1
|
Dumontel B, Rosso G, Cauda V. Natural and artificial phospholipid bilayer coatings on solid-state nanoparticles, current and future perspectives. Nanomedicine (Lond) 2024; 19:653-655. [PMID: 38406890 DOI: 10.2217/nnm-2023-0358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Affiliation(s)
- Bianca Dumontel
- Department of Applied Science & Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
| | - Giada Rosso
- Department of Applied Science & Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
| | - Valentina Cauda
- Department of Applied Science & Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin, 10129, Italy
| |
Collapse
|
2
|
Xia L, Wu T, Chen L, Mei P, Liu L, Li R, Shu M, Huan Z, Wu C, Fang B. Silicon-Based Biomaterials Modulate the Adaptive Immune Response of T Lymphocytes to Promote Osteogenesis/Angiogenesis via Epigenetic Regulation. Adv Healthc Mater 2023; 12:e2302054. [PMID: 37842937 DOI: 10.1002/adhm.202302054] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Silicon (Si)-based biomaterials are widely applied for bone regeneration. However, the underlying mechanisms of the materials function remain largely unknown. T lymphocyte-mediated adaptive immune response plays a vital role in the process of bone regeneration. In the current study, mesoporous silica (MS) is used as a model material of Si-based biomaterials. It shows that the supernatant of CD4+ T lymphocytes pretreated with MS extract significantly promotes the vascularized bone regeneration. The potential mechanism is closely related to the fact that MS extract can reduce the expression of regulatory factor X-1 (RFX-1) in CD4+ T lymphocytes. This may result in the overexpression of interleukin-17A (IL-17A) by boosting histone H3 acetylation and lowering DNA methylation and H3K9 trimethylation. Importantly, the in vivo experiments further reveal that MS particles significantly enhance bone regeneration with improved angiogenesis in the critical-sized calvarial defect mouse model accompanied by upregulation of IL-17A in peripheral blood and the proportion of Th17 cells. This study suggests that modulation of the adaptive immune response of T lymphocytes by silicate-based biomaterials plays an important role for bone regeneration.
Collapse
Affiliation(s)
- Lunguo Xia
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Tingting Wu
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Lei Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peng Mei
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Lu Liu
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Ruomei Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Mengmeng Shu
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Zhiguang Huan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bing Fang
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| |
Collapse
|
3
|
Sancho-Albero M, Rosso G, De Cola L, Cauda V. Cargo-loaded lipid-shielded breakable organosilica nanocages for enhanced drug delivery. NANOSCALE 2023; 15:14628-14640. [PMID: 37615550 DOI: 10.1039/d3nr02155f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
The recent nanomedicine advancements have introduced a variety of smart nanoparticles in cancer treatment and diagnostics. However, their application to the clinic is still hindered by several challenges related to their biocompatibility, elimination and biodistribution. Here we propose breakable organosilica mesoporous nanoparticles, i.e. nanocages, able to efficiently incorporate cargo molecules and be coated, with different lipid compositions, to enhance their biomimetic behaviour. We exploit the electrostatic interactions between the organosilica surface and the opposite charge of the lipid mixtures in order to obtain an efficient organosilica coverage. The lipid-coated nanocages are proved to have an incredibly high hemocompatibility, significantly increased with respect to pristine nanocages, and excellent colloidal stability and biocompatibility. The cargo-loaded and lipid-coated nanocages are tested and compared in vitro on two different cancer cell lines, demonstrating the key role played by the lipid coating in mediating the internalization of the nanocages, evaluated by the enhanced and rapid cellular uptake. The efficient intracellular delivery of the therapeutic agents is then assured by the destruction of the organosilica, due to the disulfide bridges, introduced into the silica framework, that in reducing media, like the intracellular one, are reduced to thiols causing the breaking of the nanoparticles. The possibility to image and effectively kill cancer cells demonstrates the potentiality of the lipid-coated nanocages as a powerful tool in anticancer research and as a promising smart theranostic platform.
Collapse
Affiliation(s)
- María Sancho-Albero
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri, 2, 20156, Milan, Italy.
| | - Giada Rosso
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy.
| | - Luisa De Cola
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri, 2, 20156, Milan, Italy.
- Department of Pharmaceutical Science, DISFARM, Università degli Studi di Milano, 20133, Milan, Italy
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy.
| |
Collapse
|
4
|
Xu J, Gu M, Hooi L, Toh TB, Thng DKH, Lim JJ, Chow EKH. Enhanced penetrative siRNA delivery by a nanodiamond drug delivery platform against hepatocellular carcinoma 3D models. NANOSCALE 2021; 13:16131-16145. [PMID: 34542130 DOI: 10.1039/d1nr03502a] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Small interfering RNA (siRNA) can cause specific gene silencing and is considered promising for treating a variety of cancers, including hepatocellular carcinoma (HCC). However, siRNA has many undesirable physicochemical properties that limit its application. Additionally, conventional methods for delivering siRNA are limited in their ability to penetrate solid tumors. In this study, nanodiamonds (NDs) were evaluated as a nanoparticle drug delivery platform for improved siRNA delivery into tumor cells. Our results demonstrated that ND-siRNA complexes could effectively be formed through electrostatic interactions. The ND-siRNA complexes allowed for efficient cellular uptake and endosomal escape that protects siRNA from degradation. Moreover, ND delivery of siRNA was more effective at penetrating tumor spheroids compared to liposomal formulations. This enhanced penetration capacity makes NDs ideal vehicles to deliver siRNA against solid tumor masses as efficient gene knockdown and decreased tumor cell proliferation were observed in tumor spheroids. Evaluation of ND-siRNA complexes within the context of a 3D cancer disease model demonstrates the potential of NDs as a promising gene delivery platform against solid tumors, such as HCC.
Collapse
Affiliation(s)
- Jingru Xu
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Mengjie Gu
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Lissa Hooi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health, National University of Singapore, 117456, Singapore
| | - Dexter Kai Hao Thng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Jhin Jieh Lim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
- The N.1 Institute for Health, National University of Singapore, 117456, Singapore
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University Singapore, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| |
Collapse
|
5
|
Mai NXD, Nguyen THT, Vong LB, Dang MHD, Nguyen TTT, Nguyen LHT, Ta HKT, Nguyen TH, Phan TB, Doan TLH. Tailoring chemical compositions of biodegradable mesoporous organosilica nanoparticles for controlled slow release of chemotherapeutic drug. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112232. [PMID: 34225873 DOI: 10.1016/j.msec.2021.112232] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/17/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
Biodegradable periodic mesoporous organosilica nanoparticles (B-PMO) are an outstanding nanocarrier due to their biodegradability and high drug load capacities. The present study describes a synthesis of a phenylene-containing tetrasulfide based B-PMO, named P4S. The incorporation of aromatic phenylene groups into the framework creates a strong interaction between nanoparticles (NPs) with aromatic rings in the cordycepin molecules. This results in the low release profile under various conditions. In addition, the replacement of this linker slowed the degradation of nanoparticles. The physicochemical properties of the nanoparticles are evaluated and compared with a biodegradable ethane-containing tetrasulfide based PMO and a non-degradable MCM-41. The biodegradability of P4S is also demonstrated in a reducing environment and the 100 nm spherical nanoparticles completely decomposed within 14 days. The porous structure of P4S has a high loading of hydrophilic cordycepin (approximately 731.52 mg g-1) with a slow releasing speed. The release rates of P4S NPs are significantly lower than other materials, such as liposomes, gelatin nanoparticles, and photo-crosslinked hyaluronic acid methacrylate hydrogels, in the same solution. This specific release behavior could guarantee drug therapeutic effects with minimum side-effects and optimized drug dosages. Most importantly, according to the in vitro cytotoxicity study, cordycepin-loaded P4S NPs could retain the toxicity against liver cancer cell (HepG2) while suppressed the cytotoxicity against normal cells (BAEC).
Collapse
Affiliation(s)
- Ngoc Xuan Dat Mai
- Center for Innovative Materials and Architectures, Ho Chi Minh City, Viet Nam; Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam; Faculty of Physics and Engineering Physics, University of Science, Ho Chi Minh City, Viet Nam
| | - Thu-Ha Thi Nguyen
- Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam; School of Biomedical Engineering, International University, Ho Chi Minh City, Viet Nam
| | - Long Binh Vong
- Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam; School of Biomedical Engineering, International University, Ho Chi Minh City, Viet Nam
| | - Minh-Huy Dinh Dang
- Center for Innovative Materials and Architectures, Ho Chi Minh City, Viet Nam; Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Trang Thi Thu Nguyen
- Center for Innovative Materials and Architectures, Ho Chi Minh City, Viet Nam; Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Linh Ho Thuy Nguyen
- Center for Innovative Materials and Architectures, Ho Chi Minh City, Viet Nam; Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Hanh Kieu Thi Ta
- Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam; Faculty of Materials Science and Technology, University of Science, Ho Chi Minh City, Viet Nam
| | - Thi-Hiep Nguyen
- Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam; School of Biomedical Engineering, International University, Ho Chi Minh City, Viet Nam
| | - Thang Bach Phan
- Center for Innovative Materials and Architectures, Ho Chi Minh City, Viet Nam; Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam.
| | - Tan Le Hoang Doan
- Center for Innovative Materials and Architectures, Ho Chi Minh City, Viet Nam; Vietnam National University-Ho Chi Minh City, Ho Chi Minh City, Viet Nam.
| |
Collapse
|
6
|
Zhou J, Ma S, Zhang Y, He Y, Mao H, Yang J, Zhang H, Luo K, Gong Q, Gu Z. Bacterium-mimicking sequentially targeted therapeutic nanocomplexes based on O-carboxymethyl chitosan and their cooperative therapy by dual-modality light manipulation. Carbohydr Polym 2021; 264:118030. [PMID: 33910720 DOI: 10.1016/j.carbpol.2021.118030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/10/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023]
Abstract
An integrated gene nanovector capable of overcoming complicated physiological barriers in one vector is desirable to circumvent the challenges imposed by the intricate tumor microenvironment. Herein, a nuclear localization signals (NLS)-decorated element and an iRGD-functionalized element based on O-carboxymethyl chitosan were synthesized, mixed, and coated onto PEI/DNA to fabricate bacterium-mimicking sequentially targeted therapeutic nanocomplexes (STNPs) which were internalized through receptor-mediated endocytosis and other pathways and achieved nuclear translocation of DNA. The endo/lysosomal membrane disruption triggered by reactive oxygen species (ROS) after short-time illumination, together with the DNA nuclear translocation, evoked an enhanced gene expression. Alternatively, the excessive ROS from long-time irradiation induced apoptosis in tumor cells, bringing about greater anti-tumor efficacy owing to the integration of gene and photodynamic therapy. Overall, these results demonstrated bacterium-mimicking STNPs could be a potential candidate for tumor treatments.
Collapse
Affiliation(s)
- Jie Zhou
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Shengnan Ma
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Yuxin Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Yiyan He
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China.
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, PR China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China; Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, 30 South Puzhu Road, Nanjing, 211816, PR China.
| |
Collapse
|
7
|
Wang Y, Wang J, Zhu D, Wang Y, Qing G, Zhang Y, Liu X, Liang XJ. Effect of physicochemical properties on in vivo fate of nanoparticle-based cancer immunotherapies. Acta Pharm Sin B 2021; 11:886-902. [PMID: 33996405 PMCID: PMC8105773 DOI: 10.1016/j.apsb.2021.03.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/25/2020] [Accepted: 01/15/2021] [Indexed: 12/14/2022] Open
Abstract
Current advances of immunotherapy have greatly changed the way of cancer treatment. At the same time, a great number of nanoparticle-based cancer immunotherapies (NBCIs) have also been explored to elicit potent immune responses against tumors. However, few NBCIs are nearly in the clinical trial which is mainly ascribed to a lack understanding of in vivo fate of nanoparticles (NPs) for cancer immunotherapy. NPs for cancer immunotherapy mainly target the immune organs or immune cells to enable efficient antitumor immune responses. The physicochemical properties of NPs including size, shape, elasticity and surface properties directly affect their interaction with immune systems as well as their in vivo fate and therapeutic effect. Hence, systematic analysis of the physicochemical properties and their effect on in vivo fate is urgently needed. In this review, we first recapitulate the fundamentals for the in vivo fate of NBCIs including physio-anatomical features of lymphatic system and strategies to modulate immune responses. Moreover, we highlight the effect of physicochemical properties on their in vivo fate including lymph nodes (LNs) drainage, cellular uptake and intracellular transfer. Challenges and opportunities for rational design of NPs for cancer immunotherapy are also discussed in detail.
Collapse
|
8
|
Khan MA, Fugate M, Rogers DT, Sambi J, Littleton JM, Rankin SE, Knutson BL. Mechanism of Mesoporous Silica Nanoparticle Interaction with Hairy Root Cultures during Nanoharvesting of Biomolecules. Adv Biol (Weinh) 2021; 5:e2000173. [PMID: 33729698 DOI: 10.1002/adbi.202000173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/11/2021] [Indexed: 11/06/2022]
Abstract
Cellular uptake and expulsion mechanisms of engineered mesoporous silica nanoparticles (MSNPs) are important in their design for novel biomolecule isolation and delivery applications such as nanoharvesting, defined as using nanocarriers to transport and isolate valuable therapeutics (secondary metabolites) out of living plant organ cultures (e.g., hairy roots). Here, temperature-dependent MSNP uptake and recovery processes in hairy roots are examined as a function of surface chemistry. MSNP uptake into hairy roots and time-dependent expulsion are quantified using Ti content (present for biomolecule binding) and fluorescence spectroscopy of fluorescently tagged MSNPs, respectively. The results suggest that functionalization and surface charge (regulated by amine group attachment) play the biggest role in the effectiveness of uptake and recovery. Comparison of MSNP interactions with hairy roots at 4 and 23 °C shows that weakly charged MSNPs functionalized only with Ti are taken up and expelled by thermally activated mechanisms, while amine-modified positively charged particles are taken up and expelled mainly by direct penetration of cell walls. Amine-functionalized MSNPs move spontaneously in and out of plant cells by dynamic exchange with a residence time of 20 ± 5 min, suggesting promise as a biomolecule nanoharvesting platform for plant organ cultures.
Collapse
Affiliation(s)
- Md Arif Khan
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY, 40506, USA
| | - Madeleine Fugate
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY, 40506, USA
| | | | | | | | - Stephen E Rankin
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY, 40506, USA
| | - Barbara L Knutson
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY, 40506, USA
| |
Collapse
|
9
|
Wang Y, Gou K, Guo X, Ke J, Li S, Li H. Advances in regulating physicochemical properties of mesoporous silica nanocarriers to overcome biological barriers. Acta Biomater 2021; 123:72-92. [PMID: 33454385 DOI: 10.1016/j.actbio.2021.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/16/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
Mesoporous silica nanoparticles (MSNs) with remarkable structural features have been proven to be an excellent platform for the delivery of therapeutic molecules. Biological barriers in various forms (e.g., mucosal barrier, cellular barrier, gastrointestinal barrier, blood-brain barrier, and blood-tumor barrier) present substantial obstacles for MSNs. The physicochemical parameters of MSNs are known to be effective and tunable not only for load and release of therapeutic molecules but also for their biological responsiveness that is beneficial for cells and tissues. This review innovatively provides a description of how and why physicochemical properties (e.g., particle size, morphology, surface charge, hydrophilic-hydrophobic property, and surface modification) of MSNs influence their ability to cross the biological barriers prior to reaching targeted sites. First, the structural and physiological features of biological barriers are outlined. Next, the recent progresses in the critical physicochemical parameters of MSNs are highlighted from physicochemical and biological aspects. Surface modification, as an important strategy for achieving rapid transport, is also reviewed with special attention to the latest findings of bioactive groups and molecular mechanisms. Furthermore, advanced designs of multifunction intelligent MSNs to surmount the blood-tumor barrier and to actively target tumor sites are demonstrated in detail. Lastly, the biodegradability and toxicity of MSNs are evaluated. With perspectives for their potential application and biosafety, the clues in summary might lead to drug delivery with high efficiency and provide useful knowledge for rational design of nanomaterials.
Collapse
|
10
|
Liu Y, Li Q, Bai Q, Jiang W. Advances of smart nano-drug delivery systems in osteosarcoma treatment. J Mater Chem B 2021; 9:5439-5450. [PMID: 34155495 DOI: 10.1039/d1tb00566a] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nanotechnology has recently become popular due to its potential for biomedical applications, especially for cancer treatment. Nanotechnology, featuring responsiveness to stimuli and stable drug release, has been widely used for the delivery of chemotherapeutic drugs, which are commonly used in the treatment of osteosarcoma. Smart stimuli-responsive nanotechnology is expected to improve the treatment of osteosarcoma. Herein, we focus on the latest research on nanomaterials in treating osteosarcoma that respond to internal and external stimuli. We also discuss nanocarriers with targeting ligands and the use of smart nanotechnology to partially reverse the multidrug resistance of osteosarcoma.
Collapse
Affiliation(s)
- Ying Liu
- Department of Molecular Pathology, Application Center for Precision Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Qing Li
- Department of Molecular Pathology, Application Center for Precision Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Qian Bai
- Department of Molecular Pathology, Application Center for Precision Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Wei Jiang
- Department of Molecular Pathology, Application Center for Precision Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
11
|
Zou Y, Huang B, Cao L, Deng Y, Su J. Tailored Mesoporous Inorganic Biomaterials: Assembly, Functionalization, and Drug Delivery Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005215. [PMID: 33251635 DOI: 10.1002/adma.202005215] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/18/2020] [Indexed: 05/06/2023]
Abstract
Infectious or immune diseases have caused serious threat to human health due to their complexity and specificity, and emerging drug delivery systems (DDSs) have evolved into the most promising therapeutic strategy for drug-targeted therapy. Various mesoporous biomaterials are exploited and applied as efficient nanocarriers to loading drugs by virtue of their large surface area, high porosity, and prominent biocompatibility. Nanosized mesoporous nanocarriers show great potential in biomedical research, and it has become the research hotspot in the interdisciplinary field. Herein, recent progress and assembly mechanisms on mesoporous inorganic biomaterials (e.g., silica, carbon, metal oxide) are summarized systematically, and typical functionalization methods (i.e., hybridization, polymerization, and doping) for nanocarriers are also discussed in depth. Particularly, structure-activity relationship and the effect of physicochemical parameters of mesoporous biomaterials, including morphologies (e.g., hollow, core-shell), pore textures (e.g., pore size, pore volume), and surface features (e.g., roughness and hydrophilic/hydrophobic) in DDS application are overviewed and elucidated in detail. As one of the important development directions, advanced stimuli-responsive DDSs (e.g., pH, temperature, redox, ultrasound, light, magnetic field) are highlighted. Finally, the prospect of mesoporous biomaterials in disease therapeutics is stated, and it will open a new spring for the development of mesoporous nanocarriers.
Collapse
Affiliation(s)
- Yidong Zou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Biaotong Huang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
- Department of Orthopedics Trauma, Shanghai Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Yonghui Deng
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
- State Key Lab of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
12
|
Volpi S, Cancelli U, Neri M, Corradini R. Multifunctional Delivery Systems for Peptide Nucleic Acids. Pharmaceuticals (Basel) 2020; 14:14. [PMID: 33375595 PMCID: PMC7823687 DOI: 10.3390/ph14010014] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The number of applications of peptide nucleic acids (PNAs)-oligonucleotide analogs with a polyamide backbone-is continuously increasing in both in vitro and cellular systems and, parallel to this, delivery systems able to bring PNAs to their targets have been developed. This review is intended to give to the readers an overview on the available carriers for these oligonucleotide mimics, with a particular emphasis on newly developed multi-component- and multifunctional vehicles which boosted PNA research in recent years. The following approaches will be discussed: (a) conjugation with carrier molecules and peptides; (b) liposome formulations; (c) polymer nanoparticles; (d) inorganic porous nanoparticles; (e) carbon based nanocarriers; and (f) self-assembled and supramolecular systems. New therapeutic strategies enabled by the combination of PNA and proper delivery systems are discussed.
Collapse
Affiliation(s)
| | | | | | - Roberto Corradini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124 Parma, Italy; (S.V.); (U.C.); (M.N.)
| |
Collapse
|
13
|
Razavi M, Primavera R, Kevadiya BD, Wang J, Ullah M, Buchwald P, Thakor AS. Controlled Nutrient Delivery to Pancreatic Islets Using Polydopamine-Coated Mesoporous Silica Nanoparticles. NANO LETTERS 2020; 20:7220-7229. [PMID: 32909757 PMCID: PMC8121116 DOI: 10.1021/acs.nanolett.0c02576] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
In the present study, we created a nanoscale platform that can deliver nutrients to pancreatic islets in a controlled manner. Our platform consists of a mesoporous silica nanoparticle (MSNP), which can be loaded with glutamine (G: an essential amino acid required for islet survival and function). To control the release of G, MSNPs were coated with a polydopamine (PD) layer. With the optimal parameters (0.5 mg/mL and 0.5 h), MSNPs were coated with a layer of PD, which resulted in a delay of G release from MSNPs over 14 d (57.4 ± 4.7% release). Following syngeneic renal subcapsule islet transplantation in diabetic mice, PDG-MSNPs improved the engraftment of islets (i.e., enhanced revascularization and reduced inflammation) as well as their function, resulting in re-establishment of glycemic control. Collectively, our data show that PDG-MSNPs can support transplanted islets by providing them with a controlled and sustained supply of nutrients.
Collapse
Affiliation(s)
- Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States; Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine and Department of Materials Science & Engineering, University of Central Florida, Orlando, Florida 32827, United States
| | - Rosita Primavera
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Bhavesh D Kevadiya
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| |
Collapse
|
14
|
Visible-light photocatalytic selective aerobic oxidation of thiols to disulfides on anatase TiO2. CHINESE JOURNAL OF CATALYSIS 2020. [DOI: 10.1016/s1872-2067(20)63640-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
15
|
Barui S, Cauda V. Multimodal Decorations of Mesoporous Silica Nanoparticles for Improved Cancer Therapy. Pharmaceutics 2020; 12:E527. [PMID: 32521802 PMCID: PMC7355899 DOI: 10.3390/pharmaceutics12060527] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
The presence of leaky vasculature and the lack of lymphatic drainage of small structures by the solid tumors formulate nanoparticles as promising delivery vehicles in cancer therapy. In particular, among various nanoparticles, the mesoporous silica nanoparticles (MSN) exhibit numerous outstanding features, including mechanical thermal and chemical stability, huge surface area and ordered porous interior to store different anti-cancer therapeutics with high loading capacity and tunable release mechanisms. Furthermore, one can easily decorate the surface of MSN by attaching ligands for active targeting specifically to the cancer region exploiting overexpressed receptors. The controlled release of drugs to the disease site without any leakage to healthy tissues can be achieved by employing environment responsive gatekeepers for the end-capping of MSN. To achieve precise cancer chemotherapy, the most desired delivery system should possess high loading efficiency, site-specificity and capacity of controlled release. In this review we will focus on multimodal decorations of MSN, which is the most demanding ongoing approach related to MSN application in cancer therapy. Herein, we will report about the recently tried efforts for multimodal modifications of MSN, exploiting both the active targeting and stimuli responsive behavior simultaneously, along with individual targeted delivery and stimuli responsive cancer therapy using MSN.
Collapse
Affiliation(s)
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy;
| |
Collapse
|
16
|
Xu H, Zhang YF, Lang X. TEMPO visible light photocatalysis: The selective aerobic oxidation of thiols to disulfides. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2019.10.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
17
|
Montaseri H, Kruger CA, Abrahamse H. Recent Advances in Porphyrin-Based Inorganic Nanoparticles for Cancer Treatment. Int J Mol Sci 2020; 21:E3358. [PMID: 32397477 PMCID: PMC7247422 DOI: 10.3390/ijms21093358] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
The application of porphyrins and their derivatives have been investigated extensively over the past years for phototherapy cancer treatment. Phototherapeutic Porphyrins have the ability to generate high levels of reactive oxygen with a low dark toxicity and these properties have made them robust photosensitizing agents. In recent years, Porphyrins have been combined with various nanomaterials in order to improve their bio-distribution. These combinations allow for nanoparticles to enhance photodynamic therapy (PDT) cancer treatment and adding additional nanotheranostics (photothermal therapy-PTT) as well as enhance photodiagnosis (PDD) to the reaction. This review examines various porphyrin-based inorganic nanoparticles developed for phototherapy nanotheranostic cancer treatment over the last three years (2017 to 2020). Furthermore, current challenges in the development and future perspectives of porphyrin-based nanomedicines for cancer treatment are also highlighted.
Collapse
Affiliation(s)
| | | | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa; (H.M.); (C.A.K.)
| |
Collapse
|
18
|
Pang YT, Ge Z, Zhang B, Xiu P, Li Q, Wang Y. Pore formation induced by nanoparticles binding to a lipid membrane. NANOSCALE 2020; 12:7902-7913. [PMID: 32227042 DOI: 10.1039/c9nr10534d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Nanoparticles (NPs) enter a cell primarily via endocytosis, during which they are engulfed by the cell and reside in lipid vesicles named endosomes. Apart from when an endosome is pinched off the plasma membrane, structural integrity of its lipid membrane is usually well maintained. Under certain circumstances, however, such structural integrity can be considerably perturbed by a nanoparticle. For instance, recent experiments [Chu et al., Sci. Rep., 2014, 4, 4495] indicate that nanodiamonds with sharp corners can escape from an endosome by piercing its lipid membrane. Nonetheless, the energetics of this behavior and how it may be controlled by membrane adhesion and NP morphology remain unclear. In this work, we employ continuum modeling to investigate membrane pore formation induced by the spontaneous binding of a sharp nanoparticle. Based on two axial-symmetric NP models, we characterize the indispensable role played by curvature heterogeneity, membrane adhesion, and the sharpness as well as the size of a nanoparticle in 'breaking' a lipid membrane. Apart from revealing a general mechanism of NP binding-induced membrane pore formation, our results provide the reference for improving the endosomal escape of nanoparticles through manipulating their morphology, a direction that can be explored either independently or combined with existing strategies targeting NP surface chemistry.
Collapse
Affiliation(s)
- Yui Tik Pang
- Department of Physics, Chinese University of Hong Kong, Shatin, N.T., Hong Kong.
| | | | | | | | | | | |
Collapse
|
19
|
Mai NXD, Birault A, Matsumoto K, Ta HKT, Intasa‐ard SG, Morrison K, Thang PB, Doan TLH, Tamanoi F. Biodegradable Periodic Mesoporous Organosilica (BPMO) Loaded with Daunorubicin: A Promising Nanoparticle-Based Anticancer Drug. ChemMedChem 2020; 15:593-599. [PMID: 32020745 PMCID: PMC7187469 DOI: 10.1002/cmdc.201900595] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/30/2020] [Indexed: 11/28/2022]
Abstract
Biodegradable periodic mesoporous organosilica (BPMO) nanoparticles have emerged as a promising type of nanocarrier for drug delivery, given the biodegradable feature is advantageous for clinical translation. In this paper, we report synthesis and characterization of daunorubicin (DNR) loaded BPMO. DNR was loaded onto rhodamine B-labeled BPMO that contain tetrasulfide bonds. Tumor spheroids and chicken egg tumor models were used to characterize the activity in biological settings. In the first experiment we examined the uptake of BPMO into tumor spheroids prepared from ovarian cancer cells. BPMO were efficiently taken up into tumor spheroids and inhibited their growth. In the chicken egg tumor model, intravenous injection of DNR-loaded BPMO led to the elimination of ovarian tumor. Lack of adverse effect on organs such as lung appears to be due to excellent tumor accumulation of BPMO. Thus, DNR-loaded BPMO represents a promising nanodrug compared with free DNR currently used in cancer therapy. OK.
Collapse
Affiliation(s)
- Ngoc Xuan Dat Mai
- Center for Innovative Materials and Architectures (INOMAR)Vietnam National University-Ho Chi Minh CityHo Chi Minh City721337Vietnam
- Faculty of Physics and Engineering Physics, University of ScienceVietnam National UniversityHo Chi Minh City700000Vietnam
| | - Albane Birault
- Institute for Integrated Cell-Material Sciences (ICeMS)Institute for Advanced Study Kyoto UniversityKyoto606 8501Japan
| | - Kotaro Matsumoto
- Institute for Integrated Cell-Material Sciences (ICeMS)Institute for Advanced Study Kyoto UniversityKyoto606 8501Japan
| | - Hanh Kieu Thi Ta
- Center for Innovative Materials and Architectures (INOMAR)Vietnam National University-Ho Chi Minh CityHo Chi Minh City721337Vietnam
- Faculty of Materials Science and Technology, University of ScienceVietnam National UniversityHo Chi Minh City700000Vietnam
| | - Soontaree Grace Intasa‐ard
- Institute for Integrated Cell-Material Sciences (ICeMS)Institute for Advanced Study Kyoto UniversityKyoto606 8501Japan
| | - Kendall Morrison
- TAE Life SciencesDrug Development DivisionSanta Monica, CA90404USA
| | - Phan Bach Thang
- Center for Innovative Materials and Architectures (INOMAR)Vietnam National University-Ho Chi Minh CityHo Chi Minh City721337Vietnam
| | - Tan Le Hoang Doan
- Center for Innovative Materials and Architectures (INOMAR)Vietnam National University-Ho Chi Minh CityHo Chi Minh City721337Vietnam
| | - Fuyuhiko Tamanoi
- Institute for Integrated Cell-Material Sciences (ICeMS)Institute for Advanced Study Kyoto UniversityKyoto606 8501Japan
| |
Collapse
|
20
|
Photochemical Internalization for Intracellular Drug Delivery. From Basic Mechanisms to Clinical Research. J Clin Med 2020; 9:jcm9020528. [PMID: 32075165 PMCID: PMC7073817 DOI: 10.3390/jcm9020528] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 02/01/2020] [Indexed: 02/06/2023] Open
Abstract
Photochemical internalisation (PCI) is a unique intervention which involves the release of endocytosed macromolecules into the cytoplasmic matrix. PCI is based on the use of photosensitizers placed in endocytic vesicles that, following light activation, lead to rupture of the endocytic vesicles and the release of the macromolecules into the cytoplasmic matrix. This technology has been shown to improve the biological activity of a number of macromolecules that do not readily penetrate the plasma membrane, including type I ribosome-inactivating proteins (RIPs), gene-encoding plasmids, adenovirus and oligonucleotides and certain chemotherapeutics, such as bleomycin. This new intervention has also been found appealing for intracellular delivery of drugs incorporated into nanocarriers and for cancer vaccination. PCI is currently being evaluated in clinical trials. Data from the first-in-human phase I clinical trial as well as an update on the development of the PCI technology towards clinical practice is presented here.
Collapse
|
21
|
Chirra S, Siliveri S, Gangalla R, Goskula S, Gujjula SR, Adepu AK, Anumula R, Sivasoorian SS, Wang LF, Narayanan V. Synthesis of new multivalent metal ion functionalized mesoporous silica and studies of their enhanced antimicrobial and cytotoxicity activities. J Mater Chem B 2019; 7:7235-7245. [PMID: 31664291 DOI: 10.1039/c9tb01736d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the present study, we have reported the synthesis of a transition metal (Me = Ti, V, and Pd) incorporated into MCM-41 mesoporous molecular sieves (Si/Me = 20) synthesized by the sol-gel method. Their physicochemical properties were studied in detail by standard techniques like low angle powder X-ray diffraction (XRD), scanning electron microscopy-energy-dispersive X-ray spectroscopy (SEM-EDXS), transmission electron microscopy (TEM), N2 adsorption/desorption studies, and thermogravimetric-differential thermal (TG-DTA) analysis and spectral studies like Fourier transform infrared spectroscopic analysis (FT-IR), diffuse reflectance ultraviolet-visible spectroscopic analysis (UV-Visible-DRS), and X-ray photoelectron spectroscopy (XPS). The XRD patterns prove that the material's phase identity is the same irrespective of metal incorporation. SEM displayed the uniform shape and size of the nanoparticles. The presence of elements such as Ti, V, Pd, Si and O in respective materials is revealed using the EDXS analysis. Around 30% weight loss arose upon calcination from room temperature to 800 °C. BET surface area analysis presented that the parent materials have a high surface area (1024 m2 g-1) which was reduced upon metal incorporation. FT-IR analysis exhibited the framework vibrations of the synthesised materials. UV-Visible-DRS analysis indicated the presence of tetrahedrally coordinated transition metal ions. The multivalent-metal-ion-functionalized mesoporous materials showed significant enhancement in potent antimicrobial and anticancer activity. The antimicrobial activity is because of its low lipophilicity, which no longer allows the materials to enter via the lipid membrane. Thus, the new materials neither obstruct the metal-binding sites nor inhibit the growth of microbe enzymes. Further, the results show that the transition metal ion-containing mesoporous materials possessing good anticancer activity arising from their excessive surface area to volume ratio provided appropriate association with a tumour cell due to the direct penetration of mesoporous materials into the cell wall, causing membrane damage and cell death.
Collapse
Affiliation(s)
- Suman Chirra
- Department of Chemistry, National Institute of Technology, Warangal 506 004, Telangana, India.
| | - Suresh Siliveri
- Department of Chemistry, National Institute of Technology, Warangal 506 004, Telangana, India.
| | - Ravi Gangalla
- Department of Microbiology, Kakatiya University, Warangal 506 009, Telangana, India
| | - Srinath Goskula
- Department of Chemistry, National Institute of Technology, Warangal 506 004, Telangana, India.
| | - Sripal Reddy Gujjula
- Department of Chemistry, National Institute of Technology, Warangal 506 004, Telangana, India.
| | - Ajay Kumar Adepu
- Inorganic & Physical Chemistry Division, CSIR - Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Rajini Anumula
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100090, China
| | - Siva Sankari Sivasoorian
- Department of Medicinal & Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Li-Fang Wang
- Department of Medicinal & Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Venkatathri Narayanan
- Department of Chemistry, National Institute of Technology, Warangal 506 004, Telangana, India.
| |
Collapse
|
22
|
Barkat A, Beg S, Panda SK, S Alharbi K, Rahman M, Ahmed FJ. Functionalized mesoporous silica nanoparticles in anticancer therapeutics. Semin Cancer Biol 2019; 69:365-375. [PMID: 31442571 DOI: 10.1016/j.semcancer.2019.08.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 11/26/2022]
Abstract
The application of nanomedicines in tumor targeting and attaining meaningful therapeutic benefits for the treatment of cancers has been going on for almost two decades. Beyond the lipidic and polymeric nanomedicines-based passive and active targeting, the quest for inventing the new generation of carriers has no end. This has lead to the evolution of some of the unique carrier systems with supramolecular assembly structures. Mesoporous nanoparticulate systems (MSNPs) are the recently explored substances with favorable potential for drug delivery and drug targeting applications especially in cancer chemotherapeutics. Notwithstanding their physical properties that makes them a suitable carrier for cancer treatment, but their outstanding ability towards chemical functionalization helps in delivering the imaging agents for diagnostic applications. MSNPs can improve the dissolution rate and systemic availability of the poorly water soluble drugs due to their mesoporous structures. Besides, guest molecules including targeting ligands, biomimetic agents, fluorescent dyes, and biocompatible polymers can be efficiently encapsulated in their tunable porous structure for targeting purpose. Some special features of the MSNPs which make them one of the highly effective nanocarrier systems include their ability to encapsulate non-crystalline drugs in their mesopores, high dispersion ability as a function of large surface area and wetting properties. For anticancer drug delivery, MSNPs are worthful to provide excellent drug loading capacity and endocytotic behavior. Moreover, the external surface of MSNPs can be precisely modified for tumor-recognition and developing sensitivity of the antitumor agents towards the cancer cells. Owing to the innumerable applications of MSNPs till now in cancer treatment, the present article particularly focuses to provide an overview account with complete details on the topic to make the readers abreast with details on physiochemical and material properties of MSNPs, their applications and current innovations for the purpose.
Collapse
Affiliation(s)
- Abul Barkat
- Department of Pharmaceutics, School of Medical & Allied Sciences, KR Mangalam University, Gurgaon, Sohna, Haryana, India
| | - Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard (Hamdard University), New Delhi, India.
| | - Sunil K Panda
- Research Director, Menovo Pharmaceuticals Research Lab, Ningbo, People's Republic of China
| | - Khalid S Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakakah, Kingdom of Saudi Arabia
| | - Mahfoozur Rahman
- Department of Pharmaceutical Sciences, SIHAS, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, UP, India.
| | - Farhan J Ahmed
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard (Hamdard University), New Delhi, India.
| |
Collapse
|
23
|
Ji Y, Li J, Zhao J, Shan S, Chu CC. A light-facilitated drug delivery system from a pseudo-protein/hyaluronic acid nanocomplex with improved anti-tumor effects. NANOSCALE 2019; 11:9987-10003. [PMID: 31080976 DOI: 10.1039/c9nr01909j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Reduction-sensitive nanomedicine is a promising strategy to achieve controlled release of payloads in response to intracellular reductive milieu. However, endolysosomal sequestration of internalized carriers and insufficient redox potential in endolysosomes may delay the release of payloads and impact their therapeutic efficacy. Photochemical internalization (PCI), which takes advantage of light-induced endolysosomal rupture, is an effective technique for endosomal escape and cytosolic release of cargos. In this study, a biodegradable and reduction-sensitive nanocomplex was developed from arginine based poly(ester amide)s and hyaluronic acid (HA), and the PCI-photosensitizer AlPcS2a was conjugated to the surface of the nanocomplex (ArgPEA-ss-HA(AP)). This nanocomplex was used for the co-delivery of both PCI-photosensitizers and therapeutic agents to eliminate the biodistribution discrepancy resulting from the separated administration of free therapeutics. The PCI effect of the ArgPEA-ss-HA(AP) nanocomplex was validated in both monolayers and 3D spheroid models of MDA-MB-231 breast cancer cells. Synergism was detected between the PCI effect and doxorubicin-loaded nanocomplex in the inhibition of MDA-MB-231 cells. In addition, the ArgPEA-ss-HA(AP) nanocomplex also provided enhanced intratumoral penetration in 3D spheroids compared to free AlPcS2a. The in vivo results suggested that the conjugation of AlPCs2a in the nanocomplex enabled the consistent and preferential accumulation of both doxorubicin and AlPcS2a in tumor sites. A light-enhanced anti-tumor effect was observed for the doxorubicin-loaded nanocomplex at well-tolerable dosage. The ArgPEA-ss-HA(AP) nanocomplex, as a reduction-responsive delivery vehicle, can hold great potential to achieve spatio-temporally controllable anti-tumor effects.
Collapse
Affiliation(s)
- Ying Ji
- Department of Fiber Science and Apparel Design, Cornell University, Ithaca, New York 14853-4401, USA.
| | - Juan Li
- Key Laboratory of Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing 100049, PR China
| | - Jihui Zhao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Shuo Shan
- Biomedical Engineering Field. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853-4401, USA
| | - Chih-Chang Chu
- Department of Fiber Science and Apparel Design, Cornell University, Ithaca, New York 14853-4401, USA. and Biomedical Engineering Field. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853-4401, USA
| |
Collapse
|
24
|
Li X, Wang Z, Xia H. Ultrasound Reversible Response Nanocarrier Based on Sodium Alginate Modified Mesoporous Silica Nanoparticles. Front Chem 2019; 7:59. [PMID: 30805332 PMCID: PMC6378627 DOI: 10.3389/fchem.2019.00059] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Mesoporous silica nanoparticles (MSN) covered by polymer coatings, cross-linked by weak coordination bonds were expected to present a reversible responsiveness under on-off ultrasound stimuli. Herein, we prepared a sodium alginate (SA) modified MSN with carboxyl-calcium (COO--Ca2+) coordination bonds in the modified layer, which could block the mesopores of MSN and effectively prevent the cargo from pre-releasing before stimulation. The coordination bonds would be destroyed under the stimulation of low intensity ultrasound (20 kHz) or high intensity focused ultrasound (HIFU, 1.1 MHz), leading to a rapid and significant cargo release, and then they could be reformed when ultrasound was turned off, resulting in an instant cargo release stopping. The reversible cleavage and reformation of this coordination bonds under on-off ultrasound stimulus were confirmed by the gel-sol transition behaviors of the SA-CaCl2 gels. An excellent real-time control of rhodamine B (RhB) release performance was obtained under the ultrasound stimuli. Obviously, the cargo release ratio could reach to nearly 40% when HIFU (80 W) was turned on for 5 min, and remained basically constant when ultrasound was turned off, which would finally reach to nearly 100% within 30 min under this on-off pulsatile status. These hybrid MSN based nanoparticles with excellent reversible ultrasound on-off responsiveness were of great interest in on-demand drug delivery applications in the future.
Collapse
Affiliation(s)
| | - Zhanhua Wang
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, Sichuan University, Chengdu, China
| | | |
Collapse
|
25
|
Abstract
Recently greater emphasis has been given to combination therapy for generating synergistic effects of treating cancer. Recent studies on thiol-sensitive nanocarriers for the delivery of drug or gene have shown promising results. In this review, we will examine the rationale and advantage in using nanocarriers for the combined delivery of different anticancer drugs and biologics. Here, we also discuss the role of nanocarriers, particularly redox-sensitive polymers in evading or inhibiting the efflux pump in cancer and how they modulate the sensitivity of cancer cells. The review aims to provide a good understanding of the new pattern of cancer treatment and key concerns for designing nanomedicine of synergistic combinations for cancer therapy.
Collapse
|
26
|
Yang B, Chen Y, Shi J. Exogenous/Endogenous-Triggered Mesoporous Silica Cancer Nanomedicine. Adv Healthc Mater 2018; 7:e1800268. [PMID: 29938917 DOI: 10.1002/adhm.201800268] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/26/2018] [Indexed: 11/12/2022]
Abstract
Recent advances in nanomedicine-based theranostic platforms have catalyzed the generation of new theranostic modalities for pathological abnormalities, such as cancer. Mesoporous silica-based nanomedicines, which feature unique physicochemical properties and specific applicability, are extensively explored for numerous oncological applications. Due to the well-defined morphology, specific surface area, and pore volume, mesoporous silica nanoparticle (MSN)-based theranostic platforms have provided unprecedented opportunities for the development of next-generation cancer nanomedicine. However, current understanding on the underlying mechanisms of how these feasible theranostic platforms interact with exogenous/endogenous triggers and how this unique responsiveness for optimized cancer therapy can be taken advantage of is still preliminary. In this progress report, efforts are made to give a comprehensive overview of the development of MSN-based "smart" theranostic platforms, from exogenous physical irradiation-triggered platforms for localized therapy to endogenous biological stimulus-triggered platforms for tumor microenvironment responsiveness. It is highly expected that these elaborately fabricated MSN-based nanoformulations will play an indispensable role in the efficient cancer therapy based on their high therapeutic outcome and reduced side effects.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Shanghai Institute of Ceramics; Chinese Academy of Sciences; Shanghai 200050 P. R. China
- University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Shanghai Institute of Ceramics; Chinese Academy of Sciences; Shanghai 200050 P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Shanghai Institute of Ceramics; Chinese Academy of Sciences; Shanghai 200050 P. R. China
| |
Collapse
|
27
|
Glutathione-depletion mesoporous organosilica nanoparticles as a self-adjuvant and Co-delivery platform for enhanced cancer immunotherapy. Biomaterials 2018; 175:82-92. [PMID: 29803106 DOI: 10.1016/j.biomaterials.2018.05.025] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/10/2018] [Accepted: 05/16/2018] [Indexed: 12/16/2022]
Abstract
Silica based nanoparticles have emerged as a promising vaccine delivery system for cancer immunotherapy, but their bio-degradability, adjuvanticity and the resultant antitumor activity remain to be largely improved. In this study, we report biodegradable glutathione-depletion dendritic mesoporous organosilica nanoparticles (GDMON) with a tetrasulfide-incorporated framework as a novel co-delivery platform in cancer immunotherapy. Functionalized GDMON are capable of co-delivering an antigen protein (ovalbumin) and a toll-like receptor 9 (TLR9) agonist into antigen presenting cells (APCs) and inducing endosome escape. Moreover, decreasing the intracellular glutathione (GSH) level through the -S-S-/GSH redox chemistry increases the ROS generation level both in vitro and in vivo, facilitating cytotoxic T lymphocyte (CTL) proliferation and reducing tumour growth in an aggressive B16-OVA melanoma tumour model. Our results have shown the potential of GDMON as a novel self-adjuvant and co-delivery nanocarrier for cancer vaccine.
Collapse
|
28
|
Bourquin J, Milosevic A, Hauser D, Lehner R, Blank F, Petri-Fink A, Rothen-Rutishauser B. Biodistribution, Clearance, and Long-Term Fate of Clinically Relevant Nanomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1704307. [PMID: 29389049 DOI: 10.1002/adma.201704307] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/20/2017] [Indexed: 05/18/2023]
Abstract
Realization of the immense potential of nanomaterials for biomedical applications will require a thorough understanding of how they interact with cells, tissues, and organs. There is evidence that, depending on their physicochemical properties and subsequent interactions, nanomaterials are indeed taken up by cells. However, the subsequent release and/or intracellular degradation of the materials, transfer to other cells, and/or translocation across tissue barriers are still poorly understood. The involvement of these cellular clearance mechanisms strongly influences the long-term fate of used nanomaterials, especially if one also considers repeated exposure. Several nanomaterials, such as liposomes and iron oxide, gold, or silica nanoparticles, are already approved by the American Food and Drug Administration for clinical trials; however, there is still a huge gap of knowledge concerning their fate in the body. Herein, clinically relevant nanomaterials, their possible modes of exposure, as well as the biological barriers they must overcome to be effective are reviewed. Furthermore, the biodistribution and kinetics of nanomaterials and their modes of clearance are discussed, knowledge of the long-term fates of a selection of nanomaterials is summarized, and the critical points that must be considered for future research are addressed.
Collapse
Affiliation(s)
- Joël Bourquin
- Adolphe Merkle InstituteUniversity of Fribourg, Chemin des Verdiers 4, 1700, Fribourg, Switzerland
| | - Ana Milosevic
- Adolphe Merkle InstituteUniversity of Fribourg, Chemin des Verdiers 4, 1700, Fribourg, Switzerland
| | - Daniel Hauser
- Adolphe Merkle InstituteUniversity of Fribourg, Chemin des Verdiers 4, 1700, Fribourg, Switzerland
| | - Roman Lehner
- Adolphe Merkle InstituteUniversity of Fribourg, Chemin des Verdiers 4, 1700, Fribourg, Switzerland
| | - Fabian Blank
- Respiratory Medicine, Department of Biomedical Research, University of Bern, Murtenstrasse 50, 3008, Bern
| | - Alke Petri-Fink
- Adolphe Merkle InstituteUniversity of Fribourg, Chemin des Verdiers 4, 1700, Fribourg, Switzerland
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700, Fribourg, Switzerland
| | | |
Collapse
|
29
|
Wang Y, Wang X, Deng F, Zheng N, Liang Y, Zhang H, He B, Dai W, Wang X, Zhang Q. The effect of linkers on the self-assembling and anti-tumor efficacy of disulfide-linked doxorubicin drug-drug conjugate nanoparticles. J Control Release 2018; 279:136-146. [PMID: 29655991 DOI: 10.1016/j.jconrel.2018.04.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/26/2018] [Accepted: 04/11/2018] [Indexed: 12/21/2022]
Abstract
Drug-drug conjugate nanoparticles (DDC NPs) is a potential method for overcoming poor solubility and nonspecific action in cancer therapy, which is based on its high drug loading efficiency and passive tumor-target properties. Our laboratory has prepared DOX-SS-DOX NPs based on disulfide-linked doxorubicin (DOX) drug-drug conjugate, which showed well physical stability and similar anti-tumor efficacy as liposomes. However, how structures of DDCs influence the self-assembling and anti-tumor efficacy is still seldom clarified and needs further investigation. Here, we discussed the role of linker types, length and linkage site in the NPs self-assembling and anti-tumor efficacy. A series of DOX prodrugs were prepared and all the prodrugs could self-assemble into NPs except DOX-SS-DOX (2), indicating the linker length played an important role during self-assembling process. The linkage sites and types of linker exhibited great influence on in vitro cytotoxicity and in vivo anti-tumor efficacy, particularly, modification on C-14 hydroxyl was more efficient for DOX release than on amino group. Besides, disulfide-bond was not cleaved and DOX-SH release did not occur in the metabolism process. The function of disulfide-bond was to enhance the release of DOX in the hydrolysis process. These findings is meaningful for effective prodrug NPs design for therapeutics.
Collapse
Affiliation(s)
- Yaoqi Wang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Feiyang Deng
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Nan Zheng
- National Drug Clinical Trial Center, Key laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yanqin Liang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| |
Collapse
|
30
|
Augspurger AE, Sun X, Trewyn BG, Fang N, Stender AS. Monitoring the Stimulated Uncapping Process of Gold-Capped Mesoporous Silica Nanoparticles. Anal Chem 2018; 90:3183-3188. [PMID: 29402079 DOI: 10.1021/acs.analchem.7b04532] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To establish a new method for tracking the interaction of nanoparticles with chemical cleaving agents, we exploited the optical effects caused by attaching 5-10 nm gold nanoparticles with molecular linkers to large mesoporous silica nanoparticles (MSN). At low levels of gold loading onto MSN, the optical spectra resemble colloidal suspensions of gold. As the gold is removed, by cleaving agents, the MSN revert to the optical spectra typical of bare silica. Time-lapse images of gold-capped MSN stationed in microchannels reveal that the rate of gold release is dependent on the concentration of the cleaving agent. The uncapping process was also monitored successfully for MSN endocytosed by A549 cancer cells, which produce the cleaving agent glutathione. These experiments demonstrate that the optical properties of MSN can be used to directly monitor cleaving kinetics, even in complex cellular settings.
Collapse
Affiliation(s)
- Ashley E Augspurger
- Department of Chemistry , Iowa State University , Ames , Iowa 50011 , United States.,The Ames Laboratory , U.S. Department of Energy , Ames , Iowa 50011 , United States
| | - Xiaoxing Sun
- Department of Chemistry , Iowa State University , Ames , Iowa 50011 , United States.,The Ames Laboratory , U.S. Department of Energy , Ames , Iowa 50011 , United States
| | - Brian G Trewyn
- Department of Chemistry , Iowa State University , Ames , Iowa 50011 , United States.,The Ames Laboratory , U.S. Department of Energy , Ames , Iowa 50011 , United States.,Department of Chemistry , Colorado School of Mines , Golden , Colorado 80401 , United States
| | - Ning Fang
- Department of Chemistry , Iowa State University , Ames , Iowa 50011 , United States.,The Ames Laboratory , U.S. Department of Energy , Ames , Iowa 50011 , United States.,Department of Chemistry , Georgia State University , Atlanta , Georgia 30302 , United States
| | - Anthony S Stender
- Department of Chemistry , Iowa State University , Ames , Iowa 50011 , United States.,The Ames Laboratory , U.S. Department of Energy , Ames , Iowa 50011 , United States.,Department of Chemistry and Biochemistry , Ohio University , Athens , Ohio 45701 , United States
| |
Collapse
|
31
|
Aftab S, Shah A, Nadhman A, Kurbanoglu S, Aysıl Ozkan S, Dionysiou DD, Shukla SS, Aminabhavi TM. Nanomedicine: An effective tool in cancer therapy. Int J Pharm 2018; 540:132-149. [PMID: 29427746 DOI: 10.1016/j.ijpharm.2018.02.007] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/30/2018] [Accepted: 02/04/2018] [Indexed: 12/24/2022]
Abstract
Various types of nanoparticles (NPs) have been used in delivering anticancer drugs to the site of action. This area has become more attractive in recent years due to optimal size and negligible undesirable side effects caused by the NPs. The focus of this review is to explore various types of NPs and their surface/chemical modifications as well as attachment of targeting ligands for tuning their properties in order to facilitate targeted delivery to the cancer sites in a rate-controlled manner. Heme compatibility, biodistribution, longer circulation time, hydrophilic lipophilic balance for high bioavailability, prevention of drug degradation and leakage are important in transporting drugs to the targeted cancer sites. The review discusses advantages of polymeric, magnetic, gold, and mesoporous silica NPs in delivering chemotherapeutic agents over the conventional dosage formulations along with their shortcomings/risks and possible solutions/alternatives.
Collapse
Affiliation(s)
- Saima Aftab
- Department of Chemistry, Quaid-i-Azam University, 45320 Islamabad, Pakistan
| | - Afzal Shah
- Department of Chemistry, Quaid-i-Azam University, 45320 Islamabad, Pakistan; Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, Tandogan, 06100 Ankara, Turkey.
| | - Akhtar Nadhman
- Department of Chemistry, Quaid-i-Azam University, 45320 Islamabad, Pakistan
| | - Sevinc Kurbanoglu
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, Tandogan, 06100 Ankara, Turkey
| | - Sibel Aysıl Ozkan
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, Tandogan, 06100 Ankara, Turkey
| | - Dionysios D Dionysiou
- Environmental Engineering and Science Program, Department of Biomedical, Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221-0012, USA
| | - Shyam S Shukla
- Department of Chemistry and Biochemistry, Lamar University, Beaumont, TX 77710, USA
| | - Tejraj M Aminabhavi
- Department of Chemistry and Biochemistry, Lamar University, Beaumont, TX 77710, USA.
| |
Collapse
|
32
|
Croissant JG, Fatieiev Y, Almalik A, Khashab NM. Mesoporous Silica and Organosilica Nanoparticles: Physical Chemistry, Biosafety, Delivery Strategies, and Biomedical Applications. Adv Healthc Mater 2018; 7. [PMID: 29193848 DOI: 10.1002/adhm.201700831] [Citation(s) in RCA: 306] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/30/2017] [Indexed: 01/08/2023]
Abstract
Predetermining the physico-chemical properties, biosafety, and stimuli-responsiveness of nanomaterials in biological environments is essential for safe and effective biomedical applications. At the forefront of biomedical research, mesoporous silica nanoparticles and mesoporous organosilica nanoparticles are increasingly investigated to predict their biological outcome by materials design. In this review, it is first chronicled that how the nanomaterial design of pure silica, partially hybridized organosilica, and fully hybridized organosilica (periodic mesoporous organosilicas) governs not only the physico-chemical properties but also the biosafety of the nanoparticles. The impact of the hybridization on the biocompatibility, protein corona, biodistribution, biodegradability, and clearance of the silica-based particles is described. Then, the influence of the surface engineering, the framework hybridization, as well as the morphology of the particles, on the ability to load and controllably deliver drugs under internal biological stimuli (e.g., pH, redox, enzymes) and external noninvasive stimuli (e.g., light, magnetic, ultrasound) are presented. To conclude, trends in the biomedical applications of silica and organosilica nanovectors are delineated, such as unconventional bioimaging techniques, large cargo delivery, combination therapy, gaseous molecule delivery, antimicrobial protection, and Alzheimer's disease therapy.
Collapse
Affiliation(s)
- Jonas G. Croissant
- Chemical and Biological Engineering; University of New Mexico; 210 University Blvd NE Albuquerque NM 87131-0001 USA
- Center for Micro-Engineered Materials; Advanced Materials Laboratory; University of New Mexico; MSC04 2790, 1001 University Blvd SE Suite 103 Albuquerque NM 87106 USA
| | - Yevhen Fatieiev
- Smart Hybrid Materials Laboratory (SHMs); Advanced Membranes and Porous Materials Center; King Abdullah University of Science and Technology; Thuwal Riyadh KSA 11442 Saudi Arabia
| | - Abdulaziz Almalik
- Life sciences and Environment Research Institute; Center of Excellence in Nanomedicine (CENM); King Abdulaziz City for Science and Technology (KACST); Riyadh 11461 Saudi Arabia
| | - Niveen M. Khashab
- Smart Hybrid Materials Laboratory (SHMs); Advanced Membranes and Porous Materials Center; King Abdullah University of Science and Technology; Thuwal Riyadh KSA 11442 Saudi Arabia
| |
Collapse
|
33
|
Zhu D, Yan H, Zhou Z, Tang J, Liu X, Hartmann R, Parak WJ, Feliu N, Shen Y. Detailed investigation on how the protein corona modulates the physicochemical properties and gene delivery of polyethylenimine (PEI) polyplexes. Biomater Sci 2018; 6:1800-1817. [DOI: 10.1039/c8bm00128f] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Given the various cationic polymers developed as non-viral gene delivery vectors, polyethylenimine (PEI) has been/is frequently used in in vitro transfection.
Collapse
Affiliation(s)
- Dingcheng Zhu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
- Fachbereich Physik
| | - Huijie Yan
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
- Fachbereich Physik
| | - Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
| | - Jianbin Tang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
| | - Xiangrui Liu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
| | | | - Wolfgang J. Parak
- Fachbereich Physik
- Philipps Universität Marburg
- Germany
- Fachbereich Physik und Chemie and CHyN
- Universität Hamburg
| | - Neus Feliu
- Fachbereich Physik
- Philipps Universität Marburg
- Germany
- Fachbereich Physik und Chemie and CHyN
- Universität Hamburg
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
| |
Collapse
|
34
|
Bae SE, Lyu SK, Kim KJ, Shin HJ, Kwon H, Huh S. Intracellular delivery of a native functional protein using cell-penetrating peptide functionalized cubic MSNs with ultra-large mesopores. J Mater Chem B 2018; 6:3456-3465. [DOI: 10.1039/c8tb00330k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pore-enlarged 3D cubic cMSNs were successfully prepared, and their surfaces were functionalized by a cell-penetrating R8-peptide through the click reaction for cytosolic delivery of a functional protein in its native form.
Collapse
Affiliation(s)
- Sang-Eun Bae
- Department of Chemistry and Protein Research Center for Bio-Industry
- Hankuk University of Foreign Studies
- Yongin 17035
- Republic of Korea
| | - Soo Kyung Lyu
- Department of Bioscience and Biotechnology and Protein Research Center for Bio-Industry
- Hankuk University of Foreign Studies
- Yongin 17035
- Republic of Korea
| | - Ki-Jung Kim
- Department of Chemistry and Protein Research Center for Bio-Industry
- Hankuk University of Foreign Studies
- Yongin 17035
- Republic of Korea
| | - Hee Joo Shin
- Department of Bioscience and Biotechnology and Protein Research Center for Bio-Industry
- Hankuk University of Foreign Studies
- Yongin 17035
- Republic of Korea
| | - Hyockman Kwon
- Department of Bioscience and Biotechnology and Protein Research Center for Bio-Industry
- Hankuk University of Foreign Studies
- Yongin 17035
- Republic of Korea
| | - Seong Huh
- Department of Chemistry and Protein Research Center for Bio-Industry
- Hankuk University of Foreign Studies
- Yongin 17035
- Republic of Korea
| |
Collapse
|
35
|
Chesson CB, Zloza A. Nanoparticles: augmenting tumor antigen presentation for vaccine and immunotherapy treatments of cancer. Nanomedicine (Lond) 2017; 12:2693-2706. [PMID: 29098928 PMCID: PMC5704090 DOI: 10.2217/nnm-2017-0254] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/18/2017] [Indexed: 02/08/2023] Open
Abstract
The major goal of immunity is maintaining host survival. Toward this, immune cells recognize and eliminate targets that pose a danger. Primarily, these are external invaders (pathogens) and internal invaders (cancers). Their recognition relies on distinguishing foreign components (antigens) from self-antigens. Since cancer cells are the host's own cells that are harmfully altered, they are difficult to distinguish from normal self. Furthermore, the antigens least resembling the host are often sequestered in parts of the tumor least accessible to immune responses. Therefore, to sufficiently boost immunity, these tumor antigens must be exposed to the immune system. Toward this, nanoparticles provide an innovating means of tumor antigen presentation and are destined to become an integral part of cancer immunotherapy.
Collapse
Affiliation(s)
- Charles B Chesson
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Andrew Zloza
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ 08903, USA
| |
Collapse
|
36
|
Martínez-Carmona M, Lozano D, Baeza A, Colilla M, Vallet-Regí M. A novel visible light responsive nanosystem for cancer treatment. NANOSCALE 2017; 9:15967-15973. [PMID: 29019495 DOI: 10.1039/c7nr05050j] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A novel singlet-oxygen sensitive drug delivery nanocarrier able to release its cargo after exposure to visible (Vis) light from a common lamp is presented. This nanodevice is based on mesoporous silica nanoparticles (MSN) decorated with porphyrin-caps grafted via reactive oxygen species (ROS)-cleavable linkages. In the presence of Vis light porphyrin-nanocaps produce singlet oxygen molecules that break the sensitive-linker, which triggers pore uncapping and therefore allows the release of the entrapped cargo (topotecan, TOP). This new system takes advantage of the non-toxicity and greater penetration capacity of Vis radiation and a double antitumor effect due to the drug release and the ROS production. In vitro tests with HOS osteosarcoma cancer cells reveal that TOP is able to be released in a controlled fashion inside the tumor cells. This research work constitutes a proof of concept that opens up promising expectations in the search for new alternatives for the treatment of cancer.
Collapse
Affiliation(s)
- M Martínez-Carmona
- Dpto. Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid. Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12. Plaza Ramón y Cajal s/n, 28040 Madrid, Spain.
| | | | | | | | | |
Collapse
|
37
|
Hu C, Huang P, Zheng Z, Yang Z, Wang X. A Facile Strategy to Prepare an Enzyme-Responsive Mussel Mimetic Coating for Drug Delivery Based on Mesoporous Silica Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:5511-5518. [PMID: 28486810 DOI: 10.1021/acs.langmuir.7b01316] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Surface functional mesoporous silica nanoparticles (MSNs) have been widely used as promosing materials for drug delivery. Herein, we reported a facile strategy to construct MSNs coated by enzyme-resposive polylysine-dopamine (PLDA) films through self-polymerization of dopamine derivative lysine-dopamine, in which the drug could be loaded and delivered efficiently. In detail, RhB or DOX was used as a drug model and loaded in functional MSNs via a one-pot procedure among MSNs, drug, and lysine-dopamine (LDA) under basic conditions. Owing to the fact that the peptide bonds between lysine and dopamine can be cleaved under triggering by pepsin, the resulting RhB/DOX@PLDA-MSNs exibit enzyme-responsive characterization. After the DOX@PLDA-MSNs enter into the cancer cells, the drug can be released effectively through degradation of peptide bonds under the influence of enzyme in cancer cells, which shows marked anticancer activity in vitro. This facile strategy may provide a new platform to construct enzyme-responsive controlled drug delivery systems.
Collapse
Affiliation(s)
- Chunlin Hu
- School of Chemistry and Chemical Technology, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Ping Huang
- School of Chemistry and Chemical Technology, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Zhen Zheng
- School of Chemistry and Chemical Technology, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Zhibiao Yang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Xinling Wang
- School of Chemistry and Chemical Technology, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , Shanghai 200240, China
| |
Collapse
|
38
|
Liu Y, Xu CF, Iqbal S, Yang XZ, Wang J. Responsive Nanocarriers as an Emerging Platform for Cascaded Delivery of Nucleic Acids to Cancer. Adv Drug Deliv Rev 2017; 115:98-114. [PMID: 28396204 DOI: 10.1016/j.addr.2017.03.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/19/2022]
Abstract
Cascades of systemic and intracellular obstacles, including low stability in blood, little tumor accumulation, weak tumor penetration, poor cellular uptake, inefficient endosomal escape and deficient disassembly in the cytoplasm, must be overcome in order to deliver nucleic acid drugs for cancer therapy. Nanocarriers that are sensitive to a variety of physiological stimuli, such as pH, redox status, and cell enzymes, are substantially changing the landscape of nucleic acid drug delivery by helping to overcome cascaded systemic and intracellular barriers. This review discusses nucleic acid-based therapeutics, systemic and intracellular barriers to efficient nucleic acid delivery, and nanocarriers responsive to extracellular and intracellular biological stimuli to overcome individual barriers. In particular, responsive nanocarriers for the cascaded delivery of nucleic acids in vivo are highlighted. Developing novel cascaded nanocarriers that transform their physicochemical properties in response to various stimuli in a timely and spatially controlled manner for nucleic acid drug delivery holds great potential for translating the promise of nucleic acid drugs and achieving clinically successful cancer therapy.
Collapse
|
39
|
Zhou M, Du X, Li W, Li X, Huang H, Liao Q, Shi B, Zhang X, Zhang M. One-pot synthesis of redox-triggered biodegradable hybrid nanocapsules with a disulfide-bridged silsesquioxane framework for promising drug delivery. J Mater Chem B 2017; 5:4455-4469. [DOI: 10.1039/c6tb03368g] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Redox-triggered biodegradable hybrid nanocapsules with the disulfide-bridged silsesquioxane framework were developed for promising drug delivery.
Collapse
Affiliation(s)
- Mengyun Zhou
- Research Center for Bioengineering and Sensing Technology
- Beijing Key Laboratory for Bioengineering and Sensing Technology
- School of Chemistry and Biological Engineering
- University of Science & Technology Beijing
- Beijing 100083
| | - Xin Du
- Research Center for Bioengineering and Sensing Technology
- Beijing Key Laboratory for Bioengineering and Sensing Technology
- School of Chemistry and Biological Engineering
- University of Science & Technology Beijing
- Beijing 100083
| | - Weike Li
- Research Center for Bioengineering and Sensing Technology
- Beijing Key Laboratory for Bioengineering and Sensing Technology
- School of Chemistry and Biological Engineering
- University of Science & Technology Beijing
- Beijing 100083
| | - Xiaoyu Li
- National Engineering Laboratory for Hydrometallurgical Cleaner Production Technology
- State Key Laboratory of Biochemical Engineering
- Key Laboratory of Green Process and Engineering
- Institute of Process Engineering
- Chinese Academic of Sciences
| | - Hongwei Huang
- School of Materials Science and Technology
- China University of Geosciences
- Beijing 100083
- China
| | - Qingliang Liao
- State Key Laboratory for Advanced Metals and Materials
- School of Materials Science and Engineering
- Beijing Municipal Key Laboratory of New Energy Materials and Technologies
- University of Science and Technology Beijing
- Beijing 100083
| | - Bingyang Shi
- Faculty of Medicine and Health Sciences
- Macquarie University
- Sydney
- Australia
| | - Xueji Zhang
- Research Center for Bioengineering and Sensing Technology
- Beijing Key Laboratory for Bioengineering and Sensing Technology
- School of Chemistry and Biological Engineering
- University of Science & Technology Beijing
- Beijing 100083
| | - Meiqin Zhang
- Research Center for Bioengineering and Sensing Technology
- Beijing Key Laboratory for Bioengineering and Sensing Technology
- School of Chemistry and Biological Engineering
- University of Science & Technology Beijing
- Beijing 100083
| |
Collapse
|
40
|
Zhou J, Li J, Wu D, Hong C. CNT-Based and MSN-Based Organic/Inorganic Hybrid Nanocomposites for Biomedical Applications. ACS SYMPOSIUM SERIES 2017. [DOI: 10.1021/bk-2017-1253.ch009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- Jiemei Zhou
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Jiaoyang Li
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Decheng Wu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Chunyan Hong
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| |
Collapse
|
41
|
Lee J, Han S, Lee J, Choi M, Kim C. Stimuli-responsive α-helical peptide gatekeepers for mesoporous silica nanocarriers. NEW J CHEM 2017. [DOI: 10.1039/c7nj00124j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A stimuli-responsive α-helical peptide, as a gatekeeper on the surface of mesoporous silica nanoparticles, efficiently controlled the release of entrapped drugs through triggered conformational conversion and effectively disrupted lipid membranes.
Collapse
Affiliation(s)
- Jeonghun Lee
- Department of Polymer Science and Engineering
- Inha University
- Incheon 22212
- Korea
| | - Seungjong Han
- Department of Polymer Science and Engineering
- Inha University
- Incheon 22212
- Korea
| | - Jinyoung Lee
- Department of Polymer Science and Engineering
- Inha University
- Incheon 22212
- Korea
| | - Minhyuek Choi
- Department of Polymer Science and Engineering
- Inha University
- Incheon 22212
- Korea
| | - Chulhee Kim
- Department of Polymer Science and Engineering
- Inha University
- Incheon 22212
- Korea
| |
Collapse
|
42
|
Zhang Y, Qu Q, Cao X, Zhao Y. NIR-absorbing dye functionalized hollow mesoporous silica nanoparticles for combined photothermal–chemotherapy. Chem Commun (Camb) 2017; 53:12032-12035. [DOI: 10.1039/c7cc07897h] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multifunctional nanocarriers consisting of hollow mesoporous silica nanoparticles loaded with doxorubicin and then capped by a complex between disulfide linked β-cyclodextrin and adamantane functionalized indocyanine dye are developed for improved anticancer efficacy through combined photothermal–chemotherapy.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Chemical Engineering, Huaihai Institute of Technology
- Lianyungang
- P. R. China
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University
- Singapore
| | - Qiuyu Qu
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University
- Singapore
- Singapore
| | - Xiang Cao
- School of Chemical Engineering, Huaihai Institute of Technology
- Lianyungang
- P. R. China
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University
- Singapore
- Singapore
- School of Materials Science and Engineering, Nanyang Technological University
- Singapore 639798
| |
Collapse
|
43
|
Bottecchia C, Erdmann N, Tijssen PMA, Milroy LG, Brunsveld L, Hessel V, Noël T. Batch and Flow Synthesis of Disulfides by Visible-Light-Induced TiO2 Photocatalysis. CHEMSUSCHEM 2016; 9:1781-1785. [PMID: 27329945 DOI: 10.1002/cssc.201600602] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Indexed: 06/06/2023]
Abstract
A mild and practical method for the preparation of disulfides through visible-light-induced photocatalytic aerobic oxidation of thiols has been developed. The method involves the use of TiO2 as a heterogeneous photocatalyst. The catalyst's high stability and recyclability makes this method highly practical. The reaction can be substantially accelerated in a continuous-flow packed-bed reactor, which enables a safe and reliable scale-up of the reaction conditions. The batch and flow protocol described herein can be applied to a diverse set of thiol substrates for the preparation of homo- and hetero-dimerized disulfides. Furthermore, biocompatible reaction conditions (i.e., room temperature, visible light, neutral buffer solution, and no additional base) have been developed, which permits the rapid and chemoselective modification of densely functionalized peptide substrates without recourse to complex purification steps.
Collapse
Affiliation(s)
- Cecilia Bottecchia
- Department of Chemical Engineering and Chemistry, Micro Flow Chemistry & Process Technology, Eindhoven University of Technology, Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Nico Erdmann
- Department of Chemical Engineering and Chemistry, Micro Flow Chemistry & Process Technology, Eindhoven University of Technology, Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Patricia M A Tijssen
- Department of Chemical Engineering and Chemistry, Micro Flow Chemistry & Process Technology, Eindhoven University of Technology, Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Lech-Gustav Milroy
- Laboratory of Chemical Biology and, Institute of Complex Molecular Systems (ICMS), Department of Biomedical Engineering, Eindhoven University of Technology, Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology and, Institute of Complex Molecular Systems (ICMS), Department of Biomedical Engineering, Eindhoven University of Technology, Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Volker Hessel
- Department of Chemical Engineering and Chemistry, Micro Flow Chemistry & Process Technology, Eindhoven University of Technology, Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Timothy Noël
- Department of Chemical Engineering and Chemistry, Micro Flow Chemistry & Process Technology, Eindhoven University of Technology, Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands.
| |
Collapse
|
44
|
Zheng X, Li Z, Chen L, Xie Z, Jing X. Self-Assembly of Porphyrin-Paclitaxel Conjugates Into Nanomedicines: Enhanced Cytotoxicity due to Endosomal Escape. Chem Asian J 2016; 11:1780-4. [DOI: 10.1002/asia.201600423] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/14/2016] [Indexed: 02/02/2023]
Affiliation(s)
- Xiaohua Zheng
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Zhensheng Li
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 China
| | - Li Chen
- Department of Chemistry; Northeast Normal University; 5628 Renmin Street Changchun 130024 P. R. China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 China
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 China
| |
Collapse
|
45
|
Zhang Y, Ang CY, Li M, Tan SY, Qu Q, Zhao Y. Polymeric Prodrug Grafted Hollow Mesoporous Silica Nanoparticles Encapsulating Near-Infrared Absorbing Dye for Potent Combined Photothermal-Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2016; 8:6869-6879. [PMID: 26937591 DOI: 10.1021/acsami.6b00376] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In this study, polymeric prodrug coated hollow mesoporous silica nanoparticles (HMSNs) with encapsulated near-infrared (NIR) absorbing dye were prepared and explored for combined photothermal-chemotherapy. A copolymer integrated with tert-butoxycarbonyl protected hydrazide groups and oligoethylene glycols was initially grafted on the surface of HMSNs via reversible addition-fragmentation chain-transfer (RAFT) polymerization followed by the deprotection to reactivate the hydrazide groups for the conjugation of anticancer drug doxorubicin (DOX). DOX was covalently bound onto the polymer substrate by acid-labile hydrazone bond and released quickly in weak acidic environment for chemotherapy. The hollow cavity of HMSNs was loaded with an NIR absorbing dye IR825 to form the final multifunctional hybrid denoted as HMSNs-DOX/IR825. The hybrid exhibited good dispersity and stability as well as high light-to-heat conversion efficiency. As revealed by confocal microscopy and flow cytometry analysis, the hybrid was efficiently taken up by cancer cells, and the conjugated DOX could be released under the cellular environment. In vitro cytotoxicity study demonstrated that anticancer activity of HMSNs-DOX/IR825 could be significantly improved by the NIR irradiation, which led to a satisfactory therapeutic efficacy through the combination treatment. Thus, the developed hybrid could be a promising candidate for the combined photothermal-chemotherapy of cancer.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, Singapore 637371, Singapore
| | - Chung Yen Ang
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, Singapore 637371, Singapore
| | - Menghuan Li
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, Singapore 637371, Singapore
| | - Si Yu Tan
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, Singapore 637371, Singapore
| | - Qiuyu Qu
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, Singapore 637371, Singapore
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, Singapore 637371, Singapore
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
46
|
Gunnoo SB, Madder A. Chemical Protein Modification through Cysteine. Chembiochem 2016; 17:529-53. [DOI: 10.1002/cbic.201500667] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Smita B. Gunnoo
- Organic & Biomimetic Chemistry Research Group; Department of Organic and Macromolecular Chemistry; Ghent University; Krijgslaan 281 9000 Gent Belgium
| | - Annemieke Madder
- Organic & Biomimetic Chemistry Research Group; Department of Organic and Macromolecular Chemistry; Ghent University; Krijgslaan 281 9000 Gent Belgium
| |
Collapse
|
47
|
Decan N, Wu D, Williams A, Bernatchez S, Johnston M, Hill M, Halappanavar S. Characterization of in vitro genotoxic, cytotoxic and transcriptomic responses following exposures to amorphous silica of different sizes. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 796:8-22. [PMID: 26778505 DOI: 10.1016/j.mrgentox.2015.11.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/18/2015] [Accepted: 11/23/2015] [Indexed: 11/25/2022]
Abstract
The objectives of the present study were to investigate the underlying mechanisms of genetic and cellular toxicity induced by silica nanoparticles (SiNPs) and determine if such toxicity is influenced by particle size. Commercially available amorphous SiNPs (12 nm, 5-10 nm, and 10-15 nm) and micrometer sized (SiP2 μm) silica were characterised for size, chemical composition, and aggregation state. Mouse lung epithelial (FE1) cells derived from Muta™Mouse were exposed to various concentrations (12.5, 25, 50, 100 μg/ml) of SiNPs and SiP2 μm. Cellular viability, clonogenic potential, oxidative stress, micronucleus formation, and mutant frequency were measured at different post-exposure time points. Cellular internalization of particles was assessed using nanoscale hyperspectral microscopy. Biological pathway and functional perturbations were assessed using DNA microarrays. Detailed characterization of particles confirmed their size, purity, and uniform dispersion in the exposure medium. Decreased cellular viability was observed acutely at 24h at concentrations higher than 25 μg/ml for all particle types, with SiNPs being the most sensitive; loss of viability was surface area dependent at the lowest concentration tested. However, only SiNP12 showed poor long-term survival. A size-dependent increase in micronucleus formation was also observed for SiNPs. In contrast to the viability results, SiP2 μm exhibited the highest potential to induce oxidative stress compared to the SiNPs at all tested concentrations. Gene ontology and biological pathway analysis revealed significant changes in the expression of genes implicated in lysosomal functions in SiNP12-treated cells, which appear closely associated with higher SiNP12 internalization and lysosomal rearrangements in the cytoplasm of these cells. These results suggest that SiNPs induce cellular and genetic toxicity in a size-dependent manner and that the observed toxicity may be the results of higher particle internalization of smaller SiNP and subsequent lysosomal overload.
Collapse
Affiliation(s)
- Nathalie Decan
- Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Canada
| | - Dongmei Wu
- Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Canada
| | - Stéphane Bernatchez
- New Substances Assessment and Control Bureau, Safe Environments Directorate, Health Canada, Ottawa, Canada
| | - Michael Johnston
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, Canada
| | - Myriam Hill
- New Substances Assessment and Control Bureau, Safe Environments Directorate, Health Canada, Ottawa, Canada
| | - Sabina Halappanavar
- Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Canada.
| |
Collapse
|
48
|
Heidegger S, Gössl D, Schmidt A, Niedermayer S, Argyo C, Endres S, Bein T, Bourquin C. Immune response to functionalized mesoporous silica nanoparticles for targeted drug delivery. NANOSCALE 2016; 8:938-48. [PMID: 26659601 DOI: 10.1039/c5nr06122a] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Multifunctional mesoporous silica nanoparticles (MSN) have attracted substantial attention with regard to their high potential for targeted drug delivery. For future clinical applications it is crucial to address safety concerns and understand the potential immunotoxicity of these nanoparticles. In this study, we assess the biocompatibility and functionality of multifunctional MSN in freshly isolated, primary murine immune cells. We show that the functionalized silica nanoparticles are rapidly and efficiently taken up into the endosomal compartment by specialized antigen-presenting cells such as dendritic cells. The silica nanoparticles showed a favorable toxicity profile and did not affect the viability of primary immune cells from the spleen in relevant concentrations. Cargo-free MSN induced only very low immune responses in primary cells as determined by surface expression of activation markers and release of pro-inflammatory cytokines such as Interleukin-6, -12 and -1β. In contrast, when surface-functionalized MSN with a pH-responsive polymer capping were loaded with an immune-activating drug, the synthetic Toll-like receptor 7 agonist R848, a strong immune response was provoked. We thus demonstrate that MSN represent an efficient drug delivery vehicle to primary immune cells that is both non-toxic and non-inflammagenic, which is a prerequisite for the use of these particles in biomedical applications.
Collapse
Affiliation(s)
- Simon Heidegger
- Center for Integrated Protein Science Munich (CIPSM), Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, 80336 Munich, Germany. and III. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Dorothée Gössl
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU), 81377 Munich, Germany.
| | - Alexandra Schmidt
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU), 81377 Munich, Germany.
| | - Stefan Niedermayer
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU), 81377 Munich, Germany.
| | - Christian Argyo
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU), 81377 Munich, Germany.
| | - Stefan Endres
- Center for Integrated Protein Science Munich (CIPSM), Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| | - Thomas Bein
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU), 81377 Munich, Germany.
| | - Carole Bourquin
- Center for Integrated Protein Science Munich (CIPSM), Division of Clinical Pharmacology, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, 80336 Munich, Germany. and Chair of Pharmacology, Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
49
|
Botella P, Muniesa C, Vicente V, Cabrera-García A. Effect of drug precursor in cell uptake and cytotoxicity of redox-responsive camptothecin nanomedicines. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 58:692-9. [DOI: 10.1016/j.msec.2015.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/04/2015] [Accepted: 09/02/2015] [Indexed: 12/29/2022]
|
50
|
Liu Y, Pei Q, Chen L, Li Z, Xie Z. Reduction-responsive fluorescence off–on BODIPY–camptothecin conjugates for self-reporting drug release. J Mater Chem B 2016; 4:2332-2337. [DOI: 10.1039/c6tb00009f] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A reduction-responsive fluorescence off–on theranostic prodrug with self-reporting drug release was constructed based on boron dipyrromethene (BODIPY) and therapeutic drug camptothecin (CPT) with a long flexible disulfide linker.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry
- Northeast Normal University
- Changchun 130024
- P. R. China
- State Key Laboratory of Polymer Physics and Chemistry
| | - Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Li Chen
- Department of Chemistry
- Northeast Normal University
- Changchun 130024
- P. R. China
| | - Zhensheng Li
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| |
Collapse
|