1
|
Yun Y, An J, Kim HJ, Choi HK, Cho HY. Recent advances in functional lipid-based nanomedicines as drug carriers for organ-specific delivery. NANOSCALE 2025; 17:7617-7638. [PMID: 40026004 DOI: 10.1039/d4nr04778h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Lipid-based nanoparticles have emerged as promising drug delivery systems for a wide range of therapeutic agents, including plasmids, mRNA, and proteins. However, these nanoparticles still encounter various challenges in drug delivery, including drug leakage, poor solubility, and inadequate target specificity. In this comprehensive review, we present an in-depth investigation of four distinct drug delivery methods: liposomes, lipid nanoparticle formulations, solid lipid nanoparticles, and nanoemulsions. Moreover, we explore recent advances in lipid-based nanomedicines (LBNs) for organ-specific delivery, employing ligand-functionalized particles that specifically target receptors in desired organs. Through this strategy, LBNs enable direct and efficient drug delivery to the intended organs, leading to superior DNA or mRNA expression outcomes compared to conventional approaches. Importantly, the development of novel ligands and their judicious combination holds promise for minimizing the side effects associated with nonspecific drug delivery. By leveraging the unique properties of lipid-based nanoparticles and optimizing their design, researchers can overcome the limitations associated with current drug delivery systems. In this review, we aim to provide valuable insights into the advancements, challenges, and future directions of lipid-based nanoparticles in the field of drug delivery, paving the way for enhanced therapeutic strategies with improved efficacy and reduced adverse effects.
Collapse
Affiliation(s)
- Yeochan Yun
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea.
| | - Jeongmin An
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea.
| | - Hyun Joong Kim
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea.
| | - Hye Kyu Choi
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, USA
| | - Hyeon-Yeol Cho
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea.
| |
Collapse
|
2
|
Kailasa SK, Makwana KP, Deshpande MP, Choi Y, Stephanie R, Park CY, Park TJ. Synthesis of trypsin-protected CsPbCl 3 fluorescent nanocrystals for hydroxyl radical sensing. Mikrochim Acta 2025; 192:217. [PMID: 40063164 PMCID: PMC11893688 DOI: 10.1007/s00604-025-07070-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025]
Abstract
Water-dispersible perovskite nanocrystals (PNCs) show promising applications in recognizing ionic and molecular species because of their excellent optical properties. However, lead halide PNCs have some limitations when they are used as probes for molecular species sensing in aqueous media. Here, we introduce trypsin (Try) as a bioligand for the synthesis of cesium lead chloride (CsPbCl3) PNCs with high water stability. The as-fabricated Try-CsPbCl3 PNCs show λEm/Ex at 433/370 nm with a quantum yield of 17.26%. The fluorescence emission spectral characteristics of Try-CsPbCl3 PNCs demonstrated that water-stable Try-CsPbCl3 PNCs acted as a promising fluorescent probe for the detection of hydroxyl radical (•OH) via turn-off mechanism. The Try-CsPbCl3 PNCs-based turn-off fluorescence approach displayed good selectivity for hydroxyl radical in water, showing a wider linear range (0.01-5 µM) with a remarkable detection limit of 3.10 nM for hydroxyl radical. The Try-CsPbCl3 PNCs were demonstrated to be a facile probe for sensing •OH in water samples, which signifies that Try-CsPbCl3 PNCs exhibited broad applications for hydroxyl radical sensing in real samples.
Collapse
Affiliation(s)
- Suresh Kumar Kailasa
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, 395 007, India
| | | | - Madhura Pradeep Deshpande
- Department of Chemistry, Research Institute of Chem-Bio Diagnostic Technology, Chung-Ang University, 84, Heukseok-Ro, Dongjak-Gu, Seoul, 06974, Republic of Korea
| | - Yoojin Choi
- Department of Chemistry, Research Institute of Chem-Bio Diagnostic Technology, Chung-Ang University, 84, Heukseok-Ro, Dongjak-Gu, Seoul, 06974, Republic of Korea
| | - Ruth Stephanie
- Department of Chemistry, Research Institute of Chem-Bio Diagnostic Technology, Chung-Ang University, 84, Heukseok-Ro, Dongjak-Gu, Seoul, 06974, Republic of Korea
| | - Chan Yeong Park
- Department of Chemistry, Research Institute of Chem-Bio Diagnostic Technology, Chung-Ang University, 84, Heukseok-Ro, Dongjak-Gu, Seoul, 06974, Republic of Korea
| | - Tae Jung Park
- Department of Chemistry, Research Institute of Chem-Bio Diagnostic Technology, Chung-Ang University, 84, Heukseok-Ro, Dongjak-Gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
3
|
Barman S, Roy SM, Kishore P, Ghosh M, Bag P, Sarkar AK, Ghatak T, Maji PS, Basu A, Mukherjee R, Ghosh SK, Chowdhury AD, Maity AR. Concurrent targeted delivery of doxorubicin and curcumin to the cancer cells using simple and versatile ligand-installed multifaceted chitosan-based nanoconjugates. J Mater Chem B 2025; 13:2490-2503. [PMID: 39834258 DOI: 10.1039/d4tb01809e] [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: 01/22/2025]
Abstract
Existing chemotherapeutic approaches against refractory cancers are ineffective due to off-target effects, inefficient delivery, and inadequate accumulation of anticancer drugs at the tumor site, which causes limited efficiency of drug treatment and toxicity to neighboring healthy cells. The development of nano-based drug delivery systems (DDSs) with the goal of delivering desired therapeutic doses to the diseased cells and has already proven to be a promising strategy to address these challenges. Our study focuses on achieving an efficient tumor-targeted delivery of a combination of drugs for therapeutic benefits by developing a versatile DDS by following a simple one-step chemical approach. We used low-molecular-weight chitosan and modified its primary amine groups with reactive forms of cholesterol and folic acid by simple chemical tools and thus prepared folic acid-chitosan-cholesterol graft copolymer. The polymer contains numerous residual primary amine groups, which offer enough water solubility and positive charge to its polymeric backbone to foster the interaction of negatively charged and/or hydrophobic drugs to load and encapsulate a wide variety of drugs within it via various non-bonding interactions. We used curcumin and doxorubicin as the combination of drugs and thus finally prepared targeted nanoconjugates (targeted NCs). In vitro cellular experiments show that our developed targeted NCs demonstrate 3-5 times higher cellular uptake than non-targeted NCs at various incubation times (2 h, 8 h, and 12 h) in KB cells where folate receptors are overexpressed. This enhanced cellular uptake of targeted NCs and the following delivery of drugs in the cytosol and its disposition to the nucleus exhibit a substantial amount of toxicity to KB cells towards an effective therapeutic strategy for treatment.
Collapse
Affiliation(s)
- Sourav Barman
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| | - Sayoni Maitra Roy
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| | - Purvi Kishore
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| | - Malabika Ghosh
- Amity Institute of Nanotechnology, Amity University, Kolkata, West Bengal, 700135, India
| | - Pousali Bag
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| | - Ankan Kumar Sarkar
- School of Materials Sciences, Indian Association for the Cultivation of Science, Kolkata, West Bengal, 700032, India
| | - Tapas Ghatak
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Partha Sona Maji
- Department of Physics, Amity Institute of Applied Sciences, Amity University, Kolkata, West Bengal, 700135, India
| | - Arnab Basu
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Belur Math, Howrah, West Bengal, 711202, India
| | - Rupam Mukherjee
- Department of Physics, Presidency University, Bangalore, Karnataka, 560064, India
| | - Surya K Ghosh
- Department of Physics, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Ankan Dutta Chowdhury
- Amity Institute of Nanotechnology, Amity University, Kolkata, West Bengal, 700135, India
| | - Amit Ranjan Maity
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
4
|
Singh P, Pandit S, Balusamy SR, Madhusudanan M, Singh H, Amsath Haseef HM, Mijakovic I. Advanced Nanomaterials for Cancer Therapy: Gold, Silver, and Iron Oxide Nanoparticles in Oncological Applications. Adv Healthc Mater 2025; 14:e2403059. [PMID: 39501968 PMCID: PMC11804848 DOI: 10.1002/adhm.202403059] [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: 08/26/2024] [Revised: 10/07/2024] [Indexed: 01/05/2025]
Abstract
Cancer remains one of the most challenging health issues globally, demanding innovative therapeutic approaches for effective treatment. Nanoparticles, particularly those composed of gold, silver, and iron oxide, have emerged as promising candidates for changing cancer therapy. This comprehensive review demonstrates the landscape of nanoparticle-based oncological interventions, focusing on the remarkable advancements and therapeutic potentials of gold, silver, and iron oxide nanoparticles. Gold nanoparticles have garnered significant attention for their exceptional biocompatibility, tunable surface chemistry, and distinctive optical properties, rendering them ideal candidates for various cancer diagnostic and therapeutic strategies. Silver nanoparticles, renowned for their antimicrobial properties, exhibit remarkable potential in cancer therapy through multiple mechanisms, including apoptosis induction, angiogenesis inhibition, and drug delivery enhancement. With their magnetic properties and biocompatibility, iron oxide nanoparticles offer unique cancer diagnosis and targeted therapy opportunities. This review critically examines the recent advancements in the synthesis, functionalization, and biomedical applications of these nanoparticles in cancer therapy. Moreover, the challenges are discussed, including toxicity concerns, immunogenicity, and translational barriers, and ongoing efforts to overcome these hurdles are highlighted. Finally, insights into the future directions of nanoparticle-based cancer therapy and regulatory considerations, are provided aiming to accelerate the translation of these promising technologies from bench to bedside.
Collapse
Affiliation(s)
- Priyanka Singh
- The Novo Nordisk FoundationCenter for BiosustainabilityTechnical University of DenmarkKogens LyngbyDK‐2800Denmark
| | - Santosh Pandit
- Systems and Synthetic Biology DivisionDepartment of Life SciencesChalmers University of TechnologyGothenburgSE‐412 96Sweden
| | - Sri Renukadevi Balusamy
- Department of Food Science and BiotechnologySejong UniversityGwangjin‐GuSeoul05006Republic of Korea
| | - Mukil Madhusudanan
- The Novo Nordisk FoundationCenter for BiosustainabilityTechnical University of DenmarkKogens LyngbyDK‐2800Denmark
| | - Hina Singh
- Division of Biomedical SciencesSchool of MedicineUniversity of CaliforniaRiversideCA92521USA
| | | | - Ivan Mijakovic
- The Novo Nordisk FoundationCenter for BiosustainabilityTechnical University of DenmarkKogens LyngbyDK‐2800Denmark
- Systems and Synthetic Biology DivisionDepartment of Life SciencesChalmers University of TechnologyGothenburgSE‐412 96Sweden
| |
Collapse
|
5
|
Alamelu S, Venkatesan KB, Srinivasan MK, Pachaiappan P. Synthesis and Characterization of Brucine Gold Nanoparticles for Targeted Breast Cancer Therapy: Mechanistic Insights Into Apoptosis and Antioxidant Disruption in MCF-7 Cells. Biotechnol Appl Biochem 2025. [PMID: 39789744 DOI: 10.1002/bab.2719] [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: 09/14/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025]
Abstract
Globally, breast cancer continues to be the leading type of cancer affecting women, with rising mortality rates projected by 2030. This highlights the importance of developing new, affordable treatments, like drug delivery systems that use nanoparticles. Gold nanoparticles (AuNPs), including their exceptional optical and physical attributes, make them an attractive vehicle for targeted treatment, allowing for accurate and focused delivery of medication directly to cancerous cells while reducing harmful side effect. This study focuses on the synthesis and characterization of brucine-gold nanoparticles (BRU-AuNPs) for targeted breast cancer therapy by evaluating their antioxidant and apoptotic mechanism. BRU-AuNPs were synthesized and characterized (UV-Vis spectroscopy, Fourier transform infrared [FTIR], scanning electron microscopy [SEM], x-ray diffraction [XRD], dynamic light scattering [DLS], and zeta potential) to confirm successful synthesis, size, and stability. In vitro studies were assessed using MCF-7 breast cancer cell lines to evaluate cell cytotoxicity, antioxidant balance, reactive oxygen species (ROS) generation, mitochondrial membrane potential, apoptosis induction, cell migration, and pro-apoptotic gene expression. Characterization results confirmed the successful synthesis of BRU-AuNPs with an average crystal size of 85.40 nm and stable surface charge. Results demonstrated that BRU-AuNPs reduced MCF-7 cell viability in a dose-dependent manner, with an IC50 value of 11.47 µg/mL. Treatment with BRU-AuNPs altered the antioxidant balance, increased ROS generation, depolarized mitochondrial membranes, and induced apoptosis. Additionally, BRU-AuNPs inhibited cell migration and upregulated pro-apoptotic gene expression. The synthesized BRU-AuNPs exhibit potential as a highly effective targeted delivery system for breast cancer treatment. Their ability to directly deliver BRU to tumor cells while reducing side effects and enhancing therapeutic efficacy underscores their promise in advancing breast cancer therapy. Further studies are warranted to explore their clinical potential and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Saravanan Alamelu
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Kamalesh Balakumar Venkatesan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Manoj Kumar Srinivasan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Pugalendhi Pachaiappan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| |
Collapse
|
6
|
Jia X, Wang Y, Qiao Y, Jiang X, Li J. Nanomaterial-based regulation of redox metabolism for enhancing cancer therapy. Chem Soc Rev 2024; 53:11590-11656. [PMID: 39431683 DOI: 10.1039/d4cs00404c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Altered redox metabolism is one of the hallmarks of tumor cells, which not only contributes to tumor proliferation, metastasis, and immune evasion, but also has great relevance to therapeutic resistance. Therefore, regulation of redox metabolism of tumor cells has been proposed as an attractive therapeutic strategy to inhibit tumor growth and reverse therapeutic resistance. In this respect, nanomedicines have exhibited significant therapeutic advantages as intensively reported in recent studies. In this review, we would like to summarize the latest advances in nanomaterial-assisted strategies for redox metabolic regulation therapy, with a focus on the regulation of redox metabolism-related metabolite levels, enzyme activity, and signaling pathways. In the end, future expectations and challenges of such emerging strategies have been discussed, hoping to enlighten and promote their further development for meeting the various demands of advanced cancer therapies. It is highly expected that these therapeutic strategies based on redox metabolism regulation will play a more important role in the field of nanomedicine.
Collapse
Affiliation(s)
- Xiaodan Jia
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Wang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Qiao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiue Jiang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Jinghong Li
- Beijing Institute of Life Science and Technology, Beijing 102206, P. R. China
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
7
|
Tripathy S, Haque S, Londhe S, Das S, Norbert CC, Chandra Y, Sreedhar B, Patra CR. ROS mediated Cu[Fe(CN) 5NO] nanoparticles for triple negative breast cancer: A detailed study in preclinical mouse model. BIOMATERIALS ADVANCES 2024; 160:213832. [PMID: 38547763 DOI: 10.1016/j.bioadv.2024.213832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 05/04/2024]
Abstract
Triple negative breast cancer (TNBC) is an aggressive form of tumor, more prevalent in younger women resulting in poor survival rate (2nd in cancer deaths) because of its asymptomatic existence. The most popular and convenient approach for the treatment of TNBC is chemotherapy which is associated with several limitations. Considering the importance of nanotechnology in health care system, in the present manuscript, we have designed and developed a simple, efficient, cost effective, and ecofriendly method for the synthesis of copper nitroprusside analogue nanoparticles (Cu[Fe(CN)5NO] which is abbreviated as CuNPANP that may be the potential anti-cancer nanomedicine for the treatment of TNBC. Copper (present in CuNPANP) is used because of its affordability, nutritional value and various biomedical applications. The CuNPANP are thoroughly characterized using several analytical techniques. The in vitro cell viability (in normal cells) and the ex vivo hemolysis assay reveal the biocompatible nature of CuNPANP. The anti-cancer activity of the CuNPANP is established in TNBC cells (MDA-MB-231 and 4T1) through several in vitro assays along with plausible mechanisms. The intraperitoneal administration of CuNPANP in orthotopic breast tumor model by transplanting 4T1 cells into the mammary fat pad of BALB/c mouse significantly inhibits the growth of breast carcinoma as well as increases the survival time of tumor-bearing mice. These results altogether potentiate the anti-cancer efficacy of CuNPANP as a smart therapeutic nanomedicine for treating TNBC in near future after bio-safety evaluation in large animals.
Collapse
Affiliation(s)
- Sanchita Tripathy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Shagufta Haque
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Swapnali Londhe
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Sourav Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Caroline Celine Norbert
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
| | - Yogesh Chandra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Bojja Sreedhar
- Department of Analytical & Structural ChemistryCSIR-Indian Institute of Chemical Technology, Uppal Road,Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India.
| |
Collapse
|
8
|
Cho H, Yoo M, Pongkulapa T, Rabie H, Muotri AR, Yin PT, Choi J, Lee K. Magnetic Nanoparticle-Assisted Non-Viral CRISPR-Cas9 for Enhanced Genome Editing to Treat Rett Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306432. [PMID: 38647391 PMCID: PMC11200027 DOI: 10.1002/advs.202306432] [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: 10/26/2023] [Revised: 03/23/2024] [Indexed: 04/25/2024]
Abstract
The CRISPR-Cas9 technology has the potential to revolutionize the treatment of various diseases, including Rett syndrome, by enabling the correction of genes or mutations in human patient cells. However, several challenges need to be addressed before its widespread clinical application. These challenges include the low delivery efficiencies to target cells, the actual efficiency of the genome-editing process, and the precision with which the CRISPR-Cas system operates. Herein, the study presents a Magnetic Nanoparticle-Assisted Genome Editing (MAGE) platform, which significantly improves the transfection efficiency, biocompatibility, and genome-editing accuracy of CRISPR-Cas9 technology. To demonstrate the feasibility of the developed technology, MAGE is applied to correct the mutated MeCP2 gene in induced pluripotent stem cell-derived neural progenitor cells (iPSC-NPCs) from a Rett syndrome patient. By combining magnetofection and magnetic-activated cell sorting, MAGE achieves higher multi-plasmid delivery (99.3%) and repairing efficiencies (42.95%) with significantly shorter incubation times than conventional transfection agents without size limitations on plasmids. The repaired iPSC-NPCs showed similar characteristics as wild-type neurons when they differentiated into neurons, further validating MAGE and its potential for future clinical applications. In short, the developed nanobio-combined CRISPR-Cas9 technology offers the potential for various clinical applications, particularly in stem cell therapies targeting different genetic diseases.
Collapse
Affiliation(s)
- Hyeon‐Yeol Cho
- Department of Chemistry and Chemical BiologyRutgers, The State University of New JerseyPiscatawayNJ08854USA
- Department of Chemical and Biomolecular EngineeringSogang UniversitySeoul04107South Korea
- Department of Bio and Fermentation Convergence TechnologyKookmin UniversitySeoul02707South Korea
| | - Myungsik Yoo
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and NeuroscienceRutgers, The State University of New JerseyPiscatawayNJ08854USA
| | - Thanapat Pongkulapa
- Department of Chemistry and Chemical BiologyRutgers, The State University of New JerseyPiscatawayNJ08854USA
| | - Hudifah Rabie
- Department of Chemistry and Chemical BiologyRutgers, The State University of New JerseyPiscatawayNJ08854USA
| | - Alysson R. Muotri
- School of MedicineDepartment of Pediatrics/Rady Children's Hospital San DiegoDepartment of Cellular and Molecular MedicineStem Cell ProgramLa JollaCA92093USA
| | - Perry T. Yin
- Department of Biomedical EngineeringRutgersThe State University of New JerseyPiscatawayNJ08854USA
| | - Jeong‐Woo Choi
- Department of Chemical and Biomolecular EngineeringSogang UniversitySeoul04107South Korea
| | - Ki‐Bum Lee
- Department of Chemistry and Chemical BiologyRutgers, The State University of New JerseyPiscatawayNJ08854USA
| |
Collapse
|
9
|
Ouyang C, Zhang W, Nie J, Yu L, Liu J, Ren L, Chen G. Nanoparticles with Active Targeting Ability and Acid Responsiveness for an Enhanced Antitumor Effect of Docetaxel. Biomacromolecules 2024; 25:213-221. [PMID: 38116982 DOI: 10.1021/acs.biomac.3c00922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Docetaxel (DOC) is commonly used in cancer treatment, especially for breast cancer. However, there are severe side effects in clinical application. In order to deliver docetaxel more effectively, a novel, active targeting acid-responsive polymer called cRGD-PAE-PEG-DSPE was developed. The polymer structure incorporated poly(ethylene glycol) (PEG) as the hydrophilic segment, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) as the hydrophobic segment, and poly(β-amino ester) (PAE) as the acid-responsive group, which was grafted onto the PEG. Furthermore, c(RGDyC) was grafted onto PAE to confer active targeting capability. Through self-assembly, docetaxel was encapsulated in RAED@DOC. Through in vitro experiments, it was confirmed that RAED@DOC had good serum stability and acid responsiveness, as well as enhanced uptake by MDA-MB-231 cells. Additionally, the antitumor efficiency in vivo and histopathological analysis showed that RAED@DOC exhibited higher antitumor activity and lower systemic toxicity in comparison to free docetaxel. These results suggested that RAED@DOC had considerable potential clinical use.
Collapse
Affiliation(s)
| | - Wei Zhang
- Nanjing Tech University, Nanjing 211816, China
| | - Junfang Nie
- Nanjing Tech University, Nanjing 211816, China
| | - Luting Yu
- Nanjing Tech University, Nanjing 211816, China
| | - Jia Liu
- Nanjing Tech University, Nanjing 211816, China
| | - Lili Ren
- Nanjing Tech University, Nanjing 211816, China
| | | |
Collapse
|
10
|
Daviu N, Portilla Y, Gómez de Cedrón M, Ramírez de Molina A, Barber DF. DMSA-coated IONPs trigger oxidative stress, mitochondrial metabolic reprograming and changes in mitochondrial disposition, hindering cell cycle progression of cancer cells. Biomaterials 2024; 304:122409. [PMID: 38052135 DOI: 10.1016/j.biomaterials.2023.122409] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/06/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023]
Abstract
There is increasing interest in modulating the redox homeostasis of tumors since high levels of reactive oxygen species (ROS) make them more vulnerable to changes in these species. Nanomedicine offers promise in this context as such applications may provoke biological responses that induce ROS production. Indeed, iron oxide nanoparticles (IONPs) can induce ROS accumulation through the so-called Fenton reaction of iron, further augmenting the ROS in tumors and overloading the antioxidant system beyond its capacity, thereby driving oxidative stress to a level that is incompatible with cell survival. Here, three different coatings for IONPs were compared to assess their intrinsic capacity to induce ROS production in cells. Of these coatings, dimercaptosuccinic acid-coated IONPs (DMSA-NPs) provoked the strongest ROS production, which was associated with the ability to reprogram the metabolism of cancer cells. This latter phenomenon involved shutting-down oxidative phosphorylation (OXPHOS), shifting mitochondrial morphology towards a more elongated phenotype, reducing the total mitochondrial mass and ultimately, blocking cell proliferation by inducing G0/G1 cell cycle arrest. Consequently, the data obtained highlights the importance of studying the chemical properties of IONPs, presenting DMSA-NPs as a novel tool to induce oxidative stress in cancer cells and alter their cell fate.
Collapse
Affiliation(s)
- Neus Daviu
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049, Madrid, Spain
| | - Yadileiny Portilla
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049, Madrid, Spain
| | - Marta Gómez de Cedrón
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, Crta. De Canto Blanco 8, 28049, Madrid, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, Crta. De Canto Blanco 8, 28049, Madrid, Spain
| | - Domingo F Barber
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049, Madrid, Spain.
| |
Collapse
|
11
|
Shete MB, Deshpande AS, Shende P. Enhancement of in-vitro anti-oral cancer activities of silymarin using dispersion of nanostructured lipid carrier in mucoadhesive in-situ gel. Int J Pharm 2023; 636:122860. [PMID: 36933584 DOI: 10.1016/j.ijpharm.2023.122860] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 02/17/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Silymarin (SME) shows multiple therapeutic actions against several cancers, however, low aqueous solubility and poor bioavailability issues restrict its clinical use. In this study, SME was loaded in nanostructured lipid carriers (NLCs) and further incorporated in mucoadhesive in-situ gel (SME-NLCs-Plx/CP-ISG) for localized treatment of oral cancer. Using a 33 Box-Behnken design (BBD), an optimized SME-NLC formula was developed with the ratios of solid lipids, surfactant concentration, and sonication time as independent variables, while particle size (PS), polydispersity index (PDI), and % encapsulation efficiency (EE) as dependent variables, resulting in 315.5 ± 0.1 nm PS, 0.341 ± 0.01 PDI, and 71.05 ± 0.05 % EE. Structural studies confirmed the formation of SME-NLCs. SME-NLCs incorporated in-situ gel demonstrated a sustained release for SME, indicating enhanced retention on the buccal mucosal membrane. The in-situ gel containing SME-NLCs showed a marked decrease in IC50 value (24.90 ± 0.45 µM) than SME-NLCs (28.40 ± 0.89 µM) and plain SME (36.60 ± 0.26 µM). The studies demonstrated that Reactive oxygen species (ROS) generation potential and SME-NLCs-Plx/CP-ISG induced apoptosis at Sub-G0 phase owing to higher penetration of SME-NLCs led to higher inhibition against human KB oral cancer cells. Therefore, SME-NLCs-Plx/CP-ISG can be the alternative to chemotherapy and surgery with site-specific delivery of SME to oral cancer patients.
Collapse
Affiliation(s)
- Meghanath B Shete
- School of Pharmacy & Technology Management, SVKM's, NMIMS, Shirpur, Maharashtra, India; Department of Pharmaceutical Quality Assurance, R C Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra, India
| | - Ashwini S Deshpande
- School of Pharmacy & Technology Management, SVKM's, NMIMS, Polepally SEZ, TSIIC Jadcherla, Hyderabad, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's, NMIMS, Vile-Parle (W), Mumbai, Maharashtra, India.
| |
Collapse
|
12
|
Jiang D, Pan L, Yang X, Ji Z, Zheng C, Meng Z, Liang B, Zhang W, Chen J, Shi C. Photo-controllable burst generation of peroxynitrite based on synergistic interactions of polymeric nitric oxide donors and IR780 for enhancing broad-spectrum antibacterial therapy. Acta Biomater 2023; 159:259-274. [PMID: 36690050 DOI: 10.1016/j.actbio.2023.01.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/08/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
The newly attractive peroxynitrite (ONOO-) therapy can prominently enhance antibacterial therapeutic efficacy. However, it is a great challenge but urgently needed to generate ONOO- with adjustable release rate and dosage in order to satisfy personalized treatments for different disease types and severities. Herein, PSNO@IR780 nanoparticles are fabricated via co-assembly of an amphiphilic PEG-b-PAASNO block copolymer grafted with abundant nitric oxide (NO) donor units and IR780 as a photothermal and photodynamic agent. Photo-controllable burst generation of ONOO- from PSNO@IR780 nanoparticles could be realized based on synergistic reactions of rapid NO release induced by increased local temperature and efficiently produced superoxide anion radical (O2•-) from IR780. The maximum ONOO- release dosage is up to 6.73 ± 0.07 µM and release rate is up to 98.1 ± 1.38 nM/s. Furthermore, the ONOO- release behavior can be precisely manipulated by varying sample concentrations, irradiated durations, output power densities, and laser switches, respectively. Ultra-efficiently generated ONOO- from biocompatible PSNO@IR780 nanoparticles significantly elevated broad spectrum antibacterial efficiency through damaging bacterial membranes. Thus, PSNO@IR780 nanoparticles may present a new insight into preparation of burst and controllable generating ONOO- materials, and provide new opportunities for antibacterial therapy. STATEMENT OF SIGNIFICANCE: 1. Polymeric NO donor (PEG-b-PAASNO) grafted with abundant NO donor units was synthesized. 2. PSNO@IR780 nanoparticles were prepared by co-assembly of IR780 and amphiphilic PEG-b-PAASNOpolymer. 3. The maximum ONOO- release dosage from PSNO@IR780 nanoparticles was 6.73 ± 0.08 µM. 4. The fastest ONOO- release rate from PSNO@IR780 nanoparticles was 98.1 ± 1.4 nM/s. 5. Ultra-efficiently generated ONOO- significantly elevated antibacterial efficiency via damaging bac-terial membranes.
Collapse
Affiliation(s)
- Dawei Jiang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Joint Center of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Luqi Pan
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Joint Center of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Xiao Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Joint Center of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Zhixiao Ji
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Joint Center of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Cheng Zheng
- Department of Critical Care Medicine, Taizhou Municipal Hospital, Taizhou, Zhejiang 318000, China
| | - Zhizhen Meng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Joint Center of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Department of Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Bin Liang
- Department of Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200030, China.
| | - Jinfei Chen
- Department of Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Changcan Shi
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Joint Center of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Department of Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
13
|
An Update on Phytochemicals in Redox Homeostasis: “Virtuous or Evil” in Cancer Chemoprevention? CHEMISTRY 2023. [DOI: 10.3390/chemistry5010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Redox homeostasis, a dynamic process ensuring a balance between cellular oxidizing and reducing reactions, is crucial for maintaining healthy cellular physiology and regulating many biological processes, requiring continuous monitoring and fine-tuning. Reactive species play a critical role in intra/intercellular signaling, and each cell has a specific system guarding cellular redox homeostasis. ROS signaling and oxidative stress are involved in cancer initiation and progression. However, the generation of reactive species beyond the threshold level inside the tumor microenvironment is considered one of the therapeutic approaches. Various studies have shown that some phytochemicals can target the redox homeostasis of the tumor microenvironment. Recent advances have focused on developing and introducing phytochemical interventions as favorable therapeutic options against cancer. However, studies have also suggested the “virtuous” and “evil” impacts of phytochemicals. Some phytochemicals enhance therapeutic efficacy by promoting intracellular oxidant accumulation. However, under certain conditions, some phytochemicals may harm the cellular microenvironment to promote cancer and tend to target different pathways for cancer initiation and development instead of targeting redox homeostasis. In this context, this review is focused on providing an overall understanding of redox homeostasis and intends to highlight the potential positive and negative impacts of phytochemicals in redox homeostasis and disease development. We also discuss the recent nanotechnology-based advancements in combating cancer development.
Collapse
|
14
|
Afzal O, Altamimi ASA, Nadeem MS, Alzarea SI, Almalki WH, Tariq A, Mubeen B, Murtaza BN, Iftikhar S, Riaz N, Kazmi I. Nanoparticles in Drug Delivery: From History to Therapeutic Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:4494. [PMID: 36558344 PMCID: PMC9781272 DOI: 10.3390/nano12244494] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/04/2022] [Accepted: 12/14/2022] [Indexed: 05/25/2023]
Abstract
Current research into the role of engineered nanoparticles in drug delivery systems (DDSs) for medical purposes has developed numerous fascinating nanocarriers. This paper reviews the various conventionally used and current used carriage system to deliver drugs. Due to numerous drawbacks of conventional DDSs, nanocarriers have gained immense interest. Nanocarriers like polymeric nanoparticles, mesoporous nanoparticles, nanomaterials, carbon nanotubes, dendrimers, liposomes, metallic nanoparticles, nanomedicine, and engineered nanomaterials are used as carriage systems for targeted delivery at specific sites of affected areas in the body. Nanomedicine has rapidly grown to treat certain diseases like brain cancer, lung cancer, breast cancer, cardiovascular diseases, and many others. These nanomedicines can improve drug bioavailability and drug absorption time, reduce release time, eliminate drug aggregation, and enhance drug solubility in the blood. Nanomedicine has introduced a new era for drug carriage by refining the therapeutic directories of the energetic pharmaceutical elements engineered within nanoparticles. In this context, the vital information on engineered nanoparticles was reviewed and conferred towards the role in drug carriage systems to treat many ailments. All these nanocarriers were tested in vitro and in vivo. In the coming years, nanomedicines can improve human health more effectively by adding more advanced techniques into the drug delivery system.
Collapse
Affiliation(s)
- Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Aqsa Tariq
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore 54000, Pakistan
| | - Bismillah Mubeen
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore 54000, Pakistan
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology (AUST), Abbottabad 22310, Pakistan
| | - Saima Iftikhar
- School of Biological Sciences, University of Punjab, Lahore 54000, Pakistan
| | - Naeem Riaz
- Department of Pharmacy, COMSATS University, Abbottabad 22020, Pakistan
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
15
|
Yu S, Chen L, Xu H, Long S, Jiang J, Wei W, Niu X, Li X. Application of nanomaterials in diagnosis and treatment of glioblastoma. Front Chem 2022; 10:1063152. [PMID: 36569956 PMCID: PMC9780288 DOI: 10.3389/fchem.2022.1063152] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Diagnosing and treating glioblastoma patients is currently hindered by several obstacles, such as tumor heterogeneity, the blood-brain barrier, tumor complexity, drug efflux pumps, and tumor immune escape mechanisms. Combining multiple methods can increase benefits against these challenges. For example, nanomaterials can improve the curative effect of glioblastoma treatments, and the synergistic combination of different drugs can markedly reduce their side effects. In this review, we discuss the progression and main issues regarding glioblastoma diagnosis and treatment, the classification of nanomaterials, and the delivery mechanisms of nanomedicines. We also examine tumor targeting and promising nano-diagnosis or treatment principles based on nanomedicine. We also summarize the progress made on the advanced application of combined nanomaterial-based diagnosis and treatment tools and discuss their clinical prospects. This review aims to provide a better understanding of nano-drug combinations, nano-diagnosis, and treatment options for glioblastoma, as well as insights for developing new tools.
Collapse
Affiliation(s)
- Shuangqi Yu
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Lijie Chen
- China Medical University, Shenyang, Liaoning, China
| | - Hongyu Xu
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Shengrong Long
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Jiazhi Jiang
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Xing Niu
- China Medical University, Shenyang, Liaoning, China
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
16
|
Li Y, Yoon B, Dey A, Nguyen VQ, Park JH. Recent progress in nitric oxide-generating nanomedicine for cancer therapy. J Control Release 2022; 352:179-198. [PMID: 36228954 DOI: 10.1016/j.jconrel.2022.10.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/26/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Nitric oxide (NO) is an endogenous, multipotent biological signaling molecule that participates in several physiological processes. Recently, exogenous supplementation of tumor tissues with NO has emerged as a potential anticancer therapy. In particular, it induces synergistic effects with other conventional therapies (such as chemo-, radio-, and photodynamic therapies) by regulating the activity of P-glycoprotein, acting as a vascular relaxant to relieve tumor hypoxia, and participating in the metabolism of reactive oxygen species. However, NO is highly reactive, and its half-life is relatively short after generation. Meanwhile, NO-induced anticancer activity is dose-dependent. Therefore, the targeted delivery of NO to the tumor is required for better therapeutic effects. In the past decade, NO-generating nanomedicines (NONs), which enable sustained and specific NO release in tumor tissues, have been developed for enhanced cancer therapy. This review describes the recent efforts and preclinical achievements in the development of NON-based cancer therapies. The chemical structures employed in the fabrication of NONs are summarized, and the strategies involved in NON-based cancer therapies are elaborated.
Collapse
Affiliation(s)
- Yuce Li
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Been Yoon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Anup Dey
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Van Quy Nguyen
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea.; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea.
| |
Collapse
|
17
|
Egorova EA, Nikitin MP. Delivery of Theranostic Nanoparticles to Various Cancers by Means of Integrin-Binding Peptides. Int J Mol Sci 2022; 23:ijms232213735. [PMID: 36430214 PMCID: PMC9696485 DOI: 10.3390/ijms232213735] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
Active targeting of tumors is believed to be the key to efficient cancer therapy and accurate, early-stage diagnostics. Active targeting implies minimized off-targeting and associated cytotoxicity towards healthy tissue. One way to acquire active targeting is to employ conjugates of therapeutic agents with ligands known to bind receptors overexpressed onto cancer cells. The integrin receptor family has been studied as a target for cancer treatment for almost fifty years. However, systematic knowledge on their effects on cancer cells, is yet lacking, especially when utilized as an active targeting ligand for particulate formulations. Decoration with various integrin-targeting peptides has been reported to increase nanoparticle accumulation in tumors ≥ 3-fold when compared to passively targeted delivery. In recent years, many newly discovered or rationally designed integrin-binding peptides with excellent specificity towards a single integrin receptor have emerged. Here, we show a comprehensive analysis of previously unreviewed integrin-binding peptides, provide diverse modification routes for nanoparticle conjugation, and showcase the most notable examples of their use for tumor and metastases visualization and eradication to date, as well as possibilities for combined cancer therapies for a synergetic effect. This review aims to highlight the latest advancements in integrin-binding peptide development and is directed to aid transition to the development of novel nanoparticle-based theranostic agents for cancer therapy.
Collapse
Affiliation(s)
- Elena A. Egorova
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sirius, Russia
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 1 Meditsinskaya Str., 603081 Nizhny Novgorod, Russia
| | - Maxim P. Nikitin
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sirius, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., 141701 Dolgoprudny, Russia
- Correspondence:
| |
Collapse
|
18
|
Acar M, Solak K, Yildiz S, Unver Y, Mavi A. Comparative heating efficiency and cytotoxicity of magnetic silica nanoparticles for magnetic hyperthermia treatment on human breast cancer cells. 3 Biotech 2022; 12:313. [PMID: 36276464 PMCID: PMC9547765 DOI: 10.1007/s13205-022-03377-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 09/17/2022] [Indexed: 11/01/2022] Open
Abstract
Magnetic hyperthermia (MHT) is a promising treatment for a variety of cancers due to its ability to increase the sensitivity of cells to other treatments, such as chemotherapy. Superparamagnetic nanoparticles (MNPs) were used for MHT treatment due to their heat generation ability under an AC magnetic field (AMF). In this study, iron oxide and zinc-doped iron oxide MNPs were produced and modified with silica to obtain eleven different types (MSNP-I to -XI) of magnetic silica nanoparticles (MSNPs). The MSNPs which show the highest heating capacity were selected to investigate their MHT ability on non-tumourigenic MCF-10A and tumourigenic MCF-7 cell lines. The cytotoxicity results indicated that the size, the content of the magnetic core and silica coating thickness were important in the heating capacity of MSNPs under AMF. After MHT treatment, selected MSNPs showed limited cytotoxicity on MCF-10A, but significant cell death on MCF-7. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03377-y.
Collapse
Affiliation(s)
- Melek Acar
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Kubra Solak
- Department of Nanoscience and Nanoengineering, Graduate School of Natural and Applied Science, Atatürk University, Erzurum, Turkey
| | - Seyda Yildiz
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Yagmur Unver
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Ahmet Mavi
- Department of Nanoscience and Nanoengineering, Graduate School of Natural and Applied Science, Atatürk University, Erzurum, Turkey
- Department of Chemistry Education, Kazim Karabekir Faculty of Education, Atatürk University, Erzurum, Turkey
| |
Collapse
|
19
|
Laser empowered ‘chemo-free’ phytotherapy: Newer approach in anticancer therapeutics delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Çelik SY, Solak K, Mavi A. Sulfanilamide Modified Magnetic Nanoparticles for Purification of Carbonic Anhydrase from Bovine Blood. Appl Biochem Biotechnol 2022; 195:3900-3913. [DOI: 10.1007/s12010-022-03983-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
|
21
|
Li X, Wang Y, Li M, Wang H, Dong X. Metal Complexes or Chelators with ROS Regulation Capacity: Promising Candidates for Cancer Treatment. Molecules 2021; 27:148. [PMID: 35011380 PMCID: PMC8746559 DOI: 10.3390/molecules27010148] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 12/25/2021] [Accepted: 12/26/2021] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) are rapidly eliminated and reproduced in organisms, and they always play important roles in various biological functions and abnormal pathological processes. Evaluated ROS have frequently been observed in various cancers to activate multiple pro-tumorigenic signaling pathways and induce the survival and proliferation of cancer cells. Hydrogen peroxide (H2O2) and superoxide anion (O2•-) are the most important redox signaling agents in cancer cells, the homeostasis of which is maintained by dozens of growth factors, cytokines, and antioxidant enzymes. Therefore, antioxidant enzymes tend to have higher activity levels to maintain the homeostasis of ROS in cancer cells. Effective intervention in the ROS homeostasis of cancer cells by chelating agents or metal complexes has already developed into an important anti-cancer strategy. We can inhibit the activity of antioxidant enzymes using chelators or metal complexes; on the other hand, we can also use metal complexes to directly regulate the level of ROS in cancer cells via mitochondria. In this review, metal complexes or chelators with ROS regulation capacity and with anti-cancer applications are collectively and comprehensively analyzed, which is beneficial for the development of the next generation of inorganic anti-cancer drugs based on ROS regulation. We expect that this review will provide a new perspective to develop novel inorganic reagents for killing cancer cells and, further, as candidates or clinical drugs.
Collapse
Affiliation(s)
- Xiang Li
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yuhui Wang
- School of Chemistry, Central China Normal University, Wuhan 430079, China; (Y.W.); (M.L.)
| | - Man Li
- School of Chemistry, Central China Normal University, Wuhan 430079, China; (Y.W.); (M.L.)
| | - Huipeng Wang
- National Local Joint Engineering Laboratory for Advanced Textile Processing and Clean Production, Wuhan Textile University, Wuhan 430073, China;
| | - Xiongwei Dong
- National Local Joint Engineering Laboratory for Advanced Textile Processing and Clean Production, Wuhan Textile University, Wuhan 430073, China;
| |
Collapse
|
22
|
Wang Y, Wang J, Gou K, Kang W, Guo X, Zhu K, Li S, Li H. pH/H 2O 2 Dual-Responsive Chiral Mesoporous Silica Nanorods Coated with a Biocompatible Active Targeting Ligand for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:35397-35409. [PMID: 34313104 DOI: 10.1021/acsami.1c08532] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nano-drug delivery systems (nano-DDSs) with an existing specific interaction to tumor cells and intelligent stimulus-triggered drug delivery performance in a tumor microenvironment (TME) remain hotspots for effective cancer therapy. Herein, multifunctional pH/H2O2 dual-responsive chiral mesoporous silica nanorods (HA-CD/DOX-PCMSRs) were creatively constructed by first grafting phenylboronic acid pinacol ester (PBAP) onto the amino-functioned nanorods, then incorporating doxorubicin (DOX) into the mesoporous structure, and finally coating with the cyclodextrin-modified hyaluronic acid conjugate (HA-CD) through a weak host-guest interaction. Under a physiological environment, the gatekeeper CD could avoid the premature leakage of DOX and minimize the side effects to normal cells. After the uptake by the tumor cells, the H2O2-sensitive moieties of PBAP were exposed and a small amount of DOX was leaked along with the shift of the supramolecular switch HA-CD under the acidic condition. Notably, the self-supplying H2O2 mediated by the released DOX in turn accelerated the PBAP disintegration, further promoted the rapid release of DOX, and increased the DOX accumulation in tumor regions. Innovatively, this nano-DDS could simultaneously achieve the tumor-targeting ability via CD44 receptor-mediated endocytosis and pH/H2O2 dual responsiveness activated by the TME and hence exhibited superior antitumor efficacy. Furthermore, HA acting as the hydrophilic shell could improve the biocompatibility of this nano-DDS.
Collapse
Affiliation(s)
- Yumei Wang
- Shenyang Pharmaceutical University, Wenhua RD103, 110016 Shenyang, Liaoning Province, China
| | - Jing Wang
- Shenyang Pharmaceutical University, Wenhua RD103, 110016 Shenyang, Liaoning Province, China
| | - Kaijun Gou
- Shenyang Pharmaceutical University, Wenhua RD103, 110016 Shenyang, Liaoning Province, China
| | - Wei Kang
- Shenyang Pharmaceutical University, Wenhua RD103, 110016 Shenyang, Liaoning Province, China
| | - Xianmou Guo
- Shenyang Pharmaceutical University, Wenhua RD103, 110016 Shenyang, Liaoning Province, China
| | - Keyu Zhu
- Shenyang Pharmaceutical University, Wenhua RD103, 110016 Shenyang, Liaoning Province, China
| | - Sanming Li
- Shenyang Pharmaceutical University, Wenhua RD103, 110016 Shenyang, Liaoning Province, China
| | - Heran Li
- China Medical University, Puhe RD77, Shenyang North New Area, 110122 Shenyang, Liaoning Province, China
| |
Collapse
|
23
|
Reyes-Ortega F, Delgado ÁV, Iglesias GR. Modulation of the Magnetic Hyperthermia Response Using Different Superparamagnetic Iron Oxide Nanoparticle Morphologies. NANOMATERIALS 2021; 11:nano11030627. [PMID: 33802441 PMCID: PMC8001085 DOI: 10.3390/nano11030627] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
The use of magnetic nanoparticles in hyperthermia, that is, heating induced by alternating magnetic fields, is gaining interest as a non-invasive, free of side effects technique that can be considered as a co-adjuvant of other cancer treatments. Having sufficient control on the field characteristics, within admissible limits, the focus is presently on the magnetic material. In the present contribution, no attempt has been made of using other composition than superparamagnetic iron oxide nanoparticles (SPION), or of applying surface functionalization, which opens a wider range of choices. We have used a hydrothermal synthesis route that allows preparing SPION nanoparticles in the 40 nm size range, with spherical, cuboidal or rod-like shapes, by minor changes in the synthesis steps. The three kinds of particles (an attempt to produce star-shaped colloids yielded hematite) were demonstrated to have the magnetite (or maghemite) crystallinity. Magnetization cycles showed virtually no hysteresis and demonstrated the superparamagnetic nature of the particles, cuboidal ones displaying saturation magnetization comparable to bulk magnetite, followed by rods and spheres. The three types were used as hyperthermia agents using magnetic fields of 20 kA/m amplitude and frequency in the range 136–205 kHz. All samples demonstrated to be able to raise the solution temperature from room values to 45 °C in a mere 60 s. Not all of them performed the same way, though. Cuboidal magnetic nanoparticles (MNPs) displayed the maximum heating power (SAR or specific absorption rate), ranging in fact among the highest reported with these geometries and raw magnetite composition.
Collapse
Affiliation(s)
- Felisa Reyes-Ortega
- Department of Applied Physics, University of Granada, 18071 Granada, Spain;
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, University of Córdoba, 14004 Córdoba, Spain
- Correspondence: or (F.R.-O.); (G.R.I.); Tel.: +34-957-736-483 (F.R.-O.); +34-958-242-734 (G.R.I.)
| | - Ángel V. Delgado
- Department of Applied Physics, University of Granada, 18071 Granada, Spain;
| | - Guillermo R. Iglesias
- Department of Applied Physics, University of Granada, 18071 Granada, Spain;
- Correspondence: or (F.R.-O.); (G.R.I.); Tel.: +34-957-736-483 (F.R.-O.); +34-958-242-734 (G.R.I.)
| |
Collapse
|
24
|
Solak K, Mavi A, Yılmaz B. Disulfiram-loaded functionalized magnetic nanoparticles combined with copper and sodium nitroprusside in breast cancer cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 119:111452. [DOI: 10.1016/j.msec.2020.111452] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/18/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
|
25
|
Anik MI, Hossain MK, Hossain I, Mahfuz AMUB, Rahman MT, Ahmed I. Recent progress of magnetic nanoparticles in biomedical applications: A review. NANO SELECT 2021. [DOI: 10.1002/nano.202000162] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Muzahidul I. Anik
- Chemical Engineering University of Rhode Island Kingston Rhode Island 02881 USA
| | - M. Khalid Hossain
- Interdisciplinary Graduate School of Engineering Science Kyushu University Fukuoka 816–8580 Japan
- Atomic Energy Research Establishment Bangladesh Atomic Energy Commission Dhaka 1349 Bangladesh
| | - Imran Hossain
- Institute for Micromanufacturing Louisiana Tech University Ruston Louisiana 71270 USA
| | - A. M. U. B. Mahfuz
- Biotechnology and Genetic Engineering University of Development Alternative Dhaka 1209 Bangladesh
| | - M. Tayebur Rahman
- Materials Science and Engineering University of Rajshahi Rajshahi 6205 Bangladesh
| | - Isteaque Ahmed
- Chemical Engineering University of Cincinnati Cincinnati Ohio 45221 USA
| |
Collapse
|
26
|
Tang SY, Wei H, Yu CY. Peptide-functionalized delivery vehicles for enhanced cancer therapy. Int J Pharm 2021; 593:120141. [DOI: 10.1016/j.ijpharm.2020.120141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/17/2020] [Accepted: 11/28/2020] [Indexed: 02/08/2023]
|
27
|
Arvapalli DM, Sheardy AT, Allado K, Chevva H, Yin Z, Wei J. Design of Curcumin Loaded Carbon Nanodots Delivery System: Enhanced Bioavailability, Release Kinetics, and Anticancer Activity. ACS APPLIED BIO MATERIALS 2020; 3:8776-8785. [DOI: 10.1021/acsabm.0c01144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Durga M. Arvapalli
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, North Carolina 27401, United States
| | - Alex T. Sheardy
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, North Carolina 27401, United States
| | - Kokougan Allado
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, North Carolina 27401, United States
| | - Harish Chevva
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, North Carolina 27401, United States
| | - Ziyu Yin
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, North Carolina 27401, United States
| | - Jianjun Wei
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, North Carolina 27401, United States
| |
Collapse
|
28
|
Banstola A, Duwa R, Emami F, Jeong JH, Yook S. Enhanced Caspase-Mediated Abrogation of Autophagy by Temozolomide-Loaded and Panitumumab-Conjugated Poly(lactic-co-glycolic acid) Nanoparticles in Epidermal Growth Factor Receptor Overexpressing Glioblastoma Cells. Mol Pharm 2020; 17:4386-4400. [DOI: 10.1021/acs.molpharmaceut.0c00856] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu 42601, South Korea
| | - Ramesh Duwa
- College of Pharmacy, Keimyung University, Daegu 42601, South Korea
| | | | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Gyeongbuk, South Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu 42601, South Korea
| |
Collapse
|
29
|
Feng Z, Guo J, Liu X, Song H, Zhang C, Huang P, Dong A, Kong D, Wang W. Cascade of reactive oxygen species generation by polyprodrug for combinational photodynamic therapy. Biomaterials 2020; 255:120210. [PMID: 32592871 DOI: 10.1016/j.biomaterials.2020.120210] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 05/29/2020] [Accepted: 06/13/2020] [Indexed: 12/13/2022]
Abstract
The redox status of cancer cells is well regulated by the balance between the reactive oxygen species (ROS) generation and elimination. Thus, the overall elevation of ROS level above the cellular tolerability threshold would lead to apoptotic or necrotic cell death. Herein, cinnamaldehyde (CA), a kind of oxidative stress amplified agent, was combined with photosensitizer pheophorbide A (PA) to promote the generation of ROS though synergistically endogenous and exogenous pathways. Firstly, acid-responsive polygalactose-co-polycinnamaldehyde polyprodrug (termed as PGCA) was synthesized, which could self-assemble into stable nanoparticles for the delivery of PA (termed as PGCA@PA NPs). The abundant expression of galactose receptor on tumor cells facilitated the positive targeting and cellular uptake efficiency of PGCA@PA NPs, after which PA could be synchronously released in company with the intracellular disassembly of PGCA NPs, due to the detaching of CA moieties under acidic microenvironment in endo/lysosomal compartment. Significantly increased ROS level was induced by the combined action of CA and PA with light irradiation, resulting in dramatically enhanced apoptosis of cancer cells. Importantly, intravenous injection of PGCA@PA NPs potently inhibited the tumor growth in hepatocellular carcinoma with negligible adverse effects. Moreover, combined with anti-programmed cell death protein 1 (anti-PD-1) therapy, PGCA@PA NPs treatment elicited anti-melanoma T-cell immune response and significantly promoted T cells infiltration in tumors. Hence, this novel polyprodrug nano delivery system was able to target and modulate the unique redox regulatory mechanisms of cancer cells through endogenous and exogenous pathways, providing a feasible approach to achieve synergetic therapeutic activity and selectivity.
Collapse
Affiliation(s)
- Zujian Feng
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, China
| | - Jinxuan Guo
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, China
| | - Xiang Liu
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, China
| | - Huijuan Song
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China.
| | - Anjie Dong
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, China.
| | - Deling Kong
- The Key Laboratory of Bioactive Materials, Ministry of Education; College of Life Science, Nankai University, Tianjin, 300071, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China.
| |
Collapse
|
30
|
Wang S, Yang L, Cho HY, Dean Chueng ST, Zhang H, Zhang Q, Lee KB. Programmed degradation of a hierarchical nanoparticle with redox and light responsivity for self-activated photo-chemical enhanced chemodynamic therapy. Biomaterials 2019; 224:119498. [PMID: 31557590 DOI: 10.1016/j.biomaterials.2019.119498] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/22/2019] [Accepted: 09/12/2019] [Indexed: 12/24/2022]
Abstract
Chemodynamic therapy (CDT) has recently emerged as a promising treatment for cancer due to the high specificity of CDT towards tumor microenvironment (TME). However, the low efficiency of reactive oxygen species (ROS) generation and the robust ROS defensive mechanisms in cancer cells remain critical hurdles for current CDT. Addressing both challenges in a single platform, we developed a novel redox and light-responsive (RLR) nanoparticle with a core-shell structure. Remarkably, our hierarchical RLR nanoparticle is composed of an ultrasmall Fe3O4 nanoparticle engineered framework of hollow carbon matrix core and a nanoflower-like MnO2 shell. Under the abundant overexpressed glutathione (GSH) and acidic nature in TME, the RLR nanoparticle was programmed to degrade and self-activate CDT-induced cancer-killing by accelerating ROS generation via overcoming the ROS defensive mechanisms based on the depletion of intracellular GSH, the sequential production of theranostic ion species (e.g., Mn2+ and Fe2+), a spatiotemporal controllable photothermal hyperthermia and a redox triggered chemotherapeutic drug release. Additionally, the carbon framework of RLR nanoparticle could collapse by leaching of iron ions. An excellent selective and near-complete tumor suppression based on the RLR nanoparticles through a strong synergy between CDT, PTT and anti-cancer drugs was demonstrated via in vitro and in vivo anti-tumoral assays.
Collapse
Affiliation(s)
- Shenqiang Wang
- MOE Key Laboratory of Material Physics and Chemistry Under Extraordinary Conditions, School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710129, China; Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Hyeon-Yeol Cho
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Sy-Tsong Dean Chueng
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Hepeng Zhang
- MOE Key Laboratory of Material Physics and Chemistry Under Extraordinary Conditions, School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Qiuyu Zhang
- MOE Key Laboratory of Material Physics and Chemistry Under Extraordinary Conditions, School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710129, China.
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA; Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, South Korea.
| |
Collapse
|