1
|
Zhang J, Liu Z, Zhang Z, Yang H, Wang H, Yang Z, Xu Y, Li S, Yang D. Recent Advances in Silica-Based Nanomaterials for Enhanced Tumor Imaging and Therapy. ACS APPLIED BIO MATERIALS 2024. [PMID: 39495482 DOI: 10.1021/acsabm.4c01318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Cancer remains a formidable challenge, inflicting profound physical, psychological, and financial burdens on patients. In this context, silica-based nanomaterials have garnered significant attention for their potential in tumor imaging and therapy owing to their exceptional properties, such as biocompatibility, customizable porosity, and versatile functionalization capabilities. This review meticulously examines the latest advancements in the application of silica-based nanomaterials for tumor imaging and therapy. It underscores their potential in enhancing various cancer imaging modalities, including fluorescence imaging, magnetic resonance imaging, computed tomography, positron emission tomography, ultrasound imaging, and multimodal imaging approaches. Moreover, the review delves into their therapeutic efficacy in chemotherapy, radiotherapy, phototherapy, immunotherapy, gas therapy, sonodynamic therapy, chemodynamic therapy, starvation therapy, and gene therapy. Critical evaluations of the biosafety profiles and degradation pathways of these nanomaterials within biological environments are also presented. By discussing the current challenges and prospects, this review aims to provide a nuanced perspective on the clinical translation of silica-based nanomaterials, thereby highlighting their promise in revolutionizing cancer diagnostics, enabling real-time monitoring of therapeutic responses, and advancing personalized medicine.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zilu Liu
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zhijing Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zhenlu Yang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Yunjian Xu
- School of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an 271000, China
- Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Shengke Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| |
Collapse
|
2
|
Liu J, Liu J, Wang Y, Chen F, He Y, Xie X, Zhong Y, Yang C. Bioactive mesoporous silica materials-assisted cancer immunotherapy. Biomaterials 2024; 315:122919. [PMID: 39481339 DOI: 10.1016/j.biomaterials.2024.122919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/12/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Immunotherapy is initially envisioned as a powerful approach to train immune cells within the tumor microenvironment (TME) and lymphoid tissues to elicit strong anti-tumor responses. However, clinical cancer immunotherapy still faces challenges, such as limited immunogenicity and insufficient immune response. Leveraging the advantages of mesoporous silica (MS) materials in controllable drug and immunomodulator release, recent efforts have focused on engineering MS with intrinsic immunoregulatory functions to promote robust, systemic, and safe anti-tumor responses. This review discusses advances in bioactive MS materials that address the challenges of immunotherapy. Beyond their role in on-demand delivery and drug release in response to the TME, we highlight the intrinsic functions of bioactive MS in orchestrating localized immune responses by inducing immunogenic cell death in tumor cells, modulating immune cell activity, and facilitating tumor-immune cell interactions. Additionally, we emphasize the advantages of bioactive MS in recruiting and activating immune cells within lymphoid tissues to initiate anti-tumor vaccination. The review also covers the challenges of MS-assisted immunotherapy, potential solutions, and future outlooks. With a deeper understanding of material-bio interactions, the rational design of MS with sophisticated bioactivities and controllable responsiveness holds great promise for enhancing the outcomes of personalized immunotherapy.
Collapse
Affiliation(s)
- Jiali Liu
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| | - Jiying Liu
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yaxin Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yan He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yiling Zhong
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China.
| | - Chao Yang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China.
| |
Collapse
|
3
|
Avendaño-Godoy J, Cattoën X, Kogan MJ, Morales Valenzuela J. Epigallocatechin-3-gallate adsorbed on core-shell gold nanorod@mesoporous silica nanoparticles, an antioxidant nanomaterial with photothermal properties. Int J Pharm 2024; 662:124507. [PMID: 39048041 DOI: 10.1016/j.ijpharm.2024.124507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/07/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Epigallocatechin-3-gallate (EGCG) exhibits several pharmacological activities with potential benefits for human health, however, it has low oral bioavailability. A promising approach is to transport EGCG in a nanostructured system to protect it until it reaches the site of action and also allow combining chemotherapy with phototherapy to improve its therapeutic efficiency. The aim of this work was to synthesize GNR@mSiO2-NH2/EGCG and characterize the adsorption process, its antioxidant activity, properties and photothermal stability, for its potential use in chemo-photothermal therapy. The nanosystem presented good encapsulation efficiency (19.2 %) and EGCG loading capacity (6.0 %). The DPPH• free radical scavenging capacity (RSA) and chelating activity of the nanosystem was 60.7 ± 6.9 % and 71.0 ± 6.4 % at an EGCG equivalent concentration of 1 µg/mL and 30 µg/mL, respectively. The core-shell NPs presented a good photothermal transduction efficiency of 17 %. EGCG free, as well as its RSA and chelating activity, remained stable after NIR irradiation (808 nm, 7 W/cm2). The morphology of GNR@mSiO2 remained intact after being irradiated with NIR, however, ultrasmall gold NPs could be observed, probably a product of photocracking of GNR. In summary, the nanosystem has good antioxidant activity, photothermal stability, and photothermal transduction ability making it potentially useful for chemo-photothermal therapy.
Collapse
Affiliation(s)
- Javier Avendaño-Godoy
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Chile; Advanced Center of Chronic Diseases (ACCDiS), Chile; Departamento de Ciencias y Tecnología Farmacéuticas, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Chile; Université Grenoble Alpes, CNRS, Grenoble INP, Intitut Néel, France
| | - Xavier Cattoën
- Université Grenoble Alpes, CNRS, Grenoble INP, Intitut Néel, France
| | - Marcelo J Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Chile; Advanced Center of Chronic Diseases (ACCDiS), Chile.
| | - Javier Morales Valenzuela
- Departamento de Ciencias y Tecnología Farmacéuticas, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Chile.
| |
Collapse
|
4
|
Chen Z, Gezginer I, Zhou Q, Tang L, Deán-Ben XL, Razansky D. Multimodal optoacoustic imaging: methods and contrast materials. Chem Soc Rev 2024; 53:6068-6099. [PMID: 38738633 PMCID: PMC11181994 DOI: 10.1039/d3cs00565h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Indexed: 05/14/2024]
Abstract
Optoacoustic (OA) imaging offers powerful capabilities for interrogating biological tissues with rich optical absorption contrast while maintaining high spatial resolution for deep tissue observations. The spectrally distinct absorption of visible and near-infrared photons by endogenous tissue chromophores facilitates extraction of diverse anatomic, functional, molecular, and metabolic information from living tissues across various scales, from organelles and cells to whole organs and organisms. The primarily blood-related contrast and limited penetration depth of OA imaging have fostered the development of multimodal approaches to fully exploit the unique advantages and complementarity of the method. We review the recent hybridization efforts, including multimodal combinations of OA with ultrasound, fluorescence, optical coherence tomography, Raman scattering microscopy and magnetic resonance imaging as well as ionizing methods, such as X-ray computed tomography, single-photon-emission computed tomography and positron emission tomography. Considering that most molecules absorb light across a broad range of the electromagnetic spectrum, the OA interrogations can be extended to a large number of exogenously administered small molecules, particulate agents, and genetically encoded labels. This unique property further makes contrast moieties used in other imaging modalities amenable for OA sensing.
Collapse
Affiliation(s)
- Zhenyue Chen
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland.
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| | - Irmak Gezginer
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland.
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| | - Quanyu Zhou
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland.
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| | - Lin Tang
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland.
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| | - Xosé Luís Deán-Ben
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland.
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| | - Daniel Razansky
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland.
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| |
Collapse
|
5
|
Ma J, Li Y, Ying Y, Wu B, Liu Y, Zhou J, Hu L. Progress of Mesoporous Silica Coated Gold Nanorods for Biological Imaging and Cancer Therapy. ChemMedChem 2024; 19:e202300374. [PMID: 37990850 DOI: 10.1002/cmdc.202300374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/19/2023] [Accepted: 11/19/2023] [Indexed: 11/23/2023]
Abstract
For unique surface plasmon absorption and fluorescence characteristics, gold nanorods have been developed and widely employed in the biomedical field. However, limitations still exist due their low specific surface area, instability and tendency agglomerate in cytoplasm. Mesoporous silica materials have been broadly applied in field of catalysts, adsorbents, nanoreactors, and drug carriers due to its unique mesoporous structure, highly comparative surface area, good stability and biocompatibility. Therefore, coating gold nanorods with a dendritic mesopore channels can effectively prevent particle agglomeration, while increasing the specific surface area and drug loading efficiency. This review discusses the advancements of GNR@MSN in synthetic process, bio-imaging technique and tumor therapy. Additionally, the further application of GNR@MSN in imaging-guided treatment modalities is explored, while its promising superior application prospect is highlighted. Finally, the issues related to in vivo studies are critically examined for facilitating the transition of this promising nanoplatform into clinical trials.
Collapse
Affiliation(s)
- Jiaying Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, University of South China, Hengyang, 421001, PR China
| | - Yongzhen Li
- Department of Pharmacy, School of Pharmacy, University of South China, Hengyang, 421001, PR China
| | - Yunfei Ying
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, University of South China, Hengyang, 421001, PR China
| | - Baibei Wu
- Department of Clinical Medicine, University of South China, Hengyang, 421001, PR China
| | - Yanmei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, University of South China, Hengyang, 421001, PR China
| | - Juan Zhou
- School of Mechanical Engineering, University of South China, Hengyang, 421001, PR China
| | - Lidan Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, University of South China, Hengyang, 421001, PR China
| |
Collapse
|
6
|
Cai B, Huang L, Zhou X, Zhou X, Lei K, Han M, Zhang Z, Li X, Li G. Black phosphorus-incorporated novel Ti-12Mo-10Zr implant for multimodal treatment of osteosarcoma. Biometals 2024; 37:131-142. [PMID: 37682402 DOI: 10.1007/s10534-023-00533-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/27/2023] [Indexed: 09/09/2023]
Abstract
The repair and reconstruction of large bone defects after bone tumor resection is still a great clinical challenge. At present, orthopedic implant reconstruction is the mainstream treatment for repairing bone defects. However, according to clinical feedback, local tumor recurrence and nonunion of bone graft are common reasons leading to the failure of bone defect repair and reconstruction after bone tumor resection, which seriously threaten the physical and mental health of patients. On this basis, here the self-developed low modulus Ti-12Mo-10Zr alloy (TMZ) was chosen as substrate material. To improve its biological activity and osteointegration, calcium, oxygen, and phosphorus co-doped microporous coating was prepared on TMZ alloy by microarc oxidation (MAO). Then, black phosphorus (BP) nanosheets were incorporated onto MAO treated TMZ alloy to obtain multifunctional composites. The obtained BP-MAO-TMZ implant exhibited excellent photothermal effects and effective ablation of osteosarcoma cancer cells under the irradiation of 808 nm near infrared laser, while no photothermal or therapeutic effects were observed for TMZ alloy. Meanwhile, the structure/component bionic coating obtained after MAO treatment as well as the P-driven in situ biomineralization performance after incorporation of BP nanosheets endowed BP-MAO-TMZ implant with synergistic promoting effect on MC3T3-E1 osteoblasts' activity, proliferation and differentiation ability. This study is expected to provide effective clinical solutions for problems of difficult bone regeneration and tumor recurrence after tumor resection in patients with bone tumors and to solve a series of medical problems such as poor prognosis and poor postoperative quality of patients life with malignant bone tumors.
Collapse
Affiliation(s)
- Bianyun Cai
- College of Medical Technology and Engineering, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Leizhen Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xueke Zhou
- College of Medical Technology and Engineering, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Xuan Zhou
- College of Medical Technology and Engineering, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Kun Lei
- College of Medical Technology and Engineering, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Meng Han
- College of Medical Technology and Engineering, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Zilin Zhang
- College of Medical Technology and Engineering, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Xiaofang Li
- College of Medical Technology and Engineering, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Guangda Li
- College of Medical Technology and Engineering, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China.
| |
Collapse
|
7
|
Song M, Aipire A, Dilxat E, Li J, Xia G, Jiang Z, Fan Z, Li J. Research Progress of Polysaccharide-Gold Nanocomplexes in Drug Delivery. Pharmaceutics 2024; 16:88. [PMID: 38258099 PMCID: PMC10820823 DOI: 10.3390/pharmaceutics16010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Clinical drug administration aims to deliver drugs efficiently and safely to target tissues, organs, and cells, with the objective of enabling their therapeutic effects. Currently, the main approach to enhance a drug's effectiveness is ensuring its efficient delivery to the intended site. Due to the fact that there are still various drawbacks of traditional drug delivery methods, such as high toxicity and side effects, insufficient drug specificity, poor targeting, and poor pharmacokinetic performance, nanocarriers have emerged as a promising alternative. Nanocarriers possess significant advantages in drug delivery due to their size tunability and surface modifiability. Moreover, nano-drug delivery systems have demonstrated strong potential in terms of prolonging drug circulation time, improving bioavailability, increasing drug retention at the tumor site, decreasing drug resistance, as well as reducing the undesirable side effects of anticancer drugs. Numerous studies have focused on utilizing polysaccharides as nanodelivery carriers, developing delivery systems based on polysaccharides, or exploiting polysaccharides as tumor-targeting ligands to enhance the precision of nanoparticle delivery. These types of investigations have become commonplace in the academic literature. This review aims to elucidate the preparation methods and principles of polysaccharide gold nanocarriers. It also provides an overview of the factors that affect the loading of polysaccharide gold nanocarriers with different kinds of drugs. Additionally, it outlines the strategies employed by polysaccharide gold nanocarriers to improve the delivery efficiency of various drugs. The objective is to provide a reference for further development of research on polysaccharide gold nanodelivery systems.
Collapse
Affiliation(s)
- Ming Song
- Institute of Materia Medica & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; (M.S.); (A.A.); (E.D.); (J.L.); (G.X.)
| | - Adila Aipire
- Institute of Materia Medica & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; (M.S.); (A.A.); (E.D.); (J.L.); (G.X.)
| | - Elzira Dilxat
- Institute of Materia Medica & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; (M.S.); (A.A.); (E.D.); (J.L.); (G.X.)
| | - Jianmin Li
- Institute of Materia Medica & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; (M.S.); (A.A.); (E.D.); (J.L.); (G.X.)
| | - Guoyu Xia
- Institute of Materia Medica & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; (M.S.); (A.A.); (E.D.); (J.L.); (G.X.)
| | - Ziwen Jiang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China;
| | - Zhongxiong Fan
- Institute of Materia Medica & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; (M.S.); (A.A.); (E.D.); (J.L.); (G.X.)
| | - Jinyao Li
- Institute of Materia Medica & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; (M.S.); (A.A.); (E.D.); (J.L.); (G.X.)
| |
Collapse
|
8
|
Liu H, Huang Z, Chen H, Zhang Y, Yu P, Hu P, Zhang X, Cao J, Zhou T. A potential strategy against clinical carbapenem-resistant Enterobacteriaceae: antimicrobial activity study of sweetener-decorated gold nanoparticles in vitro and in vivo. J Nanobiotechnology 2023; 21:409. [PMID: 37932843 PMCID: PMC10626710 DOI: 10.1186/s12951-023-02149-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Carbapenem-resistant Enterobacteriaceae (CRE) present substantial challenges to clinical intervention, necessitating the formulation of novel antimicrobial strategies to counteract them. Nanomaterials offer a distinctive avenue for eradicating bacteria by employing mechanisms divergent from traditional antibiotic resistance pathways and exhibiting reduced susceptibility to drug resistance development. Non-caloric artificial sweeteners, commonly utilized in the food sector, such as saccharin, sucralose, acesulfame, and aspartame, possess structures amenable to nanomaterial formation. In this investigation, we synthesized gold nanoparticles decorated with non-caloric artificial sweeteners and evaluated their antimicrobial efficacy against clinical CRE strains. RESULTS Among these, gold nanoparticles decorated with aspartame (ASP_Au NPs) exhibited the most potent antimicrobial effect, displaying minimum inhibitory concentrations ranging from 4 to 16 µg/mL. As a result, ASP_Au NPs were chosen for further experimentation. Elucidation of the antimicrobial mechanism unveiled that ASP_Au NPs substantially elevated bacterial reactive oxygen species (ROS) levels, which dissipated upon ROS scavenger treatment, indicating ROS accumulation within bacteria as the fundamental antimicrobial modality. Furthermore, findings from membrane permeability assessments suggested that ASP_Au NPs may represent a secondary antimicrobial modality via enhancing inner membrane permeability. In addition, experiments involving crystal violet and confocal live/dead staining demonstrated effective suppression of bacterial biofilm formation by ASP_Au NPs. Moreover, ASP_Au NPs demonstrated notable efficacy in the treatment of Galleria mellonella bacterial infection and acute abdominal infection in mice, concurrently mitigating the organism's inflammatory response. Crucially, evaluation of in vivo safety and biocompatibility established that ASP_Au NPs exhibited negligible toxicity at bactericidal concentrations. CONCLUSIONS Our results demonstrated that ASP_Au NPs exhibit promise as innovative antimicrobial agents against clinical CRE.
Collapse
Affiliation(s)
- Haifeng Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Zeyu Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Huanchang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Ying Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Pingting Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Panjie Hu
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaotuan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Jianming Cao
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China.
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
9
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|
10
|
Fernandes DA. Liposomes for Cancer Theranostics. Pharmaceutics 2023; 15:2448. [PMID: 37896208 PMCID: PMC10610083 DOI: 10.3390/pharmaceutics15102448] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/16/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is one of the most well-studied diseases and there have been significant advancements over the last few decades in understanding its molecular and cellular mechanisms. Although the current treatments (e.g., chemotherapy, radiotherapy, gene therapy and immunotherapy) have provided complete cancer remission for many patients, cancer still remains one of the most common causes of death in the world. The main reasons for the poor response rates for different cancers include the lack of drug specificity, drug resistance and toxic side effects (i.e., in healthy tissues). For addressing the limitations of conventional cancer treatments, nanotechnology has shown to be an important field for constructing different nanoparticles for destroying cancer cells. Due to their size (i.e., less than 1 μm), nanoparticles can deliver significant amounts of cancer drugs to tumors and are able to carry moieties (e.g., folate, peptides) for targeting specific types of cancer cells (i.e., through receptor-mediated endocytosis). Liposomes, composed of phospholipids and an interior aqueous core, can be used as specialized delivery vehicles as they can load different types of cancer therapy agents (e.g., drugs, photosensitizers, genetic material). In addition, the ability to load imaging agents (e.g., fluorophores, radioisotopes, MRI contrast media) enable these nanoparticles to be used for monitoring the progress of treatment. This review examines a wide variety of different liposomes for cancer theranostics, with the different available treatments (e.g., photothermal, photodynamic) and imaging modalities discussed for different cancers.
Collapse
|
11
|
Kumar PPP, Lim DK. Photothermal Effect of Gold Nanoparticles as a Nanomedicine for Diagnosis and Therapeutics. Pharmaceutics 2023; 15:2349. [PMID: 37765317 PMCID: PMC10534847 DOI: 10.3390/pharmaceutics15092349] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Gold nanoparticles (AuNPs) have received great attention for various medical applications due to their unique physicochemical properties. AuNPs with tunable optical properties in the visible and near-infrared regions have been utilized in a variety of applications such as in vitro diagnostics, in vivo imaging, and therapeutics. Among the applications, this review will pay more attention to recent developments in diagnostic and therapeutic applications based on the photothermal (PT) effect of AuNPs. In particular, the PT effect of AuNPs has played an important role in medical applications utilizing light, such as photoacoustic imaging, photon polymerase chain reaction (PCR), and hyperthermia therapy. First, we discuss the fundamentals of the optical properties in detail to understand the background of the PT effect of AuNPs. For diagnostic applications, the ability of AuNPs to efficiently convert absorbed light energy into heat to generate enhanced acoustic waves can lead to significant enhancements in photoacoustic signal intensity. Integration of the PT effect of AuNPs with PCR may open new opportunities for technological innovation called photonic PCR, where light is used to enable fast and accurate temperature cycling for DNA amplification. Additionally, beyond the existing thermotherapy of AuNPs, the PT effect of AuNPs can be further applied to cancer immunotherapy. Controlled PT damage to cancer cells triggers an immune response, which is useful for obtaining better outcomes in combination with immune checkpoint inhibitors or vaccines. Therefore, this review examines applications to nanomedicine based on the PT effect among the unique optical properties of AuNPs, understands the basic principles, the advantages and disadvantages of each technology, and understands the importance of a multidisciplinary approach. Based on this, it is expected that it will help understand the current status and development direction of new nanoparticle-based disease diagnosis methods and treatment methods, and we hope that it will inspire the development of new innovative technologies.
Collapse
Affiliation(s)
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea;
- Department of Integrative Energy Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
12
|
Tang S, Li R, Luo T, Huang T, Lu X, Wu X, Dong Y, Wu C, Xu K, Wang Y. Preparation of Gd-doped AuNBP@mSiO 2 nanocomposites for the MR imaging, drug delivery and chemo-photothermal synergistic killing of breast cancer cells. RSC Adv 2023; 13:23976-23983. [PMID: 37577100 PMCID: PMC10413050 DOI: 10.1039/d3ra03753c] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/05/2023] [Indexed: 08/15/2023] Open
Abstract
Under near-infrared (NIR) light, gold nanobipyramids (AuNBPs) exhibit a high photothermal conversion rate and photothermal stability, making them ideal mediators for photothermal therapy (PTT). In this study, highly purified AuNBPs are prepared, followed by coating their surfaces with mesoporous silica (mSiO2). The obtained AuNBP@mSiO2 nanocomplex exhibits an ellipsoidal shape with a relatively large specific surface, pore diameter and pore volume. To achieve MRI guided chemo-photothermal therapy of breast cancer cells, the nanocomplex is further coupled with the MRI contrast agent Gd-DTTA and the chemotherapeutic drug doxorubicin (DOX). The results indicated that under NIR light irradiation, AuNBPs exhibited promising PTT effects, while the cumulative release rate of DOX was significantly enhanced to 81.40%. Moreover, the chemo-photothermal therapy approach effectively eradicated 4T1 breast cancer cells. This work successfully confirms that chemo-photothermal synergistic therapy is an effective tumor treatment strategy and demonstrates the potential application of AuNBP@mSiO2 as a nano-drug delivery platform. Additionally, it introduces new ideas for the integrated study of breast cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Shiyi Tang
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
| | - Ruohan Li
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University Xuzhou Jiangsu 221002 China
| | - Tao Luo
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
| | - Tianhao Huang
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
| | - Xiaotong Lu
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
| | - Xinyao Wu
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
| | - Yulin Dong
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
| | - Changyu Wu
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
| | - Kai Xu
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University Xuzhou Jiangsu 221002 China
| | - Yong Wang
- School of Medical Imaging, Xuzhou Medical University Xuzhou Jiangsu 221004 China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University Xuzhou Jiangsu 221002 China
| |
Collapse
|
13
|
Xu C, Dobson HE, Yu M, Gong W, Sun X, Park KS, Kennedy A, Zhou X, Xu J, Xu Y, Tai AW, Lei YL, Moon JJ. STING agonist-loaded mesoporous manganese-silica nanoparticles for vaccine applications. J Control Release 2023; 357:84-93. [PMID: 36948420 PMCID: PMC10164691 DOI: 10.1016/j.jconrel.2023.03.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/25/2023] [Accepted: 03/19/2023] [Indexed: 03/24/2023]
Abstract
Cyclic dinucleotides (CDNs), as one type of Stimulator of Interferon Genes (STING) pathway agonist, have shown promising results for eliciting immune responses against cancer and viral infection. However, the suboptimal drug-like properties of conventional CDNs, including their short in vivo half-life and poor cellular permeability, compromise their therapeutic efficacy. In this study, we have developed a manganese-silica nanoplatform (MnOx@HMSN) that enhances the adjuvant effects of CDN by achieving synergy with Mn2+ for vaccination against cancer and SARS-CoV-2. MnOx@HMSN with large mesopores were efficiently co-loaded with CDN and peptide/protein antigens. MnOx@HMSN(CDA) amplified the activation of the STING pathway and enhanced the production of type-I interferons and other proinflammatory cytokines from dendritic cells. MnOx@HMSN(CDA) carrying cancer neoantigens elicited robust antitumor T-cell immunity with therapeutic efficacy in two different murine tumor models. Furthermore, MnOx@HMSN(CDA) loaded with SARS-CoV-2 antigen achieved strong and durable (up to one year) humoral immune responses with neutralizing capability. These results demonstrate that MnOx@HMSN(CDA) is a versatile nanoplatform for vaccine applications.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Pharmaceutical Science, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hannah E Dobson
- Department of Pharmaceutical Science, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mengjie Yu
- Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Wang Gong
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoqi Sun
- Department of Pharmaceutical Science, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kyung Soo Park
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Andrew Kennedy
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Xingwu Zhou
- Department of Pharmaceutical Science, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jin Xu
- Department of Pharmaceutical Science, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yao Xu
- Department of Pharmaceutical Science, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrew W Tai
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yu Leo Lei
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Science, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Hsu JC, Tang Z, Eremina OE, Sofias AM, Lammers T, Lovell JF, Zavaleta C, Cai W, Cormode DP. Nanomaterial-based contrast agents. NATURE REVIEWS. METHODS PRIMERS 2023; 3:30. [PMID: 38130699 PMCID: PMC10732545 DOI: 10.1038/s43586-023-00211-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 12/23/2023]
Abstract
Medical imaging, which empowers the detection of physiological and pathological processes within living subjects, has a vital role in both preclinical and clinical diagnostics. Contrast agents are often needed to accompany anatomical data with functional information or to provide phenotyping of the disease in question. Many newly emerging contrast agents are based on nanomaterials as their high payloads, unique physicochemical properties, improved sensitivity and multimodality capacity are highly desired for many advanced forms of bioimaging techniques and applications. Here, we review the developments in the field of nanomaterial-based contrast agents. We outline important nanomaterial design considerations and discuss the effect on their physicochemical attributes, contrast properties and biological behaviour. We also describe commonly used approaches for formulating, functionalizing and characterizing these nanomaterials. Key applications are highlighted by categorizing nanomaterials on the basis of their X-ray, magnetic, nuclear, optical and/or photoacoustic contrast properties. Finally, we offer our perspectives on current challenges and emerging research topics as well as expectations for future advancements in the field.
Collapse
Affiliation(s)
- Jessica C. Hsu
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhongmin Tang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Olga E. Eremina
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Cristina Zavaleta
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - David P. Cormode
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
15
|
Bordeianu G, Filip N, Cernomaz A, Veliceasa B, Hurjui LL, Pinzariu AC, Pertea M, Clim A, Marinca MV, Serban IL. The Usefulness of Nanotechnology in Improving the Prognosis of Lung Cancer. Biomedicines 2023; 11:biomedicines11030705. [PMID: 36979684 PMCID: PMC10045176 DOI: 10.3390/biomedicines11030705] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Lung cancer remains a major public health problem both in terms of incidence and specific mortality despite recent developments in terms of prevention, such as smoking reduction policies and clinical management advances. Better lung cancer prognosis could be achieved by early and accurate diagnosis and improved therapeutic interventions. Nanotechnology is a dynamic and fast-developing field; various medical applications have been developed and deployed, and more exist as proofs of concepts or experimental models. We aim to summarize current knowledge relevant to the use of nanotechnology in lung cancer management. Starting from the chemical structure-based classification of nanoparticles, we identify and review various practical implementations roughly organized as diagnostic or therapeutic in scope, ranging from innovative contrast agents to targeted drug carriers. Available data are presented starting with standards of practice and moving to highly experimental methods and proofs of concept; particularities, advantages, limits and future directions are explored, focusing on the potential impact on lung cancer clinical prognosis.
Collapse
Affiliation(s)
- Gabriela Bordeianu
- Department of Morpho-Functional Sciences (II), Discipline of Biochemistry, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Nina Filip
- Department of Morpho-Functional Sciences (II), Discipline of Biochemistry, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (N.F.); (A.C.)
| | - Andrei Cernomaz
- III-rd Medical Department, Discipline of Pneumology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (N.F.); (A.C.)
| | - Bogdan Veliceasa
- Department of Orthopedics and Traumatology, Surgical Science (II), Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Loredana Liliana Hurjui
- Department of Morpho-Functional Sciences (II), Discipline of Physiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Alin Constantin Pinzariu
- Department of Morpho-Functional Sciences (II), Discipline of Physiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihaela Pertea
- Department of Plastic Surgery and Reconstructive Microsurgery, “Sf. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Andreea Clim
- Department of Morpho-Functional Sciences (II), Discipline of Physiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihai Vasile Marinca
- III-rd Medical Department, Discipline of Oncology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ionela Lacramioara Serban
- Department of Morpho-Functional Sciences (II), Discipline of Physiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
16
|
Yin B, Ho WKH, Xia X, Chan CKW, Zhang Q, Ng YM, Lam CYK, Cheung JCW, Wang J, Yang M, Wong SHD. A Multilayered Mesoporous Gold Nanoarchitecture for Ultraeffective Near-Infrared Light-Controlled Chemo/Photothermal Therapy for Cancer Guided by SERS Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206762. [PMID: 36593512 DOI: 10.1002/smll.202206762] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/10/2022] [Indexed: 06/17/2023]
Abstract
Surface-enhanced Raman scattering (SERS) imaging has emerged as a promising tool for guided cancer diagnosis and synergistic therapies, such as combined chemotherapy and photothermal therapy (chemo-PTT). Yet, existing therapeutic agents often suffer from low SERS sensitivity, insufficient photothermal conversion, or/and limited drug loading capacity. Herein, a multifunctional theragnostic nanoplatform consisting of mesoporous silica-coated gold nanostar with a cyclic Arg-Gly-Asp (RGD)-coated gold nanocluster shell (named RGD-pAS@AuNC) is reported that exhibits multiple "hot spots" for pronouncedly enhanced SERS signals and improved near-infrared (NIR)-induced photothermal conversion efficiency (85.5%), with a large capacity for high doxorubicin (DOX) loading efficiency (34.1%, named RGD/DOX-pAS@AuNC) and effective NIR-triggered DOX release. This nanoplatform shows excellent performance in xenograft tumor model of HeLa cell targeting, negligible cytotoxicity, and good stability both in vitro and in vivo. By SERS imaging, the optimal temporal distribution of injected RGD/DOX-pAS@AuNCs at the tumor site is identified for NIR-triggered local chemo-PTT toward the tumor, achieving ultraeffective therapy in tumor cells and tumor-bearing mouse model with 5 min of NIR irradiation (0.5 W cm-2 ). This work offers a promising approach to employing SERS imaging for effective noninvasive tumor treatment by on-site triggered chemo-PTT.
Collapse
Affiliation(s)
- Bohan Yin
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
| | - Willis Kwun Hei Ho
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
| | - Xinyue Xia
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong, 999077, China
| | - Cecilia Ka Wing Chan
- Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong, 999077, China
| | - Qin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
| | - Yip Ming Ng
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
| | - Ching Ying Katherine Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
| | - James Chung Wai Cheung
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
| | - Jianfang Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong, 999077, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
17
|
Haemmerich D, Ramajayam KK, Newton DA. Review of the Delivery Kinetics of Thermosensitive Liposomes. Cancers (Basel) 2023; 15:cancers15020398. [PMID: 36672347 PMCID: PMC9856714 DOI: 10.3390/cancers15020398] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
Thermosensitive liposomes (TSL) are triggered nanoparticles that release the encapsulated drug in response to hyperthermia. Combined with localized hyperthermia, TSL enabled loco-regional drug delivery to tumors with reduced systemic toxicities. More recent TSL formulations are based on intravascular triggered release, where drug release occurs within the microvasculature. Thus, this delivery strategy does not require enhanced permeability and retention (EPR). Compared to traditional nanoparticle drug delivery systems based on EPR with passive or active tumor targeting (typically <5%ID/g tumor), TSL can achieve superior tumor drug uptake (>10%ID/g tumor). Numerous TSL formulations have been combined with various drugs and hyperthermia devices in preclinical and clinical studies over the last four decades. Here, we review how the properties of TSL dictate delivery and discuss the advantages of rapid drug release from TSL. We show the benefits of selecting a drug with rapid extraction by tissue, and with quick cellular uptake. Furthermore, the optimal characteristics of hyperthermia devices are reviewed, and impact of tumor biology and cancer cell characteristics are discussed. Thus, this review provides guidelines on how to improve drug delivery with TSL by optimizing the combination of TSL, drug, and hyperthermia method. Many of the concepts discussed are applicable to a variety of other triggered drug delivery systems.
Collapse
Affiliation(s)
- Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
- Correspondence:
| | - Krishna K. Ramajayam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Danforth A. Newton
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
18
|
Florensa M, Llenas M, Medina-Gutiérrez E, Sandoval S, Tobías-Rossell G. Key Parameters for the Rational Design, Synthesis, and Functionalization of Biocompatible Mesoporous Silica Nanoparticles. Pharmaceutics 2022; 14:2703. [PMID: 36559195 PMCID: PMC9788600 DOI: 10.3390/pharmaceutics14122703] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/11/2022] Open
Abstract
Over the last few years, research on silica nanoparticles has rapidly increased. Particularly on mesoporous silica nanoparticles (MSNs), as nanocarriers for the treatment of various diseases because of their physicochemical properties and biocompatibility. The use of MSNs combined with therapeutic agents can provide better encapsulation and effective delivery. MSNs as nanocarriers might also be a promising tool to lower the therapeutic dosage levels and thereby to reduce undesired side effects. Researchers have explored several routes to conjugate both imaging and therapeutic agents onto MSNs, thus expanding their potential as theranostic platforms, in order to allow for the early diagnosis and treatment of diseases. This review introduces a general overview of recent advances in the field of silica nanoparticles. In particular, the review tackles the fundamental aspects of silicate materials, including a historical presentation to new silicates and then focusing on the key parameters that govern the tailored synthesis of functional MSNs. Finally, the biomedical applications of MSNs are briefly revised, along with their biocompatibility, biodistribution and degradation. This review aims to provide the reader with the tools for a rational design of biocompatible MSNs for their application in the biomedical field. Particular attention is paid to the role that the synthesis conditions have on the physicochemical properties of the resulting MSNs, which, in turn, will determine their pharmacological behavior. Several recent examples are highlighted to stress the potential that MSNs hold as drug delivery systems, for biomedical imaging, as vaccine adjuvants and as theragnostic agents.
Collapse
Affiliation(s)
| | | | | | - Stefania Sandoval
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Barcelona, Spain
| | - Gerard Tobías-Rossell
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Barcelona, Spain
| |
Collapse
|
19
|
Zeng Y, Dou T, Ma L, Ma J. Biomedical Photoacoustic Imaging for Molecular Detection and Disease Diagnosis: "Always-On" and "Turn-On" Probes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202384. [PMID: 35773244 PMCID: PMC9443455 DOI: 10.1002/advs.202202384] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/25/2022] [Indexed: 05/05/2023]
Abstract
Photoacoustic (PA) imaging is a nonionizing, noninvasive imaging technique that combines optical and ultrasonic imaging modalities to provide images with excellent contrast, spatial resolution, and penetration depth. Exogenous PA contrast agents are created to increase the sensitivity and specificity of PA imaging and to offer diagnostic information for illnesses. The existing PA contrast agents are categorized into two groups in this review: "always-on" and "turn-on," based on their ability to be triggered by target molecules. The present state of these probes, their merits and limitations, and their future development, is explored.
Collapse
Affiliation(s)
- Yun Zeng
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi Province, 710126, P. R. China
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment and Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi Province, 7100126, P. R. China
| | - Taotao Dou
- Neurosurgery Department, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710054, P. R. China
| | - Lei Ma
- Vascular Intervention Department, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710054, P. R. China
| | - Jingwen Ma
- Radiology Department, CT and MRI Room, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710054, P. R. China
| |
Collapse
|
20
|
Chang Y, Wang Q, Xu W, Huang X, Xu X, Han FY, Qiao R, Ediriweera GR, Peng H, Fu C, Liu K, Whittaker AK. Low-Fouling Gold Nanorod Theranostic Agents Enabled by a Sulfoxide Polymer Coating. Biomacromolecules 2022; 23:3866-3874. [PMID: 35977724 DOI: 10.1021/acs.biomac.2c00696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Gold nanorods (GNRs) are widely used in various biomedical applications such as disease imaging and therapy due to their unique plasmonic properties. To improve their bioavailability, GNRs often need to be coated with hydrophilic polymers so as to impart stealth properties. Poly(ethylene glycol) (PEG) has been long used as such a coating material for GNRs. However, there is increasing acknowledgement that the amphiphilic nature of PEG facilitates its interaction with protein molecules, leading to immune recognition and consequent side effects. This has motivated the search for new classes of low-fouling polymers with high hydrophilicity as alternative low-fouling surface coating materials for GNRs. Herein, we report the synthesis, characterization, and application of GNRs coated with highly hydrophilic sulfoxide-containing polymers. We investigated the effect of the sulfoxide polymer coating on the cellular uptake and in vivo circulation time of the GNRs and compared these properties with pegylated GNR counterparts. The photothermal effect and photoacoustic imaging of these polymer-coated GNRs were also explored, and the results show that these GNRs are promising as nanotheranostic particles for the treatment of cancer.
Collapse
Affiliation(s)
- Yixin Chang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Qiaoyun Wang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Weizhi Xu
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xumin Huang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Xin Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Felicity Y Han
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Ruirui Qiao
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Gayathri R Ediriweera
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Hui Peng
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Changkui Fu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kun Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
21
|
Sargazi S, Laraib U, Barani M, Rahdar A, Fatima I, Bilal M, Pandey S, Sharma RK, Kyzas GZ. Recent trends in mesoporous silica nanoparticles of rode-like morphology for cancer theranostics: A review. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
22
|
Xu X, Li T, Jin K. Bioinspired and Biomimetic Nanomedicines for Targeted Cancer Therapy. Pharmaceutics 2022; 14:1109. [PMID: 35631695 PMCID: PMC9147382 DOI: 10.3390/pharmaceutics14051109] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
Undesirable side effects and multidrug resistance are the major obstacles in conventional chemotherapy towards cancers. Nanomedicines provide alternative strategies for tumor-targeted therapy due to their inherent properties, such as nanoscale size and tunable surface features. However, the applications of nanomedicines are hampered in vivo due to intrinsic disadvantages, such as poor abilities to cross biological barriers and unexpected off-target effects. Fortunately, biomimetic nanomedicines are emerging as promising therapeutics to maximize anti-tumor efficacy with minimal adverse effects due to their good biocompatibility and high accumulation abilities. These bioengineered agents incorporate both the physicochemical properties of diverse functional materials and the advantages of biological materials to achieve desired purposes, such as prolonged circulation time, specific targeting of tumor cells, and immune modulation. Among biological materials, mammalian cells (such as red blood cells, macrophages, monocytes, and neutrophils) and pathogens (such as viruses, bacteria, and fungi) are the functional components most often used to confer synthetic nanoparticles with the complex functionalities necessary for effective nano-biointeractions. In this review, we focus on recent advances in the development of bioinspired and biomimetic nanomedicines (such as mammalian cell-based drug delivery systems and pathogen-based nanoparticles) for targeted cancer therapy. We also discuss the biological influences and limitations of synthetic materials on the therapeutic effects and targeted efficacies of various nanomedicines.
Collapse
Affiliation(s)
- Xiaoqiu Xu
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tong Li
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ke Jin
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
Wang Y, Bai H, Miao Y, Weng J, Huang Z, Fu J, Zhang Y, Lin J, Ye D. Tailoring a Near‐Infrared Macrocyclization Scaffold Allows the Control of In Situ Self‐Assembly for Photoacoustic/PET Bimodal Imaging. Angew Chem Int Ed Engl 2022; 61:e202200369. [DOI: 10.1002/anie.202200369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Yuqi Wang
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering Chemistry and Biomedicine Innovation Center (ChemBIC) Nanjing University Nanjing 210023 China
| | - He Bai
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering Chemistry and Biomedicine Innovation Center (ChemBIC) Nanjing University Nanjing 210023 China
| | - Yinxing Miao
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering Chemistry and Biomedicine Innovation Center (ChemBIC) Nanjing University Nanjing 210023 China
| | - Jianhui Weng
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering Chemistry and Biomedicine Innovation Center (ChemBIC) Nanjing University Nanjing 210023 China
| | - Zheng Huang
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering Chemistry and Biomedicine Innovation Center (ChemBIC) Nanjing University Nanjing 210023 China
| | - Jiayu Fu
- NHC Key Laboratory of Nuclear Medicine Jiangsu Key Laboratory of Molecular Nuclear Medicine Jiangsu Institute of Nuclear Medicine Wuxi 214063 China
| | - Yan Zhang
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering Chemistry and Biomedicine Innovation Center (ChemBIC) Nanjing University Nanjing 210023 China
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine Jiangsu Key Laboratory of Molecular Nuclear Medicine Jiangsu Institute of Nuclear Medicine Wuxi 214063 China
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Science School of Chemistry and Chemical Engineering Chemistry and Biomedicine Innovation Center (ChemBIC) Nanjing University Nanjing 210023 China
| |
Collapse
|
24
|
Chen Z, Zeng Y, Chen N, Zhang M, Wang Y, Pan Z, Yuan J, Ye Z, Li X, Bian W, Li H, Zhang K, He Y, Liu X. A Facile and Universal Method for Preparing Polyethylene Glycol-Metal Hybrid Nanoparticles and Their Application in Tumor Theranostics. Adv Healthc Mater 2022; 11:e2200044. [PMID: 35192244 DOI: 10.1002/adhm.202200044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/29/2022] [Indexed: 12/19/2022]
Abstract
Metal ions are of widespread interest owing to their brilliant biomedical functions. However, a simple and universal nanoplatform designed for assembling a range of functional metal ions has not been explored. In this study, a concept of polyethylene glycol (PEG)-mediated transport of metal ions is proposed. 31 types of PEG-metal hybrid nanoparticles (P-MNPs) are successfully synthesized through anionic ring-opening polymerization (ROP), "thiol-ene" click reaction, and subsequent incorporation with multiple metal ions. Compared with other methods, the facile method proposed in this study can provide a feasible approach to design MNPs (mostly <200 nm) containing different metal ions and thus to explore their potential for cancer theranostics. As a proof-of-concept demonstration, four types P-MNPs, i.e., PEG-metal hybrid copper nanoparticles (PEG-Cu NPs), ruthenium nanoparticles (PEG-Ru NPs), and manganese nanoparticles (PEG-Mn NPs) or gadolinium nanoparticles (PEG-Gd NPs), are proven to be tailored for chemodynamic therapy, photothermal therapy, and magnetic resonance imaging of tumors, respectively. Overall, this study provides several metal ions-based nanomaterials with versatile functions for broad applications in cancer theranostics. Furthermore, it offers a promising tool that can be utilized for processing other metal-based nanoparticles and exploring their potential in the biomedical field.
Collapse
Affiliation(s)
- Zefeng Chen
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Yaoxun Zeng
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Niping Chen
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Mingxia Zhang
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Yakun Wang
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Zhenxing Pan
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Jiongpeng Yuan
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Zhaoyi Ye
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Xiaojing Li
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Wangqing Bian
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Haihong Li
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Kun Zhang
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Yan He
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Xujie Liu
- Conney Laboratory for Anticancer Research School of Biomedical and Pharmaceutical Sciences Guangdong University of Technology Guangzhou Guangdong 510006 P. R. China
| |
Collapse
|
25
|
Yu A, Dai X, Wang Z, Chen H, Guo B, Huang L. Recent Advances of Mesoporous Silica as a Platform for Cancer Immunotherapy. BIOSENSORS 2022; 12:109. [PMID: 35200369 PMCID: PMC8869707 DOI: 10.3390/bios12020109] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 05/06/2023]
Abstract
Immunotherapy is a promising modality of treatment for cancer. Immunotherapy is comprised of systemic and local treatments that induce an immune response, allowing the body to fight back against cancer. Systemic treatments such as cancer vaccines harness antigen presenting cells (APCs) to activate T cells with tumor-associated antigens. Small molecule inhibitors can be employed to inhibit immune checkpoints, disrupting tumor immunosuppression and immune evasion. Despite the current efficacy of immunotherapy, improvements to delivery can be made. Nanomaterials such as mesoporous silica can facilitate the advancement of immunotherapy. Mesoporous silica has high porosity, decent biocompatibility, and simple surface functionalization. Mesoporous silica can be utilized as a versatile carrier of various immunotherapeutic agents. This review gives an introduction on mesoporous silica as a nanomaterial, briefly covering synthesis and biocompatibility, and then an overview of the recent progress made in the application of mesoporous silica to cancer immunotherapy.
Collapse
Affiliation(s)
- Albert Yu
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Xiaoyong Dai
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Zixian Wang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Huaqing Chen
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| | - Bing Guo
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China;
| | - Laiqiang Huang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (A.Y.); (X.D.); (Z.W.); (H.C.)
- Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, State Key Laboratory of Health Sciences and Technology, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
26
|
Wang Y, Bai H, Miao Y, Weng J, Huang Z, Fu J, Zhang Y, Lin J, Ye D. Tailoring a Near‐Infrared Macrocyclization Scaffold Allows the Control of In Situ Self‐assembly for Photoacoustic/PET Bimodal Imaging. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202200369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yuqi Wang
- Nanjing University School of Chemistry and Chemical Engineering CHINA
| | - He Bai
- Nanjing University School of Chemistry and Chemical Engineering CHINA
| | - Yinxing Miao
- Nanjing University School of Chemistry and Chemical Engineering CHINA
| | - Jianhui Weng
- Nanjing University School of Chemistry and Chemical Engineering CHINA
| | - Zheng Huang
- Nanjing University School of Chemistry and Chemical Engineering CHINA
| | - Jiayu Fu
- Jiangsu Institute of Nuclear Medicine Molecular Nuclear Medicine CHINA
| | - Yan Zhang
- Nanjing University School of Chemistry and Chemical Engineering CHINA
| | - Jianguo Lin
- Jiangsu Institute of Nuclear Medicine Molecular Nuclear Medicine CHINA
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Chemistry 163 Xianlin Road, 210023 Nanjing CHINA
| |
Collapse
|
27
|
Wang Q, Qu B, Li J, Liu Y, Dong J, Peng X, Zhang R. Multifunctional MnO 2/Ag 3SbS 3 Nanotheranostic Agent for Single-Laser-Triggered Tumor Synergistic Therapy in the NIR-II Biowindow. ACS APPLIED MATERIALS & INTERFACES 2022; 14:4980-4994. [PMID: 35050589 DOI: 10.1021/acsami.1c21752] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Regulating the level of reactive oxygen species (ROS) in a tumor is an efficient and innovative anticancer strategy. However, the therapeutic efficacy of ROS-based therapies, such as chemodynamic therapy (CDT) and photodynamic therapy (PDT), offers finite outcomes due to the oxygen dependence and limited concentration of hydrogen peroxide (H2O2) and overexpression of glutathione (GSH) within the tumor microenvironment (TME), so a single therapeutic strategy is insufficient to completely eliminate tumors. Therefore, we demonstrated an omnipotent nanoplatform MnO2/Ag3SbS3 (abbreviated as MA) with strong optical absorbance in the NIR-II biowindow and oxygen self-sufficient ROS-mediated ability, which not only relieves tumor hypoxia significantly but also enhances the photothermal therapy (PTT)/PDT/CDT efficacy. By 1064 nm laser irradiation, MnO2/Ag3SbS3 nanoparticles (NPs) reveal a favorable photothermal conversion efficiency of 23.15% and achieve a single-laser-triggered NIR-II PTT/PDT effect, resulting in effective tumor elimination. Once internalized into the tumor, MnO2/Ag3SbS3 NPs will be degraded to Mn2+ and Ag3SbS3. The released Ag3SbS3 NPs as a NIR-II phototherapy agent could be utilized for photoacoustic imaging-guided NIR-II PDT/PTT. Mn2+ could be used as a Fenton-like catalyst to continuously catalyze endogenous H2O2 for generating highly virulent hydroxyl radicals (•OH) for CDT and O2 for PDT, enhancing the efficiency of PDT and CDT, respectively. Meanwhile, Mn2+ realizes magnetic resonance imaging-guided accurate tumor therapy. Moreover, the MnO2/Ag3SbS3 NPs could deplete intracellular GSH in TME to promote oxidative stress of the tumor, further strengthening ROS-mediated antitumor treatment efficacy. Overall, this work presents a distinctive paradigm of TME-responsive PDT/CDT/PTT in the second near-infrared biowindow by depleting GSH and decomposing H2O2 for efficient and precise cancer treatment.
Collapse
Affiliation(s)
- Qian Wang
- General Surgery Department, The Radiology Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan 030032, China
| | - Botao Qu
- General Surgery Department, The Radiology Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan 030032, China
- School of Basic Medical Sciences, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Juan Li
- School of Basic Medical Sciences, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Yuqin Liu
- School of Basic Medical Sciences, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Jie Dong
- General Surgery Department, The Radiology Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan 030032, China
| | - Xiaoyang Peng
- School of Basic Medical Sciences, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- General Surgery Department, The Radiology Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan 030032, China
- School of Basic Medical Sciences, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
28
|
Chemically engineered mesoporous silica nanoparticles-based intelligent delivery systems for theranostic applications in multiple cancerous/non-cancerous diseases. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214309] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
29
|
Dual gate-keeping and reversible on-off switching drug release for anti-cancer therapy with pH- and NIR light-responsive mesoporous silica-coated gold nanorods. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.10.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
30
|
Farooq A, Sabah S, Dhou S, Alsawaftah N, Husseini G. Exogenous Contrast Agents in Photoacoustic Imaging: An In Vivo Review for Tumor Imaging. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:393. [PMID: 35159738 PMCID: PMC8840344 DOI: 10.3390/nano12030393] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022]
Abstract
The field of cancer theranostics has grown rapidly in the past decade and innovative 'biosmart' theranostic materials are being synthesized and studied to combat the fast growth of cancer metastases. While current state-of-the-art oncology imaging techniques have decreased mortality rates, patients still face a diminished quality of life due to treatment. Therefore, improved diagnostics are needed to define in vivo tumor growths on a molecular level to achieve image-guided therapies and tailored dosage needs. This review summarizes in vivo studies that utilize contrast agents within the field of photoacoustic imaging-a relatively new imaging modality-for tumor detection, with a special focus on imaging and transducer parameters. This paper also details the different types of contrast agents used in this novel diagnostic field, i.e., organic-based, metal/inorganic-based, and dye-based contrast agents. We conclude this review by discussing the challenges and future direction of photoacoustic imaging.
Collapse
Affiliation(s)
- Afifa Farooq
- Biomedical Engineering Graduate Program, American University of Sharjah, Sharjah 26666, United Arab Emirates; (A.F.); (S.S.); (N.A.)
| | - Shafiya Sabah
- Biomedical Engineering Graduate Program, American University of Sharjah, Sharjah 26666, United Arab Emirates; (A.F.); (S.S.); (N.A.)
| | - Salam Dhou
- Biomedical Engineering Graduate Program, American University of Sharjah, Sharjah 26666, United Arab Emirates; (A.F.); (S.S.); (N.A.)
- Department of Computer Science and Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Nour Alsawaftah
- Biomedical Engineering Graduate Program, American University of Sharjah, Sharjah 26666, United Arab Emirates; (A.F.); (S.S.); (N.A.)
| | - Ghaleb Husseini
- Biomedical Engineering Graduate Program, American University of Sharjah, Sharjah 26666, United Arab Emirates; (A.F.); (S.S.); (N.A.)
- Department of Chemical Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| |
Collapse
|
31
|
Nanomaterials-based hyperthermia: A literature review from concept to applications in chemistry and biomedicine. J Therm Biol 2022; 104:103201. [DOI: 10.1016/j.jtherbio.2022.103201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 10/19/2022]
|
32
|
Guo Y, Liu X. Radionanomedicine: Advanced Strategy for Precision Theranostics of Breast Cancer. J Biomed Nanotechnol 2022; 18:50-60. [PMID: 35180899 DOI: 10.1166/jbn.2022.3226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Breast carcinoma remains one of the most common and fatal cancers, and even though a series of general therapeutic approaches have been used to treat breast cancer, their outcomes are significantly affected by a variety of side effects. However, nanomedicine could offer novel strategies for dealing with breast carcinoma. In fact, an increasing number of radionanomedicine approaches have recently been used in both diagnostics and therapy. To highlight this trend, the aim of the current review is to systemically summarize the latest advances in radionanomedicine, including single-modular imaging, multiple-modular imaging, and nanomedicine-based theranostics. Barriers to clinical application, the development of next-generation radionanomedicine, and challenges associated with future design are also discussed.
Collapse
Affiliation(s)
- Yang Guo
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaoying Liu
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
33
|
Smart Nanocarriers as an Emerging Platform for Cancer Therapy: A Review. MOLECULES (BASEL, SWITZERLAND) 2021; 27:molecules27010146. [PMID: 35011376 PMCID: PMC8746670 DOI: 10.3390/molecules27010146] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023]
Abstract
Cancer is a group of disorders characterized by uncontrolled cell growth that affects around 11 million people each year globally. Nanocarrier-based systems are extensively used in cancer imaging, diagnostics as well as therapeutics; owing to their promising features and potential to augment therapeutic efficacy. The focal point of research remains to develop new-fangled smart nanocarriers that can selectively respond to cancer-specific conditions and deliver medications to target cells efficiently. Nanocarriers deliver loaded therapeutic cargos to the tumour site either in a passive or active mode, with the least drug elimination from the drug delivery systems. This review chiefly focuses on current advances allied to smart nanocarriers such as dendrimers, liposomes, mesoporous silica nanoparticles, quantum dots, micelles, superparamagnetic iron-oxide nanoparticles, gold nanoparticles and carbon nanotubes, to list a few. Exhaustive discussion on crucial topics like drug targeting, surface decorated smart-nanocarriers and stimuli-responsive cancer nanotherapeutics responding to temperature, enzyme, pH and redox stimuli have been covered.
Collapse
|
34
|
Recent advancements and future submissions of silica core-shell nanoparticles. Int J Pharm 2021; 609:121173. [PMID: 34627997 DOI: 10.1016/j.ijpharm.2021.121173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022]
Abstract
The core-shell silica-based nanoparticles (CSNPs) possess outstanding properties for developing next-generation therapeutics. CSNPs provide greater surface area owing to their mesoporous structure, which offers a high opportunity for surface modification. This review highlights the potential of core-shell silica-based nanoparticle (CSNP) based injectable nanotherapeutics (INT); its role in drug delivery, biomedical imaging, light-triggered phototherapy, Plasmonic enhancers, gene delivery, magnetic hyperthermia, immunotherapy, and potential as next-generation theragnostic. Specifically, the conceptual crosstalk on modern synthetic strategies, biodistribution profiles with a mechanistic view on the therapeutics loading and release modeling are dealt in detail. The manuscript also converses the challenges associated with CSNPs, regulatory hurdles, and their current market position.
Collapse
|
35
|
Wang K, Lu J, Li J, Gao Y, Mao Y, Zhao Q, Wang S. Current trends in smart mesoporous silica-based nanovehicles for photoactivated cancer therapy. J Control Release 2021; 339:445-472. [PMID: 34637819 DOI: 10.1016/j.jconrel.2021.10.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
Photoactivated therapeutic strategies (photothermal therapy and photodynamic therapy), due to the adjusted therapeutic area, time and light dosage, have prevailed for the fight against tumors. Currently, the monotherapy with limited treatment effect and undesired side effects is gradually replaced by multimodal and multifunctional nanosystems. Mesoporous silica nanoparticles (MSNs) with unique physicochemical advantages, such as huge specific surface area, controllable pore size and morphology, functionalized modification, satisfying biocompatibility and biodegradability, are considered as promising candidates for multimodal photoactivated cancer therapy. Excitingly, the innovative nanoplatforms based on the mesoporous silica nanoparticles provide more and more effective treatment strategies and display excellent antitumor potential. Given the rapid development of antitumor strategies based on MSNs, this review summarizes the current progress in MSNs-based photoactivated cancer therapy, mainly consists of (1) photothermal therapy-related theranostics; (2) photodynamic therapy-related theranostics; (3) multimodal synergistic therapy, such as chemo-photothermal-photodynamic therapy, phototherapy-immunotherapy and phototherapy-radio therapy. Based on the limited penetration of irradiation light in photoactivated therapy, the challenges faced by deep-seated tumor therapy are fully discussed, and future clinical translation of MSNs-based photoactivated cancer therapy are highlighted.
Collapse
Affiliation(s)
- Kaili Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Junya Lu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jiali Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Yinlu Gao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Yuling Mao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| |
Collapse
|
36
|
Wang H, Zhang Y, Xu X, Wang A. An injectable mesoporous silica-based analgesic delivery system prolongs the duration of sciatic nerve block in mice with minimal toxicity. Acta Biomater 2021; 135:638-649. [PMID: 34520884 DOI: 10.1016/j.actbio.2021.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 12/16/2022]
Abstract
The major limitation of traditional local anesthetics is the finite duration of a single injection. The present study developed two kinds of novel injectable anesthetic nanocomposites based on mesoporous silica, and evaluated their long-lasting analgesic effect and biosafety. The nanoparticulate carriers, mesoporous silica nanoparticles (MSNs) and mesoporous silica-coated gold nanorods (GNR@MSN), were firstly constructed using the oil-water biphase reaction approach and then ropivacaine (RPC), a local anesthetic, was loaded into the mesoporous carriers by vacuum suction. Transmission electron microscopic images showed the well-ordered mesoporous structure for drug loading. RPC-loaded MSNs and RPC-loaded GNR@MSN exhibited a sustained-release pattern in vitro, and the latter also showed a controlled-release manner triggered by near-infrared (NIR) irradiation. RPC-loaded MSNs and RPC-loaded GNR@MSN caused an initial sensory blockade in mice that lasted for 6 h, almost 2.5 folds of that from free RPC solution. Furthermore, upon NIR irradiation, the latter induced three additional periods of the blockade. Neither of them showed motor nerve block, which may be due to the sustained release manner. The low myotoxicity and low neurotoxicity of the two nanocomposites were presented both in vitro and in vivo. These results demonstrate the potential of the mesoporous silica-based analgesic nanocomposites in effectively controlling postoperative pain, maybe RPC-loaded MSNs for moderate pain and RPC-loaded GNR@MSN for severe pain. STATEMENT OF SIGNIFICANCE: Adequate postoperative analgesia helps early functional exercise after surgery and accelerates rapid recovery, while uncontrolled postoperative pain probably develops chronic post-surgical pain that impacts the life quality of patients for a long time. However, postoperative pain management is still a challenge. The current treatment drugs are always accompanied by some side effects due to their systemic effect. Opioids have risks of addiction and respiratory depression, and nonsteroidal anti-inflammatory drugs can lead to gastrointestinal reaction. Therefore, the long-lasting local anesthetic formulation with good biocompatibility is the most promising solution to manage post-surgical pain. The present study developed novel injectable anesthetic nanocomposites based on mesoporous silica, providing long-lasting pain relief in mice with minimal toxicity.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, China
| | - Yu Zhang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, China
| | - Xiaotao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, China
| | - Aizhong Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, China.
| |
Collapse
|
37
|
Chang Y, Bai Q, Wang M, Ma Y, Yu K, Lu H, Lu T, Lin H, Qu F. Plasmonic Bi nanoparticles encapsulated by N-Carbon for dual-imaging and photothermal/photodynamic/chemo-therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112546. [DOI: 10.1016/j.msec.2021.112546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/29/2021] [Accepted: 11/07/2021] [Indexed: 11/28/2022]
|
38
|
Development of theranostic dual-layered Au-liposome for effective tumor targeting and photothermal therapy. J Nanobiotechnology 2021; 19:262. [PMID: 34481489 PMCID: PMC8418714 DOI: 10.1186/s12951-021-01010-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/23/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Photothermal therapy (PTT) is an emerging anti-cancer therapeutic strategy that generates hyperthermia to ablate cancer cells under laser irradiation. Gold (Au) coated liposome (AL) was reported as an effective PTT agent with good biocompatibility and excretory property. However, exposed Au components on liposomes can cause instability in vivo and difficulty in further functionalization. RESULTS Herein, we developed a theranostic dual-layered nanomaterial by adding liposomal layer to AL (LAL), followed by attaching polyethylene glycol (PEG) and radiolabeling. Functionalization with PEG improves the in vivo stability of LAL, and radioisotope labeling enables in vivo imaging of LAL. Functionalized LAL is stable in physiological conditions, and 64Cu labeled LAL (64Cu-LAL) shows a sufficient blood circulation property and an effective tumor targeting ability of 16.4%ID g-1 from in vivo positron emission tomography (PET) imaging. Also, intravenously injected LAL shows higher tumor targeting, temperature elevation in vivo, and better PTT effect in orthotopic breast cancer mouse model compared to AL. The tumor growth inhibition rate of LAL was 3.9-fold higher than AL. CONCLUSION Based on these high stability, in vivo imaging ability, and tumor targeting efficiency, LAL could be a promising theranostic PTT agent.
Collapse
|
39
|
Rizwan Younis M, He G, Gurram B, Lin J, Huang P. Recent Advances in Gold Nanorods‐Based Cancer Theranostics. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Muhammad Rizwan Younis
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province College of Optoelectronic Engineering Shenzhen University Shenzhen 518060 China
| | - Gang He
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Bhaskar Gurram
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province College of Optoelectronic Engineering Shenzhen University Shenzhen 518060 China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| |
Collapse
|
40
|
Hu X, Li F, Xia F, Wang Q, Lin P, Wei M, Gong L, Low LE, Lee JY, Ling D. Dynamic nanoassembly-based drug delivery system (DNDDS): Learning from nature. Adv Drug Deliv Rev 2021; 175:113830. [PMID: 34139254 DOI: 10.1016/j.addr.2021.113830] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/19/2021] [Accepted: 06/10/2021] [Indexed: 12/18/2022]
Abstract
Dynamic nanoassembly-based drug delivery system (DNDDS) has evolved from being a mere curiosity to emerging as a promising strategy for high-performance diagnosis and/or therapy of various diseases. However, dynamic nano-bio interaction between DNDDS and biological systems remains poorly understood, which can be critical for precise spatiotemporal and functional control of DNDDS in vivo. To deepen the understanding for fine control over DNDDS, we aim to explore natural systems as the root of inspiration for researchers from various fields. This review highlights ingenious designs, nano-bio interactions, and controllable functionalities of state-of-the-art DNDDS under endogenous or exogenous stimuli, by learning from nature at the molecular, subcellular, and cellular levels. Furthermore, the assembly strategies and response mechanisms of tailor-made DNDDS based on the characteristics of various diseased microenvironments are intensively discussed. Finally, the current challenges and future perspectives of DNDDS are briefly commented.
Collapse
|
41
|
Abu-Dief A, Alsehli M, Al-Enizi A, Nafady A. Recent Advances in Mesoporous Silica Nanoparticles for Targeted Drug Delivery applications. Curr Drug Deliv 2021; 19:436-450. [PMID: 34238185 DOI: 10.2174/1567201818666210708123007] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/15/2021] [Accepted: 04/17/2021] [Indexed: 11/22/2022]
Abstract
Nanotechnology provides the means to design and fabricate delivery vehicles capable of overcoming physiologically imposed obstacles and undesirable side effects of systemic drug delivery. This protocol allows maximal targeting effectiveness and therefore enhances therapeutic efficiency. In recent years, mesoporous silica nanoparticles (MSNPs) have sparked interest in the nanomedicine research community, particularly for their promising applications in cancer treatment. The intrinsic physio-chemical stability, facile functionalization, high surface area, low toxicity, and great loading capacity for a wide range of chemotherapeutic agents make MSNPs very appealing candidates for controllable drug delivery systems. Importantly, the peculiar nanostructures of MSNPs enabled them to serve as an effective drug, gene, protein, and antigen delivery vehicle for a variety of therapeutic regimens. For these reasons, in this review article, we underscore the recent progress in the design and synthesis of MSNPs and the parameters influencing their characteristic features and activities. In addition, the process of absorption, dissemination, and secretion by injection or oral management of MSNPs are also discussed, as they are key directions for the potential utilization of MSNPs. Factors influencing the in vivo fate of MSNPs will also be highlighted, with the main focus on particle size, morphology, porosity, surface functionality, and oxidation. Given that combining other functional materials with MSNPs may increase their biological compatibility, monitor drug discharge, or improve absorption by tumor cells coated MSNPs; these aspects are also covered and discussed herein.
Collapse
Affiliation(s)
- Ahmed Abu-Dief
- Chemistry Department, Faculty of Science, Taibah University, Madinah, Saudi Arabia
| | - Mosa Alsehli
- Chemistry Department, Faculty of Science, Taibah University, Madinah, Saudi Arabia
| | - Abdullah Al-Enizi
- Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ayman Nafady
- Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
42
|
Lee J, Ha JW. Influence of oxygen plasma treatment on structural and spectral changes in silica-coated gold nanorods studied using total internal reflection microscopy and spectroscopy. Analyst 2021; 146:4125-4129. [PMID: 34076657 DOI: 10.1039/d1an00592h] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This paper shows how oxygen plasma treatment affects the structural, localized surface plasmon resonance (LSPR) spectral, and spatial orientation changes in single gold nanorods coated with a mesoporous silica shell (AuNRs@SiO2) in comparison with bare AuNRs with the same aspect ratio (AR). Single AuNRs@SiO2 subjected to different plasma treatment times were characterized using scanning electron microscopy and total internal reflection scattering (TIRS) microscopy and spectroscopy. The AR of the single AuNRs without a silica shell was decreased by structural deformation, while their LSPR linewidth was increased with increasing plasma treatment time. In contrast, single AuNRs@SiO2 showed much higher structural and spectral stability due to the silica shell under the energetic plasma treatment. Furthermore, there was no noticeable variation in the three-dimensional (3D) orientations of single AuNR cores in the silica shell before and after the plasma treatment. The results support that no significant structural and spectral changes occur in single AuNRs@SiO2 and that the silica coating enhances the stability of AuNR cores against oxygen plasma treatment. Therefore, fundamental information on the relationship among plasma treatment time, structural change, LSPR damping, and defocused orientation patterns is provided at the single-particle level.
Collapse
Affiliation(s)
- Jaeran Lee
- Energy Harvest-Storage Research Center (EHSRC), University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan 44610, Republic of Korea.
| | - Ji Won Ha
- Energy Harvest-Storage Research Center (EHSRC), University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan 44610, Republic of Korea. and Department of Chemistry, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan 44610, Republic of Korea
| |
Collapse
|
43
|
Zhang Y, Jiang B, Guo C, Liu L, Xu J, Wang Y, Shen C, Zhu J, Shen G, Jiang H, Zhu J, Tao J. Multifunctional Cu xS- and DOX-loaded AuNR@mSiO 2 platform for combined melanoma therapy with inspired antitumor immunity. Biomater Sci 2021; 9:4086-4098. [PMID: 33913979 DOI: 10.1039/d1bm00373a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Combined antitumor therapies based on nanomedicines have shown efficacy in various tumor models in recent years, overcoming the disadvantages of inefficiency and undesired toxicity of traditional therapies. Herein, we present a copper sulfide- and doxorubicin-loaded gold nanorods@mesoporous SiO2 multifunctional nanocomposite (AuNR@mSiO2@DOX-CuxS-PEG) to integrate chemotherapy, the photothermal properties of AuNRs, and the photodynamic properties of CuxS into a single nanoplatform based on hydrophobic interaction and electrostatic attraction. Upon near-infrared light irradiation, the AuNR@mSiO2@DOX-CuxS-PEG nanocomposites exhibit a synergistic therapeutic effect and inhibit the in situ tumor growth and lung metastasis in a melanoma model. This occurs because of the high photothermal conversion efficiency, boosted intracellular reactive oxygen species production, and excellent doxorubicin (DOX) release, as well as an induced tumor-specific immune response. The inspired antitumor immunity was confirmed by elevated infiltration of activated T cells in tumor tissues and improved maturation and activation of dendritic cells in tumor-draining lymph nodes. This study highlights the superior antitumor therapeutic effect elicited by a multifunctional nanoplatform for skin with in situ melanoma and lung metastasis inhibition, indicating its satisfactory clinical application prospects.
Collapse
Affiliation(s)
- Yamin Zhang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Biling Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Chen Guo
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, HUST, Wuhan 430074, China.
| | - Liping Liu
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, HUST, Wuhan 430074, China.
| | - Jian Xu
- Department of Hematology, Union Hospital, Tongji Medical College, HUST, Wuhan 430022, China
| | - Yujue Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Chen Shen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Jinjin Zhu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, HUST, Wuhan 430022, China
| | - Hao Jiang
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, HUST, Wuhan 430074, China.
| | - Jintao Zhu
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, HUST, Wuhan 430074, China.
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China.
| |
Collapse
|
44
|
Wang J, Zhang B, Sun J, Hu W, Wang H. Recent advances in porous nanostructures for cancer theranostics. NANO TODAY 2021; 38:101146. [PMID: 33897805 PMCID: PMC8059603 DOI: 10.1016/j.nantod.2021.101146] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Porous nanomaterials with high surface area, tunable porosity, and large mesopores have recently received particular attention in cancer therapy and imaging. Introduction of additional pores to nanostructures not only endows the tunability of optoelectronic and optical features optimal for tumor treatment, but also modulates the loading capacity and controlled release of therapeutic agents. In recognition, increasing efforts have been made to fabricate various porous nanomaterials and explore their potentials in oncology applications. Thus, a systematic and comprehensive summary is necessary to overview the recent progress, especially in last ten years, on the development of various mesoporous nanomaterials for cancer treatment as theranostic agents. While outlining their individual synthetic mechanisms after a brief introduction of the structures and properties of porous nanomaterials, the current review highlighted the representative applications of three main categories of porous nanostructures (organic, inorganic, and organic-inorganic nanomaterials). In each category, the synthesis, representative examples, and interactions with tumors were further detailed. The review was concluded with deliberations on the key challenges and future outlooks of porous nanostructures in cancer theranostics.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey, 07030, United States
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, 300401, Tianjin, PR China
| | - Beilu Zhang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey, 07030, United States
| | - Jingyu Sun
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey, 07030, United States
| | - Wei Hu
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey, 07030, United States
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey, 07030, United States
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey, 07030, United States
| |
Collapse
|
45
|
Granja A, Pinheiro M, Sousa CT, Reis S. Gold nanostructures as mediators of hyperthermia therapies in breast cancer. Biochem Pharmacol 2021; 190:114639. [PMID: 34077740 DOI: 10.1016/j.bcp.2021.114639] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022]
Abstract
Breast cancer is the leading cause of cancer-related deaths among women. Due to the limitations of the current therapeutics, new treatment options are needed. Hyperthermia is a promising approach to improve breast cancer therapy, particularly when combined with chemo and radiotherapy. This area has gained more attention following association with nanotechnology, with the emergence of modalities, such as photothermal therapy (PTT). PTT is a simple, minimally invasive technique that requires a near infrared (NIR) light source and a PTT agent. Gold nanostructures are excellent PTT agents as they offer biocompatibility, versatility, high photothermal conversion efficiency, imaging contrast and an easily-modified surface. In this review, we describe the molecular basis and the current clinical aspects of hyperthermia-based therapies. The emergent area of nanoparticle-induced hyperthermia will be explored, in particular gold nanostructure-mediated PTT, focusing on recent preclinical studies for breast cancer management.
Collapse
Affiliation(s)
- Andreia Granja
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Marina Pinheiro
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Célia T Sousa
- IFIMUP and Dep. Física e Astronomia, Faculdade de Ciências, Universidade do Porto, Rua Campo Alegre 687, 4169 - 007 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| |
Collapse
|
46
|
Synthesis of Mesoporous Silica Coated Gold Nanorods Loaded with Methylene Blue and Its Potentials in Antibacterial Applications. NANOMATERIALS 2021; 11:nano11051338. [PMID: 34069626 PMCID: PMC8160648 DOI: 10.3390/nano11051338] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/10/2021] [Accepted: 05/16/2021] [Indexed: 02/07/2023]
Abstract
In this work, the successful preparation and characterization of gold nanorods (AuNRs) coated with a mesoporous silica shell (AuNRs@Simes) was achieved. Conjugation with methylene blue (MB) as a model drug using ultrasound-stimulated loading has been explored for further application in light-mediated antibacterial studies. Lyophilization of this conjugated nanosystem was analyzed using trehalose (TRH) as a cryogenic protector. The obtained stable dry formulation shows potent antimicrobial activity against Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) bacteria after a simple post-treatment irradiation method with a red laser during a short time period.
Collapse
|
47
|
Sun B, Zhen X, Jiang X. Development of mesoporous silica-based nanoprobes for optical bioimaging applications. Biomater Sci 2021; 9:3603-3620. [PMID: 34008597 DOI: 10.1039/d1bm00204j] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A mesoporous silica nanoparticle (MSN)-based nanoplatform has attracted growing attention in the biomedical field due to the unique characteristics of MSNs including a high surface area, tunable pore sizes, colloidal stability, ease of functionalization, and desirable biocompatibility. Typically, MSNs are designed as nanocarriers for the incorporation of a variety of contrast agents for bioimaging, which can address the intrinsic drawbacks of contrast agents, including poor solubility in water, rapid photobleaching, and low stability. This review summarizes the recent advances in the field of MSN-based nanoprobes for fluorescence imaging and photoacoustic (PA) imaging applications. The approaches for the incorporation of contrast agents into MSN-based nanoplatforms including encapsulating contrast agents within MSNs, covalently conjugating contrast agents on the surface or pores of MSNs, physically absorbing contrast agents in the pores of MSNs, and doping contrast agents in the framework of MSNs are introduced. MSN-based nanoprobes for fluorescence imaging and PA imaging are discussed. The enhanced fluorescence imaging and PA imaging performances of MSN-based nanoprobes relative to the bare contrast agents are introduced and the underlying mechanisms are discussed in detail. Finally, current challenges and perspectives of MSN-based nanoprobes in the bioimaging field are discussed.
Collapse
Affiliation(s)
- Bo Sun
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China. and Department of Chemical and Biomolecular Engineering, Faculty of Engineering, National University of Singapore, 10 Kent Ridge Crescent, Singapore 119260
| | - Xu Zhen
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China.
| | - Xiqun Jiang
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China.
| |
Collapse
|
48
|
Zhang M, Qin X, Xu W, Wang Y, Song Y, Garg S, Luan Y. Engineering of a dual-modal phototherapeutic nanoplatform for single NIR laser-triggered tumor therapy. J Colloid Interface Sci 2021; 594:493-501. [PMID: 33774405 DOI: 10.1016/j.jcis.2021.03.050] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/27/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022]
Abstract
Theranostic nanoplatforms integrating simultaneously photodynamic therapy (PDT) and photothermal therapy (PTT) exhibit intrinsic advantages in tumor therapy due to distinct mechanisms of action. However, it is challenging to achieve PDT and PTT under single near-infrared (NIR) laser irradiation with a nanoplatform utilizing conventional organic photodynamic agent and inorganic photothermal agent owing to the difference in inherent excitation wavelengths. Particularly, the single NIR light (660 nm)-triggered PTT and PDT nanoplatform, constructed from chlorin e6 (Ce6) and copper sulfide (CuS) nanoparticles (NPs), has never been reported. Herein, we, for the first time, designed and established a dual-modal phototherapeutic nanoplatform that achieved both PTT and PDT under single NIR laser (660 nm) irradiation for Ce6 and CuS NPs with the strategy of core-shell structured CuS@Carbon integrated with Ce6. Introducing of carbon shell not only endows small CuS NPs with excellent tumor accumulation, but also significantly strengthens the photothermal performance of CuS NPs, realizing efficient photothermal performance under 660 nm laser irradiation. Moreover, Ce6 in carbon shell endowed the nanoplatform with photodynamic effect under 660 nm laser irradiation. The as-prepared Ce6/CuS@Carbon nanoplatform thus achieved dual-modal phototherapy under single NIR laser irradiation, significantly inhibiting tumor growth with minimal adverse effects and superior biosafety.
Collapse
Affiliation(s)
- Mengzhu Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaohan Qin
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wei Xu
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong 250014, China
| | - Yibing Wang
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong 250014, China.
| | - Yunmei Song
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Sanjay Garg
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Yuxia Luan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
49
|
Lv Z, He S, Wang Y, Zhu X. Noble Metal Nanomaterials for NIR-Triggered Photothermal Therapy in Cancer. Adv Healthc Mater 2021; 10:e2001806. [PMID: 33470542 DOI: 10.1002/adhm.202001806] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/06/2021] [Indexed: 12/24/2022]
Abstract
It is of great significance to develop anticancer therapeutic agents or technologies with high degree of specificity and patient compliance, while low toxicity. The emerging photothermal therapy (PTT) has become a new and powerful therapeutic technology due to its noninvasiveness, high specificity, low side effects to normal tissues and strong anticancer efficacy. Noble metal nanomaterials possess strong surface plasmon resonance (SPR) effect and synthetic tunability, which make them facile and effective PTT agents with superior optical and photothermal characteristics, such as high absorption cross-section, incomparable optical-thermal conversion efficiency in the near infrared (NIR) region, as well as the potential of bioimaging. By incorporating with various functional reagents such as antibodies, peptides, biocompatible polymers, chemo-drug and immune factors, noble metal nanomaterials have presented strong potential in multifunctional cancer therapy. Herein, the recent development regarding the application of noble metal nanomaterials for NIR-triggered PTT in cancer treatment is summarized. A variety of studies with good therapeutic effects against cancer from impressive photothermal efficacy of noble metal nanomaterials are concluded. Intelligent nanoplatforms through ingenious fabrication showing potential of multifunctional PTT, combined with chemo-therapy, immunotherapy, photodynamic therapy (PDT), as well as simultaneous imaging modality are also demonstrated.
Collapse
Affiliation(s)
- Zhuoqian Lv
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Sijia He
- Cancer Center Shanghai General Hospital Shanghai Jiao Tong University School of Medicine 650 Xinsongjiang Road Shanghai 201620 China
| | - Youfu Wang
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| |
Collapse
|
50
|
Ouyang R, Cao P, Jia P, Wang H, Zong T, Dai C, Yuan J, Li Y, Sun D, Guo N, Miao Y, Zhou S. Bistratal Au@Bi 2S 3 nanobones for excellent NIR-triggered/multimodal imaging-guided synergistic therapy for liver cancer. Bioact Mater 2021; 6:386-403. [PMID: 32954056 PMCID: PMC7481884 DOI: 10.1016/j.bioactmat.2020.08.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/17/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
To fabricate a highly biocompatible nanoplatform enabling synergistic therapy and real-time imaging, novel Au@Bi2S3 core shell nanobones (NBs) (Au@Bi2S3 NBs) with Au nanorods as cores were synthesized. The combination of Au nanorods with Bi2S3 film made the Au@Bi2S3 NBs exhibit ultrahigh photothermal (PT) conversion efficiency, remarkable photoacoustic (PA) imaging and high computed tomography (CT) performance; these Au@Bi2S3 NBs thus are a promising nanotheranostic agent for PT/PA/CT imaging. Subsequently, poly(N-vinylpyrrolidone)-modified Au@Bi2S3 NBs (Au@Bi2S3-PVP NBs) were successfully loaded with the anticancer drug doxorubicin (DOX), and a satisfactory pH sensitive release profile was achieved, thus revealing the great potential of Au@Bi2S3-PVP NBs in chemotherapy as a drug carrier to deliver DOX into cancer cells. Both in vitro and in vivo investigations demonstrated that the Au@Bi2S3-PVP NBs possessed multiple desired features for cancer therapy, including extremely low toxicity, good biocompatibility, high drug loading ability, precise tumor targeting and effective accumulation. Highly efficient ablation of the human liver cancer cell HepG2 was achieved through Au@Bi2S3-PVP NB-mediated photothermal therapy (PTT). As both a contrast enhancement probe and therapeutic agent, Au@Bi2S3-PVP NBs provided outstanding NIR-triggered multi-modal PT/PA/CT imaging-guided PTT and effectively inhibited the growth of HepG2 liver cancer cells via synergistic chemo/PT therapy.
Collapse
Affiliation(s)
- Ruizhuo Ouyang
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Penghui Cao
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Pengpeng Jia
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Hui Wang
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Tianyu Zong
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Chenyu Dai
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jie Yuan
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuhao Li
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Dong Sun
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, China
| | - Ning Guo
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuqing Miao
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Shuang Zhou
- Cancer Institute, Tongji University School of Medicine, Shanghai, 200092, China
| |
Collapse
|