a critical role in sustained therapeutic efficacy and long-term imaging capabilities. Factors influencing cellular retention include nanoparticle size, surface chemistry, and the cellular microenvironment. Strategies for prolonging cellular retention are discussed, including surface modifications and encapsulation techniques. In conclusion, a comprehensive understanding of the mechanisms governing cellular uptake, subcellular localization, and cellular retention of NMs is essential for advancing their application in disease diagnosis and therapy. This review provides insights into the intricate interplay between NMs and biological systems, offering a foundation for the rational design of next-generation nanomedicines.
Collapse
Affiliation(s)
- Yue Peng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhengshuang Yang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Hui Sun
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiuwan Lan
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
15
|
Gu Z, Tian X, Guang S, Wei G, Mao Y, Xu H. POSS engineering of squaraine nanoparticle with high photothermal conversion efficiency for photothermal therapy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 306:123576. [PMID: 37922849 DOI: 10.1016/j.saa.2023.123576] [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: 07/24/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023]
Abstract
Photothermal therapy (PTT) has been extensively studied due to its promising therapeutic effects and potential for development in cancer treatments. Central to PTT is the development of photothermal agents (PTAs). This study presents a novel nanoparticle called POSS-SQ, which satisfies the necessary conditions to function as a PTA. Comprised of squaraine (SQ) and polyhedral oligomeric sesquisiloxane (POSS), POSS-SQ NPs exhibit strong near-infrared (NIR) absorption and high photothermal conversion efficiency (PCE) attributable to the intermolecular electron transfer in SQ. Furthermore, POSS when modified with polyethylene glycol (PEG) through "click" chemistry, effectively enhances cell permeability and biocompatibility of the nanoparticles. Photothermal experiments reveal that POSS-SQ NPs demonstrate concentration and laser power dependence, with a PCE of 67.2%. In vitro and in vivo experiments confirm the excellent biosafety and tumor growth inhibition potential of POSS-SQ NPs under laser irradiation, attributed to the synergistic effects of enhanced cell permeability and exceptional photothermal properties. This research highlights the possibility of obtaining PTAs with high PCE and excellent biocompatibility by combining SQ-N and POSS, offering a new approach for designing and developing more efficient PTAs to enhance better PTT outcomes.
Collapse
Affiliation(s)
- Zhengye Gu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, and College of Materials Science and Engineering & Research Center for Analysis and Measurement, Donghua University, Shanghai 201620, China
| | - Xiaoyong Tian
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Shanyi Guang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Gang Wei
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Yanfei Mao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Hongyao Xu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, and College of Materials Science and Engineering & Research Center for Analysis and Measurement, Donghua University, Shanghai 201620, China.
| |
Collapse
|
16
|
Pan L, Peng H, Lee B, Zhao J, Shen X, Yan X, Hua Y, Kim J, Kim D, Lin M, Zhang S, Li X, Yi X, Yao F, Qin Z, Du J, Chi Y, Nam JM, Hyeon T, Liu J. Cascade Catalytic Nanoparticles Selectively Alkalize Cancerous Lysosomes to Suppress Cancer Progression and Metastasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305394. [PMID: 37643367 DOI: 10.1002/adma.202305394] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/16/2023] [Indexed: 08/31/2023]
Abstract
Lysosomes are critical in modulating the progression and metastasis for various cancers. There is currently an unmet need for lysosomal alkalizers that can selectively and safely alter the pH and inhibit the function of cancer lysosomes. Here an effective, selective, and safe lysosomal alkalizer is reported that can inhibit autophagy and suppress tumors in mice. The lysosomal alkalizer consists of an iron oxide core that generates hydroxyl radicals (•OH) in the presence of excessive H+ and hydrogen peroxide inside cancer lysosomes and cerium oxide satellites that capture and convert •OH into hydroxide ions. Alkalized lysosomes, which display impaired enzyme activity and autophagy, lead to cancer cell apoptosis. It is shown that the alkalizer effectively inhibits both local and systemic tumor growth and metastasis in mice. This work demonstrates that the intrinsic properties of nanoparticles can be harnessed to build effective lysosomal alkalizers that are both selective and safe.
Collapse
Affiliation(s)
- Limin Pan
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Haibao Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Bowon Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jiaxu Zhao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xiulian Shen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ximei Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yipeng Hua
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jeonghyun Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dokyoon Kim
- Department of Bionano Engineering, Hanyang University, Ansan, 15588, Republic of Korea
| | - Mouhong Lin
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Shengjian Zhang
- Department of Radiology, Cancer Hospital/Institute and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaohui Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xueying Yi
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Feibai Yao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yudan Chi
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jwa-Min Nam
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jianan Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
17
|
Wang H, Mills J, Sun B, Cui H. Therapeutic Supramolecular Polymers: Designs and Applications. Prog Polym Sci 2024; 148:101769. [PMID: 38188703 PMCID: PMC10769153 DOI: 10.1016/j.progpolymsci.2023.101769] [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] [Indexed: 01/09/2024]
Abstract
The self-assembly of low-molecular-weight building motifs into supramolecular polymers has unlocked a new realm of materials with distinct properties and tremendous potential for advancing medical practices. Leveraging the reversible and dynamic nature of non-covalent interactions, these supramolecular polymers exhibit inherent responsiveness to their microenvironment, physiological cues, and biomolecular signals, making them uniquely suited for diverse biomedical applications. In this review, we intend to explore the principles of design, synthesis methodologies, and strategic developments that underlie the creation of supramolecular polymers as carriers for therapeutics, contributing to the treatment and prevention of a spectrum of human diseases. We delve into the principles underlying monomer design, emphasizing the pivotal role of non-covalent interactions, directionality, and reversibility. Moreover, we explore the intricate balance between thermodynamics and kinetics in supramolecular polymerization, illuminating strategies for achieving controlled sizes and distributions. Categorically, we examine their exciting biomedical applications: individual polymers as discrete carriers for therapeutics, delving into their interactions with cells, and in vivo dynamics; and supramolecular polymeric hydrogels as injectable depots, with a focus on their roles in cancer immunotherapy, sustained drug release, and regenerative medicine. As the field continues to burgeon, harnessing the unique attributes of therapeutic supramolecular polymers holds the promise of transformative impacts across the biomedical landscape.
Collapse
Affiliation(s)
- Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jason Mills
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Boran Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
18
|
Raza F, Zafar H, Jiang L, Su J, Yuan W, Qiu M, Paiva-Santos AC. Progress of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. Biomater Sci 2023; 12:57-91. [PMID: 37902579 DOI: 10.1039/d3bm01170d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
In recent years, considerable attention has been given to phototherapy, including photothermal and photodynamic therapy to kill tumor cells by producing heat or reactive oxygen species (ROS). It has the high merits of noninvasiveness and limited drug resistance. To fully utilize this therapy, an extraordinary nanovehicle is required to target phototherapeutic agents in the tumor cells. Nanovesicles embody an ideal strategy for drug delivery applications. Cell membrane-derived biomimetic nanovesicles represent a developing type of nanocarrier. Combining this technique with cancer phototherapy could enable a novel strategy. Herein, efforts are made to describe a comprehensive overview of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. The description in this review is mainly based on representative examples of exosome-derived biomimetic nanomedicine research, ranging from their comparison with traditional nanocarriers to extensive applications in cancer phototherapy. Additionally, the challenges and future prospectives for translating these for clinical application are discussed.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Liangdi Jiang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Weien Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
- LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
19
|
Li X, Gao Y, Li H, Majoral JP, Shi X, Pich A. Smart and bioinspired systems for overcoming biological barriers and enhancing disease theranostics. PROGRESS IN MATERIALS SCIENCE 2023; 140:101170. [DOI: 10.1016/j.pmatsci.2023.101170] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
20
|
Chen S, Cao R, Xiang L, Li Z, Chen H, Zhang J, Feng X. Research progress in nucleus-targeted tumor therapy. Biomater Sci 2023; 11:6436-6456. [PMID: 37609783 DOI: 10.1039/d3bm01116j] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The nucleus is considered the most important organelle in the cell as it plays a central role in controlling cell reproduction, metabolism, and the cell cycle. The successful delivery of drugs into the nucleus can achieve excellent therapeutic effects, which reveals the potential of nucleus-targeted therapy in precision medicine. However, the transportation of therapeutics into the nucleus remains a significant challenge due to various biological barriers. Herein, we summarize the recent progress in the nucleus-targeted drug delivery system (NDDS). The structures of the nucleus and nuclear envelope are first described in order to understand the mechanisms by which drugs cross the nuclear envelope. Then, various drug delivery strategies based on the mechanisms and their applications are discussed. Finally, the challenges and solutions in the field of nucleus-targeted drug delivery are raised for developing a more efficient NDDS and promoting its clinical transformation.
Collapse
Affiliation(s)
- Shaofeng Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Rumeng Cao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Ling Xiang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Ziyi Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Hui Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Jiumeng Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Xuli Feng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| |
Collapse
|
21
|
Jiang L, Cai H, Qin W, Li Z, Zhang L, Bi H. Meticulously Designed Carbon Dots as Photo-Triggered RNA-Destroyer for Evoking Pyroptosis. Bioconjug Chem 2023; 34:1387-1397. [PMID: 37534892 DOI: 10.1021/acs.bioconjchem.3c00278] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
An ideal photosensitizer for photodynamic therapy should not only possess high reactive oxygen species (ROS) generation efficiency but also maximize utilization of the in situ produced ROS species, where the latter is closely related to its intracellular location. However, rational design of such photosensitizer without tedious conjugation procedures remains a grand challenge. Here, we report the one-pot preparation of carbon dots (CDs)-based photosensitizer from levofloxacin and neutral red featuring both high 1O2 quantum yield (φΔ = 38.85%) and superior RNA selectivity. Moreover, the φΔ value shows a further 40% improvement and reaches 54.33% in response to RNA binding. Owing to these combined attributes, the CDs could exert great damage to the cellular RNA system (termed the RNA-destroyer) under extremely low dosage of light irradiation (15 mW cm-2, 1 min). It induces pyroptotic cell death and causes rapid release of different cytokines that served as molecular markers in photodynamic immunotherapy. This work represents the meticulously designed CDs with high ROS generation and utilization efficiency via good organization of the photosensitive and targeting modularity. Moreover, it is the first CDs-based pyroptosis inducer to the best of our knowledge.
Collapse
Affiliation(s)
- Lei Jiang
- School of Chemistry and Chemical Engineering, Anhui University, Hefei, Anhui 230601, China
| | - Hao Cai
- School of Materials Science and Engineering, Anhui University, Hefei, Anhui 230601, China
| | - Weixia Qin
- School of Materials Science and Engineering, Anhui University, Hefei, Anhui 230601, China
| | - Zijian Li
- School of Materials Science and Engineering, Anhui University, Hefei, Anhui 230601, China
| | - Liang Zhang
- Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road, Hefei, Anhui 230027, China
| | - Hong Bi
- School of Materials Science and Engineering, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|
22
|
Zhang K, Zhu J, Wang R, Zhu W, Zhang Z, Gong L, Feng F, Liu W, Han L, Qu W. Mitochondria-Anchoring Self-assembled Nanoparticles for Multi-Path Energy Depletion: A "Nano Bomb" in Chemo-co-Starvation Therapy. Int J Pharm 2023:123180. [PMID: 37364784 DOI: 10.1016/j.ijpharm.2023.123180] [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: 02/17/2023] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
As the main systemic treatment for triple-negative breast cancer (TNBC), the bleak medical prognosis of chemotherapy resulted in impaired life quality by tumor recurrence and metastasis. The feasible cancer starvation therapy could inhibit tumor progression by blocking energy supplements, however, the mono-therapeutic modality showed limited curing efficacy due to heterogeneity and abnormal energy metabolism of TNBC. Thus, the development of a synergistic nano-therapeutic modality involving different anti-tumor mechanisms to simultaneously transport medicines to the organelle where metabolism took place, might remarkably improve curing efficacy, targeting ability, and bio-safety. Herein, the hybrid BLG@TPGS NPs were prepared by doping multi-path energy inhibitors Berberine (BBR) and Lonidamine (LND) as well as the chemotherapeutic agent Gambogic acid (GA). Our research indicated that Nanobomb\mathord{-} BLG@TPGS NPs inherited the mitochondria targeting ability from BBR to accumulate precisely at the "energy factory" mitochondria, and then induce starvation therapy to efficiently eradicated cancer cells by coordinately powered off tumor cells via a "three-prone strategy" to cut off mitochondrial respiration, glycolysis, and glutamine metabolism. The inhibition of tumor proliferation and migration was enlarged by the synergistic combination with chemotherapy. Besides, apoptosis via mitochondria pathway and mitochondria fragmentation supported the hypothesis that NPs eliminated MDA-MB-231 cells by violently attacking MDA-MB-231 cells and especially the mitochondria. In summary, this synergistic chemo-co-starvation nanomedicine proposed an innovative site-specific targeting strategy for improved tumor treatment and decreased toxicity to normal tissues, which provided an option for clinical TNBC-sensitive treatment.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiaxin Zhu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Ruyi Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Wanfang Zhu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China; College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Zhongtao Zhang
- Tumor Precise Intervention and Translational Medicine Laboratory, The affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, China
| | - Liangping Gong
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China; Nanjing Medical University, Nanjing, 211198, China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 211198, China
| | - Lingfei Han
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
23
|
Qian L, Lin X, Gao X, Khan RU, Liao JY, Du S, Ge J, Zeng S, Yao SQ. The Dawn of a New Era: Targeting the "Undruggables" with Antibody-Based Therapeutics. Chem Rev 2023. [PMID: 37186942 DOI: 10.1021/acs.chemrev.2c00915] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.
Collapse
Affiliation(s)
- Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544
| |
Collapse
|
24
|
Zhong X, Wei G, Liu B, Wang C, Wang J, Lu Y, Cui W, Guo H. Polyhedral Oligomeric Silsesquioxane-Based Nanoparticles for Efficient Chemotherapy of Glioblastoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207248. [PMID: 36725316 DOI: 10.1002/smll.202207248] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/10/2023] [Indexed: 05/04/2023]
Abstract
Glioblastoma (GBM) is the most common lethal brain tumor with dismal treatment outcomes and poor response to chemotherapy. As the regulatory center of cytogenetics and metabolism, most tumor chemotherapeutic molecules exert therapeutic effects in the nucleus. Nanodrugs showing the nuclear aggregation effect are expected to eliminate and fundamentally suppress tumor cells. In this study, a nanodrug delivery system based on polyhedral oligomeric silsesquioxane (POSS) is introduced to deliver drugs into the nuclei of GBM cells, effectively enhancing the therapeutic efficacy of chemotherapy. The nanoparticles are modified with folic acid and iRGD peptides molecules to improve their tumor cell targeting and uptake via receptor-mediated endocytosis. Nuclear aggregation allows for the direct delivery of chemotherapeutic drug temozolomide (TMZ) to the tumor cell nuclei, resulting in more significant DNA damage and inhibition of tumor cell proliferation. Herein, TMZ-loaded POSS nanoparticles can significantly improve the survival of GBM-bearing mice. Therefore, the modified POSS nanoparticles may serve as a promising drug-loaded delivery platform to improve chemotherapy outcomes in GBM patients.
Collapse
Affiliation(s)
- Xiangyang Zhong
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Gang Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Boyang Liu
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Chenyang Wang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Yong Lu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Hongbo Guo
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
25
|
Goyal P, Malviya R. Advances in nuclei targeted delivery of nanoparticles for the management of cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188881. [PMID: 36965678 DOI: 10.1016/j.bbcan.2023.188881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/16/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
A carrier is inserted into the appropriate organelles (nucleus) in successful medication transport, crucial to achieving very effective illness treatment. Cell-membrane targeting is the major focus of using nuclei to localize delivery. It has been demonstrated that high quantities of anticancer drugs can be injected directly into the nuclei of cancer cells, causing the cancer cells to die and increasing the effectiveness of chemotherapy. There are several effective ways to functionalize Nanoparticles (NPs), including changing their chemical makeup or attaching functional groups to their surface to increase their ability to target organelles. To cause tumor cells to apoptosis, released medicines must engage with molecular targets on particular organelles when their concentration is high enough. Targeted medication delivery studies will increasingly focus on organelle-specific delivery.
Collapse
Affiliation(s)
- Priyanshi Goyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
26
|
Zhang M, Hu S, Liu L, Dang P, Liu Y, Sun Z, Qiao B, Wang C. Engineered exosomes from different sources for cancer-targeted therapy. Signal Transduct Target Ther 2023; 8:124. [PMID: 36922504 PMCID: PMC10017761 DOI: 10.1038/s41392-023-01382-y] [Citation(s) in RCA: 190] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/31/2023] [Accepted: 02/22/2023] [Indexed: 03/17/2023] Open
Abstract
Exosome is a subgroup of extracellular vesicles, which has been serving as an efficient therapeutic tool for various diseases. Engineered exosomes are the sort of exosomes modified with surface decoration and internal therapeutic molecules. After appropriate modification, engineered exosomes are able to deliver antitumor drugs to tumor sites efficiently and precisely with fewer treatment-related adverse effects. However, there still exist many challenges for the clinical translation of engineered exosomes. For instance, what sources and modification strategies could endow exosomes with the most efficient antitumor activity is still poorly understood. Additionally, how to choose appropriately engineered exosomes in different antitumor therapies is another unresolved problem. In this review, we summarized the characteristics of engineered exosomes, especially the spatial and temporal properties. Additionally, we concluded the recent advances in engineered exosomes in the cancer fields, including the sources, isolation technologies, modification strategies, and labeling and imaging methods of engineered exosomes. Furthermore, the applications of engineered exosomes in different antitumor therapies were summarized, such as photodynamic therapy, gene therapy, and immunotherapy. Consequently, the above provides the cancer researchers in this community with the latest ideas on engineered exosome modification and new direction of new drug development, which is prospective to accelerate the clinical translation of engineered exosomes for cancer-targeted therapy.
Collapse
Affiliation(s)
- Menghui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Lin Liu
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.,Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Pengyuan Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, 450001, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China. .,Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Bingbing Qiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Chengzeng Wang
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China. .,Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
27
|
Zhang X, Yi C, Zhang L, Zhu X, He Y, Lu H, Li Y, Tang Y, Zhao W, Chen G, Wang C, Huang S, Ouyang G, Yu D. Size-optimized nuclear-targeting phototherapy enhances the type I interferon response for "cold" tumor immunotherapy. Acta Biomater 2023; 159:338-352. [PMID: 36669551 DOI: 10.1016/j.actbio.2023.01.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023]
Abstract
There is growing interest in the effect of innate immune silencing in "cold" tumors, which always fail in the immune checkpoint blockade monotherapy using PD-L1 monoclonal antibodies (aPD-L1). Combination of aPD-L1 with photodynamic therapy, i.e., photoimmunotherapy, is a promising strategy to improve the mono immunotherapy. Nuclear-targeting nanoparticles could elicit a type I interferon (IFN)-mediated innate immune response and reverse the immunosuppressive microenvironment for long-term immunotherapy of "cold" tumors. Photosensitizers such as zinc phthalocyanine (ZnPc) have limited ability to target the nucleus and activate innate sensing pathways to minimize tumor recurrence. Additionally, the relationship between nanoparticle size and nuclear entry capacity remains unclear. Herein, graphene quantum dots (GQDs) were employed as aPD-L1 and ZnPc carriers. Three particle sizes (200 nm, 32 nm and 5 nm) of aPD-L1/ZnPc/GQD-PEG (PZGE) were synthesized and tested. The 5 nm nanoparticles achieved the best nuclear enrichment capacity contributing to their ultrasmall size. Notably, 5 nm PZGE-based photodynamic therapy enabled an amplification of the type I IFN-mediated innate immune response and could convert "immune-cold" tumors into "immune-hot" ones. Utilizing their size advantage to target the nucleus, 5 nm nanoparticles induced DNA damage and activated the type I IFN-mediated innate immune response, subsequently promoting cytotoxic T-lymphocyte infiltration and reversing negative PD-L1 expression. Furthermore, the nanoplatform we designed is promising for the effective suppression of distant oral squamous cell carcinoma. Thus, for the first time, this study presents a size design strategy for nuclear-targeted photo-controlled immune adjuvants and the nuclear-targeted phototherapy-mediated immunomodulatory functions of type I IFN innate immune signalling for "immune-cold" tumors. STATEMENT OF SIGNIFICANCE: The potential of commonly used photosensitizers to activate innate sensing pathways for producing type I IFNs is limited due to the lack of nuclear targeting. Facilitating the nuclear-targeting of photosensitizers to enhance innate immune response and execute long-term tumor killing effect would be a promising strategy for "cold" tumor photoimmunotherapy. Herein, we report an optimal size of PZGE nanoparticles that enable the nuclear-targeting of ZnPc, which reinforces the type I IFN-mediated innate immune response, synergistically reversing "cold tumors" to "hot tumors" for effective primary and distant tumor photoimmunotherapy. This work highlights the marked efficacy of ultrasmall nuclear-located nanocarriers and offers new insight into "immune-cold tumors" via prominent innate immune activation mediated by nuclear-targeting photoimmunotherapy.
Collapse
Affiliation(s)
- Xiliu Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Chen Yi
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Lejia Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Xinyu Zhu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Yi He
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Huanzi Lu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Yiming Li
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Yuquan Tang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Wei Zhao
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China
| | - Guosheng Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Cheng Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China.
| | - Siming Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Gangfeng Ouyang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Dongsheng Yu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Guangzhou, 510055, China.
| |
Collapse
|
28
|
Gao B, Wang X, Wang M, Liu W, Li Y, Xia S, Zhang W, Feng Y. "Intercellular Mass Transport" Mimic Enables ASO Entry Completely into the Cell Nucleus for Enhanced Ischemia Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12777-12786. [PMID: 36854063 DOI: 10.1021/acsami.2c21691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Currently, the development of a new therapeutic technology is focused on antisense oligonucleotides (ASOs), where ASOs are used to complementarily pair with DNA, messenger RNA, or long noncoding RNA (lncRNA) to regulate the cell behavior by inhibiting the target gene expression. However, the targeted regulation toward nuclear genes still faces great challenges in ASO delivery for clinical applications, i.e., two essential criteria (high nuclear entry and delivery vehicle safety/simplification) generally compromise each other and are not simultaneously satisfactory. Herein, for the first time, inspired by "intercellular-mass-transport", we report an important discovery that the cell membrane of endothelial cells (ECs) serving as the biointerface enables ASOs to rapidly and completely enter the EC nucleus. Thereby, we innovatively fabricate a nanosystem only by sequential self-assembly of natural/off-the-shelf biomaterials to well overcome the above-mentioned contradiction. The efficacy is strikingly superior to that of the previous delivery vehicles. Furthermore, our technology is applied to successfully silence lncRNA MEG3 in the EC nucleus, significantly augmenting EC morphogenesis. More importantly, this nanosystem is applicable for in vivo intramuscular injection to enhance the therapeutic outcome in a critical limb ischemia mouse model. This work brings a new hope for the technological innovation of ASO nuclear delivery and opens a new avenue to explore natural/off-the-shelf materials for cargo delivery into subcellular compartments.
Collapse
Affiliation(s)
- Bin Gao
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Xiaoyu Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Meiyu Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Wen Liu
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Ying Li
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Shihai Xia
- Department of Hepatopancreatobiliary and Splenic Medicine, Affiliated Hospital, Logistics University of People's Armed Police Force, 220 Chenglin Road, Tianjin 300162, China
| | - Wencheng Zhang
- Department of Physiology and Pathophysiology, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Weijin Road 92, Tianjin 300072, China
| |
Collapse
|
29
|
Zhao Y, Sun S, Lyu Y, Gao M, Lin H, Yang C. A near-infrared fluorescent nanoprobe for senescence-associated β-galactosidase sensing in living cells. Chem Commun (Camb) 2023; 59:2974-2977. [PMID: 36807350 DOI: 10.1039/d2cc05550c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
A near-infrared fluorescent nanoprobe based on semiconducting polymer nanoparticles (SPNs) for the detection of senescence-associated β-gal (SA-β-gal) is developed. Benefiting from the intrinsic lysosome-locating feature, this probe can be successfully used for the visualization of SA-β-gal in living cells.
Collapse
Affiliation(s)
- Ye Zhao
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China.
| | - Shan Sun
- International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China.
| | - Yan Lyu
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China.
| | - Mengyao Gao
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China.
| | - Hengwei Lin
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China. .,International Joint Research Center for Photo-responsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China.
| | - Cheng Yang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
30
|
Chen Y, Xing Y, Wang Z, Li L, Wang H, Tang S, Cai K, Zhang J. Dual factor coactivatable fluorescent nanosensor with boosted cytoplasmic biomarker accessibility toward selective tumor imaging. Biosens Bioelectron 2023; 223:115026. [PMID: 36565544 DOI: 10.1016/j.bios.2022.115026] [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: 10/19/2022] [Revised: 12/12/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Fluorescent nanosensor-based tumor imaging holds great promise in cancer diagnosis and treatment assistance, yet the signal contrast is heavily hampered by the unspecific/unwanted activation at microscopic regions with a highly restricted local abundance of biomarkers. Herein, we developed an activation boosting strategy by the integration and manipulation of dual-factor coactivation of sensing and lysosome escape facilitated the rise of cytosolic biomarker accessibility. By employing hybrid DNA probes on gold nanoquenchers, ATP sensing initiated conformation switch of the corresponding aptamer units triggered the exposure of a hidden toehold in a loop structure. Sequentially, miRNA-21 sensing was triggered by toehold-mediated strand displacement and detachment of the binding complexes. The application of lysosomotropic agent chloroquine at optimized time interval facilitated the release of nanosensors into the cytosol and a ∼10.5-fold increment of intracellular fluorescence in vitro, while coactivation improved the cancer-to-normal cell signal ratio by ∼5.9 times. The synergy effects led to a high tumor-to-normal tissue ratio value of ∼7.9 in the in vivo imaging results. This strategy establishes a new paradigm of fluorescent nanosensors for selective and specific tumor imaging.
Collapse
Affiliation(s)
- Yuhua Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, PR China
| | - Yuxin Xing
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, PR China
| | - Zhenqiang Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, PR China
| | - Lin Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, PR China
| | - Hailing Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, PR China
| | - Shuqi Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, PR China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, PR China
| | - Jixi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, PR China.
| |
Collapse
|
31
|
Zhao Y, Yang J, Hao D, Xie R, Jia L, Yang M, Ma H, Wang P, Yang W, Sui F, Zhao H, Chen Y, Zhao Q. Infection Microenvironment-Sensitive Photothermal Nanotherapeutic Platform to Inhibit Methicillin-Resistant Staphylococcus aureus Infection. Macromol Biosci 2023; 23:e2200430. [PMID: 36478660 DOI: 10.1002/mabi.202200430] [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: 10/12/2022] [Revised: 11/27/2022] [Indexed: 12/12/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) can induce multiple inflammations. The biofilm formed by MRSA is resistant to a variety of antibiotics and is extremely difficult to cure, which seriously threatens human health. Herein, a nanoparticle encapsulating berberine with polypyrrole core and pH-sensitive shell to provide chemo-photothermal dual therapy for MRSA infection is reported. By integrating photothermal agent polypyrrole, berberine, acid-degradable crosslinker, and acid-induced charge reversal polymer, the nanoparticle exhibited highly efficient MRSA infection treatment. In normal uninfected areas and bloodstream, nanoparticles showed negatively charged, demonstrating high biocompatibility and excellent hemocompatibility. However, once arriving at the MRSA infection site, the nanoparticle can penetrate and accumulate in the biofilm within 2 h. Simultaneously, berberine can be released into biofilm rapidly. Under the combined effect of photothermal response and berberine inhibition, 88.7% of the biofilm is removed at 1000 µg mL-1 . Moreover, the nanoparticles have an excellent inhibitory effect on biofilm formation, the biofilm inhibition capacity can reach up to 90.3%. Taken together, this pH-tunable nanoparticle can be employed as a new generation treatment strategy to fight against the fast-growing MRSA infection.
Collapse
Affiliation(s)
- Yu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiaying Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Danli Hao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ran Xie
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lingyu Jia
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Miyi Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Hai Ma
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Pengqian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Feng Sui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanjun Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qinghe Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| |
Collapse
|
32
|
Jiang Q, Lu S, Xu X, Bai C, Yan Q, Fang M, Huang L, Jin C, Zhang Y, Sun J, He Z, Zhao C, Qin F, Wang Y, Zhang T. Inhibition of alanine-serine-cysteine transporter 2-mediated auto-enhanced photodynamic cancer therapy of co-nanoassembly between V-9302 and photosensitizer. J Colloid Interface Sci 2023; 629:773-784. [PMID: 36195017 DOI: 10.1016/j.jcis.2022.05.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/28/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
The efficiency of reactive oxygen species (ROS)-based photodynamic therapy (PDT) is far from satisfactory, because cancer cells can adapt to PDT by upregulating glutathione (GSH) levels. The GSH levels in tumor cells are determined based on glutamine availability via alanine-serine-cysteine transporter 2 (ASCT2)-mediated entry into cells. Herein, we develop co-assembled nanoparticles (PPa/V-9302 NPs) of the photosensitizer pyropheophorbide a (PPa) and V-9302 (a known inhibitor of ASCT2) in a 1:1 M ratio using a one-step precipitation method to auto-enhance photodynamic therapy. The computational simulations revealed that PPa and V-9302 could self-assemble through different driving forces, such as π-π stacking, hydrophobic interactions, and ionic bonds. Such PPa/V-9302 NPs could disrupt the intracellular redox homeostasis due to enhanced ROS production via PPa-induced PDT and reduced GSH synthesis via inhibition of the ASCT2-mediated glutamine flux by V-9302. The in vivo assays reveal that PPa/V-9302 NPs could increase the drug accumulation in tumor sites and suppress tumor growth in BALB/c mice bearing mouse breast carcinoma (4 T1) tumor. Our findings provide a new paradigm for the rational design of the PDT-based combinational cancer therapy.
Collapse
Affiliation(s)
- Qikun Jiang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Sirun Lu
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaolan Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chenxia Bai
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qing Yan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mengna Fang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Li Huang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | | | - Yunran Zhang
- Shanghai Institute of Pharmacists, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 315615, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chunyang Zhao
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110002, China.
| | - Feng Qin
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yongjun Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Tianhong Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
33
|
Yang J, Griffin A, Qiang Z, Ren J. Organelle-targeted therapies: a comprehensive review on system design for enabling precision oncology. Signal Transduct Target Ther 2022; 7:379. [PMID: 36402753 PMCID: PMC9675787 DOI: 10.1038/s41392-022-01243-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer is a major threat to human health. Among various treatment methods, precision therapy has received significant attention since the inception, due to its ability to efficiently inhibit tumor growth, while curtailing common shortcomings from conventional cancer treatment, leading towards enhanced survival rates. Particularly, organelle-targeted strategies enable precise accumulation of therapeutic agents in organelles, locally triggering organelle-mediated cell death signals which can greatly reduce the therapeutic threshold dosage and minimize side-effects. In this review, we comprehensively discuss history and recent advances in targeted therapies on organelles, specifically including nucleus, mitochondria, lysosomes and endoplasmic reticulum, while focusing on organelle structures, organelle-mediated cell death signal pathways, and design guidelines of organelle-targeted nanomedicines based on intervention mechanisms. Furthermore, a perspective on future research and clinical opportunities and potential challenges in precision oncology is presented. Through demonstrating recent developments in organelle-targeted therapies, we believe this article can further stimulate broader interests in multidisciplinary research and technology development for enabling advanced organelle-targeted nanomedicines and their corresponding clinic translations.
Collapse
Affiliation(s)
- Jingjing Yang
- grid.24516.340000000123704535Institute of Nano and Biopolymeric Materials, School of Materials Science and Engineering, Tongji University, 201804 Shanghai, China
| | - Anthony Griffin
- grid.267193.80000 0001 2295 628XSchool of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406 USA
| | - Zhe Qiang
- grid.267193.80000 0001 2295 628XSchool of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406 USA
| | - Jie Ren
- grid.24516.340000000123704535Institute of Nano and Biopolymeric Materials, School of Materials Science and Engineering, Tongji University, 201804 Shanghai, China
| |
Collapse
|
34
|
Liu S, Zhang Q, He H, Yi M, Tan W, Guo J, Xu B. Intranuclear Nanoribbons for Selective Killing of Osteosarcoma Cells. Angew Chem Int Ed Engl 2022; 61:e202210568. [PMID: 36102872 PMCID: PMC9869109 DOI: 10.1002/anie.202210568] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Indexed: 01/26/2023]
Abstract
Herein, we show intranuclear nanoribbons formed upon dephosphorylation of leucine-rich L- or D-phosphopeptide catalyzed by alkaline phosphatase (ALP) to selectively kill osteosarcoma cells. Being dephosphorylated by ALP, the peptides are first transformed into micelles and then converted into nanoribbons. The peptides/assemblies first aggregate on cell membranes, then enter cells via endocytosis, and finally accumulate in nuclei (mainly in nucleoli). Proteomics analysis suggests that the assemblies interact with histone proteins. The peptides kill osteosarcoma cells rapidly and are nontoxic to normal cells. Moreover, the repeated stimulation of the osteosarcoma cells by the peptides sensitizes the cancer cells rather than inducing resistance. This work not only illustrates a novel mechanism for nucleus targeting, but may also pave a new way for selectively killing osteosarcoma cells and minimizing drug resistance.
Collapse
Affiliation(s)
- Shuang Liu
- School of Materials Science and Engineering, Wuhan University of Technology, 122 Luoshi Road, Wuhan, Hubei, 430070, China
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Meihui Yi
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| |
Collapse
|
35
|
Wang W, Gao Y, Chen Y, Wang W, Li Q, Huang Z, Zhang J, Xiang Q, Wu Z. Outward Movement of Targeting Ligands from a Built-In Reserve Pool in Nuclease-Resistant 3D Hierarchical DNA Nanocluster for in Vivo High-Precision Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203698. [PMID: 36253152 PMCID: PMC9685459 DOI: 10.1002/advs.202203698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/14/2022] [Indexed: 06/16/2023]
Abstract
Nanostructures made entirely of DNAs display great potential as chemotherapeutic drug carriers but so far cannot achieve sufficient clinic therapy outcomes due to off-target toxicity. In this contribution, an aptamer-embedded hierarchical DNA nanocluster (Apt-eNC) is constructed as an intelligent carrier for cancer-targeted drug delivery. Specifically, Apt-eNC is designed to have a built-in reserve pool in the interior cavity from which aptamers may move outward to function as needed. When surface aptamers are degraded, ones in reserve pool can move outward to offer the compensation, thereby magically preserving tumor-targeting performance in vivo. Even if withstanding extensive aptamer depletion, Apt-eNC displays a 115-fold enhanced cell targeting compared with traditional counterparts and at least 60-fold improved tumor accumulation. Moreover, one Apt-eNC accommodates 5670 chemotherapeutic agents. As such, when systemically administrated into HeLa tumor-bearing BALB/c nude mouse model, drug-loaded Apt-eNC significantly inhibits tumor growth without systemic toxicity, holding great promise for high precision therapy.
Collapse
Affiliation(s)
- Weijun Wang
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
| | - Yansha Gao
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
| | - Yaxin Chen
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
| | - Wenqing Wang
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
| | - Qian Li
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
| | - Zhiyi Huang
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
| | - Jingjing Zhang
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
| | - Qi Xiang
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
- Key Laboratory of Laboratory MedicineMinistry of Education of ChinaZhejiang Provincial Key Laboratory of Medicine GeneticsSchool of Laboratory Medicine and Life SciencesInstitute of Functional Nucleic Acids and Personalized Cancer TheranosticsWenzhou Medical UniversityWenzhou325035China
| | - Zai‐Sheng Wu
- Cancer Metastasis Alert and Prevention CenterFujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and ChemotherapyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou UniversityFuzhou350108China
| |
Collapse
|
36
|
In situ generation of micrometer-sized tumor cell-derived vesicles as autologous cancer vaccines for boosting systemic immune responses. Nat Commun 2022; 13:6534. [PMID: 36319625 PMCID: PMC9626595 DOI: 10.1038/s41467-022-33831-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 10/05/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer vaccine, which can promote tumor-specific immunostimulation, is one of the most important immunotherapeutic strategies and holds tremendous potential for cancer treatment/prevention. Here, we prepare a series of nanoparticles composed of doxorubicin- and tyrosine kinase inhibitor-loaded and hyaluronic acid-coated dendritic polymers (termed HDDT nanoparticles) and find that the HDDT nanoparticles can convert various cancer cells to micrometer-sized vesicles (1.6-3.2 μm; termed HMVs) with ~100% cell-to-HMV conversion efficiency. We confirm in two tumor-bearing mouse models that the nanoparticles can restrain tumor growth, induce robust immunogenic cell death, and convert the primary tumor into an antigen depot by producing HMVs in situ to serve as personalized vaccines for cancer immunotherapy. Furthermore, the HDDT-healed mice show a strong immune memory effect and the HDDT treatment can realize long-term protection against tumor rechallenge. Collectively, the present work provides a general strategy for the preparation of tumor-associated antigen-containing vesicles and the development of personalized cancer vaccines.
Collapse
|
37
|
Chen Y, Wang Z, Wang X, Su M, Xu F, Yang L, Jia L, Zhang Z. Advances in Antitumor Nano-Drug Delivery Systems of 10-Hydroxycamptothecin. Int J Nanomedicine 2022; 17:4227-4259. [PMID: 36134205 PMCID: PMC9482956 DOI: 10.2147/ijn.s377149] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/25/2022] [Indexed: 01/10/2023] Open
Abstract
10-Hydroxycamptothecin (HCPT) is a natural plant alkaloid from Camptotheca that shows potent antitumor activity by targeting intracellular topoisomerase I. However, factors such as instability of the lactone ring and insolubility in water have limited the clinical application of this drug. In recent years, unprecedented advances in biomedical nanotechnology have facilitated the development of nano drug delivery systems. It has been found that nanomedicine can significantly improve the stability and water solubility of HCPT. NanoMedicines with different diagnostic and therapeutic functions have been developed to significantly improve the anticancer effect of HCPT. In this paper, we collected reports on HCPT nanomedicines against tumors in the past decade. Based on current research advances, we dissected the current status and limitations of HCPT nanomedicines development and looked forward to future research directions.
Collapse
Affiliation(s)
- Yukun Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Zhenzhi Wang
- Shaanxi University of Chinese Medicine, Xianyang, 712046, People's Republic of China
| | - Xiaofan Wang
- Department of Oncology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People's Republic of China
| | - Mingliang Su
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Fan Xu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Lian Yang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People's Republic of China
| |
Collapse
|
38
|
Kumar A, Ahmad A, Ansari MM, Gowd V, Rashid S, Chaudhary AA, Rudayni HA, Alsalamah SA, Khan R. Functionalized-DNA nanostructures as potential targeted drug delivery systems for cancer therapy. Semin Cancer Biol 2022; 86:54-68. [PMID: 36087856 DOI: 10.1016/j.semcancer.2022.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 09/01/2022] [Accepted: 09/03/2022] [Indexed: 01/14/2023]
Abstract
Seeman's pioneer idea has led to the foundation of DNA nanostructures, resulting in a remarkable advancement in DNA nanotechnology. Over the last few decades, remarkable advances in drug delivery techniques have resulted in the self-assembly of DNA for encapsulating candidate drug molecules. The nuclear targeting capability of DNA nanostructures is lies within their high spatial addressability and tremendous potential for active targeting. However, effective programming and assembling those DNA molecules remains a challenge, making the path to DNA nanostructures for real-world applications difficult. Because of their small size, most nanostructures are self-capable of infiltrating into the tumor cellular environment. Furthermore, to enable controlled and site-specific delivery of encapsulated drug molecules, DNA nanostructures are functionalized with special moieties that allow them to bind specific targets and release cargo only at targeted sites rather than non-specific sites, resulting in the prevention/limitation of cellular toxicity. In light of this, the current review seeks to shed light on the versatility of the DNA molecule as a targeting and encapsulating moiety for active drugs in order to achieve controlled and specific drug release with spatial and temporal precision. Furthermore, this review focused on the challenges associated with the construction of DNA nanostructures as well as the most recent advances in the functionalization of DNA nanostructures using various materials for controlled and targeted delivery of medications for cancer therapy.
Collapse
Affiliation(s)
- Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Anas Ahmad
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Md Meraj Ansari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Vemana Gowd
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, Riyadh, 11623, Saudi Arabia
| | - Hassan Ahmed Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, Riyadh, 11623, Saudi Arabia
| | - Sulaiman A Alsalamah
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, Riyadh, 11623, Saudi Arabia
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali 140306, Punjab, India.
| |
Collapse
|
39
|
Jiang Z, Tang H, Xiong Q, Li M, Dai Y, Zhou Z. Placental cell translocation of folate-conjugated pullulan acetate non-spherical nanoparticles. Colloids Surf B Biointerfaces 2022; 216:112553. [PMID: 35598508 DOI: 10.1016/j.colsurfb.2022.112553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/26/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Due to the adverse effects of free drugs on the fetus, placental-mediated pregnancy complications still lack effective pharmacotherapy. This study aims to construct a non-spherical drug delivery system based on folate-conjugated pullulan acetate (FPA) for placental targeting and translocation. By adjusting the initial solvent system, FPA nanoparticles with different morphologies were prepared using dialysis method without a surfactant. Cytotoxicity and lactate dehydrogenase release assays indicated the good biocompatibility of FPA nanoparticles in BeWo b30 cells. Cellular uptake and in vitro placental barrier transportation studies showed that FPA nanoparticles entered the cells and transported across the cell monolayer, benefiting from the active target effect mediated by the folate receptor. Moreover, non-spherical FPA nanoparticles showed nuclear translocation due to their shape effect. These findings provide a novel aspect in placental-mediated pregnancy treatment and applications in the obstetrics field of drug use during pregnancy.
Collapse
Affiliation(s)
- Ziwen Jiang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Hongbo Tang
- Department of Pharmacy, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China.
| | - Qingqing Xiong
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Min Li
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| | - Yinmei Dai
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China.
| | - Zhimin Zhou
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China.
| |
Collapse
|
40
|
Long X, Zhang X, Chen Q, Liu M, Xiang Y, Yang Y, Xiao Z, Huang J, Wang X, Liu C, Nan Y, Huang Q. Nucleus-Targeting Phototherapy Nanodrugs for High-Effective Anti-Cancer Treatment. Front Pharmacol 2022; 13:905375. [PMID: 35645841 PMCID: PMC9130747 DOI: 10.3389/fphar.2022.905375] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 04/19/2022] [Indexed: 12/30/2022] Open
Abstract
DNA is always one of the most important targets for cancer therapy due to its leading role in the proliferation of cancer cells. Phototherapy kills cancer cells by generating reactive oxygen species (ROS) and local hyperthermia under light. It has attracted extensive interest in the clinical treatment of tumors because of many advantages such as non-invasiveness, high patient compliance, and low toxicity and side effects. However, the short ROS diffusion distance and limited thermal diffusion rate make it difficult for phototherapy to damage DNA deep in the nucleus. Therefore, nucleus-targeting phototherapy that can destroy DNAs via in-situ generation of ROS and high temperature can be a very effective strategy to address this bottleneck. Recently, some emerging nucleus-targeting phototherapy nanodrugs have demonstrated extremely effective anticancer effects. However, reviews in the field are still rarely reported. Here, we comprehensively summarized recent advances in nucleus-targeting phototherapy in recent years. We classified nucleus-targeting phototherapy into three categories based on the characteristics of these nucleus-targeting strategies. The first category is the passive targeting strategy, which mainly targets the nucleus by adjusting the physicochemical characteristics of phototherapy nanomedicines. The second category is to mediate the phototherapy nanodrugs into the nucleus by modifying functional groups that actively target the nucleus. The third category is to assist nanodrugs enter into the nucleus in a light-controlled way. Finally, we provided our insights and prospects for nucleus-targeting phototherapy nanodrugs. This minireview provides unique insights and valuable clues in the design of phototherapy nanodrugs and other nucleus-targeting drugs.
Collapse
Affiliation(s)
- Xingyu Long
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xiaojie Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Min Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuting Xiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yuqi Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Jia Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xiaoyuan Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Chong Liu
- Departments of Clinical Pharmacology and Pharmacy, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Institute of Clinical Pharmacology, Ministry of Education, Central South University, Changsha, China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
41
|
Du W, Zhang L, Li X, Ling G, Zhang P. Nuclear targeting Subcellular-delivery nanosystems for precise cancer treatment. Int J Pharm 2022; 619:121735. [DOI: 10.1016/j.ijpharm.2022.121735] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 12/20/2022]
|
42
|
Yang F, Ji Q, Liao R, Li S, Wang Y, Zhang X, Zhang S, Zhang H, Kan Q, Sun J, He Z, Sun B, Luo C. Precisely engineering a dual-drug cooperative nanoassembly for proteasome inhibition-potentiated photodynamic therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.11.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
43
|
Wang Z, Wu F. Emerging Single-Atom Catalysts/Nanozymes for Catalytic Biomedical Applications. Adv Healthc Mater 2022; 11:e2101682. [PMID: 34729955 DOI: 10.1002/adhm.202101682] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/10/2021] [Indexed: 12/29/2022]
Abstract
Single-atom catalysts (SACs) are a type of atomically dispersed nanozymes with the highest atom utilization, which employ low-coordinated single atoms as the catalytically active sites. SACs not only inherit the merits of traditional nanozymes, but also hold high catalytic activity and superb catalytic selectivity, which ensure their tremendous application potential in environmental remediation, energy storage and conversion, chemical industry, nanomedicine, etc. Nevertheless, undesired aggregation effect of single atoms during preactivation and reaction processes is significantly enhanced owing to the high surface free energy of single atoms. In this case, appropriate substrates are requisite to prevent the aggregation event through the powerful interactions between the single atoms and the substrates, thereby stabilizing the high catalytic activity of the catalysts. In this review, the synthetic methods and characterization approaches of SACs are first described. Then the application cases of SACs in nanomedicine are summarized. Finally, the current challenges and future opportunities of the SACs in nanomedicine are outlined. It is hoped that this review may have implications for furthering the development of new SACs with improved biophysicochemical properties and broadened biomedical applications.
Collapse
Affiliation(s)
- Zihao Wang
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering Southeast University 2 Sipailou Road Nanjing 210096 P. R. China
| | - Fu‐Gen Wu
- State Key Laboratory of Bioelectronics School of Biological Science and Medical Engineering Southeast University 2 Sipailou Road Nanjing 210096 P. R. China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University) Ministry of Education 22 Shuangyong Road Nanning 530022 P. R. China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor 22 Shuangyong Road Nanning 530022 P. R. China
| |
Collapse
|
44
|
Xiang Y, Zheng S, Yuan S, Wang J, Wu Y, Zhu X. Near-infrared mediated orthogonal bioimaging and intracellular tracking of upconversion nanophotosensitizers. Mikrochim Acta 2022; 189:120. [PMID: 35201432 DOI: 10.1007/s00604-022-05218-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
Abstract
Although upconversion photodynamic therapy (PDT) has gained extensive interests in disease treatment, the intracellular migration pathway of upconversion photosensitizers and underlying cell-particle interaction mechanism is still largely unexplored. In this work photoswitchable upconversion nanoparticles (UCNPs) are reported that can release orthogonal emissions excited by two near-infrared lights, i.e., red color of 980-nm and green color of 808-nm light excitation. Taking advantage of the dual-emissive property, a methodology based on Pearson's correlation analysis is proposed to verify the accuracy of upconversion luminescence signals under different excitation lights, which has been previously neglected. Meanwhile, we have designed a near-infrared mediated bioimaging nanoplatform that can generate reactive oxygen species (ROS) using one light and simultaneously track the location of upconversion photosensitizers using another excitation light. Our study not only depicts the migration pathway of upconversion photosensitizers, but also demonstrates the organelle escape of these upconversion nanoparticles via PCI (photochemical internalization) process. It is believed that our results inspire more efficient synergistic therapy by combining PDT with other modalities in a programmable manner.
Collapse
Affiliation(s)
- Yi Xiang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Shanshan Zheng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Shanshan Yuan
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Jing Wang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yihan Wu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Xiaohui Zhu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
45
|
Zhao H, Xu J, Wang Y, Sun C, Bao L, Zhao Y, Yang X, Zhao Y. A Photosensitizer Discretely Loaded Nanoaggregate with Robust Photodynamic Effect for Local Treatment Triggers Systemic Antitumor Responses. ACS NANO 2022; 16:3070-3080. [PMID: 35038865 DOI: 10.1021/acsnano.1c10590] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT), is a rising star for suppression of in situ and metastatic tumors, yet it is impeded by low ROS production and off-target phototoxicity. Herein, an aggregation degree editing strategy, inspired by gene editing, was accomplished by the coordination of an aggregation degree editor, p(MEO2MA160-co-OEGMA40)-b-pSS30 [POEGS; MEO2MA = 2-(2-methoxyethoxy)ethyl methacrylate, OEGMA = oligo(ethylene glycol) methacrylate; pSS = poly(styrene sulfonate)] and indocyanine green (ICG) to nontoxic Mg2+, forming an ICG discretely loaded nanoaggregate (ICG-DNA). Optimization of the ICG aggregation degree [POEGS/ICG (P/I) = 6.55] was achieved by tuning the P/I ratio, alleviating aggregation-caused-quenching (ACQ) and photobleaching concurrently. The process boosts the PDT efficacy, spurring robust immunogenic cell death (ICD) and systemic antitumor immunity against primary and metastatic immunogenic "cold" 4T1 tumors via intratumoral administration. Moreover, the temperature-sensitive phase-transition property facilitates intratumoral long-term retention of ICG-DNA, reducing undesired phototoxicity to normal tissues; meanwhile, the photothermal-induced tumor oxygenation further leads to an augmented PDT outcome. Thus, this simple strategy improves PDT efficacy, boosting the singlet oxygen quantum yield (ΦΔ)-dependent ICD effect and systemic antitumor responses via local treatment.
Collapse
Affiliation(s)
- Hao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | | | - Yuqiao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | | | - Lin Bao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | | | - Xiangliang Yang
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510530, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510530, China
| |
Collapse
|
46
|
Gbetuwa M, Lu LS, Wang TJ, Chen YJ, Chiou JF, Su TY, Yang TS. Nucleus Near-Infrared (nNIR) Irradiation of Single A549 Cells Induces DNA Damage and Activates EGFR Leading to Mitochondrial Fission. Cells 2022; 11:cells11040624. [PMID: 35203275 PMCID: PMC8870661 DOI: 10.3390/cells11040624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/29/2022] [Accepted: 02/08/2022] [Indexed: 12/10/2022] Open
Abstract
There has been great interest in identifying the biological substrate for light-cell interaction and their relations to cancer treatment. In this study, a near-infrared (NIR) laser is focused into the nucleus (nNIR) or cytoplasm (cNIR) of a single living cell by a high numerical aperture condenser to dissect the novel role of cell nucleus in mediating NIR effects on mitochondrial dynamics of A549 non-small cell lung cancer cells. Our analysis showed that nNIR, but not cNIR, triggered mitochondrial fission in 10 min. In contrast, the fission/fusion balance of mitochondria directly exposed to cNIR does not change. While the same phenomenon is also triggered by single molecular interactions between epidermal growth factor (EGF) and its receptor EGFR, pharmacological studies with cetuximab, PD153035, and caffeine suggest EGF signaling crosstalk to DNA damaging response to mediate rapid mitochondrial fission as a result of nNIR irradiation. These results suggest that nuclear DNA integrity is a novel biological target for cellular response to NIR.
Collapse
Affiliation(s)
- Momoh Gbetuwa
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110, Taiwan; (M.G.); (L.-S.L.); (Y.-J.C.)
| | - Long-Sheng Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110, Taiwan; (M.G.); (L.-S.L.); (Y.-J.C.)
- International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan;
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Center for Cell Therapy, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
- International PhD Program for Cell Therapy and Regeneration, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-Jen Wang
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110, Taiwan; (M.G.); (L.-S.L.); (Y.-J.C.)
- International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan;
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Jeng-Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan;
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiology, School of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Tai-Yuan Su
- Department of Electrical Engineering, Yuan-Ze University, Chung-Li 32003, Taiwan;
| | - Tzu-Sen Yang
- International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei 110, Taiwan
- School of Dental Technology, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Biomedical Device, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: ; Tel.: +886-2-27361661 (ext. 5206)
| |
Collapse
|
47
|
Wu H, Xia F, Zhang L, Fang C, Lee J, Gong L, Gao J, Ling D, Li F. A ROS-Sensitive Nanozyme-Augmented Photoacoustic Nanoprobe for Early Diagnosis and Therapy of Acute Liver Failure. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108348. [PMID: 34839560 DOI: 10.1002/adma.202108348] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/25/2021] [Indexed: 06/13/2023]
Abstract
Early diagnosis of acute liver failure (ALF) is critical for curable treatment of patients, because most existing ALF therapies have narrow therapeutic time windows after disease onset. Reactive oxygen species (ROS), which lead to the sequential occurrences of hepatocyte necrosis and the leakage of alanine aminotransferase (ALT), represent early biomarkers of ALF. Photoacoustic imaging is emerging as a powerful tool for in vivo imaging of ROS. However, high-performance imaging probes that can boost the photoacoustic signals of the short-lived ROS of ALF are yet to be developed, and there remains a great challenge for ROS-based imaging of ALF. Herein, a ROS-sensitive nanozyme-augmented photoacoustic nanoprobe for successful in vivo imaging of ALF is presented. The deep-penetrating photoacoustic signals of the nanoprobe can be activated by the overexpressed ROS in ALF due to the synergy between nanocatalytic bubbles generation and thermoelastic expansion. Impressively, the nanozyme-augmented ROS imaging enables earlier diagnosis of ALF than the clinical ALT method, and the ROS-activated catalytic activity of nanoprobe permits timely nanocatalytic therapy of ALF.
Collapse
Affiliation(s)
- Haibin Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Fan Xia
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Lingxiao Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Chunyan Fang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jiyoung Lee
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Linji Gong
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Daishun Ling
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310012, P. R. China
| | - Fangyuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310012, P. R. China
| |
Collapse
|
48
|
Li C, Wang Q, Li D, Liu Y, Hu B, Feng Y, Zhang H, He Z, Luo C, Sun J. Molecular recognition-driven supramolecular nanoassembly of a hydrophobic uracil prodrug and hydrophilic cytarabine for precise combination treatment of solid and non-solid tumors. NANOSCALE HORIZONS 2022; 7:235-245. [PMID: 35048915 DOI: 10.1039/d1nh00590a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Combination chemotherapy has shown distinct therapeutic advantages over monotherapy in clinical cancer treatment, especially for two chemotherapeutic drugs with different mechanisms of action. However, how to achieve efficient co-delivery of two or more drugs with different physicochemical and pharmacokinetic properties for synergistic therapy is still a huge challenge. In particular, it is even more difficult to efficiently co-deliver a hydrophilic drug and a hydrophobic drug into one nanosystem. Herein, inspired by the natural Watson-Crick base pair molecular recognition in nucleic acids, a reduction-sensitive uracil prodrug of doxorubicin (U-SS-DOX) is synthesized and performs supramolecular co-assembly with cytarabine (Ara-C). Interestingly, the hydrophilic Ara-C molecules could readily co-assemble with U-SS-DOX, and multiple hydrogen bonds are found in the nanoassembly with an ultra-high drug loading rate. Moreover, 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide (DiR) is used as a fluorescent probe to investigate the pharmacokinetics of U : C NPs. It turns out that the DiR-labeled U : C NPs significantly prolong the systemic circulation and promote the tumor-specific accumulation of DiR when compared with DiR solution. Furthermore, the supramolecular nanoassembly demonstrates potent satisfactory therapeutic effects in treating both solid and non-solid tumors in vivo. This study provides a novel molecular co-assembly nanoplatform for efficient co-delivery of hydrophilic and hydrophobic drugs.
Collapse
Affiliation(s)
- Chang Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China.
| | - Qiu Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China.
| | - Dan Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China.
| | - Yubo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China.
| | - Baichun Hu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Yao Feng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Haotian Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China.
| | - Cong Luo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China.
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China.
| |
Collapse
|
49
|
Zhang Y, Pei Q, Yue Y, Xie Z. Binary dimeric prodrug nanoparticles for self-boosted drug release and synergistic chemo-photodynamic therapy. J Mater Chem B 2022; 10:880-886. [PMID: 35043826 DOI: 10.1039/d1tb02638k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chemotherapy is the major strategy for cancer therapy, but its limited therapeutic efficiency and serious toxicity to normal tissues greatly restrict its clinical performance. Herein, we develop carrier-free self-activated prodrug nanoparticles combining chemotherapy and photodynamic therapy to enhance the antitumor efficiency. Reactive oxygen species (ROS)-responsive paclitaxel and porphyrin prodrugs are synthesized and co-assembled into nanoparticles without the addition of any adjuvants, which improves the drug content and reduces carrier-associated toxicity. After entering cancer cells, the obtained co-assembled nanoparticles can generate sufficient ROS upon light irradiation not only for photodynamic therapy, but also triggering on-demand drug release for chemotherapy, thus realizing self-enhanced prodrug activation and synergistic chemo-photodynamic therapy. This simple and effective carrier-free prodrug nanoplatform unifies the distinct traits of on-demand drug release and combination therapy, thus possessing great potential in advancing cancer treatment.
Collapse
Affiliation(s)
- Youwei Zhang
- Department of Gynecological Oncology, The First Hospital of Jilin University, Xinmin Street, Changchun, Jilin 130021, P. R. China.
| | - Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
| | - Ying Yue
- Department of Gynecological Oncology, The First Hospital of Jilin University, Xinmin Street, Changchun, Jilin 130021, P. R. China.
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
| |
Collapse
|
50
|
Sarker P, Sajib MSJ, Tao X, Wei T. Multiscale Simulation of Protein Corona Formation on Silver Nanoparticles: Study of Ovispirin-1 Peptide Adsorption. J Phys Chem B 2022; 126:601-608. [PMID: 35026946 DOI: 10.1021/acs.jpcb.1c08267] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The exposure of nanoparticles (NPs) to biofluids leads to the rapid coverage of proteins, named protein corona, which alters the NPs' chemicophysical and biological properties. Fundamental studies of the protein corona are thus critical to the increasing applications of NPs in nanotechnology and nanomedicines. The present work utilizes multiscale simulations of a model biological system, small ovispirin-1 peptides, and bare silver nanoparticles (AgNPs) to examine the NPs' size and surface hydrophilicity effects on formation dynamics and the structure of the peptide corona. Our simulations revealed the different adsorption dynamics of ovispirin-1 peptides on the NPs, including the direct adsorption of a single peptide and peptide aggregates and multistep adsorption, as well as an intermediate cycle of desorption and readsorption. Notably, the whole process of peptide adsorption on hydrophilic AgNP surfaces can be generalized as three stages: diffusion to the surface, initial landing via hydrophilic residues, and the final attachment. The decrease in AgNP's size leads to faster adsorption with more heterogeneous peptide interfacial dynamics, a denser and inhomogeneous peptide packing structure, and a wider distribution of adsorption orientations. Subsequent atomistic molecular dynamics simulations demonstrated that on the hydrophilic AgNP surfaces, adsorbed peptides display moderate changes in their secondary structure, resulting in further changes of corona composition, i.e., amino acid residue distribution on the surface.
Collapse
Affiliation(s)
- Pranab Sarker
- Department of Chemical Engineering, Howard University, Washington, D.C. 20059, United States
| | - Md Symon Jahan Sajib
- Department of Chemical Engineering, Howard University, Washington, D.C. 20059, United States
| | - Xiuping Tao
- Department of Chemistry, Winston-Salem State University, Winston-Salem, North Carolina 27110, United States
| | - Tao Wei
- Department of Chemical Engineering, Howard University, Washington, D.C. 20059, United States
| |
Collapse
|