1
|
Wang F, Liu Z, Liu Y, Zhang J, Xu W, Liu B, Sun Z, Chu H. A Spatiotemporally Controlled Gene-Regulation Strategy for Combined Tumor Therapy Based on Upconversion Hybrid Nanosystem. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405640. [PMID: 39207039 PMCID: PMC11515897 DOI: 10.1002/advs.202405640] [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: 05/23/2024] [Revised: 08/09/2024] [Indexed: 09/04/2024]
Abstract
The lack of precise spatiotemporal gene modulation and therapy impedes progress in medical applications. Herein, a 980 nm near-infrared (NIR) light-controlled nanoplatform, namely URMT, is developed, which can allow spatiotemporally controlled photodynamic therapy and trigger the enzyme-activated gene expression regulation in tumors. URMT is constructed by engineering an enzyme-activatable antisense oligonucleotide, which combined with an upconversion nanoparticle (UCNP)-based photodynamic nanosystem, followed by the surface functionalization of triphenylphosphine (TPP), a mitochondria-targeting ligand. URMT allows for the 980 nm NIR light-activated generation of reactive oxygen species, which can induce the translocation of a DNA repair enzyme (namely apurinic/apyrimidinic endonuclease 1, APE1) from the nucleus to mitochondria. APE1 can recognize the basic apurinic/apyrimidinic (AP) sites in DNA double-strands and perform cleavage, thereby releasing the functional single-strands for gene regulation. Overall, an augmented antitumor effect is observed due to NIR light-controlled mitochondrial damage and enzyme-activated gene regulation. Altogether, the approach reported in this study offers high spatiotemporal precision and shows the potential to achieve precise and specific gene regulation for targeted tumor treatment.
Collapse
Affiliation(s)
- Fang Wang
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Zechao Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Yuechen Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Jiayi Zhang
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Weizhe Xu
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Bei Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Zhaogang Sun
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Hongqian Chu
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| |
Collapse
|
2
|
Wang W, Li C, Luo S, Wu ZS. Spherical Nucleic Acid-Mediated Spatial Matching-Guided Nonenzymatic DNA Circuits for the Prediction and Prevention of Malignant Tumor Invasion. Anal Chem 2024; 96:7091-7100. [PMID: 38663871 DOI: 10.1021/acs.analchem.4c00476] [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: 05/08/2024]
Abstract
Detection of intracellular miRNAs, especially sensitive imaging of in vivo miRNAs, is vital to the precise prediction and timely prevention of tumorgenesis but remains a technical challenge in terms of nuclease resistance and signal amplification. Here, we demonstrate a gold nanoparticle-based spherical nucleic acid-mediated spatial matching-guided nonenzymatic DNA circuit (SSDC) for efficient screening of intracellular miRNAs and, in turn, finding cancerous tissues in living organisms before the appearance of clinical symptoms. Due to the substantially enhanced nuclease resistance, the false positive signal is avoided even in a complex biological medium. Target miRNA can straighten out the hairpin DNA probe to be linear, allowing the probe to penetrate into the internal region of a core/shell DNA-functionalized signal nanoampfilier and initiate a strand displacement reaction, generating an amplified fluorescence signal. The detection limit is as low as 17 pM, and miRNA imaging is in good accordance with the gold standard polymerase chain reaction method. The ability to image intracellular miRNAs is substantially superior to that of conventional fluorescence in situ hybridization techniques, making in vivo SSDC-based imaging competent for the precise prediction of tumorigenesis. By intratumoral chemotherapy guided by SSDC-based imaging, tumorigenesis and progression are efficiently controlled before the onset of clinical symptoms.
Collapse
Affiliation(s)
- Weijun Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
- 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
- College of Chemistry and Food Science, Nanchang Normal University, Nanchang 330032, China
| | - Congcong Li
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
- 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
| | - Shasha Luo
- 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
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
- 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
|
3
|
Gu L, Duan Z, Li X, Li X, Li Y, Li X, Xu G, Gao P, Zhang H, Gu Z, Chen J, Gong Q, Luo K. Enzyme-triggered deep tumor penetration of a dual-drug nanomedicine enables an enhanced cancer combination therapy. Bioact Mater 2023; 26:102-115. [PMID: 36875053 PMCID: PMC9974368 DOI: 10.1016/j.bioactmat.2023.02.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 02/27/2023] Open
Abstract
Cancer cells could be eradicated by promoting generation of excessive intracellular reactive oxygen species (ROS) via emerging nanomedicines. However, tumor heterogeneity and poor penetration of nanomedicines often lead to diverse levels of ROS production in the tumor site, and ROS at a low level promote tumor cell growth, thus diminishing the therapeutic effect of these nanomedicines. Herein, we construct an amphiphilic and block polymer-dendron conjugate-derived nanomedicine (Lap@pOEGMA-b-p(GFLG-Dendron-Ppa), GFLG-DP/Lap NPs) that incorporates a photosensitizer, Pyropheophorbide a (Ppa), for ROS therapy and Lapatinib (Lap) for molecular targeted therapy. Lap, an epidermal growth factor receptor (EGFR) inhibitor that plays a role in inhibiting cell growth and proliferation, is hypothesized to synergize with ROS therapy for effectively killing cancer cells. Our results suggest that the enzyme-sensitive polymeric conjugate, pOEGMA-b-p(GFLG-Dendron-Ppa) (GFLG-DP), releases in response to cathepsin B (CTSB) after entering the tumor tissue. Dendritic-Ppa has a strong adsorption capacity to tumor cell membranes, which promotes efficient penetration and long-term retention. Lap can also be efficiently delivered to internal tumor cells to play its role due to the increased vesicle activity. Laser irradiation of Ppa-containing tumor cells results in production of intracellular ROS that is sufficient for inducing cell apoptosis. Meanwhile, Lap efficiently inhibits proliferation of remaining viable cells even in deep tumor regions, thus generating a significant synergistic anti-tumor therapeutic effect. This novel strategy can be extended to the development of efficient membrane lipid-based therapies to effectively combat tumors.
Collapse
Affiliation(s)
- Lei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xue Li
- Liver Transplant Center, Organ Transplant Center, Breast Center, Laboratory of Stem Cell Biology, Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Li
- Liver Transplant Center, Organ Transplant Center, Breast Center, Laboratory of Stem Cell Biology, Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yinggang Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoling Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Xu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.,Liver Transplant Center, Organ Transplant Center, Breast Center, Laboratory of Stem Cell Biology, Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Peng Gao
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.,Liver Transplant Center, Organ Transplant Center, Breast Center, Laboratory of Stem Cell Biology, Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Jie Chen
- Liver Transplant Center, Organ Transplant Center, Breast Center, Laboratory of Stem Cell Biology, Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.,Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361000, Fujian, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
4
|
Cui X, Ruan Q, Zhuo X, Xia X, Hu J, Fu R, Li Y, Wang J, Xu H. Photothermal Nanomaterials: A Powerful Light-to-Heat Converter. Chem Rev 2023. [PMID: 37133878 DOI: 10.1021/acs.chemrev.3c00159] [Citation(s) in RCA: 184] [Impact Index Per Article: 184.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
All forms of energy follow the law of conservation of energy, by which they can be neither created nor destroyed. Light-to-heat conversion as a traditional yet constantly evolving means of converting light into thermal energy has been of enduring appeal to researchers and the public. With the continuous development of advanced nanotechnologies, a variety of photothermal nanomaterials have been endowed with excellent light harvesting and photothermal conversion capabilities for exploring fascinating and prospective applications. Herein we review the latest progresses on photothermal nanomaterials, with a focus on their underlying mechanisms as powerful light-to-heat converters. We present an extensive catalogue of nanostructured photothermal materials, including metallic/semiconductor structures, carbon materials, organic polymers, and two-dimensional materials. The proper material selection and rational structural design for improving the photothermal performance are then discussed. We also provide a representative overview of the latest techniques for probing photothermally generated heat at the nanoscale. We finally review the recent significant developments of photothermal applications and give a brief outlook on the current challenges and future directions of photothermal nanomaterials.
Collapse
Affiliation(s)
- Ximin Cui
- State Key Laboratory of Radio Frequency Heterogeneous Integration, College of Electronics and Information Engineering, Shenzhen University, Shenzhen 518060, China
| | - Qifeng Ruan
- Ministry of Industry and Information Technology Key Lab of Micro-Nano Optoelectronic Information System & Guangdong Provincial Key Laboratory of Semiconductor Optoelectronic Materials and Intelligent Photonic Systems, Harbin Institute of Technology, Shenzhen 518055, China
| | - Xiaolu Zhuo
- Guangdong Provincial Key Lab of Optoelectronic Materials and Chips, School of Science and Engineering, The Chinese University of Hong Kong (Shenzhen), Shenzhen 518172, China
| | - Xinyue Xia
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Jingtian Hu
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Runfang Fu
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Yang Li
- State Key Laboratory of Radio Frequency Heterogeneous Integration, College of Electronics and Information Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jianfang Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Hongxing Xu
- School of Physics and Technology and School of Microelectronics, Wuhan University, Wuhan 430072, Hubei, China
- Henan Academy of Sciences, Zhengzhou 450046, Henan, China
- Wuhan Institute of Quantum Technology, Wuhan 430205, Hubei, China
| |
Collapse
|
5
|
Zhang H, Zhu T, Li M. Quantitative Analysis of the Shape Effect of Thermoplasmonics in Gold Nanostructures. J Phys Chem Lett 2023; 14:3853-3860. [PMID: 37067229 DOI: 10.1021/acs.jpclett.3c00632] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The shape effect of thermoplasmonic properties of Au nanostructures remains largely unexplored. Herein, we report a systematic investigation on the photothermal effects of Au nanoparticles (NPs) of different shapes: nanosphere, nanocube, nanorod, nanostar, and nanobipyramid. The Joule (Jo) number (absorption cross section normalized by the particulate volume) is utilized for quantitatively assessing the photothermal properties of these different shaped Au NPs. It is shown that the Jo number of Au NPs greatly varies with the geometric shape and localized surface plasmon resonance (LSPR) wavelength. Specifically, the Jo number decreases with the red-shifting of the LSPR wavelength in these Au NPs, and the Au NPs of sharp structural features such as Au nanorod, nanostar and nanobipyramid have a much larger Jo number, indicative of their exceptional light-to-heat conversion ability. We further demonstrate the close correlation of the Jo number of Au NPs of different shapes with their optical absorption power density.
Collapse
Affiliation(s)
- Huiming Zhang
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Ting Zhu
- School of Physics and Electronic Information, Yunnan Normal University, Kunming, Yunnan 650500, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
6
|
Kim HJ, Sritandi W, Xiong Z, Ho JS. Bioelectronic devices for light-based diagnostics and therapies. BIOPHYSICS REVIEWS 2023; 4:011304. [PMID: 38505817 PMCID: PMC10903427 DOI: 10.1063/5.0102811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 12/28/2022] [Indexed: 03/21/2024]
Abstract
Light has broad applications in medicine as a tool for diagnosis and therapy. Recent advances in optical technology and bioelectronics have opened opportunities for wearable, ingestible, and implantable devices that use light to continuously monitor health and precisely treat diseases. In this review, we discuss recent progress in the development and application of light-based bioelectronic devices. We summarize the key features of the technologies underlying these devices, including light sources, light detectors, energy storage and harvesting, and wireless power and communications. We investigate the current state of bioelectronic devices for the continuous measurement of health and on-demand delivery of therapy. Finally, we highlight major challenges and opportunities associated with light-based bioelectronic devices and discuss their promise for enabling digital forms of health care.
Collapse
Affiliation(s)
| | - Weni Sritandi
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
| | | | - John S. Ho
- Author to whom correspondence should be addressed:
| |
Collapse
|
7
|
Pan Q, Lu Y, Xie L, Wu D, Liu R, Gao W, Luo K, He B, Pu Y. Recent Advances in Boosting EGFR Tyrosine Kinase Inhibitors-Based Cancer Therapy. Mol Pharm 2023; 20:829-852. [PMID: 36588471 DOI: 10.1021/acs.molpharmaceut.2c00792] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epidermal growth factor receptor (EGFR) plays a key role in signal transduction pathways associated with cell proliferation, growth, and survival. Its overexpression and aberrant activation in malignancy correlate with poor prognosis and short survival. Targeting inhibition of EGFR by small-molecular tyrosine kinase inhibitors (TKIs) is emerging as an important treatment model besides of chemotherapy, greatly reshaping the landscape of cancer therapy. However, they are still challenged by the off-targeted toxicity, relatively limited cancer types, and drug resistance after long-term therapy. In this review, we summarize the recent progress of oral, pulmonary, and injectable drug delivery systems for enhanced and targeting TKI delivery to tumors and reduced side effects. Importantly, EGFR-TKI-based combination therapies not only greatly broaden the applicable cancer types of EGFR-TKI but also significantly improve the anticancer effect. The mechanisms of TKI resistance are summarized, and current strategies to overcome TKI resistance as well as the application of TKI in reversing chemotherapy resistance are discussed. Finally, we provide a perspective on the future research of EGFR-TKI-based cancer therapy.
Collapse
Affiliation(s)
- Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Yao Lu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Li Xie
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Di Wu
- Meat Processing Key Laboratory of Sichuan Province, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Rong Liu
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and Molecular Imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
8
|
Prasad R, Conde J. Bioinspired soft nanovesicles for site-selective cancer imaging and targeted therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1792. [PMID: 35318815 DOI: 10.1002/wnan.1792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/17/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
Cell-to-cell communication within the heterogeneous solid tumor environment plays a significant role in the uncontrolled metastasis of cancer. To inhibit the metastasis and growth of cancer cells, various chemically designed and biologically derived nanosized biomaterials have been applied for targeted cancer therapeutics applications. Over the years, bioinspired soft nanovesicles have gained tremendous attention for targeted cancer therapeutics due to their easy binding with tumor microenvironment, natural targeting ability, bio-responsive nature, better biocompatibility, high cargo capacity for multiple therapeutics agents, and long circulation time. These cell-derived nanovesicles guard their loaded cargo molecules from immune clearance and make them site-selective to cancer cells due to their natural binding and delivery abilities. Furthermore, bioinspired soft nanovesicles prevent cell-to-cell communication and secretion of cancer cell markers by delivering the therapeutics agents predominantly. Cell-derived vesicles, namely, exosomes, extracellular vesicles, and so forth have been recognized as versatile carriers for therapeutic biomolecules. However, low product yield, poor reproducibility, and uncontrolled particle size distribution have remained as major challenges of these soft nanovesicles. Furthermore, the surface biomarkers and molecular contents of these vesicles change with respect to the stage of disease and types. Here in this review, we have discussed numerous examples of bioinspired soft vesicles for targeted imaging and cancer therapeutic applications with their advantages and limitations. Importance of bioengineered soft nanovesicles for localized therapies with their clinical relevance has also been addressed in this article. Overall, cell-derived nanovesicles could be considered as clinically relevant platforms for cancer therapeutics. This article is categorized under: Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Rajendra Prasad
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - João Conde
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
9
|
Navrátil O, Lizoňová D, Slonková K, Mašková L, Zadražil A, Sedmidubský D, Štěpánek F. Antibiotic depot system with radiofrequency controlled drug release. Colloids Surf B Biointerfaces 2022; 217:112618. [PMID: 35738076 DOI: 10.1016/j.colsurfb.2022.112618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/20/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Abstract
Drug depot systems have traditionally relied on the spontaneous dissolution and diffusion of drugs or prodrugs from a reservoir with constant exposure to the surrounding physiological fluids. While this is appropriate for clinical scenarios that require constant plasma concentration of the drug over time, there are also situations where multiple bursts of the drug at well-defined time intervals are preferred. This work presents a drug depot system that enables repeated on-demand release of antibiotics in precise doses, controlled by an external radiofrequency magnetic field. The remotely controlled depot system consists of composite microcapsules with a core-shell structure. The core contains micronized drug particles embedded in a low-melting hydrophobic matrix. The shell is formed by a hydrogel with immobilised magnetic nanoparticles that facilitate local heat dissipation after exposure to a radiofrequency magnetic field. When the melting point of the core material is locally exceeded, the embedded drug particles are mobilised and their surface is exposed to the external aqueous phase. It is shown that drug release can be controlled in an on/off manner by a chosen sequence and duration of radiofrequency pulses. The capacity of the depot system is shown to be significantly higher than that of purely diffusion-controlled systems containing a pre-dissolved drug. The functionality of the depot system is demonstrated in vitro for the specific case of norfloxacin acting on E. coli.
Collapse
Affiliation(s)
- Ondřej Navrátil
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Denisa Lizoňová
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Karolína Slonková
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Lucie Mašková
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Aleš Zadražil
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - David Sedmidubský
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - František Štěpánek
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| |
Collapse
|
10
|
Hafiz S, Xavierselvan M, Gokalp S, Labadini D, Barros S, Duong J, Foster M, Mallidi S. Eutectic Gallium-Indium Nanoparticles for Photodynamic Therapy of Pancreatic Cancer. ACS APPLIED NANO MATERIALS 2022; 5:6125-6139. [PMID: 35655927 PMCID: PMC9150699 DOI: 10.1021/acsanm.1c04353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/12/2022] [Indexed: 05/04/2023]
Abstract
Developing a cancer theranostic nanoplatform with diagnosis and treatment capabilities to effectively treat tumors and reduce side effects is of great significance. Herein, we present a drug delivery strategy for photosensitizers based on a new liquid metal nanoplatform that leverages the tumor microenvironment to achieve photodynamic therapeutic effects in pancreatic cancer. Eutectic gallium indium (EGaIn) nanoparticles were successfully conjugated with a water-soluble cancer targeting ligand, hyaluronic acid, and a photosensitizer, benzoporphyrin derivative, creating EGaIn nanoparticles (EGaPs) via a simple green sonication method. The prepared sphere-shaped EGaPs, with a core-shell structure, presented high biocompatibility and stability. EGaPs had greater cellular uptake, manifested targeting competence, and generated significantly higher intracellular ROS. Further, near-infrared light activation of EGaPs demonstrated their potential to effectively eliminate cancer cells due to their single oxygen generation capability. Finally, from in vivo studies, EGaPs caused tumor regression and resulted in 2.3-fold higher necrosis than the control, therefore making a good vehicle for photodynamic therapy. The overall results highlight that EGaPs provide a new way to assemble liquid metal nanomaterials with different ligands for enhanced cancer therapy.
Collapse
Affiliation(s)
- Sabrina
S. Hafiz
- Department
of Chemistry, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
| | - Marvin Xavierselvan
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Sumeyra Gokalp
- Department
of Chemistry, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
| | - Daniela Labadini
- Department
of Chemistry, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
| | - Sebastian Barros
- Department
of Chemistry, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
| | - Jeanne Duong
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Michelle Foster
- Department
of Chemistry, University of Massachusetts
Boston, Boston, Massachusetts 02125, United States
| | - Srivalleesha Mallidi
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
11
|
Field RD, Jakus MA, Chen X, Human K, Zhao X, Chitnis PV, Sia SK. Ultrasound-Responsive Aqueous Two-Phase Microcapsules for On-Demand Drug Release. Angew Chem Int Ed Engl 2022; 61:e202116515. [PMID: 35233907 DOI: 10.1002/anie.202116515] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Indexed: 12/21/2022]
Abstract
Traditional implanted drug delivery systems cannot easily change their release profile in real time to respond to physiological changes. Here we present a microfluidic aqueous two-phase system to generate microcapsules that can release drugs on demand as triggered by focused ultrasound (FUS). The biphasic microcapsules are made of hydrogels with an outer phase of mixed molecular weight (MW) poly(ethylene glycol) diacrylate that mitigates premature payload release and an inner phase of high MW dextran with payload that breaks down in response to FUS. Compound release from microcapsules could be triggered as desired; 0.4 μg of payload was released across 16 on-demand steps over days. We detected broadband acoustic signals amidst low heating, suggesting inertial cavitation as a key mechanism for payload release. Overall, FUS-responsive microcapsules are a biocompatible and wirelessly triggerable structure for on-demand drug delivery over days to weeks.
Collapse
Affiliation(s)
- Rachel D Field
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Margaret A Jakus
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Xiaoyu Chen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kelia Human
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Xuanhe Zhao
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Parag V Chitnis
- Department of Bioengineering, George Mason University, 4400 University Drive, Fairfax, VA 22030, USA
| | - Samuel K Sia
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| |
Collapse
|
12
|
Yang Y, Zheng X, Chen L, Gong X, Yang H, Duan X, Zhu Y. Multifunctional Gold Nanoparticles in Cancer Diagnosis and Treatment. Int J Nanomedicine 2022; 17:2041-2067. [PMID: 35571258 PMCID: PMC9094645 DOI: 10.2147/ijn.s355142] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/20/2022] [Indexed: 12/18/2022] Open
Abstract
Cancer is the second leading cause of death in the world, behind only cardiovascular diseases, and is one of the most serious diseases threatening human health nowadays. Cancer patients’ lives are being extended by the use of contemporary medical technologies, such as surgery, radiotherapy, and chemotherapy. However, these treatments are not always effective in extending cancer patients’ lives. Simultaneously, these approaches are often accompanied with a series of negative consequences, such as the occurrence of adverse effects and an increased risk of relapse. As a result, the development of a novel cancer-eradication strategy is still required. The emergence of nanomedicine as a promising technology brings a new avenue for the circumvention of limitations of conventional cancer therapies. Gold nanoparticles (AuNPs), in particular, have garnered extensive attention due to their many specific advantages, including customizable size and shape, multiple and useful physicochemical properties, and ease of functionalization. Based on these characteristics, many therapeutic and diagnostic applications of AuNPs have been exploited, particularly for malignant tumors, such as drug and nucleic acid delivery, photodynamic therapy, photothermal therapy, and X-ray-based computed tomography imaging. To leverage the potential of AuNPs, these applications demand a comprehensive and in-depth overview. As a result, we discussed current achievements in AuNPs in anticancer applications in a more methodical manner in this review. Also addressed in depth are the present status of clinical trials, as well as the difficulties that may be encountered when translating some basic findings into the clinic, in order to serve as a reference for future studies.
Collapse
Affiliation(s)
- Yan Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Xi Zheng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Lu Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Xuefeng Gong
- POWERCHINA Chengdu Engineering Corporation Limited, Chengdu, 611130, People’s Republic of China
| | - Hao Yang
- POWERCHINA Chengdu Engineering Corporation Limited, Chengdu, 611130, People’s Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Yuxuan Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
- Correspondence: Yuxuan Zhu, Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China, Email
| |
Collapse
|
13
|
Schirato A, Moretti L, Yang Z, Mazzanti A, Cerullo G, Pileni MP, Maiuri M, Della Valle G. Chemically-Controlled Ultrafast Photothermal Response in Plasmonic Nanostructured Assemblies. THE JOURNAL OF PHYSICAL CHEMISTRY. C, NANOMATERIALS AND INTERFACES 2022; 126:6308-6317. [PMID: 35449522 PMCID: PMC9014708 DOI: 10.1021/acs.jpcc.2c00364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/15/2022] [Indexed: 06/14/2023]
Abstract
Plasmonic nanoparticles are renowned as efficient heaters due to their capability to resonantly absorb and concentrate electromagnetic radiation, trigger excitation of highly energetic (hot) carriers, and locally convert their excess energy into heat via ultrafast nonradiative relaxation processes. Furthermore, in assembly configurations (i.e., suprastructures), collective effects can even enhance the heating performance. Here, we report on the dynamics of photothermal conversion and the related nonlinear optical response from water-soluble nanoeggs consisting of a Au nanocrystal assembly trapped in a water-soluble shell of ferrite nanocrystals (also called colloidosome) of ∼250-300 nm in size. This nanoegg configuration of the plasmonic assembly enables control of the size of the gold suprastructure core by changing the Au concentration in the chemical synthesis. Different metal concentrations are analyzed by means of ultrafast pump-probe spectroscopy and semiclassical modeling of photothermal dynamics from the onset of hot-carrier photogeneration (few picosecond time scale) to the heating of the matrix ligands in the suprastructure core (hundreds of nanoseconds). Results show the possibility to design and tailor the photothermal properties of the nanoeggs by acting on the core size and indicate superior performances (both in terms of peak temperatures and thermalization speed) compared to conventional (unstructured) nanoheaters of comparable size and chemical composition.
Collapse
Affiliation(s)
- Andrea Schirato
- Dipartimento
di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
- Istituto
Italiano di Tecnologia, via Morego 30, I-16163 Genova, Italy
| | - Luca Moretti
- Dipartimento
di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
| | - Zhijie Yang
- Key
Laboratory of Colloid and Interface Chemistry, Ministry of Education,
School of Chemistry and Chemical Engineering, Shandong University, 250100 Jinan, China
| | - Andrea Mazzanti
- Dipartimento
di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
| | - Giulio Cerullo
- Dipartimento
di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
- Istituto
di Fotonica e Nanotecnologie - Consiglio Nazionale delle Ricerche, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
| | | | - Margherita Maiuri
- Dipartimento
di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
- Istituto
di Fotonica e Nanotecnologie - Consiglio Nazionale delle Ricerche, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
| | - Giuseppe Della Valle
- Dipartimento
di Fisica, Politecnico di Milano, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
- Istituto
di Fotonica e Nanotecnologie - Consiglio Nazionale delle Ricerche, Piazza Leonardo da Vinci, 32, I-20133 Milano, Italy
| |
Collapse
|
14
|
Jiang Y, Zhao W, Zhou H, Zhang Q, Zhang S. ATP-Triggered Intracellular In Situ Aggregation of a Gold-Nanoparticle-Equipped Triple-Helix Molecular Switch for Fluorescence Imaging and Photothermal Tumor Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:3755-3764. [PMID: 35291761 DOI: 10.1021/acs.langmuir.1c03331] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Isotropic gold nanoparticles (AuNPs) can generate a plasma-plasma interaction when aggregating and can also produce ideal photothermal effects. Some studies have designed ATP-responsive nanodrug delivery systems by taking advantage of the differences between internal and external ATP in tumor cells, but few studies have focused on the photothermal effects of ATP-induced AuNP aggregation in tumors. Here, a triple-helix probe (THP) molecular switch and MUC1 aptamer-functionalized AuNPs were constructed for fluorescence imaging analysis and photothermal therapy (PTT). The MUC1 aptamer guides THP-AuNP targeting in tumor cells, followed by the high concentration of ATP inducing structural changes in triple-helix probes and causing the intracellular aggregation of AuNPs, which cannot escape from the tumor site, enabling tumor imaging while performing PTT. Therefore, the designed THP-AuNPs have promising applications in fluorescence imaging and PTT.
Collapse
Affiliation(s)
- Yao Jiang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, P. R. China
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Wenjing Zhao
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Huimin Zhou
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Qiuqi Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Shusheng Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| |
Collapse
|
15
|
Field RD, Jakus MA, Chen X, Human K, Zhao X, Chitnis PV, Sia SK. Ultrasound‐Responsive Aqueous Two‐Phase Microcapsules for On‐Demand Drug Release. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202116515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Rachel D. Field
- Department of Biomedical Engineering Columbia University 351 Engineering Terrace, 1210 Amsterdam Avenue New York NY 10027 USA
| | - Margaret A. Jakus
- Department of Biomedical Engineering Columbia University 351 Engineering Terrace, 1210 Amsterdam Avenue New York NY 10027 USA
| | - Xiaoyu Chen
- Department of Mechanical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Kelia Human
- Department of Biomedical Engineering Columbia University 351 Engineering Terrace, 1210 Amsterdam Avenue New York NY 10027 USA
| | - Xuanhe Zhao
- Department of Mechanical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
- Department of Civil and Environmental Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Parag V. Chitnis
- Department of Bioengineering George Mason University 4400 University Drive Fairfax VA 22030 USA
| | - Samuel K. Sia
- Department of Biomedical Engineering Columbia University 351 Engineering Terrace, 1210 Amsterdam Avenue New York NY 10027 USA
| |
Collapse
|
16
|
Upadhyay A, Kundu P, Ramu V, Kondaiah P, Chakravarty AR. BODIPY-Tagged Platinum(II) Curcumin Complexes for Endoplasmic Reticulum-Targeted Red Light PDT. Inorg Chem 2022; 61:1335-1348. [PMID: 34990135 DOI: 10.1021/acs.inorgchem.1c02745] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
[Pt(RB)(Cur)]NO3 (RBC), [Pt(IRB)(Cur)]NO3 (IRBC), and [Pt(L)(Cur)]NO3 (PBC), where HCur is curcumin, L is 1-benzyl-2-(2-pyridyl)benzimidazole, and RB and IRB are red-light-active non-iodo and diiodo-BODIPY tagged to L, respectively, were synthesized and characterized, and their anticancer activities were studied (BODIPY, boron-dipyrromethene). RBC and IRBC displayed BODIPY-centered absorption bands within 615-635 nm along with the respective curcumin bands at 445 and 492 nm in 10% dimethyl sulfoxide (DMSO)-Dulbecco's phosphate-buffered saline (DPBS). Emission bands were observed at 723 and 845 nm for RBC and IRBC, respectively, in 10% DMSO-DPBS. RBC (ΦΔ, 0.27) and IRBC (ΦΔ, 0.40) generated singlet oxygen in red light (λ = 642 nm) as evidenced from 1,3-diphenylisobenzofuran (DPBF) titrations. The formation of 1O2 from BODIPY and HO• from the curcumin was evidenced from the mechanistic pUC19 DNA photocleavage studies. The BODIPY complexes showed photocytotoxicity in A549, HeLa, and MDA-MB-231 cells while being less toxic in the dark [IC50: 1.3-6.9 μM, red light; 7.2-12.8 μM, 400-700 nm visible light]. The emissive RBC displayed localization in the endoplasmic reticulum (ER). Apoptotic cell death was evidenced from the Annexin-V/fluorescein isothiocyanate (FITC)/propidium iodide (PI) assay and green fluorescence in red light in the Fluo-4 AM assay due to ER stress, and mitochondrial dysfunction was evidenced from the 5,5,6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1) assay in A549 cells.
Collapse
|
17
|
Zhang L, Guan X, Xiao X, Chai Y, Chen Z, Zhou G, Fan Y. Near-infrared triggered injectable ferrimagnetic chitosan thermosensitive hydrogel for photo hyperthermia and precisely controlled drug release in tumor ablation. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2021.110879] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
18
|
Qian C, Al-Hamyari B, Tang X, Hou B, Yang S, Zhang G, Lv H, Yang Z, Wang Z, Shi Y. Interface-Engineered Paclitaxel-Based Hollow Mesoporous Organosilica Nanoplatforms for Photothermal-Enhanced Chemotherapy of Tumor. Mol Pharm 2021; 18:4531-4542. [PMID: 34739255 DOI: 10.1021/acs.molpharmaceut.1c00735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Having benefited from the combination of different therapeutic modalities, functionalized nanoplatforms with synergistic strategies have aroused great interest in anticancer treatment. Herein, an engineered, a biodegradable hollow mesoporous organosilica nanoparticle (HMON)-based nanoplatform was fabricated for photothermal-enhanced chemotherapy of tumor. For the first time, we demonstrated that HMONs could serve as nanocarriers for co-delivering of both the paclitaxel and photothermal agent new indocyanine green (IR820), denoted as Paclitaxel/IR820@ HMONs-PEG. The as-prepared nanosystem exhibited a high paclitaxel-loading capacity of 28.4%, much higher than most paclitaxel-loaded nanoformulations. Furthermore, incorporating thioether bonds (S-S) into the HMONs' framework endowed them with GSH-responsive biodegradation behavior, leading to the controllable release of drugs under a tumor reducing microenvironment, and hindered the premature release of paclitaxel. Upon being irradiated with an NIR laser, the obtained co-delivery nanosystem exhibited great photothermal properties generated from IR820. The fabricated nanocomposites could significantly suppress tumor growth under NIR laser irradiation, as validated by in vitro and in vivo assessments. Combined with outstanding biocompatibility, the constructed nanosystem holds great potential in combinational antitumor therapy.
Collapse
Affiliation(s)
- Chunxi Qian
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| | - Bandar Al-Hamyari
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| | - Xiaofei Tang
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| | - Bo Hou
- School of Physics and Astronomy, Cardiff University, Cardiff, Wales CF243AA, United Kingdom
| | - Shuaibo Yang
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| | - Guifang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| | - Huijuan Lv
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| | - Zhigang Yang
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| | - Zhaohui Wang
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| | - Yanbin Shi
- School of Pharmacy, Lanzhou University, Lanzhou 730030, Gansu, P.R. China
| |
Collapse
|
19
|
Rinoldi C, Zargarian SS, Nakielski P, Li X, Liguori A, Petronella F, Presutti D, Wang Q, Costantini M, De Sio L, Gualandi C, Ding B, Pierini F. Nanotechnology-Assisted RNA Delivery: From Nucleic Acid Therapeutics to COVID-19 Vaccines. SMALL METHODS 2021; 5:e2100402. [PMID: 34514087 PMCID: PMC8420172 DOI: 10.1002/smtd.202100402] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/04/2021] [Indexed: 05/07/2023]
Abstract
In recent years, the main quest of science has been the pioneering of the groundbreaking biomedical strategies needed for achieving a personalized medicine. Ribonucleic acids (RNAs) are outstanding bioactive macromolecules identified as pivotal actors in regulating a wide range of biochemical pathways. The ability to intimately control the cell fate and tissue activities makes RNA-based drugs the most fascinating family of bioactive agents. However, achieving a widespread application of RNA therapeutics in humans is still a challenging feat, due to both the instability of naked RNA and the presence of biological barriers aimed at hindering the entrance of RNA into cells. Recently, material scientists' enormous efforts have led to the development of various classes of nanostructured carriers customized to overcome these limitations. This work systematically reviews the current advances in developing the next generation of drugs based on nanotechnology-assisted RNA delivery. The features of the most used RNA molecules are presented, together with the development strategies and properties of nanostructured vehicles. Also provided is an in-depth overview of various therapeutic applications of the presented systems, including coronavirus disease vaccines and the newest trends in the field. Lastly, emerging challenges and future perspectives for nanotechnology-mediated RNA therapies are discussed.
Collapse
Affiliation(s)
- Chiara Rinoldi
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Seyed Shahrooz Zargarian
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Pawel Nakielski
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Xiaoran Li
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Anna Liguori
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
| | - Francesca Petronella
- Institute of Crystallography CNR‐ICNational Research Council of ItalyVia Salaria Km 29.300Monterotondo – Rome00015Italy
| | - Dario Presutti
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Qiusheng Wang
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Marco Costantini
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Luciano De Sio
- Department of Medico‐Surgical Sciences and BiotechnologiesResearch Center for BiophotonicsSapienza University of RomeCorso della Repubblica 79Latina04100Italy
- CNR‐Lab. LicrylInstitute NANOTECArcavacata di Rende87036Italy
| | - Chiara Gualandi
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
- Interdepartmental Center for Industrial Research on Advanced Applications in Mechanical Engineering and Materials TechnologyCIRI‐MAMUniversity of BolognaViale Risorgimento 2Bologna40136Italy
| | - Bin Ding
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Filippo Pierini
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| |
Collapse
|
20
|
Yang Y, Wang H. The Golden Age: Shining the Light on Theragnostics. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Yamin Yang
- Department of Biomedical Engineering Nanjing University of Aeronautics and Astronautics Nanjing Jiangsu 211106 China
| | - Hongjun Wang
- Department of Biomedical Engineering Stevens Institute of Technology Hoboken NJ 07030 USA
- Department of Chemistry and Chemical Biology Stevens Institute of Technology Hoboken NJ 07030 USA
| |
Collapse
|
21
|
Yang Y, Feng Q, Ding C, Kang W, Xiao X, Yu Y, Zhou Q. Controllable Drug Delivery by Na+/K+ ATPase α1 Targeting Peptide Conjugated DSPE-PEG Nanocarriers for Breast Cancer. Technol Cancer Res Treat 2021; 20:15330338211027898. [PMID: 34180301 PMCID: PMC8243088 DOI: 10.1177/15330338211027898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although Epirubicin (EPI) is a commonly used anthracycline for the treatment of
breast cancer in clinic, the serious side effects limit its long-term
administration including myelosuppression and cardiomyopathy. Nanomedicines have
been widely utilized as drug delivery vehicles to achieve precise targeting of
breast cancer cells. Herein, we prepared a DSPE-PEG nanocarrier conjugated a
peptide, which targeted the breast cancer overexpression protein Na+/K+ ATPase
α1 (NKA-α1). The nanocarrier encapsulated the EPI and grafted with the NKA-α1
targeting peptide through the click reaction between maleimide and thiol groups.
The EPI was slowly released from the nanocarrier after entering the breast
cancer cells with the guidance of the targeting NKA-α1 peptide. The precise and
controllable delivery and release of the EPI into the breast cancer cells
dramatically inhibited the cells proliferation and migration in
vitro and suppressed the tumor volume in vivo.
These results demonstrate significant prospects for this nanocarrier as a
promising platform for numerous chemotherapy drugs.
Collapse
Affiliation(s)
- Yayan Yang
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Qian Feng
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Chuanfeng Ding
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Kang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China.,Ningbo Institute of Dalian University of Technology, Ningbo, Zhejiang, China
| | - Xiufeng Xiao
- Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Yongsheng Yu
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qian Zhou
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
NIR light-responsive nanocarriers for controlled release. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY C: PHOTOCHEMISTRY REVIEWS 2021. [DOI: 10.1016/j.jphotochemrev.2021.100420] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Xue C, Hu S, Gao ZH, Wang L, Luo MX, Yu X, Li BF, Shen Z, Wu ZS. Programmably tiling rigidified DNA brick on gold nanoparticle as multi-functional shell for cancer-targeted delivery of siRNAs. Nat Commun 2021; 12:2928. [PMID: 34006888 PMCID: PMC8131747 DOI: 10.1038/s41467-021-23250-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
Small interfering RNA (siRNA) is an effective therapeutic to regulate the expression of target genes in vitro and in vivo. Constructing a siRNA delivery system with high serum stability, especially responsive to endogenous stimuli, remains technically challenging. Herein we develop anti-degradation Y-shaped backbone-rigidified triangular DNA bricks with sticky ends (sticky-YTDBs) and tile them onto a siRNA-packaged gold nanoparticle in a programmed fashion, forming a multi-functional three-dimensional (3D) DNA shell. After aptamers are arranged on the exterior surface, a biocompatible siRNA-encapsulated core/shell nanoparticle, siRNA/Ap-CS, is achieved. SiRNAs are internally encapsulated in a 3D DNA shell and are thus protected from enzymatic degradation by the outermost layer of YTDB. The siRNAs can be released by endogenous miRNA and execute gene silencing within tumor cells, causing cell apoptosis higher than Lipo3000/siRNA formulation. In vivo treatment shows that tumor growth is completely (100%) inhibited, demonstrating unique opportunities for next-generation anticancer-drug carriers for targeted cancer therapies.
Collapse
Affiliation(s)
- Chang Xue
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Shuyao Hu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zhi-Hua Gao
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medicine Genetics, School of Laboratory Medicine and Life Sciences, Institute of Functional Nucleic Acids and Personalized Cancer Theranostics, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lei Wang
- Hunan Provincial Key Laboratory of Phytohormones and Growth Development, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Meng-Xue Luo
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xin Yu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Bi-Fei Li
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zhifa Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medicine Genetics, School of Laboratory Medicine and Life Sciences, Institute of Functional Nucleic Acids and Personalized Cancer Theranostics, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
24
|
Chen W, Cheng CA, Xiang D, Zink JI. Expanding nanoparticle multifunctionality: size-selected cargo release and multiple logic operations. NANOSCALE 2021; 13:5497-5506. [PMID: 33687426 DOI: 10.1039/d1nr00642h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Physically stimulated nanoparticles that deliver size-selected cargo and function as logic gates are reported. To achieve this goal the particle requires multiple components, and we recognized early on that the components, not just the released cargo, could be used to demonstrate logic operations (OR and AND logic). For stimuli, we chose two non-invasive types, red light and alternating magnetic fields (AMF), because they both have potential biological relevance. To realize cargo delivery with size selection and logic operations, we mechanized the surface of core@shell nanoparticles with a superparamagnetic core that generates localized heating when exposed to an AMF, and a mesoporous silica shell into which cargo molecules with different sizes were loaded. We demonstrate the core@shell nanoparticles can load the dual cargos with different sizes and subsequently release the smaller (∼0.5 nm) and bigger (∼2 nm) cargos in succession when stimulated by a red light followed by an AMF. Finally, we demonstrate that the multi-component nanoparticles could function as nanoparticle-based Boolean logic gates where AMF and red light served as the two inputs and the release of small cargo, and free cyclodextrin served as the outputs. The construction of two Boolean logic gates (OR, and AND) was realized.
Collapse
Affiliation(s)
- Wei Chen
- Department of Chemistry & Biochemistry, University of California Los Angeles, Los Angeles, California, 90095, USA. and California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, 90095, USA
| | - Chi-An Cheng
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, 90095, USA and Department of Bioengineering, University of California Los Angeles, Los Angeles, California, 90095, USA
| | - Danlei Xiang
- Department of Chemistry & Biochemistry, University of California Los Angeles, Los Angeles, California, 90095, USA.
| | - Jeffrey I Zink
- Department of Chemistry & Biochemistry, University of California Los Angeles, Los Angeles, California, 90095, USA. and California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, 90095, USA
| |
Collapse
|
25
|
The Issue of Tissue: Approaches and Challenges to the Light Control of Drug Activity. CHEMPHOTOCHEM 2021; 5:611-618. [DOI: 10.1002/cptc.202100001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
26
|
Nicolson F, Kircher MF. Theranostics: Agents for Diagnosis and Therapy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
27
|
Affiliation(s)
- Na Liu
- 2nd Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569 Stuttgart, Germany
- Max Planck Institute for Solid State Research, Heisenbergstrasse 1, 70569 Stuttgart, Germany
| |
Collapse
|
28
|
Nicolson F, Ali A, Kircher MF, Pal S. DNA Nanostructures and DNA-Functionalized Nanoparticles for Cancer Theranostics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001669. [PMID: 33304747 PMCID: PMC7709992 DOI: 10.1002/advs.202001669] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/27/2020] [Indexed: 05/12/2023]
Abstract
In the last two decades, DNA has attracted significant attention toward the development of materials at the nanoscale for emerging applications due to the unparalleled versatility and programmability of DNA building blocks. DNA-based artificial nanomaterials can be broadly classified into two categories: DNA nanostructures (DNA-NSs) and DNA-functionalized nanoparticles (DNA-NPs). More importantly, their use in nanotheranostics, a field that combines diagnostics with therapy via drug or gene delivery in an all-in-one platform, has been applied extensively in recent years to provide personalized cancer treatments. Conveniently, the ease of attachment of both imaging and therapeutic moieties to DNA-NSs or DNA-NPs enables high biostability, biocompatibility, and drug loading capabilities, and as a consequence, has markedly catalyzed the rapid growth of this field. This review aims to provide an overview of the recent progress of DNA-NSs and DNA-NPs as theranostic agents, the use of DNA-NSs and DNA-NPs as gene and drug delivery platforms, and a perspective on their clinical translation in the realm of oncology.
Collapse
Affiliation(s)
- Fay Nicolson
- Department of ImagingDana‐Farber Cancer Institute & Harvard Medical SchoolBostonMA02215USA
- Center for Molecular Imaging and NanotechnologyMemorial Sloan Kettering Cancer CenterNew YorkNY10065USA
| | - Akbar Ali
- Department of ChemistryIndian Institute of Technology‐ BhilaiRaipurChhattisgarh492015India
| | - Moritz F. Kircher
- Department of ImagingDana‐Farber Cancer Institute & Harvard Medical SchoolBostonMA02215USA
- Center for Molecular Imaging and NanotechnologyMemorial Sloan Kettering Cancer CenterNew YorkNY10065USA
- Department of RadiologyBrigham and Women's Hospital & Harvard Medical SchoolBostonMA02215USA
| | - Suchetan Pal
- Department of ChemistryIndian Institute of Technology‐ BhilaiRaipurChhattisgarh492015India
| |
Collapse
|
29
|
Bhattacharjee S, Brayden DJ. Addressing the challenges to increase the efficiency of translating nanomedicine formulations to patients. Expert Opin Drug Discov 2020; 16:235-254. [PMID: 33108229 DOI: 10.1080/17460441.2021.1826434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nanotechnology is in a growth phase for drug delivery and medical imaging. Nanomaterials with unique properties present opportunities for encapsulation of therapeutics and imaging agents, along with conjugation to ligands for targeting. Favorable chemistry of nanomaterials can create formulations that address critical challenges for therapeutics, such as insolubility and a low capacity to cross the blood-brain-barrier (BBB) and intestinal wall. AREAS COVERED The authors investigate challenges faced during translation of nanomedicines while suggesting reasons as to why some nanoformulations have under-performed in clinical trials. They assess physiological barriers such as the BBB and gut mucus that nanomedicines must overcome to deliver cargos. They also provide an overview with examples of how nanomedicines can be designed to improve localization and site-specific delivery (e.g., encapsulation, bioconjugation, and triggered-release). EXPERT OPINION There are examples where nanomedicines have demonstrated improved efficacy of payload in humans; however, most of the advantages conferred were in improved pharmacokinetics and reduced toxicity. Problematic data show susceptibility of nanoformulations against natural protective mechanisms present in the body, including distribution impediment by physiological barriers and activation of the reticuloendothelial system. Further initiatives should address current challenges while expanding the scope of nanomedicine into advanced biomedical imaging and antibiotic delivery.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland
| | - David J Brayden
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin, Ireland
| |
Collapse
|
30
|
Lee HL, Wei H, Kim K, Choe HS, Park H, Yu T, Lee C, Kim JH, Kim JH. Versatile Yolk-Shell Encapsulation: Catalytic, Photothermal, and Sensing Demonstration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002311. [PMID: 32743935 DOI: 10.1002/smll.202002311] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/18/2020] [Indexed: 05/28/2023]
Abstract
Here, a novel, versatile synthetic strategy to fabricate a yolk-shell structured material that can encapsulate virtually any functional noble metal or metal oxide nanocatalysts of any morphology in a free suspension fashion is reported. This strategy also enables encapsulation of more than one type of nanoparticle inside a single shell, including paramagnetic iron oxide used for magnetic separation. The mesoporous organosilica shell provides efficient mass transfer of small target molecules, while serving as a size exclusion barrier for larger interfering molecules. Major structural and functional advantages of this material design are demonstrated by performing three proof-of-concept applications. First, effective encapsulation of plasmonic gold nanospheres for localized photothermal heating and heat-driven reaction inside the shell is shown. Second, hydrogenation catalysis is demonstrated under spatial confinement driven by palladium nanocubes. Finally, the surface-enhanced Raman spectroscopic detection of model pollutant by gold nanorods is presented for highly sensitive environmental sensing with size exclusion.
Collapse
Affiliation(s)
- Hak-Lae Lee
- Department of Chemical and Environmental Engineering, Pusan National University, Busan, 46241, Korea
| | - Haoran Wei
- Department of Chemical and Environmental Engineering and Nanosystems Engineering Research Center for Nanotechnology-Enabled Water Treatment (NEWT), Yale University, New Haven, CT, 06511, USA
- Environmental Chemistry and Technology and Department of Civil and Environmental Engineering, University of Wisconsin-Madison, 660 N Park Street, Madison, WI, 53706, USA
| | - Kiyoon Kim
- Department of Chemical Engineering, Kyung Hee University, Yongin, 17140, Korea
| | - Hyun-Seok Choe
- Department of Chemical and Environmental Engineering, Pusan National University, Busan, 46241, Korea
| | - Hyun Park
- Department of Naval Architecture and Ocean Engineering, Pusan National University, Busan, 46241, Korea
| | - Taekyung Yu
- Department of Chemical Engineering, Kyung Hee University, Yongin, 17140, Korea
| | - Changha Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Korea
| | - Jae-Hong Kim
- Department of Chemical and Environmental Engineering and Nanosystems Engineering Research Center for Nanotechnology-Enabled Water Treatment (NEWT), Yale University, New Haven, CT, 06511, USA
| | - Jae-Hyuk Kim
- Department of Chemical and Environmental Engineering, Pusan National University, Busan, 46241, Korea
| |
Collapse
|
31
|
Bromma K, Bannister A, Kowalewski A, Cicon L, Chithrani DB. Elucidating the fate of nanoparticles among key cell components of the tumor microenvironment for promoting cancer nanotechnology. Cancer Nanotechnol 2020; 11:8. [PMID: 32849921 PMCID: PMC7437649 DOI: 10.1186/s12645-020-00064-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Successful integration of nanotechnology into the current paradigm of cancer therapy requires proper understanding of the interface between nanoparticles (NPs) and cancer cells, as well as other key components within the tumor microenvironment (TME), such as normal fibroblasts (FBs) and cancer-associated FBs (CAFs). So far, much focus has been on cancer cells, but FBs and CAFs also play a critical role: FBs suppress the tumor growth while CAFs promote it. It is not yet known how NPs interact with FBs and CAFs compared to cancer cells. Hence, our goal was to elucidate the extent of NP uptake, retention, and toxicity in cancer cells, FBs, and CAFs to further understand the fate of NPs in a real tumor-like environment. The outcome of this would guide designing of NP-based delivery systems to fully exploit the TME for a better therapeutic outcome. We used gold nanoparticles as our model NP system due to their numerous applications in cancer therapy, including radiotherapy and chemotherapy. A cervical cancer cell line, HeLa, and a triple-negative breast cancer cell line, MDA-MB-231 were chosen as cancer cell lines. For this study, a clinically feasible 0.2 nM concentration of GNPs was employed. According to our results, the cancer cells and CAFs had over 25- and 10-fold higher NP uptake per unit cell volume compared to FBs, respectively. Further, the cancer cells and CAFs had over 30% higher NP retention compared to FBs. There was no observed significant toxicity due to GNPs in all the cell lines studied. Higher uptake and retention of NPs in cancer cells and CAFs vs FBs is very important in promoting NP-based applications in cancer therapy. Our results show potential in modulating uptake and retention of GNPs among key components of TME, in an effort to develop NP-based strategies to suppress the tumor growth. An ideal NP-based platform would eradicate tumor cells, protect FBs, and deactivate CAFs. Therefore, this study lays a road map to exploit the TME for the advancement of "smart" nanomedicines that would constitute the next generation of cancer therapeutics.
Collapse
Affiliation(s)
- Kyle Bromma
- Department of Physics and Astronomy, University of Victoria, Victoria, BC Canada
| | - Aaron Bannister
- Department of Physics and Astronomy, University of Victoria, Victoria, BC Canada
| | | | - Leah Cicon
- Department of Physics and Astronomy, University of Victoria, Victoria, BC Canada
| | - Devika B. Chithrani
- Department of Physics and Astronomy, University of Victoria, Victoria, BC Canada
- Centre for Advanced Materials and Related Technologies (CAMTEC), Victoria, BC Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC Canada
| |
Collapse
|
32
|
Tomitaka A, Arami H, Ahmadivand A, Pala N, McGoron AJ, Takemura Y, Febo M, Nair M. Magneto-plasmonic nanostars for image-guided and NIR-triggered drug delivery. Sci Rep 2020; 10:10115. [PMID: 32572041 PMCID: PMC7308341 DOI: 10.1038/s41598-020-66706-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/15/2020] [Indexed: 11/10/2022] Open
Abstract
Smart multifunctional nanoparticles with magnetic and plasmonic properties assembled on a single nanoplatform are promising for various biomedical applications. Owing to their expanding imaging and therapeutic capabilities in response to external stimuli, they have been explored for on-demand drug delivery, image-guided drug delivery, and simultaneous diagnostic and therapeutic (i.e. theranostic) applications. In this study, we engineered nanoparticles with unique morphology consisting of a superparamagnetic iron oxide core and star-shaped plasmonic shell with high-aspect-ratio gold branches. Strong magnetic and near-infrared (NIR)-responsive plasmonic properties of the engineered nanostars enabled multimodal quantitative imaging combining advantageous functions of magnetic resonance imaging (MRI), magnetic particle imaging (MPI), photoacoustic imaging (PAI), and image-guided drug delivery with a tunable drug release capacity. The model drug molecules bound to the core-shell nanostars were released upon NIR illumination due to the heat generation from the core-shell nanostars. Moreover, our simulation analysis showed that the specific design of the core-shell nanostars demonstrated a pronounced multipolar plasmon resonance, which has not been observed in previous reports. The multimodal imaging and NIR-triggered drug release capabilities of the proposed nanoplatform verify their potential for precise and controllable drug release with different applications in personalized medicine.
Collapse
Affiliation(s)
- Asahi Tomitaka
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199, USA
| | - Hamed Arami
- Molecular Imaging Program at Stanford (MIPS), The James H Clark Center, Stanford University, Stanford, California, 94305, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, California, 94305, USA
| | - Arash Ahmadivand
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, 77005, USA
| | - Nezih Pala
- Department of Electrical and Computer Engineering, Florida International University, Miami, Florida, 33174, USA
| | - Anthony J McGoron
- Department of Biomedical Engineering, Florida International University, Miami, Florida, 33174, USA
| | - Yasushi Takemura
- Department of Electrical and Computer Engineering, Yokohama National University, Yokohama, 240-8501, Japan
| | - Marcelo Febo
- Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, 32611, USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199, USA.
| |
Collapse
|
33
|
Gao C, Kwong CHT, Sun C, Li Z, Lu S, Yang YW, Lee SMY, Wang R. Selective Decoating-Induced Activation of Supramolecularly Coated Toxic Nanoparticles for Multiple Applications. ACS APPLIED MATERIALS & INTERFACES 2020; 12:25604-25615. [PMID: 32406668 DOI: 10.1021/acsami.0c05013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In spite of the rapid emergence of numerous nanoparticles (NPs) for biomedical applications, it is often challenging to precisely control, or effectively tame, the bioactivity/toxicity of NPs, thereby exhibiting limited applications in biomedical areas. Herein, we report the construction of hyaluronic acid (HA)-laminated, otherwise toxic methylviologen (MV), NPs via ternary host-guest complexation among cucurbit[8]uril, trans-azobenzene-conjugated HA, and MV-functionalized polylactic acid NPs (MV-NPs). The high, nonspecific toxicity of MV-NPs was effectively shielded (turned off) by HA lamination, as demonstrated in cells, zebrafish, and mouse models. The supramolecular host-guest interaction-mediated HA coating offered several HA-MV-NP modalities, including hyaluronidase locally and photoirradiation remotely, to precisely remove HA lamination on demand, thereby endowing materials with the capability of selective decoating-induced activation (DIA) for applications as a user-friendly herbicide, a selective antibacterial agent, or an anticancer nanomedicine. This work offers facile supramolecular coating and DIA strategies to effectively tame and precisely control the bioactivity and toxicity of functional nanomaterials for diverse applications.
Collapse
Affiliation(s)
- Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao 999078, China
| | - Cheryl H T Kwong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao 999078, China
| | - Chen Sun
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao 999078, China
| | - Zheng Li
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, International Joint Research Laboratory of Nano-Micro Architecture Chemistry (NMAC), College of Chemistry, Jilin University, Changchun 130012, China
| | - Siyu Lu
- Green Catalysis Center, College of Chemistry, Zhengzhou University, 100 Kexue Road, Zhengzhou 450001, China
| | - Ying-Wei Yang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, International Joint Research Laboratory of Nano-Micro Architecture Chemistry (NMAC), College of Chemistry, Jilin University, Changchun 130012, China
| | - Simon M Y Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao 999078, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao 999078, China
| |
Collapse
|
34
|
Abstract
The current therapies against cancer showed limited success. Nanotechnology is a promising strategy for cancer tracking, diagnosis, and therapy. The hybrid nanotechnology assembled several materials in a multimodal system to develop multifunctional approaches to cancer treatment. The quantum dot and polymer are some of these hybrid nanoparticle platforms. The quantum dot hybrid system possesses photonic and magnetic properties, allowing photothermal therapy and live multimodal imaging of cancer. These quantum dots were used to convey medicines to cancer cells. Hybrid polymer nanoparticles were utilized for the systemic delivery of small interfering RNA to malignant tumors and metastasis. They allowed non-invasive imaging to track in real-time the biodistribution of small interfering RNA in the whole body. They offer an opportunity to treat cancers by specifically silencing target genes. This review highlights the major nanotechnology approaches to effectively treat cancer and metastasis.
Collapse
|
35
|
De Matteis V, Cascione M, Toma CC, Rinaldi R. Engineered Gold Nanoshells Killing Tumor Cells: New Perspectives. Curr Pharm Des 2020; 25:1477-1489. [PMID: 31258061 DOI: 10.2174/1381612825666190618155127] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/11/2019] [Indexed: 12/30/2022]
Abstract
The current strategies to treat different kinds of cancer are mainly based on chemotherapy, surgery and radiation therapy. Unfortunately, these approaches are not specific and rather invasive as well. In this scenario, metal nano-shells, in particular gold-based nanoshells, offer interesting perspectives in the effort to counteract tumor cells, due to their unique ability to tune Surface Plasmon Resonance in different light-absorbing ranges. In particular, the Visible and Near Infrared Regions of the electromagnetic spectrum are able to penetrate through tissues. In this way, the light absorbed by the gold nanoshell at a specific wavelength is converted into heat, inducing photothermal ablation in treated cancer cells. Furthermore, inert gold shells can be easily functionalized with different types of molecules in order to bind cellular targets in a selective manner. This review summarizes the current state-of-art of nanosystems embodying gold shells, regarding methods of synthesis, bio-conjugations, bio-distribution, imaging and photothermal effects (in vitro and in vivo), providing new insights for the development of multifunctional antitumor drugs.
Collapse
Affiliation(s)
- Valeria De Matteis
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| | - Mariafrancesca Cascione
- Dipartimento di Scienze Biomediche e Oncologia Umana, Universita degli Studi di Bari "Aldo Moro", p.zza G. Cesare, c/o Policlinico, 70124 Bari, Italy
| | - Chiara C Toma
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| | - Rosaria Rinaldi
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| |
Collapse
|
36
|
Fan X, Yuan Z, Shou C, Fan G, Wang H, Gao F, Rui Y, Xu K, Yin P. cRGD-Conjugated Fe 3O 4@PDA-DOX Multifunctional Nanocomposites for MRI and Antitumor Chemo-Photothermal Therapy. Int J Nanomedicine 2019; 14:9631-9645. [PMID: 31824156 PMCID: PMC6901060 DOI: 10.2147/ijn.s222797] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/07/2019] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Photothermal therapy (PTT) has great potential in the clinical treatment of tumors. However, most photothermal materials are difficult to apply due to their insufficient photothermal conversion efficiencies (PCEs), poor photostabilities and short circulation times. Furthermore, tumor recurrence is likely to occur using PTT only. In the present study, we prepared cyclo (Arg-Gly-Asp-d-Phe-Cys) [c(RGD)] conjugated doxorubicin (DOX)-loaded Fe3O4@polydopamine (PDA) nanoparticles to develop a multifunctional-targeted nanocomplex for integrated tumor diagnosis and treatment. MATERIALS AND METHODS Cytotoxicity of Fe3O4@PDA-PEG-cRGD-DOX against HCT-116 cells was determined by cck-8 assay. Cellular uptake was measured by confocal laser scanning microscope (CLSM). Pharmacokinetic performance of DOX was evaluated to compare the differences between free DOX and DOX in nanocarrier. Performance in magnetic resonance imaging (MRI) and antitumor activity of complex nanoparticles were evaluated in tumor-bearing nude mice. RESULTS Fe3O4@PDA-PEG-cRGD-DOX has a particle size of 200-300 nm and a zeta potential of 22.7 mV. Further studies in vitro and in vivo demonstrated their excellent capacity to target tumor cells and promote drug internalization, and significantly higher cytotoxicity with respect to that seen in a control group was shown for the nanoparticles. In addition, they have good thermal stability, photothermal conversion efficiencies (PCEs) and pH responsiveness, releasing more DOX in a mildly acidic environment, which is very conducive to their chemotherapeutic effectiveness in the tumor microenvironment. Fe3O4@PDA-PEG-cRGD-DOX NPs were used in a subcutaneous xenograft tumor model of nude mouse HCT-116 cells showed clear signal contrast in T2-weighted images and effective anti-tumor chemo-photothermal therapy under NIR irradiation. CONCLUSION According to our results, Fe3O4@PDA-PEG-cRGD-DOX had a satisfactory antitumor effect on colon cancer in nude mice and could be further developed as a potential integrated platform for the diagnosis and treatment of cancer to improve its antitumor activity against colon cancer.
Collapse
Affiliation(s)
- Xi Fan
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Zeting Yuan
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Chenting Shou
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai, People’s Republic of China
| | - Guohua Fan
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Hong Wang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Feng Gao
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai, People’s Republic of China
| | - Yuanpeng Rui
- Department of Image, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Ke Xu
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Anhui, People’s Republic of China
| |
Collapse
|
37
|
Marino A, Camponovo A, Degl'Innocenti A, Bartolucci M, Tapeinos C, Martinelli C, De Pasquale D, Santoro F, Mollo V, Arai S, Suzuki M, Harada Y, Petretto A, Ciofani G. Multifunctional temozolomide-loaded lipid superparamagnetic nanovectors: dual targeting and disintegration of glioblastoma spheroids by synergic chemotherapy and hyperthermia treatment. NANOSCALE 2019; 11:21227-21248. [PMID: 31663592 PMCID: PMC6867905 DOI: 10.1039/c9nr07976a] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Aiming at finding new solutions for fighting glioblastoma multiforme, one of the most aggressive and lethal human cancer, here an in vitro validation of multifunctional nanovectors for drug delivery and hyperthermia therapy is proposed. Hybrid magnetic lipid nanoparticles have been fully characterized and tested on a multi-cellular complex model resembling the tumor microenvironment. Investigations of cancer therapy based on a physical approach (namely hyperthermia) and on a pharmaceutical approach (by exploiting the chemotherapeutic drug temozolomide) have been extensively carried out, by evaluating its antiproliferative and pro-apoptotic effects on 3D models of glioblastoma multiforme. A systematic study of transcytosis and endocytosis mechanisms has been moreover performed with multiple complimentary investigations, besides a detailed description of local temperature increments following hyperthermia application. Finally, an in-depth proteomic analysis corroborated the obtained findings, which can be summarized in the preparation of a versatile, multifunctional, and effective nanoplatform able to overcome the blood-brain barrier and to induce powerful anti-cancer effects on in vitro complex models.
Collapse
Affiliation(s)
- Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy.
| | - Alice Camponovo
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Andrea Degl'Innocenti
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy.
| | - Martina Bartolucci
- IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy
| | - Christos Tapeinos
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy.
| | - Chiara Martinelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy.
| | - Daniele De Pasquale
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy. and Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Francesca Santoro
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for Health Care, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Valentina Mollo
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for Health Care, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Satoshi Arai
- Kanazawa University, Nano Life Science Institute (WPI-NanoLSI), Kakuma-Machi, 920-1192 Kanazawa, Japan and Waseda University, Research Institute for Science and Engineering, 3-4-1 Ohkubo, Shinjuku-ku, 169-8555 Tokyo, Japan
| | - Madoka Suzuki
- Osaka University, Institute for Protein Research, 3-2 Yamadaoka, Suita-Shi, 565-0871 Osaka, Japan and PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, 332-0012 Saitama, Japan
| | - Yoshie Harada
- Osaka University, Institute for Protein Research, 3-2 Yamadaoka, Suita-Shi, 565-0871 Osaka, Japan
| | - Andrea Petretto
- IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy. and Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
38
|
Liao S, Zhao M, Luo J, Luo K, Wu J, Liu R, Wang S, Jia P, Bai Y, Zheng X. The interaction mechanism between alkaloids and pepsin based on lum-AuNPs in the chemiluminescence analysis. RSC Adv 2019; 9:25569-25575. [PMID: 35530091 PMCID: PMC9070008 DOI: 10.1039/c9ra02978h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 07/20/2019] [Indexed: 12/14/2022] Open
Abstract
Herein, novel luminol functional gold nanoparticles (lum-AuNPs) were quickly prepared in an alkaline luminol solution with HAuCl4, which had the unique characteristics of uniform size and excellent luminescence properties. A self-made flow injection-chemiluminescence (FI-CL) system was established to study the interaction between pepsin (Pep) and five alkaloids (anisodamine, berberine, reserpine, jatrorrhizine and matrine) using lum-AuNPs as the CL probe. Based on the abovementioned home-made CL system, the possible interaction mechanisms of Pep with five alkaloids have been comprehensively discussed by molecular docking simulation, chemical thermodynamics and kinetic studies. The results indicated that there were obvious CL enhancement and inhibition effects on the lum-AuNPs CL system for the Pep and the complex of Pep/alkaloids, respectively. The possible mechanism for the interaction of Pep-five alkaloids was mainly mediated by the hydrophobic force. The binding constant K and binding site n for the Pep-alkaloid interaction are consistent with the list of Ber > Res > Ani, Jat > Mat, which is relative to the potential of groups of alkaloids interacting with the active site of Pep.
Collapse
Affiliation(s)
- Sha Liao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University Xi'an 710069 China
| | - Meimei Zhao
- School of Pharmacy, Shaanxi Institute of International Trade & Commerce Xi'an 712046 China
| | - Jing Luo
- Shaanxi Traditional Chinese Medicine Hospital Xi'an 710004 China
| | - Kai Luo
- Department of Chemistry, Fudan University Shanghai 200438 China
| | - Jingni Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University Xi'an 710069 China
| | - Ruimin Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University Xi'an 710069 China
| | - Shixiang Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University Xi'an 710069 China
| | - Pu Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University Xi'an 710069 China
| | - Yajun Bai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University Xi'an 710069 China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University Xi'an 710069 China
| |
Collapse
|
39
|
Swearer DF, Robatjazi H, Martirez JMP, Zhang M, Zhou L, Carter EA, Nordlander P, Halas NJ. Plasmonic Photocatalysis of Nitrous Oxide into N 2 and O 2 Using Aluminum-Iridium Antenna-Reactor Nanoparticles. ACS NANO 2019; 13:8076-8086. [PMID: 31244036 DOI: 10.1021/acsnano.9b02924] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Photocatalysis with optically active "plasmonic" nanoparticles is a growing field in heterogeneous catalysis, with the potential for substantially increasing efficiencies and selectivities of chemical reactions. Here, the decomposition of nitrous oxide (N2O), a potent anthropogenic greenhouse gas, on illuminated aluminum-iridium (Al-Ir) antenna-reactor plasmonic photocatalysts is reported. Under resonant illumination conditions, N2 and O2 are the only observable decomposition products, avoiding the problematic generation of NOx species observed using other approaches. Because no appreciable change to the apparent activation energy was observed under illumination, the primary reaction enhancement mechanism for Al-Ir is likely due to photothermal heating rather than plasmon-induced hot-carrier contributions. This light-based approach can induce autocatalysis for rapid N2O conversion, a process with highly promising potential for applications in N2O abatement technologies, satellite propulsion, or emergency life-support systems in space stations and submarines.
Collapse
|
40
|
White BD, Duan C, Townley HE. Nanoparticle Activation Methods in Cancer Treatment. Biomolecules 2019; 9:E202. [PMID: 31137744 PMCID: PMC6572460 DOI: 10.3390/biom9050202] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 12/25/2022] Open
Abstract
In this review, we intend to highlight the progress which has been made in recent years around different types of smart activation nanosystems for cancer treatment. Conventional treatment methods, such as chemotherapy or radiotherapy, suffer from a lack of specific targeting and consequent off-target effects. This has led to the development of smart nanosystems which can effect specific regional and temporal activation. In this review, we will discuss the different methodologies which have been designed to permit activation at the tumour site. These can be divided into mechanisms which take advantage of the differences between healthy cells and cancer cells to trigger activation, and those which activate by a mechanism extrinsic to the cell or tumour environment.
Collapse
Affiliation(s)
- Benjamin D White
- Department of Engineering Science, Oxford University, Parks Road, OX1 3PJ, Oxford, UK.
| | - Chengchen Duan
- Nuffield department of Women's and Reproductive Health, Oxford University John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK.
| | - Helen E Townley
- Department of Engineering Science, Oxford University, Parks Road, OX1 3PJ, Oxford, UK.
- Nuffield department of Women's and Reproductive Health, Oxford University John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK.
| |
Collapse
|
41
|
Annesi F, Pane A, Losso MA, Guglielmelli A, Lucente F, Petronella F, Placido T, Comparelli R, Guzzo MG, Curri ML, Bartolino R, De Sio L. Thermo-Plasmonic Killing of Escherichia coli TG1 Bacteria. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E1530. [PMID: 31083308 PMCID: PMC6539421 DOI: 10.3390/ma12091530] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/27/2019] [Accepted: 05/06/2019] [Indexed: 11/16/2022]
Abstract
Plasmonic photo-thermal therapy (PPTT) is a minimally invasive, drug-free, therapy based on the properties of noble metal nanoparticles, able to convert a bio-transparent electromagnetic radiation into heat. PPTT has been used against cancer and other diseases. Herein, we demonstrate an antimicrobial methodology based on the properties of gold nanorods (GNRs). Under a resonant laser irradiation GNRs become highly efficient light to heat nano-converters extremely useful for PPTT applications. The concept here is to assess the antimicrobial effect of easy to synthesize, suitably purified, water-dispersible GNRs on Escherichia coli bacteria. A control on the GNRs concentration used for the process has been demonstrated critical in order to rule out cytotoxic effects on the cells, and still to be able to generate, under a near infrared illumination, an adequate amount of heat suited to increase the temperature up to ≈50 °C in about 5 min. Viability experiments evidenced that the proposed system accomplished a killing efficiency suitable to reducing the Escherichia coli population of about 2 log CFU (colony-forming unit).
Collapse
Affiliation(s)
- Ferdinanda Annesi
- CNR-Lab. Licryl, Institute NANOTEC, 87036 Arcavacata di Rende, Italy.
| | - Alfredo Pane
- CNR-Lab. Licryl, Institute NANOTEC, 87036 Arcavacata di Rende, Italy.
| | - Maria Adele Losso
- Department DiBEST (Biology, Ecology and Earth Sciences), University of Calabria, 87036 Arcavacata di Rende, Italy.
| | - Alexa Guglielmelli
- Department of Physics, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy.
- CNR-Lab. Licryl, Institute NANOTEC, 87036 Arcavacata di Rende, Italy.
| | - Fabrizio Lucente
- Department DiBEST (Biology, Ecology and Earth Sciences), University of Calabria, 87036 Arcavacata di Rende, Italy.
| | - Francesca Petronella
- CNR-IPCF, National Research Council of Italy, Institute for Physical and Chemical Processes-Bari Division, Via Orabona 4, I-70126 Bari, Italy.
| | - Tiziana Placido
- CNR-IPCF, National Research Council of Italy, Institute for Physical and Chemical Processes-Bari Division, Via Orabona 4, I-70126 Bari, Italy.
| | - Roberto Comparelli
- CNR-IPCF, National Research Council of Italy, Institute for Physical and Chemical Processes-Bari Division, Via Orabona 4, I-70126 Bari, Italy.
| | - Maria Grazia Guzzo
- Department DiBEST (Biology, Ecology and Earth Sciences), University of Calabria, 87036 Arcavacata di Rende, Italy.
| | - Maria Lucia Curri
- CNR-IPCF, National Research Council of Italy, Institute for Physical and Chemical Processes-Bari Division, Via Orabona 4, I-70126 Bari, Italy.
- Department of Chemistry, "A. Moro" University of Bari, Via Orabona 4, I-70126 Bari, Italy.
| | - Roberto Bartolino
- Department of Physics, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy.
- CNR-Lab. Licryl, Institute NANOTEC, 87036 Arcavacata di Rende, Italy.
| | - Luciano De Sio
- Department of Medico-surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy.
- CNR-Lab. Licryl, Institute NANOTEC, 87036 Arcavacata di Rende, Italy.
| |
Collapse
|
42
|
Liu Y, Bhattarai P, Dai Z, Chen X. Photothermal therapy and photoacoustic imaging via nanotheranostics in fighting cancer. Chem Soc Rev 2019; 48:2053-2108. [PMID: 30259015 PMCID: PMC6437026 DOI: 10.1039/c8cs00618k] [Citation(s) in RCA: 1622] [Impact Index Per Article: 324.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The nonradiative conversion of light energy into heat (photothermal therapy, PTT) or sound energy (photoacoustic imaging, PAI) has been intensively investigated for the treatment and diagnosis of cancer, respectively. By taking advantage of nanocarriers, both imaging and therapeutic functions together with enhanced tumour accumulation have been thoroughly studied to improve the pre-clinical efficiency of PAI and PTT. In this review, we first summarize the development of inorganic and organic nano photothermal transduction agents (PTAs) and strategies for improving the PTT outcomes, including applying appropriate laser dosage, guiding the treatment via imaging techniques, developing PTAs with absorption in the second NIR window, increasing photothermal conversion efficiency (PCE), and also increasing the accumulation of PTAs in tumours. Second, we introduce the advantages of combining PTT with other therapies in cancer treatment. Third, the emerging applications of PAI in cancer-related research are exemplified. Finally, the perspectives and challenges of PTT and PAI for combating cancer, especially regarding their clinical translation, are discussed. We believe that PTT and PAI having noteworthy features would become promising next-generation non-invasive cancer theranostic techniques and improve our ability to combat cancers.
Collapse
Affiliation(s)
- Yijing Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pravin Bhattarai
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
Wang Y, Zhang Y, Wang J, Liang XJ. Aggregation-induced emission (AIE) fluorophores as imaging tools to trace the biological fate of nano-based drug delivery systems. Adv Drug Deliv Rev 2019; 143:161-176. [PMID: 30529308 DOI: 10.1016/j.addr.2018.12.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/18/2018] [Accepted: 12/03/2018] [Indexed: 01/10/2023]
Abstract
The vigorous development of nanotechnology has been accompanied by an equally strong interest and research efforts in nano-based drug delivery systems (NDDSs). However, only a few NDDSs have been translated into clinic thus far. One of the important hurdles is the lack of tools to comprehensively and directly trace the biological fate of NDDSs. Recently, aggregation-induced emission (AIE) fluorophores have emerged as attractive bioimaging tools due to flexible controllability, negligible toxicity and superior photostability. Herein, we recapitulate the current advances in the application of AIE fluorophores to monitor NDDSs both in vitro and in vivo. Particularly, we discuss the cellular fates of self-indicating and stimuli-responsive NDDSs with AIE fluorophores. Moreover, we highlight the in vivo application of AIE agents on the long-term tracking of therapeutics and the multi-modal monitoring of diagnostics in NDDSs. Challenges and opportunities in AIE-guided exploration of NDDSs are also discussed in detail.
Collapse
Affiliation(s)
- Yufei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuxuan Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinjin Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
44
|
Zhou Z, Yan Y, Wang L, Zhang Q, Cheng Y. Melanin-like nanoparticles decorated with an autophagy-inducing peptide for efficient targeted photothermal therapy. Biomaterials 2019; 203:63-72. [PMID: 30852424 DOI: 10.1016/j.biomaterials.2019.02.023] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 02/09/2023]
Abstract
Photothermal therapy efficiently ablates tumors via hyperthermia but inevitably induces serious side effects including thermal damage to normal tissues, inflammations and enhanced risk of tumor metastasis. In this study, we fabricated a dual peptide decorated melanin-like nanoparticle for tumor-targeted and autophagy-promoted photothermal therapy in pursuit of improved cancer treatment. The multifunctional nanoparticle was composed of dual peptide RGD- and beclin 1-modified and PEGylated melanin-like polydopamine nanoparticles. Beclin 1-derived peptide modified on the nanoparticle up-regulated autophagy in cancer cells and further sensitized the tumors to photothermal ablation. RGD decorated on the particle surface enhanced the selectivity and cellular uptake of polydopamine nanoparticles by breast cancer cells. In vivo therapeutic experiments revealed that the tumor-targeted and autophagy promotion-associated photothermal therapy efficiently regressed tumors at a low temperature around 43 °C. The study provides a novel and efficient strategy to improve the efficiency of photothermal therapy via the up-regulation of autophagy in tumor cells.
Collapse
Affiliation(s)
- Zhengjie Zhou
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, PR China
| | - Yang Yan
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, PR China
| | - Li Wang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, PR China
| | - Qiang Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, PR China.
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, PR China.
| |
Collapse
|
45
|
Li H, Li H, Yu W, Huang S, Liu Y, Zhang N, Yuan J, Xu X, Duan S, Hu Y. PEGylated hyaluronidase/NIR induced drug controlled release system for synergetic chemo-photothermal therapy of hepatocellular carcinoma. Eur J Pharm Sci 2019; 133:127-136. [PMID: 30779981 DOI: 10.1016/j.ejps.2019.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 01/26/2019] [Accepted: 02/15/2019] [Indexed: 01/10/2023]
Abstract
In recent years, cancer treatment has been facing the challenge of increasing antitumor efficiency and avoiding severe adverse effects simultaneously. In this study, we designed a controlled release drug delivery system, doxorubicin (Dox)-loaded and hyaluronic acid (HA)-modified PEGylated gold nanocages (AuNCs), which was designated as PEG-HAn-AuNCs/Dox (n represented 10n HA repeating units were modified on each AuNC). In this system, AuNCs were applied as the photothermal cores, Dox was employed as the model drug, HA was applied as the tumor-microenvironment responsive switch to achieve controlled release, and poly (ethylene glycol) (PEG) was used as the stealth polymer to prolong systemic circulation time. Firstly, we evaluated the physical and chemical properties of the PEG-HAn-AuNCs/Dox with different ratios of HA to AuNC and found that PEG-HA4-AuNCs/Dox was the optimal. Secondly, PEG-HA4-AuNCs/Dox revealed the feature of controlled release, namely, the drug release was triggered by hyaluronidase (HAase) and accelerated by the acidic pH and near-infrared (NIR) irradiation. And then PEG-HA4-AuNCs/Dox could be effectively delivered to a cultured SMMC-7721 cell line in vitro and the tumor tissues of the subcutaneous mouse models of hepatocellular carcinoma (HCC) in vivo. Finally, the results demonstrated the synergetic therapy, namely the combination of chemotherapy and photothermal therapy (PTT) (defined as chemo-photothermal therapy) mediated by PEG-HA4-AuNCs/Dox, could efficiently inhibit the tumor growth both in vitro and in vivo. Therefore, the advantages of PEG-HA4-AuNCs/Dox endowed it as a great potential candidate for HCC treatment.
Collapse
Affiliation(s)
- Huili Li
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Huanjie Li
- General Hospital of Xuzhou Mining Group, Quanshan District Coal Road No. 32, Xuzhou, 221006, PR China
| | - Wei Yu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Shengnan Huang
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Ying Liu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Ningxia Zhang
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Jinxiu Yuan
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Xin Xu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Shaofeng Duan
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, PR China; Henan International Joint Laboratory for Nuclear Protein Regulation, School of Medical Sciences, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, PR China.
| | - Yurong Hu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China; Key Laboratory of Key Technology of Drug Preparation, Ministry of Education, Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China.
| |
Collapse
|
46
|
Zhu D, Roy S, Liu Z, Weller H, Parak WJ, Feliu N. Remotely controlled opening of delivery vehicles and release of cargo by external triggers. Adv Drug Deliv Rev 2019; 138:117-132. [PMID: 30315833 DOI: 10.1016/j.addr.2018.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/23/2018] [Accepted: 10/08/2018] [Indexed: 01/11/2023]
Abstract
Tremendous efforts have been devoted to the development of future nanomedicines that can be specifically designed to incorporate responsive elements that undergo modification in structural properties upon external triggers. One potential use of such stimuli-responsive materials is to release encapsulated cargo upon excitation by an external trigger. Today, such stimuli-response materials allow for spatial and temporal tunability, which enables the controlled delivery of compounds in a specific and dose-dependent manner. This potentially is of great interest for medicine (e.g. allowing for remotely controlled drug delivery to cells, etc.). Among the different external exogenous and endogenous stimuli used to control the desired release, light and magnetic fields offer interesting possibilities, allowing defined, real time control of intracellular releases. In this review we highlight the use of stimuli-responsive controlled release systems that are able to respond to light and magnetic field triggers for controlling the release of encapsulated cargo inside cells. We discuss established approaches and technologies and describe prominent examples. Special attention is devoted towards polymer capsules and polymer vesicles as containers for encapsulated cargo molecules. The advantages and disadvantages of this methodology in both, in vitro and in vivo models are discussed. An overview of challenges associate with the successful translation of those stimuli-responsive materials towards future applications in the direction of potential clinical use is given.
Collapse
Affiliation(s)
- Dingcheng Zhu
- Fachbereich Physik, CHyN, Universität Hamburg, Hamburg, Germany
| | - Sathi Roy
- Fachbereich Physik, CHyN, Universität Hamburg, Hamburg, Germany
| | - Ziyao Liu
- Fachbereich Physik, CHyN, Universität Hamburg, Hamburg, Germany
| | - Horst Weller
- Fachbereich Chemie, Universität Hamburg, Hamburg, Germany
| | - Wolfgang J Parak
- Fachbereich Physik, CHyN, Universität Hamburg, Hamburg, Germany; Fachbereich Chemie, Universität Hamburg, Hamburg, Germany
| | - Neus Feliu
- Fachbereich Physik, CHyN, Universität Hamburg, Hamburg, Germany; Experimental Cancer Medicine, Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
47
|
M SM, Veeranarayanan S, Maekawa T, D SK. External stimulus responsive inorganic nanomaterials for cancer theranostics. Adv Drug Deliv Rev 2019; 138:18-40. [PMID: 30321621 DOI: 10.1016/j.addr.2018.10.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/03/2018] [Accepted: 10/08/2018] [Indexed: 01/21/2023]
Abstract
Cancer is a highly intelligent system of cells, that works together with the body to thrive and subsequently overwhelm the host in order for its survival. Therefore, treatment regimens should be equally competent to outsmart these cells. Unfortunately, it is not the case with current therapeutic practices, the reason why it is still one of the most deadly adversaries and an imposing challenge to healthcare practitioners and researchers alike. With rapid nanotechnological interventions in the medical arena, the amalgamation of diagnostic and therapeutic functionalities into a single platform, theranostics provides a never before experienced hope of enhancing diagnostic accuracy and therapeutic efficiency. Additionally, the ability of these nanotheranostic agents to perform their actions on-demand, i.e. can be controlled by external stimulus such as light, magnetic field, sound waves and radiation has cemented their position as next generation anti-cancer candidates. Numerous reports exist of such stimuli-responsive theranostic nanomaterials against cancer, but few have broken through to clinical trials, let alone clinical practice. This review sheds light on the pros and cons of a few such theranostic nanomaterials, especially inorganic nanomaterials which do not require any additional chemical moieties to initiate the stimulus. The review will primarily focus on preclinical and clinical trial approved theranostic agents alone, describing their success or failure in the respective stages.
Collapse
Affiliation(s)
- Sheikh Mohamed M
- Bio-Nano Electronics Research Centre, Toyo University, Kawagoe, 350-8585, Japan; Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe 350-8585, Japan
| | | | - Toru Maekawa
- Bio-Nano Electronics Research Centre, Toyo University, Kawagoe, 350-8585, Japan; Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe 350-8585, Japan.
| | - Sakthi Kumar D
- Bio-Nano Electronics Research Centre, Toyo University, Kawagoe, 350-8585, Japan; Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe 350-8585, Japan.
| |
Collapse
|
48
|
Macháček M, Carter KA, Kostelanský F, Miranda D, Seffouh A, Ortega J, Šimůnek T, Zimčík P, Lovell JF. Binding of an amphiphilic phthalocyanine to pre-formed liposomes confers light-triggered cargo release. J Mater Chem B 2018; 6:7298-7305. [PMID: 30984399 PMCID: PMC6456075 DOI: 10.1039/c8tb01602j] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Liposomes are able to load a range of cargos and have been used for drug delivery applications, including for stimuli-triggered drug release. Here, we describe an approach for imparting near infrared (NIR) light-triggered release to pre-formed liposomes, using a newly-synthesized cationic, amphiphilic phthalocyanine. When simply mixed in aqueous solution with cargo-loaded liposomes, the cationic amphiphilic phthalocyanine, but not a cationic hydrophilic azaphthalocyanine, spontaneously incorporates into the liposome bilayer. This enables subsequent release of loaded cargo (doxorubcin or basic orange) upon irradiation with NIR light. The rate of release could be altered by varying the amount of photosensitizer added to the liposomes. In the absence of NIR light exposure, stable cargo loading of the liposomes was maintained. Introduction.
Collapse
Affiliation(s)
- Miloslav Macháček
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
- Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, 500 05 Hradec Králové, Czech Republic
| | - Kevin A Carter
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Filip Kostelanský
- Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, 500 05 Hradec Králové, Czech Republic
| | - Dyego Miranda
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Amal Seffouh
- Department of Anatomy and Cell Biology, McGill University Montreal, Quebec, H3A 0C7, Canada
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology, McGill University Montreal, Quebec, H3A 0C7, Canada
| | - Tomáš Šimůnek
- Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, 500 05 Hradec Králové, Czech Republic
| | - Petr Zimčík
- Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, 500 05 Hradec Králové, Czech Republic
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| |
Collapse
|
49
|
Zhou EY, Knox HJ, Reinhardt CJ, Partipilo G, Nilges MJ, Chan J. Near-Infrared Photoactivatable Nitric Oxide Donors with Integrated Photoacoustic Monitoring. J Am Chem Soc 2018; 140:11686-11697. [PMID: 30198716 PMCID: PMC7331458 DOI: 10.1021/jacs.8b05514] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Photoacoustic (PA) tomography is a noninvasive technology that utilizes near-infrared (NIR) excitation and ultrasonic detection to image biological tissue at centimeter depths. While several activatable small-molecule PA sensors have been developed for various analytes, the use of PA molecules for deep-tissue analyte delivery and monitoring remains an underexplored area of research. Herein, we describe the synthesis, characterization, and in vivo validation of photoNOD-1 and photoNOD-2, the first organic, NIR-photocontrolled nitric oxide (NO) donors that incorporate a PA readout of analyte release. These molecules consist of an aza-BODIPY dye appended with an aryl N-nitrosamine NO-donating moiety. The photoNODs exhibit chemostability to various biological stimuli, including redox-active metals and CYP450 enzymes, and demonstrate negligible cytotoxicity in the absence of irradiation. Upon single-photon NIR irradiation, photoNOD-1 and photoNOD-2 release NO as well as rNOD-1 or rNOD-2, PA-active products that enable ratiometric monitoring of NO release. Our in vitro studies show that, upon irradiation, photoNOD-1 and photoNOD-2 exhibit 46.6-fold and 21.5-fold ratiometric turn-ons, respectively. Moreover, unlike existing NIR NO donors, the photoNODs do not require encapsulation or multiphoton activation for use in live animals. In this study, we use PA tomography to monitor the local, irradiation-dependent release of NO from photoNOD-1 and photoNOD-2 in mice after subcutaneous treatment. In addition, we use a murine model for breast cancer to show that photoNOD-1 can selectively affect tumor growth rates in the presence of NIR light stimulation following systemic administration.
Collapse
Affiliation(s)
- Effie Y. Zhou
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| | - Hailey J. Knox
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| | - Christopher J. Reinhardt
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| | - Gina Partipilo
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| | - Mark J. Nilges
- Illinois EPR Research Center, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, United States
| | - Jefferson Chan
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, Urbana, Illinois 61801, United States
| |
Collapse
|
50
|
Mitra K, Lyons CE, Hartman MCT. A Platinum(II) Complex of Heptamethine Cyanine for Photoenhanced Cytotoxicity and Cellular Imaging in Near-IR Light. Angew Chem Int Ed Engl 2018; 57:10263-10267. [PMID: 29939482 PMCID: PMC6548462 DOI: 10.1002/anie.201806911] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Indexed: 12/28/2022]
Abstract
Controlled generation of cytotoxic agents with near-IR light is a current focus of photoactivated cancer therapy, including that involving cytotoxic platinum species. A heptamethine cyanine scaffolded PtII complex, IR797-Platin exhibits unprecedented Pt-O bond scission and enhancement in DNA platination in near-IR light. This complex also displayed significant singlet oxygen quantum yield thereby qualifying as a near-IR photodynamic therapeutic agent. The complex showed 30-60 fold enhancement of cytotoxicity in near-IR light in various cancer cell lines. The cellular imaging properties were also leveraged to observe its significant co-localization in cytoplasmic organelles. This is the first demonstration of a near-IR light-initiated therapy involving the cytotoxic effects of both active cisplatin and singlet oxygen.
Collapse
Affiliation(s)
- Koushambi Mitra
- Department of Chemistry, Virginia Commonwealth University, 1001 West Main Street, P. O. Box 842006, Richmond, VA 23284, USA.
- Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, VA 23298, USA
| | - Charles E. Lyons
- Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, VA 23298, USA
| | - Matthew C. T. Hartman
- Department of Chemistry, Virginia Commonwealth University, 1001 West Main Street, P. O. Box 842006, Richmond, VA 23284, USA.
- Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, VA 23298, USA
| |
Collapse
|