101
|
Tanaka H, Takata N, Sakurai Y, Yoshida T, Inoue T, Tamagawa S, Nakai Y, Tange K, Yoshioka H, Maeki M, Tokeshi M, Akita H. Delivery of Oligonucleotides Using a Self-Degradable Lipid-Like Material. Pharmaceutics 2021; 13:pharmaceutics13040544. [PMID: 33924589 PMCID: PMC8070490 DOI: 10.3390/pharmaceutics13040544] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/05/2021] [Accepted: 04/05/2021] [Indexed: 11/16/2022] Open
Abstract
The world-first success of lipid nanoparticle (LNP)-based siRNA therapeutics (ONPATTRO®) promises to accelerate developments in siRNA therapeutics/gene therapy using LNP-type drug delivery systems (DDS). In this study, we explore the optimal composition of an LNP containing a self-degradable material (ssPalmO-Phe) for the delivery of oligonucleotides. siRNA or antisense oligonucleotides (ASO) were encapsulated in LNP with different lipid compositions. The hepatic knockdown efficiency of the target genes and liver toxicity were evaluated. The optimal compositions for the siRNA were different from those for ASO, and different from those for mRNA that were reported in a previous study. Extracellular stability, endosomal escape and cellular uptake appear to be the key processes for the successful delivery of mRNA, siRNA and ASO, respectively. Moreover, the compositions of the LNPs likely contribute to their toxicity. The lipid composition of the LNP needs to be optimized depending on the type of nucleic acids under consideration if the applications of LNPs are to be further expanded.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba 260-0856, Japan; (H.T.); (N.T.); (Y.S.)
| | - Nae Takata
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba 260-0856, Japan; (H.T.); (N.T.); (Y.S.)
| | - Yu Sakurai
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba 260-0856, Japan; (H.T.); (N.T.); (Y.S.)
| | - Tokuyuki Yoshida
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan; (T.Y.); (T.I.)
| | - Takao Inoue
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan; (T.Y.); (T.I.)
| | - Shinya Tamagawa
- DDS Research Laboratory, NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan; (S.T.); (Y.N.); (K.T.); (H.Y.)
| | - Yuta Nakai
- DDS Research Laboratory, NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan; (S.T.); (Y.N.); (K.T.); (H.Y.)
| | - Kota Tange
- DDS Research Laboratory, NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan; (S.T.); (Y.N.); (K.T.); (H.Y.)
| | - Hiroki Yoshioka
- DDS Research Laboratory, NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan; (S.T.); (Y.N.); (K.T.); (H.Y.)
| | - Masatoshi Maeki
- Division of Applied Chemistry, Faculty of Engineering, Hokkaido University, Kita 13, Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan; (M.M.); (M.T.)
| | - Manabu Tokeshi
- Division of Applied Chemistry, Faculty of Engineering, Hokkaido University, Kita 13, Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan; (M.M.); (M.T.)
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba 260-0856, Japan; (H.T.); (N.T.); (Y.S.)
- Correspondence: ; Tel.: +81-43-226-2893
| |
Collapse
|
102
|
Olim F, Neves AR, Vieira M, Tomás H, Sheng R. Self‐Assembly of Cholesterol‐Doxorubicin and TPGS into Prodrug‐Based Nanoparticles with Enhanced Cellular Uptake and Lysosome‐Dependent Pathway in Breast Cancer Cells. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Filipe Olim
- CQM – Centro de Química da Madeira, MMRG Universidade da Madeira Campus da Penteada Funchal 9020‐105 Portugal
| | - Ana Rute Neves
- CQM – Centro de Química da Madeira, MMRG Universidade da Madeira Campus da Penteada Funchal 9020‐105 Portugal
| | - Mariana Vieira
- CQM – Centro de Química da Madeira, MMRG Universidade da Madeira Campus da Penteada Funchal 9020‐105 Portugal
| | - Helena Tomás
- CQM – Centro de Química da Madeira, MMRG Universidade da Madeira Campus da Penteada Funchal 9020‐105 Portugal
| | - Ruilong Sheng
- CQM – Centro de Química da Madeira, MMRG Universidade da Madeira Campus da Penteada Funchal 9020‐105 Portugal
| |
Collapse
|
103
|
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: 0.8] [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
|
104
|
Lipid Nanoparticle-Mediated Lymphatic Delivery of Immunostimulatory Nucleic Acids. Pharmaceutics 2021; 13:pharmaceutics13040490. [PMID: 33916667 PMCID: PMC8103501 DOI: 10.3390/pharmaceutics13040490] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/22/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022] Open
Abstract
Lymphatic delivery of a vaccine can be achieved using a dendritic cell (DC)-targeted delivery system that can cause DC to migrate to lymph nodes upon activation by an adjuvant. Here, we designed a mannose-modified cationic lipid nanoparticle (M-NP) to deliver the nucleic acid adjuvant, polyinosinic:polycytidylic acid (PIC). PIC-loaded M-NP (PIC/M-NP) showed stable lipoplexes regardless of the ligand ratio and negligible cytotoxicity in bone marrow-derived DC. DC uptake of PIC/M-NP was demonstrated, and an increased mannose ligand ratio improved DC uptake efficiency. PIC/M-NP significantly promoted the maturation of bone marrow-derived DC, and local injection of PIC/M-NP to mice facilitated lymphatic delivery and activation (upon NP uptake) of DC. Our results support the potential of PIC/M-NP in delivering a nucleic acid adjuvant for the vaccination of antigens.
Collapse
|
105
|
Fisher RK, West PC, Mattern-Schain SI, Best MD, Kirkpatrick SS, Dieter RA, Arnold JD, Buckley MR, McNally MM, Freeman MB, Grandas OH, Mountain DJH. Advances in the Formulation and Assembly of Non-Cationic Lipid Nanoparticles for the Medical Application of Gene Therapeutics. NANOMATERIALS 2021; 11:nano11030825. [PMID: 33807086 PMCID: PMC8004789 DOI: 10.3390/nano11030825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022]
Abstract
Lipid nanoparticles have become increasingly popular delivery platforms in the field of gene therapy, but bench-to-bedside success has been limited. Many liposomal gene vectors are comprised of synthetic cationic lipids, which are associated with lipid-induced cytotoxicity and immunogenicity. Natural, non-cationic PEGylated liposomes (PLPs) demonstrate favorable biocompatibility profiles but are not considered viable gene delivery vehicles due to inefficient nucleic acid loading and reduced cellular uptake. PLPs can be modified with cell-penetrating peptides (CPPs) to enhance the intracellular delivery of liposomal cargo but encapsulate leakage upon CPP-PLP assembly is problematic. Here, we aimed to identify parameters that overcome these performance barriers by incorporating nucleic acid condensers during CPP-PLP assembly and screening variable ethanol injection parameters for optimization. CPP-PLPs were formed with R8-amphiphiles via pre-insertion, post-insertion and post-conjugation techniques and liposomes were characterized for size, surface charge, homogeneity, siRNA encapsulation efficiency and retention and cell associative properties. Herein we demonstrate that pre-insertion of stearylated R8 into PLPs is an efficient method to produce non-cationic CPP-PLPs and we provide additional assembly parameter specifications for a modified ethanol injection technique that is optimized for siRNA encapsulation/retention and enhanced cell association. This assembly technique could provide improved clinical translation of liposomal based gene therapy applications.
Collapse
Affiliation(s)
- Richard K. Fisher
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Phillip C. West
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Samuel I. Mattern-Schain
- Department of Chemistry, University of Tennessee Knoxville, 1420 Circle Drive, Knoxville, TN 37996, USA; (S.I.M.-S.); (M.D.B.)
| | - Michael D. Best
- Department of Chemistry, University of Tennessee Knoxville, 1420 Circle Drive, Knoxville, TN 37996, USA; (S.I.M.-S.); (M.D.B.)
| | - Stacy S. Kirkpatrick
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Raymond A. Dieter
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Joshua D. Arnold
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Michael R. Buckley
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Michael M. McNally
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Michael B. Freeman
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Oscar H. Grandas
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
| | - Deidra J. H. Mountain
- Department of Surgery, University of Tennessee Graduate School of Medicine, 1924 Alcoa Highway Box U-11, Knoxville, TN 37920, USA; (R.K.F.III); (P.C.W.); (S.S.K.); (R.A.D.III); (J.D.A.); (M.R.B.); (M.M.M.); (M.B.F.); (O.H.G.)
- Correspondence: ; Tel.: +1-865-305-9160
| |
Collapse
|
106
|
Yuba E, Korenaga T, Harada A. Hydrophilic Hyperbranched Polymer-Coated siRNA/Polyamidoamine Dendron-Bearing Lipid Complexes Preparation for High Colloidal Stability and Efficient RNAi. Bioconjug Chem 2021; 32:563-571. [PMID: 33660999 DOI: 10.1021/acs.bioconjchem.1c00035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
RNA interference (RNAi) using siRNA has gained much attention for use in therapies for cancer and genetic disorders. To establish RNAi-based therapeutics, the development of efficient siRNA nanocarriers is desired. Earlier, we developed polyamidoamine dendron-bearing lipids able to form complexes with nucleic acids as gene vectors. Especially, dendron lipids with unsaturated alkyl chains (DL-G1-U2) induced efficient endosomal escape by membrane fusion, leading to efficient transfection in vitro. For this study, dendron lipids having oleyl/linoleyl groups (DL-G1-U3) were designed to increase membrane fusogenic activity further. Indeed, DL-G1-U3/siRNA complexes achieved higher membrane fusogenic activity and knockdown of the target gene more efficiently than conventional DL-G1-U2/siRNA complexes did. A hydrophilic polymer, hyperbranched polyglycidol lauryl ester (HPG-Lau), was modified further on the surface of DL-G1-U3/siRNA complexes to provide colloidal stability. Surface modification of HPG-Lau increased the colloidal stability in a physiological condition more than complexes without HPG-Lau. Importantly, HPG-Lau-coated DL/siRNA complexes showed identical RNAi effects to those of parental DL/siRNA complexes, whereas the RNAi activity of poly(ethylene glycol)-bearing lipid (PEG-PE)-modified DL/siRNA complexes was hindered completely. Introduction of unsaturated bonds into dendron lipids and selection of suitable hydrophilic polymers for nanocarrier modification are important for obtaining efficient siRNA vectors toward in vivo siRNA delivery.
Collapse
Affiliation(s)
- Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan
| | - Takashi Korenaga
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan
| | - Atsushi Harada
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan
| |
Collapse
|
107
|
Nguyen KT, Go G, Jin Z, Darmawan BA, Yoo A, Kim S, Nan M, Lee SB, Kang B, Kim C, Li H, Bang D, Park J, Choi E. A Magnetically Guided Self-Rolled Microrobot for Targeted Drug Delivery, Real-Time X-Ray Imaging, and Microrobot Retrieval. Adv Healthc Mater 2021; 10:e2001681. [PMID: 33506630 DOI: 10.1002/adhm.202001681] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/22/2020] [Indexed: 12/19/2022]
Abstract
Targeted drug delivery using a microrobot is a promising technique capable of overcoming the limitations of conventional chemotherapy that relies on body circulation. However, most studies of microrobots used for drug delivery have only demonstrated simple mobility rather than precise targeting methods and prove the possibility of biodegradation of implanted microrobots after drug delivery. In this study, magnetically guided self-rolled microrobot that enables autonomous navigation-based targeted drug delivery, real-time X-ray imaging, and microrobot retrieval is proposed. The microrobot, composed of a self-rolled body that is printed using focused light and a surface with magnetic nanoparticles attached, demonstrates the loading of doxorubicin and an X-ray contrast agent for cancer therapy and X-ray imaging. The microrobot is precisely mobilized to the lesion site through automated targeting using magnetic field control of an electromagnetic actuation system under real-time X-ray imaging. The photothermal effect using near-infrared light reveals rapid drug release of the microrobot located at the lesion site. After drug delivery, the microrobot is recovered without potential toxicity by implantation or degradation using a magnetic-field-switchable coiled catheter. This microrobotic approach using automated control method of the therapeutic agents-loaded microrobot has potential use in precise localized drug delivery systems.
Collapse
Affiliation(s)
- Kim Tien Nguyen
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- School of Mechanical Engineering Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| | - Gwangjun Go
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- School of Mechanical Engineering Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| | - Zhen Jin
- School of Biomedical Engineering Xinxiang Medical University Xinxiang Henan 453003 China
| | - Bobby Aditya Darmawan
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- School of Mechanical Engineering Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| | - Ami Yoo
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
| | - Seokjae Kim
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
| | - Minghui Nan
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- School of Mechanical Engineering Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| | - Sang Bong Lee
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
| | - Byungjeon Kang
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- College of AI convergence Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| | - Chang‐Sei Kim
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- School of Mechanical Engineering Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| | - Hao Li
- Department of Mechanical Engineering Yanbian University Yanji 133002 China
| | - Doyeon Bang
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- College of AI convergence Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| | - Jong‐Oh Park
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- School of Mechanical Engineering Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| | - Eunpyo Choi
- Korea Institute of Medical Microrobotics 43‐26, Cheomdangwagi‐ro 208‐beon‐gil, Buk‐gu Gwangju 61011 South Korea
- School of Mechanical Engineering Chonnam National University 77 Yongbong‐ro, Buk‐gu Gwangju 61186 South Korea
| |
Collapse
|
108
|
Lu Z, Laney VEA, Hall R, Ayat N. Environment-Responsive Lipid/siRNA Nanoparticles for Cancer Therapy. Adv Healthc Mater 2021; 10:e2001294. [PMID: 33615743 DOI: 10.1002/adhm.202001294] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/12/2020] [Indexed: 12/14/2022]
Abstract
RNA interference (RNAi) is a promising technology to regulate oncogenes for treating cancer. The primary limitation of siRNA for clinical application is the safe and efficacious delivery of therapeutic siRNA into target cells. Lipid-based delivery systems are developed to protect siRNA during the delivery process and to facilitate intracellular uptake. There is a significant progress in lipid nanoparticle systems that utilize cationic and protonatable amino lipid systems to deliver siRNA to tumors. Among these lipids, environment-responsive lipids are a class of novel lipid delivery systems that are capable of responding to the environment changes during the delivery process and demonstrate great promise for clinical translation for siRNA therapeutics. Protonatable or ionizable amino lipids and switchable lipids as well as pH-sensitive multifunctional amino lipids are the presentative environment-responsive lipids for siRNA delivery. These lipids are able to respond to environmental changes during the delivery process to facilitate efficient cytosolic siRNA delivery. Environment-responsive lipid/siRNA nanoparticles (ERLNP) are developed with the lipids and are tested for efficient delivery of therapeutic siRNA into the cytoplasm of cancer cells to silence target genes for cancer treatment in preclinical development. This review summarizes the recent developments in environment-response lipids and nanoparticles for siRNA delivery in cancer therapy.
Collapse
Affiliation(s)
- Zheng‐Rong Lu
- Department of Biomedical Engineering Case Western Reserve University Cleveland OH 44106 USA
| | - Victoria E. A. Laney
- Department of Biomedical Engineering Case Western Reserve University Cleveland OH 44106 USA
| | - Ryan Hall
- Department of Biomedical Engineering Case Western Reserve University Cleveland OH 44106 USA
| | - Nadia Ayat
- Department of Biomedical Engineering Case Western Reserve University Cleveland OH 44106 USA
| |
Collapse
|
109
|
Chen C, Shen M, Liao H, Guo Q, Fu H, Yu J, Duan Y. A paclitaxel and microRNA-124 coloaded stepped cleavable nanosystem against triple negative breast cancer. J Nanobiotechnology 2021; 19:55. [PMID: 33632232 PMCID: PMC7905927 DOI: 10.1186/s12951-021-00800-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background Triple negative breast cancer (TNBC) is one of the most biologically aggressive breast cancers and lacks effective treatment options, resulting in a poor prognosis. Therefore, studies aiming to explore new therapeutic strategies for advanced TNBC are urgently needed. According to recent studies, microRNA-124 (miR124) not only inhibits tumour growth but also increases the sensitivity of TNBC to paclitaxel (PTX), suggesting that a platform combining PTX and miR124 may be an advanced solution for TNBC. Results Herein, we constructed a stepped cleavable calcium phosphate composite lipid nanosystem (CaP/LNS) to codeliver PTX and miR124 (PTX/miR124-NP). PTX/miR124-NP exhibited superior tumor microenvironment responsive ability, in which the surface PEG layer was shed in the mildly acidic environment of tumor tissues and exposed oligomeric hyaluronic acid (o-HA) facilitated the cellular uptake of CaP/LNS by targeting the CD44 receptor on the surface of tumor cells. Inside tumour cells, o-HA detached from CaP/LNS due to the reduction of disulfide bonds by glutathione (GSH) and inhibited tumour metastasis. Then, PTX and miR124 were sequentially released from CaP/LNS and exerted synergistic antitumour effects by reversing the Epithelial-Mesenchymal Transition (EMT) process in MDA-MB-231 cells. Moreover, PTX/miR124-NP showed significant antitumour efficiency and excellent safety in mice bearing MDA-MB-231 tumours. Conclusion Based on these results, the codelivery of PTX and miR124 by the CaP/LNS nanosystem might be a promising therapeutic strategy for TNBC.![]()
Collapse
Affiliation(s)
- Chuanrong Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Ming Shen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China. .,NHC Key Laboratory of Reproduction Regulation, (Shanghai Institute of Planned Parenthood Research), Fudan University, and Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai, 200032, China.
| | - Hongze Liao
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Qianqian Guo
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jian Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
110
|
Oz UC, Bolat ZB, Ozkose UU, Gulyuz S, Kucukturkmen B, Khalily MP, Ozcubukcu S, Yilmaz O, Telci D, Esendagli G, Sahin F, Bozkir A. A robust optimization approach for the breast cancer targeted design of PEtOx-b-PLA polymersomes. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:111929. [PMID: 33812571 DOI: 10.1016/j.msec.2021.111929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/19/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
The equipping of nanoparticles with the peptide moiety recognizing a particular receptor, enables cell or tissue-specific targeting, therefore the optimization of the targeted nanoparticles is a key factor in the formulation design process. In this paper, we report the optimization concept of Doxorubicin encapsulating PEtOx-b-PLA polymersome formulation equipped with Peptide18, which is a breast cancer recognizing tumor homing peptide, and the unveiling of the cell-specific delivery potential. The most dominant formulation parameters, which are the polymer to Doxorubicin mass ratio (w/w) and the aqueous to organic phase ratio (v/v), were optimized using Central Composite Design (CCD) based Response Surface Methodology. The characteristics of optimum polymersome formulation were determined as the hydrodynamic diameter of 146.35 nm, the PDI value of 0.136, and the encapsulation efficiency of 57.11% and TEM imaging, which are in agreement with the DLS data, showed the spherical morphology of the polymersomes. In order to demonstrate the breast cancer-specific delivery of targeted polymersomes, the flow cytometry and confocal microscopy analyses were carried out. The targeted polymersomes were accumulated 8 times higher in AU565 cells compared to MCF10A cells and the intracellular Doxorubicin was almost 10 times higher in AU565 cells. The CCD-mediated optimized targeted polymersomes proposed in this report holds the promise of targeted therapy for breast cancer and can be potentially used for the development of novel treatments.
Collapse
Affiliation(s)
- Umut Can Oz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Yenimahalle, 06560, Ankara, Turkey
| | - Zeynep Busra Bolat
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 26 Agustos Campus, 34755 Istanbul, Turkey; Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Kucukcekmece, 34303, Istanbul, Turkey
| | - Umut Ugur Ozkose
- Materials Institute, Marmara Research Center, TUBITAK, Gebze 41470, Kocaeli, Turkey; Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Maslak 34469, Istanbul, Turkey; Department of Chemistry, Faculty of Science and Letters, Piri Reis University, Tuzla, 34940, Istanbul, Turkey
| | - Sevgi Gulyuz
- Materials Institute, Marmara Research Center, TUBITAK, Gebze 41470, Kocaeli, Turkey; Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Maslak 34469, Istanbul, Turkey
| | - Berrin Kucukturkmen
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Yenimahalle, 06560, Ankara, Turkey
| | - Melek Parlak Khalily
- Department of Chemistry, Faculty of Science and Letters, Yozgat Bozok University, Yozgat 66200, Turkey
| | - Salih Ozcubukcu
- Department of Chemistry, Faculty of Science, Middle East Technical University, Ankara 06800, Turkey
| | - Ozgur Yilmaz
- Materials Institute, Marmara Research Center, TUBITAK, Gebze 41470, Kocaeli, Turkey
| | - Dilek Telci
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 26 Agustos Campus, 34755 Istanbul, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 26 Agustos Campus, 34755 Istanbul, Turkey
| | - Asuman Bozkir
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Yenimahalle, 06560, Ankara, Turkey.
| |
Collapse
|
111
|
Evaluation of Nanotargeted 111In-Cyclic RGDfK-Liposome in a Human Melanoma Xenotransplantation Model. Int J Mol Sci 2021; 22:ijms22031099. [PMID: 33499267 PMCID: PMC7866009 DOI: 10.3390/ijms22031099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Nanotargeted liposomes may be modified with targeting peptide on the surface of a prepared liposome to endow specificity and elevate targeting efficiency. The aim of this study was to develop a radioactive targeted nanoparticle, the 111In-cyclic RGDfK-liposome, and its advantage of recognizing the αVβ3 integrin was examined. The cyclic RGDfK modified liposomes were demonstrated the ability to bind the αVβ3 integrin expressed on the surface of human melanoma cell in vitro and in vivo. The effects of the cyclic RGDfK-liposome on the functioning of phagocytes was also examined, showing no considerable negative effects on the engulfment of bacteria and the generation of reactive oxygen species. Based upon these findings, the cyclic RGDfK- liposome is said to be a promising agent for tumor imaging.
Collapse
|
112
|
Wang H, Hu H, Yang H, Li Z. Hydroxyethyl starch based smart nanomedicine. RSC Adv 2021; 11:3226-3240. [PMID: 35424303 PMCID: PMC8694170 DOI: 10.1039/d0ra09663f] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/28/2020] [Indexed: 12/28/2022] Open
Abstract
In the past decades, the vigorous development of nanomedicine has opened up a new world for drug delivery. Hydroxyethyl starch (HES), a clinical plasma volume expander which has been widely used for years, is playing an attracting role as drug carriers. Compared with all other polysaccharides, HES has proven its unique characteristics for drug delivery platforms, including good manufacture practice, biodegradability, biocompatibility, abundant groups for chemical modification, excellent water solubility, and tailorability. In this review, an overview of various types of HES based drug delivery systems is provided, including HES-drug conjugates, HES-based nano-assemblies, HES-based nanocapsules, and HES-based hydrogels. In addition, the current challenges and future opportunities for design and application of HES based drug delivery systems are also discussed. The available studies show that HES based drug delivery systems has significant potential for clinical translation.
Collapse
Affiliation(s)
- Huimin Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
| | - Hang Hu
- National and Local Joint Engineering Research Center for High-efficiency Refining and High-quality Utilization of Biomass, School of Pharmaceutical Engineering and Life Sciences, Changzhou University Changzhou 213164 People's Republic of China
| | - Hai Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan 430074 China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology Wuhan 430074 China
| |
Collapse
|
113
|
Mori A, Kobayashi Y, Nirasawa K, Negishi Y, Asayama S. Structure-Activity Relationship of Mono-Ion Complexes for Plasmid DNA Delivery by Muscular Injection. Pharmaceutics 2021; 13:pharmaceutics13010078. [PMID: 33430003 PMCID: PMC7828051 DOI: 10.3390/pharmaceutics13010078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/04/2021] [Accepted: 01/04/2021] [Indexed: 11/16/2022] Open
Abstract
The structure-activity relationship of mono-ion complexes (MICs) for plasmid DNA (pDNA) delivery by muscular injection is demonstrated. MICs were formed between pDNA and monocationic poly(ethylene glycol) (PEG) macromolecules. As monocationic PEGs, the ω-amide-pentylimidazolium (APe-Im) end-modified PEGs with a stable amide (Am) and hydrolytic ester (Es) bond, that is, APe-Im-Am-PEG and APe-Im-Es-PEG, respectively, are synthesized. The difference between the APe-Im-Am-PEG and APe-Im-Es-PEG was only a spacer structure between a terminal cation and a PEG chain. The resulting pDNA MICs with APe-Im-Am-PEG at a charge ratio (+/-) of 32 or 64 were more stable than those with APe-Im-Es-PEG in the presence of serum proteins. The highest gene expression by muscular injection was achieved using the APe-Im-Am-PEG/pDNA MIC at a charge ratio (+/-) of 32 with a smaller particle diameter of approximately 50 nm, as compared to that charge ratio of 64. Consequently, the pDNA MIC with the monocationic PEG with a stable amide spacer, as compared to a hydrolytic ester spacer, is considered to be suitable for the highest gene expression by muscular injection.
Collapse
Affiliation(s)
- Amika Mori
- Department of Applied Chemistry, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan; (A.M.); (Y.K.)
| | - Yuki Kobayashi
- Department of Applied Chemistry, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan; (A.M.); (Y.K.)
| | - Kei Nirasawa
- Department of Drug Delivery and Molecular Biopharmaceutics, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan; (K.N.); (Y.N.)
| | - Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan; (K.N.); (Y.N.)
| | - Shoichiro Asayama
- Department of Applied Chemistry, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan; (A.M.); (Y.K.)
- Correspondence: ; Tel.: +81-42-677-1111 (ext. 4976)
| |
Collapse
|
114
|
Miyazaki T, Nakagawa Y, Cabral H. Strategies for ligand-installed nanocarriers. HANDBOOK OF NANOTECHNOLOGY APPLICATIONS 2021:633-655. [DOI: 10.1016/b978-0-12-821506-7.00024-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
115
|
Sato Y. Development of Lipid Nanoparticles for the Delivery of Macromolecules Based on the Molecular Design of pH-Sensitive Cationic Lipids. Chem Pharm Bull (Tokyo) 2021; 69:1141-1159. [PMID: 34853281 DOI: 10.1248/cpb.c21-00705] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Considerable efforts have been made on the development of lipid nanoparticles (LNPs) for delivering of nucleic acids in LNP-based medicines, including a first-ever short interfering RNA (siRNA) medicine, Onpattro, and the mRNA vaccines against the coronavirus disease 2019 (COVID-19), which have been approved and are currently in use worldwide. The successful rational design of ionizable cationic lipids was a major breakthrough that dramatically increased delivery efficiency in this field. The LNPs would be expected to be useful as a platform technology for the delivery of various therapeutic modalities for genome editing and even for undiscovered therapeutic mechanisms. In this review, the current progress of my research, including the molecular design of pH-sensitive cationic lipids, their applications for various tissues and cell types, and for delivering various macromolecules, including siRNA, antisense oligonucleotide, mRNA, and the clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) system will be described. Mechanistic studies regarding relationships between the physicochemical properties of LNPs, drug delivery, and biosafety are also summarized. Furthermore, current issues that need to be addressed for next generation drug delivery systems are discussed.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University
| |
Collapse
|
116
|
Qiu N, Wang G, Wang J, Zhou Q, Guo M, Wang Y, Hu X, Zhou H, Bai R, You M, Zhang Z, Chen C, Liu Y, Shen Y. Tumor-Associated Macrophage and Tumor-Cell Dually Transfecting Polyplexes for Efficient Interleukin-12 Cancer Gene Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006189. [PMID: 33270281 DOI: 10.1002/adma.202006189] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/03/2020] [Indexed: 05/26/2023]
Abstract
Interleukin 12 (IL12) is a potent pro-inflammatory chemokine with multifunction, including promoting cytotoxic T-cell-mediated killing of cancer cells. IL12-based cancer gene therapy can overcome IL12's life-threatening adverse effects, but its clinical translation has been limited by the lack of systemic gene-delivery vectors capable of efficiently transfecting tumors to produce sufficient local IL12. Macrophages inherently excrete IL12, and tumor-associated macrophages (TAMs) are the major tumor component taking up a large fraction of the vectors arriving in the tumor. It is thus hypothesized that a gene vector efficiently transfecting both cancer cells and TAMs would make the tumor to produce sufficient IL12; however, gene transfection of TAMs is challenging due to their inherent strong degradation ability. Herein, an IL12 gene-delivery vector is designed that efficiently transfects both cancer cells and TAMs to make them as a factory for IL12 production, which efficiently activates anticancer immune responses and remodels the tumor microenvironment, for instance, increasing the M1/M2 ratio by more than fourfold. Therefore, the intravenously administered vector retards tumor growth and doubles survival in three animal models' with negligible systemic toxicities. This work reports the first nonviral IL12 gene delivery system that effectively makes use of both macrophages and tumor cells.
Collapse
Affiliation(s)
- Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Guowei Wang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Jinqiang Wang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Yaling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Xuhao Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Ru Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Min You
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Zhen Zhang
- Research and Development Division, Hainan Poly Pharm. CO., Ltd., Hangzhou, 310027, P. R. China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100090, P. R. China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
- Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, P. R. China
| |
Collapse
|
117
|
Liu C, Xie Y, Li X, Yao X, Wang X, Wang M, Li Z, Cao F. Folic Acid/Peptides Modified PLGA-PEI-PEG Polymeric Vectors as Efficient Gene Delivery Vehicles: Synthesis, Characterization and Their Biological Performance. Mol Biotechnol 2021; 63:63-79. [PMID: 33141343 DOI: 10.1007/s12033-020-00285-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2020] [Indexed: 01/08/2023]
Abstract
Polymeric vectors are safer alternatives for gene delivery owing to their advantages as compared to viral vectors. To improve the stability and transfection efficiency of poly(lactic-co-glycolic acid) (PLGA)- and poly(ethylenimine) (PEI)-based vectors, poly(ethylene glycol) (PEG), folic acid (FA), arginylglycylaspartic acid (RGD) peptides and isoleucine-lysine-valine-alanine-valine (IKVAV) peptides were employed and PLGA-PEI-PEG-FA and PLGA-PEI-PEG-RGD copolymers were synthesized. PLGA-PEI-PEG-FA/DNA, PLGA-PEI-PEG-RGD/DNA and PLGA-PEI-PEG-RGD/IKVAV/DNA nanocomplexes (NCs) were formed through bulk mixing. The structure and properties, including morphology, particle size, surface charge and DNA encapsulation, of NCs were studied. Robust NCs with spherical shape, uniform size distribution and slightly positive charge were able to completely bind DNA above their respective N/P ratios. The critical N/P ratio for PLGA-PEI-PEG-FA/DNA, PLGA-PEI-PEG-RGD/DNA and PLGA-PEI-PEG-RGD/IKVAV/DNA NCs was identified to be 12:1, 8:1 and 10:1, respectively. The covalent modification of PEI through a combination of biodegradable PLGA, hydrophilic PEG and targeting motifs significantly decreased the cytotoxicity of PEI. The developed NCs showed both N/P ratio and cell type-dependent transfection efficiency. An increase in N/P ratio resulted in increased transfection efficiency, and much improved transfection efficiency of NCs was observed above their respective critical N/P ratios. This study provides a promising means to produce polymeric vectors for gene delivery.
Collapse
Affiliation(s)
- Chaoyu Liu
- Department of Research and Development, Shiningbiotek Co., Ltd, Shenzhen, 518055, People's Republic of China
| | - Yuancai Xie
- Department of Thoracic, Peking University Shenzhen Hospital, Shenzhen, 518036, People's Republic of China
| | - Xiaohua Li
- Department of Research and Development, Shiningbiotek Co., Ltd, Shenzhen, 518055, People's Republic of China
| | - Xumei Yao
- Department of Research and Development, Shiningbiotek Co., Ltd, Shenzhen, 518055, People's Republic of China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Shiyan, 442000, People's Republic of China
| | - Min Wang
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam, Hong Kong, People's Republic of China
| | - Zongxian Li
- Department of Oncology, Weihai Central Hospital, Weihai, People's Republic of China.
| | - Fengjun Cao
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
| |
Collapse
|
118
|
Tsukigawa K, Imoto S, Yamasaki K, Nishi K, Tsutsumi T, Yokoyama S, Ishima Y, Otagiri M. Synthesis and In Vitro Assessment of pH-Sensitive Human Serum Albumin Conjugates of Pirarubicin. Pharmaceuticals (Basel) 2020; 14:ph14010022. [PMID: 33396604 PMCID: PMC7823624 DOI: 10.3390/ph14010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/24/2020] [Accepted: 12/27/2020] [Indexed: 12/15/2022] Open
Abstract
In a previous study, we reported on the development of a synthetic polymer conjugate of pirarubicin (THP) that was formed via an acid-labile hydrazone bond between the polymer and the THP. However, the synthetic polymer itself was non-biodegradable, which could lead to unexpected adverse effects. Human serum albumin (HSA), which has a high biocompatibility and good biodegradability, is also a potent carrier for delivering antitumor drugs. The objective of this study was to develop pH-sensitive HSA conjugates of THP (HSA-THP), and investigate the release of THP and the cytotoxicity under acidic conditions in vitro for further clinical development. HSA-THP was synthesized by conjugating maleimide hydrazone derivatives of THP with poly-thiolated HSA using 2-iminothiolane, via a thiol-maleimide coupling reaction. We synthesized two types of HSA-THP that contained different amounts of THP (HSA-THP2 and HSA-THP4). Free THP was released from both of the HSA conjugates more rapidly at an acidic pH, and the rates of release for HSA-THP2 and HSA-THP4 were similar. Moreover, both HSA-THPs exhibited a higher cytotoxicity at acidic pH than at neutral pH, which is consistent with the effective liberation of free THP under acidic conditions. These findings suggest that these types of HSA-THPs are promising candidates for further development.
Collapse
Affiliation(s)
- Kenji Tsukigawa
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan; (K.T.); (S.I.); (K.Y.); (K.N.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Shuhei Imoto
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan; (K.T.); (S.I.); (K.Y.); (K.N.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan; (K.T.); (S.I.); (K.Y.); (K.N.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Koji Nishi
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan; (K.T.); (S.I.); (K.Y.); (K.N.)
| | - Toshihiko Tsutsumi
- School of Pharmaceutical Sciences, Kyushu University of Health and Welfare, 1714-1 Yoshino-machi, Nobeoka, Miyazaki 882-8508, Japan; (T.T.); (S.Y.)
| | - Shoko Yokoyama
- School of Pharmaceutical Sciences, Kyushu University of Health and Welfare, 1714-1 Yoshino-machi, Nobeoka, Miyazaki 882-8508, Japan; (T.T.); (S.Y.)
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima 770-8505, Japan;
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan; (K.T.); (S.I.); (K.Y.); (K.N.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
- Correspondence: ; Tel.: +81-96-326-3887
| |
Collapse
|
119
|
Li J, Kataoka K. Chemo-physical Strategies to Advance the in Vivo Functionality of Targeted Nanomedicine: The Next Generation. J Am Chem Soc 2020; 143:538-559. [PMID: 33370092 DOI: 10.1021/jacs.0c09029] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The past few decades have witnessed an evolution of nanomedicine from biologically inert entities to more smart systems, aimed at advancing in vivo functionality. However, we should recognize that most systems still rely on reasonable explanation-including some over-explanation-rather than definitive evidence, which is a watershed radically determining the speed and extent of advancing nanomedicine. Probing nano-bio interactions and desirable functionality at the tissue, cellular, and molecular levels is most frequently overlooked. Progress toward answering these questions will provide instructive insight guiding more effective chemo-physical strategies. Thus, in the next generation, we argue that much effort should be made to provide definitive evidence for proof-of-mechanism, in lieu of creating many new and complicated systems for similar proof-of-concept.
Collapse
Affiliation(s)
- Junjie Li
- Innovation Center of NanoMedicne, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicne, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan.,Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
120
|
Cho R, Sakurai Y, Jones HS, Akita H, Hisaka A, Hatakeyama H. Silencing of VEGFR2 by RGD-Modified Lipid Nanoparticles Enhanced the Efficacy of Anti-PD-1 Antibody by Accelerating Vascular Normalization and Infiltration of T Cells in Tumors. Cancers (Basel) 2020; 12:cancers12123630. [PMID: 33291555 PMCID: PMC7761875 DOI: 10.3390/cancers12123630] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/18/2020] [Accepted: 12/02/2020] [Indexed: 12/31/2022] Open
Abstract
Simple Summary siRNA delivery to tumor endothelial cells was achieved using arginyl-glycyl-aspartic acid (RGD)-modified lipid nanoparticles containing a novel pH-sensitive and biodegradable lipid. The anti-tumor efficacy of an immune checkpoint inhibitor was improved by the silencing of VEGFR2 using the delivery system, because the combination therapy induced vascular normalization and increased CD8+ T cell infiltration into tumors. The efficient delivery of nucleic acids is a promising strategy to improve therapeutic outcomes in immune checkpoint inhibitor-resistant cancers. Abstract Despite the promising anticancer effects of immune checkpoint inhibitors, their low objective response rate remains to be resolved; thus, combination therapies have been investigated. We investigated the combination of an anti-programmed cell death 1 (aPD-1) monoclonal antibody with the knockdown of vascular endothelial factor receptor 2 (VEGFR2) on tumor endothelial cells to overcome resistance to immune checkpoint inhibitors and improve the objective response rate. The successful delivery of small interfering RNA to tumor endothelial cells was achieved by RGD peptide-modified lipid nanoparticles composed of a novel, pH-sensitive, and biodegradable ssPalmO-Phe. RGD-modified lipid nanoparticles efficiently induced the knockdown of VEGFR2 in tumor endothelial cells (TECs), which induced vascular normalization. The combination of a PD-1 monoclonal antibody with Vegfr2 knockdown enhanced CD8+ T cell infiltration into tumors and successfully suppressed tumor growth and improved response rate compared with monotherapy. Our combination approach provides a promising strategy to improve therapeutic outcomes in immune checkpoint inhibitor-resistant cancers.
Collapse
Affiliation(s)
- Riki Cho
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan; (R.C.); (H.S.J.); (A.H.)
| | - Yu Sakurai
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan; (Y.S.); (H.A.)
| | - Haleigh Sakura Jones
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan; (R.C.); (H.S.J.); (A.H.)
| | - Hidetaka Akita
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan; (Y.S.); (H.A.)
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan; (R.C.); (H.S.J.); (A.H.)
| | - Hiroto Hatakeyama
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan; (R.C.); (H.S.J.); (A.H.)
- Correspondence: ; Tel.: +81-43-226-2789
| |
Collapse
|
121
|
Abasian P, Shakibi S, Maniati MS, Nouri Khorasani S, Khalili S. Targeted delivery, drug release strategies, and toxicity study of polymeric drug nanocarriers. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.5168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Payam Abasian
- Department of Chemical Engineering Isfahan University of Technology Isfahan Iran
| | - Sepideh Shakibi
- Department of Textile Engineering Amirkabir University of Technology (Tehran Polytechnique) Tehran Iran
| | - Mohammad Saeed Maniati
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
| | | | - Shahla Khalili
- Department of Chemical Engineering Isfahan University of Technology Isfahan Iran
| |
Collapse
|
122
|
Takayama Y, Kusamori K, Tsukimori C, Shimizu Y, Hayashi M, Kiyama I, Katsumi H, Sakane T, Yamamoto A, Nishikawa M. Anticancer drug-loaded mesenchymal stem cells for targeted cancer therapy. J Control Release 2020; 329:1090-1101. [PMID: 33098911 DOI: 10.1016/j.jconrel.2020.10.037] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/10/2020] [Accepted: 10/16/2020] [Indexed: 12/27/2022]
Abstract
Mesenchymal stem cells (MSCs) have a tumor-homing ability-they accumulate inside tumors after systemic injection, and may thus be useful as carriers for tumor-targeting therapy. To use MSCs effectively as an anti-cancer therapy, they must first be functionalized with a large amount of anti-cancer drugs without causing any significant changes to their tumor-tropism. In the present study, we attempted to modify the cell surface of MSCs with doxorubicin-loaded liposomes (DOX-Lips), using the avidin-biotin complex method, and evaluated delivery efficiency and anti-tumor efficacy of DOX-Lip-modified MSCs. The amount of DOX in DOX-Lip-modified C3H10T1/2 cells, a murine mesenchymal stem cell line, was approximately 21.5 pg per cell, with no significant changes to the tumor-tropism of C3H10T1/2 cells. Notably, DOX-Lip-modified C3H10T1/2 cells significantly suppressed the proliferation of firefly luciferase-expressing murine colon adenocarcinoma colon26/fluc cells, compared to DOX-Lips alone. Fluorescent DOX accumulated at the cell contact surface and inside green fluorescence protein-expressing colon26 (colon26/GFP) in co-cultures of DOX-Lip-modified C3H10T1/2 and colon26/GFP cells. This localized distribution was not observed when only DOX-Lips was added to colon26/GFP cells. These results suggest that DOX-Lips are efficiently delivered from DOX-Lip-modified C3H10T1/2 cells to the neighboring colon26 cells. Furthermore, DOX-Lip-modified C3H10T1/2 cells suppressed tumor growth in subcutaneous tumor-bearing mice, and in a lung metastasis mouse model. Taken together, these results indicate that the intercellular delivery of DOX may be enhanced using DOX-Lip-modified MSCs as an efficient carrier system for targeted tumor therapy.
Collapse
Affiliation(s)
- Yukiya Takayama
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| | - Chihiro Tsukimori
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Yosuke Shimizu
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Mika Hayashi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Ikumi Kiyama
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Toshiyasu Sakane
- Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Japan
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
123
|
Li W, Li Y, Sun P, Zhang N, Zhao Y, Qin S, Zhao Y. Antimicrobial peptide-modified silver nanoparticles for enhancing the antibacterial efficacy. RSC Adv 2020; 10:38746-38754. [PMID: 35518403 PMCID: PMC9057333 DOI: 10.1039/d0ra05640e] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Antibiotic-resistant bacteria are becoming a serious threat to public health worldwide. To address this problem, we have developed multifunctional peptide (MFP)-coated silver nanoparticles (MFP@AgNPs) for antibacterial studies. MFPs, which can physically adsorb to AgNPs via electrostatic interactions are comprised of a matrix metalloproteinase (MMP) cleavable sequence (PVGLIG), an antimicrobial peptide (tachyplesin-1), and a target peptide (PGP-PEG). The resulting MFP@AgNPs were characterized by various technologies, including UV-vis spectrophotometry, zeta potential analyzer, circular dichroism (CD) spectroscopy, attenuated total reflection-Fourier-transform infrared spectroscopy (ATR-FTIR), and transmission electron microscopy (TEM). The MIC and MBC were investigated against both Gram-positive bacteria and Gram-negative bacteria. The antibacterial activity in vivo was evaluated on MDR-AB (multidrug-resistant Acinetobacter baumannii) infected mice. We found that MFP@AgNPs exhibited antibacterial activity against both Gram-positive bacteria and Gram-negative bacteria. Compared to bare AgNPs, MFP@AgNPs-1 killed MDR-AB faster and more efficiently. SEM images showed that MFP@AgNPs-1 induced cell disruption via cell membrane damage. In vivo studies further confirmed the enhanced antibacterial activity against MDR-AB infections. The developed MFP@AgNPs-1 reduced the cytotoxicity of AgNPs and enhanced the antibacterial activity against MDR-AB in vitro and in vivo, providing a possible solution against multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Wenxi Li
- Zhengzhou Traditional Chinese Hospital of Orthopaedics Zhengzhou Henan 450004 PR China
| | - Yongchun Li
- School of Pharmaceutical Science, Zhengzhou University Zhengzhou Henan 450001 PR China +86 0371 67739546 +86037167739165
| | - Pengchao Sun
- Institute for Biological Interfaces 1, Karlsruhe Institute of Technology 76344 Eggenstein-Leopoldshafen Germany
| | - Nan Zhang
- School of Pharmaceutical Science, Zhengzhou University Zhengzhou Henan 450001 PR China +86 0371 67739546 +86037167739165
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou Henan 450001 PR China
| | - Yidan Zhao
- School of Pharmaceutical Science, Zhengzhou University Zhengzhou Henan 450001 PR China +86 0371 67739546 +86037167739165
| | - Shangshang Qin
- School of Pharmaceutical Science, Zhengzhou University Zhengzhou Henan 450001 PR China +86 0371 67739546 +86037167739165
| | - Yongxing Zhao
- School of Pharmaceutical Science, Zhengzhou University Zhengzhou Henan 450001 PR China +86 0371 67739546 +86037167739165
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou Henan 450001 PR China
| |
Collapse
|
124
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
125
|
Yadav D, Dewangan HK. PEGYLATION: an important approach for novel drug delivery system. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 32:266-280. [PMID: 32942961 DOI: 10.1080/09205063.2020.1825304] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PEGylation is the covalent addition of PEG to one more molecule. PEGylation can improve the maintenance time of the therapeutics similar to proteins, liposomes, and nanoparticle through shielding them beside different debasing mechanisms dynamic in a body that improve beneficial properties. This skill is used to get better half-life and other pharmaceutical properties of a protein, peptide, or non-peptide molecule. Polyethylene glycol is harmless, non-immunogenic, non-antigenic, and extremely soluble in water and FDA accepted polymer. It shows a significant role in drug delivery. A variety of PEG-based formulations are available in the market. This paper represents the benefits of PEGylation over non-PEGylated products. Now a day, PEGylation plays an important role in the drug delivery system. PEGylation increases the therapeutic potential of drugs.
Collapse
Affiliation(s)
- Deepa Yadav
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | | |
Collapse
|
126
|
Wang Y, Wagner E. Non-Viral Targeted Nucleic Acid Delivery: Apply Sequences for Optimization. Pharmaceutics 2020; 12:E888. [PMID: 32961908 PMCID: PMC7559072 DOI: 10.3390/pharmaceutics12090888] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/09/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
In nature, genomes have been optimized by the evolution of their nucleic acid sequences. The design of peptide-like carriers as synthetic sequences provides a strategy for optimizing multifunctional targeted nucleic acid delivery in an iterative process. The optimization of sequence-defined nanocarriers differs for different nucleic acid cargos as well as their specific applications. Supramolecular self-assembly enriched the development of a virus-inspired non-viral nucleic acid delivery system. Incorporation of DNA barcodes presents a complementary approach of applying sequences for nanocarrier optimization. This strategy may greatly help to identify nucleic acid carriers that can overcome pharmacological barriers and facilitate targeted delivery in vivo. Barcode sequences enable simultaneous evaluation of multiple nucleic acid nanocarriers in a single test organism for in vivo biodistribution as well as in vivo bioactivity.
Collapse
Affiliation(s)
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, D-81377 Munich, Germany;
| |
Collapse
|
127
|
Kannaka K, Sano K, Nakahara H, Munekane M, Hagimori M, Yamasaki T, Mukai T. Inverse Electron Demand Diels-Alder Reactions in the Liposomal Membrane Accelerates Release of the Encapsulated Drugs. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:10750-10755. [PMID: 32830502 DOI: 10.1021/acs.langmuir.0c01525] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Bio-orthogonal inverse electron demand Diels-Alder (IEDDA) reactions between liposomes containing a tetrazine-based (Tz) compound and 2-norbornene (2-NB) could be a novel trigger for accelerating drug release from the liposomes via temporary membrane destabilization, as shown in our previous report. Herein, we evaluated the in vitro drug release using NB derivatives with carboxyl groups [5-norbornene-2-carboxylic acid (NBCOOH) and 5-norbornene-2,3-dicarboxylic acid (NB(COOH)2)] to investigate the effects of substituents at the NB backbone on the drug release rate. First, POTz-liposome composed of a Tz compound (2-hexadecyl-N-(6-(6-(pyridin-2-yl)-1,2,4,5-tetrazin-3-yl)pyridin-3-yl)octadecanamide) and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphatidylcholine (POPC) were prepared. The mass spectrometry analysis revealed the binding of NB derivatives to the Tz compound via the IEDDA reaction after the POTz-liposome reacted with the NB derivatives. Indium-111-labeled diethylenetriaminepentaacetic acid (111In-DTPA) was encapsulated inside the liposomes, and the drug release rate was quantified by measuring radioactivity. At 24 h after incubation with 2-NB, NBCOOH, and NB(COOH)2, the release rates of 111In-DTPA from POTz-liposome were 21.0, 80.8, and 23.3%, respectively, which were significantly higher than those of POTz-liposome that was not treated with NB derivatives (4.2%), indicating the involvement of the IEDDA reaction for prompting drug release. Additionally, a thermodynamic evaluation using Langmuir monolayers was conducted to explore the mechanism of the accelerated drug release. An increase in membrane fluidity and a reduction in intermolecular repulsion between POPC and the Tz compound were observed after the reaction with NB derivatives, especially for NBCOOH. Thus, the IEDDA reaction in the liposomal membrane could be a potent trigger for accelerating the release of encapsulated drugs by regulating membrane fluidity and intermolecular repulsion in the liposomal membrane.
Collapse
Affiliation(s)
- Kento Kannaka
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| | - Hiromichi Nakahara
- Department of Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan
| | - Masayuki Munekane
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| | - Masayori Hagimori
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada-ku, Kobe 658-8558, Japan
- Laboratory of Analytical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Koshien Kyuban-cho, Nishinomiya 663-8179, Japan
| | - Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada-ku, Kobe 658-8558, Japan
| |
Collapse
|
128
|
Yamada Y, Sato Y, Nakamura T, Harashima H. Evolution of drug delivery system from viewpoint of controlled intracellular trafficking and selective tissue targeting toward future nanomedicine. J Control Release 2020; 327:533-545. [PMID: 32916227 PMCID: PMC7477636 DOI: 10.1016/j.jconrel.2020.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Due to the rapid changes that have occurred in the field of drug discovery and the recent developments in the early 21st century, the role of drug delivery systems (DDS) has become increasingly more important. For the past 20 years, our laboratory has been developing gene delivery systems based on lipid-based delivery systems. One of our efforts has been directed toward developing a multifunctional envelope-type nano device (MEND) by modifying the particle surface with octaarginine, which resulted in a remarkably enhanced cellular uptake and improved intracellular trafficking of plasmid DNA (pDNA). When we moved to in vivo applications, however, we were faced with the PEG-dilemma and we shifted our strategy to the incorporation of ionizable cationic lipids into our system. This resulted in some dramatic improvements over our original design and this can be attributed to the development of a new lipid library. We have also developed a mitochondrial targeting system based on a membrane fusion mechanism using a MITO-Porter, which can deliver nucleic acids/pDNA into the matrix of mitochondria. After the appearance of antibody medicines, Opdivo, an immune checkpoint inhibitor, has established cancer immunology as the 4th strategy in cancer therapy. Our DDS technologies can also be applied to this new field of cancer therapy to cure cancer by controlling our immune mechanisms. The latest studies are summarized in this review article.
Collapse
Affiliation(s)
- Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
129
|
Enzyme-responsive polymeric micelles of cabazitaxel for prostate cancer targeted therapy. Acta Biomater 2020; 113:501-511. [PMID: 32562805 DOI: 10.1016/j.actbio.2020.06.019] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
Abstract
Cabazitaxel, a novel tubulin inhibitor with poor affinity for P-glycoprotein, is a second-generation taxane holding great promise for the treatment of metastatic castration-resistant prostate cancer. However, its poor solubility and lack of target-ability limit its therapeutic applications. Herein, we develop a biodegradable, enzyme-responsive, and targeted polymeric micelle for cabazitaxel. The micelle is formed from two amphiphilic block copolymers. The first block copolymer consists of PEG, an enzyme-responsive peptide, and cholesterol; whereas the second block copolymer consists of a targeting ligand, PEG and cholesterol. The enzyme-responsive peptide is cleavable in the presence of matrixmetaloproteinase-2 (MMP-2), which is overexpressed in the tumor microenvironment of prostate cancer. The micelle showed a very low critical micelle concentration (CMC), high drug loading, and high entrapment efficiency. Release of cabazitaxel from the micelle is dependent on the cleavage of the enzyme-responsive peptide. Moreover, the micelle showed dramatically higher cellular uptake in prostate cancer cells compared to free cabazitaxel. Importantly, the ligand-coupled polymeric micelle demonstrated better inhibition of tumor growth in mice bearing prostate cancer xenografts compared to unmodified micelle and free cabazitaxel. Taken together, these findings suggest that the enzyme-responsive cabazitaxel micelle is a potent and promising drug delivery system for advanced prostate cancer therapy. STATEMENT OF SIGNIFICANCE: Herein, we develop a biodegradable, enzyme-responsive, and actively targeted polymer micelle for cabazitaxel, which is a novel tubulin inhibitor with poor affinity for P-glycoprotein. Despite cabazitaxel's great promise for metastatic castration-resistant prostate cancer, its poor solubility, lack of target-ability, and high systemic toxicity limit its therapeutic applications, and therefore a targeted delivery system is highly needed for cabazitaxel. Our results demonstrate the importance of active targeting in targeted prostate cancer therapy. Encapsulating cabazitaxel in the micelle increases its activity and is expected to reduce its systemic toxicity, which is a major hurdle in its clinical applications. Moreover, the polymeric micelle may servers as a promising nanoscale platform for the targeted delivery of other chemotherapeutic agents to prostate cancer.
Collapse
|
130
|
Hamida RS, Ali MA, Redhwan A, Bin-Meferij MM. Cyanobacteria - A Promising Platform in Green Nanotechnology: A Review on Nanoparticles Fabrication and Their Prospective Applications. Int J Nanomedicine 2020; 15:6033-6066. [PMID: 32884261 PMCID: PMC7434529 DOI: 10.2147/ijn.s256134] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
Green synthesis of nanoparticles (NPs) is a global ecofriendly method to develop and produce nanomaterials with unique biological, physical, and chemical properties. Recently, attention has shifted toward biological synthesis, owing to the disadvantages of physical and chemical synthesis, which include toxic yields, time and energy consumption, and high cost. Many natural sources are used in green fabrication processes, including yeasts, plants, fungi, actinomycetes, algae, and cyanobacteria. Cyanobacteria are among the most beneficial natural candidates used in the biosynthesis of NPs, due to their ability to accumulate heavy metals from their environment. They also contain a variety of bioactive compounds, such as pigments and enzymes, that may act as reducing and stabilizing agents. Cyanobacteria-mediated NPs have potential antibacterial, antifungal, antialgal, anticancer, and photocatalytic activities. The present review paper highlights the characteristics and applications in various fields of NPs produced by cyanobacteria-mediated synthesis.
Collapse
Affiliation(s)
- Reham Samir Hamida
- Molecular Biology Unit, Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mohamed Abdelaal Ali
- Biotechnology Unit, Department of Plant Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Alya Redhwan
- Department of Health, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | |
Collapse
|
131
|
Kargozar S, Baino F, Hamzehlou S, Hamblin MR, Mozafari M. Nanotechnology for angiogenesis: opportunities and challenges. Chem Soc Rev 2020; 49:5008-5057. [PMID: 32538379 PMCID: PMC7418030 DOI: 10.1039/c8cs01021h] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis plays a critical role within the human body, from the early stages of life (i.e., embryonic development) to life-threatening diseases (e.g., cancer, heart attack, stroke, wound healing). Many pharmaceutical companies have expended huge efforts on both stimulation and inhibition of angiogenesis. During the last decade, the nanotechnology revolution has made a great impact in medicine, and regulatory approvals are starting to be achieved for nanomedicines to treat a wide range of diseases. Angiogenesis therapies involve the inhibition of angiogenesis in oncology and ophthalmology, and stimulation of angiogenesis in wound healing and tissue engineering. This review aims to summarize nanotechnology-based strategies that have been explored in the broad area of angiogenesis. Lipid-based, carbon-based and polymeric nanoparticles, and a wide range of inorganic and metallic nanoparticles are covered in detail. Theranostic and imaging approaches can be facilitated by nanoparticles. Many preparations have been reported to have a bimodal effect where they stimulate angiogenesis at low dose and inhibit it at higher doses.
Collapse
Affiliation(s)
- Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, 917794-8564 Mashhad, Iran
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 101 29 Torino, Italy
| | - Sepideh Hamzehlou
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Masoud Mozafari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
132
|
Charge-reversible lipid derivative: A novel type of pH-responsive lipid for nanoparticle-mediated siRNA delivery. Int J Pharm 2020; 585:119479. [DOI: 10.1016/j.ijpharm.2020.119479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 12/15/2022]
|
133
|
He X, Yang X, Li D, Cao Z. Red and NIR Light-Responsive Polymeric Nanocarriers for On-Demand Drug Delivery. Curr Med Chem 2020; 27:3877-3887. [DOI: 10.2174/0929867326666190215113522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/16/2018] [Accepted: 12/04/2018] [Indexed: 11/22/2022]
Abstract
Red and NIR light-responsive polymeric nanocarriers capable of on-demand drug delivery
have gained tremendous attention for their great potential in cancer therapy. Various strategies have
been applied to fabricate such nanocarriers, and they have demonstrated significant therapeutic efficacy
and minimal toxicity to normal tissues. Here, we will review the current developments in various
red and NIR light-responsive polymeric nanocarriers with respect to their use in on-demand drug
delivery, including facilitation of drug internalization and boosting of drug release at targeted sites.
We summarize their components and design strategies, and highlight the mechanisms by which the
photoactivatable variations enhance drug uptake and drug release. We attempt to provide new insights
into the fabrication of red and NIR light-responsive polymeric nanocarriers for on-demand
drug delivery.
Collapse
Affiliation(s)
- Xinyu He
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Xianzhu Yang
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Dongdong Li
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Ziyang Cao
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| |
Collapse
|
134
|
Pereira-Silva M, Jarak I, Alvarez-Lorenzo C, Concheiro A, Santos AC, Veiga F, Figueiras A. Micelleplexes as nucleic acid delivery systems for cancer-targeted therapies. J Control Release 2020; 323:442-462. [DOI: 10.1016/j.jconrel.2020.04.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/09/2023]
|
135
|
Piotrowski-Daspit AS, Kauffman AC, Bracaglia LG, Saltzman WM. Polymeric vehicles for nucleic acid delivery. Adv Drug Deliv Rev 2020; 156:119-132. [PMID: 32585159 PMCID: PMC7736472 DOI: 10.1016/j.addr.2020.06.014] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/09/2020] [Accepted: 06/13/2020] [Indexed: 12/20/2022]
Abstract
Polymeric vehicles are versatile tools for therapeutic gene delivery. Many polymers-when assembled with nucleic acids into vehicles-can protect the cargo from degradation and clearance in vivo, and facilitate its transport into intracellular compartments. Design options in polymer synthesis yield a comprehensive range of molecules and resulting vehicle formulations. These properties can be manipulated to achieve stronger association with nucleic acid cargo and cells, improved endosomal escape, or sustained delivery depending on the application. Here, we describe current approaches for polymer use and related strategies for gene delivery in preclinical and clinical applications. Polymer vehicles delivering genetic material have already achieved significant therapeutic endpoints in vitro and in animal models. From our perspective, with preclincal assays that better mimic the in vivo environment, improved strategies for target specificity, and scalable techniques for polymer synthesis, the impact of this therapeutic approach will continue to expand.
Collapse
Affiliation(s)
| | - Amy C Kauffman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States of America; Corning Life Sciences, Kennebunk, ME 04043, United States of America
| | - Laura G Bracaglia
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States of America
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States of America; Department of Chemical & Environmental Engineering, Yale University, New Haven, CT 06511, United States of America; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, United States of America; Department of Dermatology, Yale School of Medicine, New Haven, CT 06510, United States of America.
| |
Collapse
|
136
|
Liu Y, Long T, Zhang N, Qiao B, Yang Q, Luo Y, Cao J, Luo J, Yuan D, Sun Y, Li Y, Yang Z, Wang ZG. Ultrasound-Mediated Long-Circulating Nanopolymer Delivery of Therapeutic siRNA and Antisense MicroRNAs Leads to Enhanced Paclitaxel Sensitivity in Epithelial Ovarian Cancer Chemotherapy. ACS Biomater Sci Eng 2020; 6:4036-4050. [PMID: 33463352 DOI: 10.1021/acsbiomaterials.0c00330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Epithelial ovarian cancer (EOC) is one of the leading malignant tumors that seriously threaten women's health. The development of new drugs or increasing the sensitivities of current chemotherapy drugs is critically needed. The purpose of this study was to assess the synergistic effects of two silencing RNAs [salt-inducible kinase 2 (SIK2) siRNA and antisense-microRNA21 (anti-miR21)] encapsulated in long-circulating folate-lipid-poly(lactic-co-glycolic acid) (PLGA) hybrid nanopolymers (FaLPHNPs) administered using an ultrasound- and microbubble (US-MB)-mediated approach to sensitize human EOC xenografts to paclitaxel (PTX). In the in vitro assays, this lipid-PLGA hybrid nanopolymer exhibited an extended circulation profile (t1/2: ∼8.5 h); US-MB-mediated complementary delivery of FaLPHNPs resulted in a significant reduction in EOC cell (OVCR3, A2780, and SKOV3) proliferation. In vivo, there was a 2.5-fold increase (p < 0.05) in RNA delivery in EOC xenografts, which resulted in a notable inhibition of tumor growth compared with that in the non-ultrasound-mediated and PTX alone-treated controls. We validated the therapeutic roles of SIK2, the target gene in treating advanced ovarian cancer, and anti-miR21 by evaluating the significant inhibition of tumor growth upon SIK2 silencing and inhibition of endogenous miR21 function. In summary, the results of this study revealed that US-MB-mediated codelivery of SIK2 siRNA, and anti-miR21 encapsulated in a folate-lipid-PLGA hybrid polymer nanoparticle could significantly improve the sensitivity of EOC tumors to PTX and is a highly effective approach for treating EOC in complementary experiments. Further research of this strategy could lead to better treatment results for patients with EOC.
Collapse
Affiliation(s)
- Yi Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Tengfei Long
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ni Zhang
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Department of Oncology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Bin Qiao
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qiang Yang
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yuanli Luo
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jin Cao
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jing Luo
- Department of Pathology, Chongqing Medical University, Chongqing 400010, China
| | - Dong Yuan
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yixuan Sun
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yanxi Li
- Department of Reproductive Medicine Center, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Zhu Yang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Z G Wang
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
137
|
Ibaraki H, Kanazawa T, Chien WY, Nakaminami H, Aoki M, Ozawa K, Kaneko H, Takashima Y, Noguchi N, Seta Y. The effects of surface properties of liposomes on their activity against Pseudomonas aeruginosa PAO-1 biofilm. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
138
|
Zhao Y, Cai F, Shen X, Su H. A high stable pH-temperature dual-sensitive liposome for tuning anticancer drug release. Synth Syst Biotechnol 2020; 5:103-110. [PMID: 32596520 PMCID: PMC7301211 DOI: 10.1016/j.synbio.2020.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/03/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
In order to improve the targeting and availability of liposomes to cancer cells, the temperature sensitivity of 1, 2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and the pH sensitivity of PASP in PASP-g-C8 are incorporated in a drug delivery system. A composite pH-temperature dual-sensitive liposomes (CPTLPs) was obtained as an efficient drug delivery system. The bionic bilayer is self-assembled by cholesterol/cationic temperature-sensitive lipids as base layer and pH-sensitive octylamine grafted poly aspartic acid (PASP-g-C8) as anchors coated outside. Cytarabine (CYT) was chosen as a model drug. SEM and DLS were used to observe the morphology characteristics of CPTLPs in different micro environment. The results demonstrated that the CPTLPs remained active in both normal (pH7.4 and 37 °C) and tumor tissues (pH 5.0 and 42 °C). As a stable colloidal system, the zeta potential of CPTSLs was -41.6 mV. In vitro drug-release experiments, the CTY encapsulated dual-sensitive liposomes, CPTSLs(+), not only have significant pH-temperature sensitivity but have more prolonged release in vitro than control groups. MTT tests results indicated that the cell apoptotic effects induced by CPTSLs(+) were nearly 30% higher than the naked drug CTY in HepG2 cells, and 20% lower apoptotic in vero cells. The CPTSLs(+) sustained a stable emulsion form, less toxic effects on normal cells, and exhibited a good pH-temperature sensitivity, thus expected to be a promising tumor targeting drug delivery.
Collapse
Affiliation(s)
- Yilin Zhao
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology(BUCT), 15 BeiSanhuan East Road, ChaoYang District, Beijing, 100029, PR China
| | - Fuli Cai
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology(BUCT), 15 BeiSanhuan East Road, ChaoYang District, Beijing, 100029, PR China
| | - Xiangyi Shen
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology(BUCT), 15 BeiSanhuan East Road, ChaoYang District, Beijing, 100029, PR China
| | - Haijia Su
- Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology(BUCT), 15 BeiSanhuan East Road, ChaoYang District, Beijing, 100029, PR China
| |
Collapse
|
139
|
Li S, Lin L, Wang W, Yan X, Chen B, Jiang S, Liu S, Ma X, Tian H, Yu X. Aza-crown ether locked on polyethyleneimine: solving the contradiction between transfection efficiency and safety during in vivo gene delivery. Chem Commun (Camb) 2020; 56:5552-5555. [PMID: 32297607 DOI: 10.1039/c9cc10041e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We proposed a method using an aza-crown ether derivative to lock a hyperbranched polyethyleneimine, which endows the PEI25k with tumor targeting ability, anti-serum ability and extended circulation in the blood meanwhile retaining the high gene complexation and high transfection efficiency. The method we proposed here simultaneously endows cationic materials with high transfection efficiency and high safety, which greatly pushed the cationic materials to be applied in in vivo gene delivery.
Collapse
Affiliation(s)
- Shengran Li
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Xia X, Pollock N, Zhou J, Rossi J. Tissue-Specific Delivery of Oligonucleotides. Methods Mol Biol 2020; 2036:17-50. [PMID: 31410789 DOI: 10.1007/978-1-4939-9670-4_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
From the initial discovery of short-interfering RNA (siRNA) and antisense oligonucleotides for specific gene knockdown at the posttranscriptional level to the current CRISPR-Cas9 system offering gene editing at the genomic level, oligonucleotides, in addition to their biological functions in storing and conveying genetic information, provide the most prominent solutions to targeted gene therapies. Nonetheless, looking into the future of curing cancer and acute diseases, researchers are only cautiously optimistic as the cellular delivery of these polyanionic biomacromolecules is still the biggest hurdle for their therapeutic realization. To overcome the delivery obstacle, oligonucleotides have been encapsulated within or conjugated with delivery vehicles for enhanced membrane penetration, improved payload, and tissue-specific delivery. Such delivery systems include but not limited to virus-based vehicles, gold-nanoparticle vehicles, formulated liposomes, and synthetic polymers. In this chapter, delivery challenges imposed by biological barriers are briefly discussed; followed by recent advances in tissue-specific oligonucleotide delivery utilizing both viral and nonviral delivery vectors, discussing their advantages, and how judicious design and formulation could improve and expand their potential as delivery vehicles.
Collapse
Affiliation(s)
- Xin Xia
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Nicolette Pollock
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Jiehua Zhou
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - John Rossi
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
141
|
Vaughan HJ, Green JJ, Tzeng SY. Cancer-Targeting Nanoparticles for Combinatorial Nucleic Acid Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901081. [PMID: 31222852 PMCID: PMC6923623 DOI: 10.1002/adma.201901081] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/13/2019] [Indexed: 05/03/2023]
Abstract
Nucleic acids are a promising type of therapeutic for the treatment of a wide range of conditions, including cancer, but they also pose many delivery challenges. For efficient and safe delivery to cancer cells, nucleic acids must generally be packaged into a vehicle, such as a nanoparticle, that will allow them to be taken up by the target cells and then released in the appropriate cellular compartment to function. As with other types of therapeutics, delivery vehicles for nucleic acids must also be designed to avoid unwanted side effects; thus, the ability of such carriers to target their cargo to cancer cells is crucial. Classes of nucleic acids, hurdles that must be overcome for effective intracellular delivery, types of nonviral nanomaterials used as delivery vehicles, and the different strategies that can be employed to target nucleic acid delivery specifically to tumor cells are discussed. Additonally, nanoparticle designs that facilitate multiplexed delivery of combinations of nucleic acids are reviewed.
Collapse
Affiliation(s)
- Hannah J Vaughan
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| |
Collapse
|
142
|
Li T, Huang L, Yang M. Lipid-based Vehicles for siRNA Delivery in Biomedical Field. Curr Pharm Biotechnol 2020; 21:3-22. [PMID: 31549951 DOI: 10.2174/1389201020666190924164152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/04/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Genetic drugs have aroused much attention in the past twenty years. RNA interference (RNAi) offers novel insights into discovering potential gene functions and therapies targeting genetic diseases. Small interference RNA (siRNA), typically 21-23 nucleotides in length, can specifically degrade complementary mRNA. However, targeted delivery and controlled release of siRNA remain a great challenge. METHODS Different types of lipid-based delivery vehicles have been synthesized, such as liposomes, lipidoids, micelles, lipoplexes and lipid nanoparticles. These carriers commonly have a core-shell structure. For active targeting, ligands may be conjugated to the surface of lipid particles. RESULTS Lipid-based drug delivery vehicles can be utilized in anti-viral or anti-tumor therapies. They can also be used to tackle genetic diseases or discover novel druggable genes. CONCLUSION In this review, the structures of lipid-based vehicles and possible surface modifications are described, and applications of delivery vehicles in biomedical field are discussed.
Collapse
Affiliation(s)
- Tianzhong Li
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Linfeng Huang
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Mengsu Yang
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| |
Collapse
|
143
|
Pei X, Zhu Z, Gan Z, Chen J, Zhang X, Cheng X, Wan Q, Wang J. PEGylated nano-graphene oxide as a nanocarrier for delivering mixed anticancer drugs to improve anticancer activity. Sci Rep 2020; 10:2717. [PMID: 32066812 PMCID: PMC7026168 DOI: 10.1038/s41598-020-59624-w] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/03/2020] [Indexed: 02/05/2023] Open
Abstract
Due to their high specific surface area, graphene oxide and graphene oxide-base nanoparticles have great potential both in dual-drug delivery and combination chemotherapy. Herein, we developed cisplatin (Pt) and doxorubicin (DOX) dual-drug-loaded PEGylated nano-graphene oxide (pGO) to facilitate combined chemotherapy in one system. In this study, nano-sized pGO-Pt/DOX ranged around 161.50 nm was fabricated and characterized using zeta-potential, AFM, TEM, Raman, UV-Vis, and FTIR analyses. The drug delivery efficacy of Pt was enhanced through the introduction of pGO, and the final weight ratio of DOX: Pt: pGO was optimized to 0.376: 0.376: 1. In vitro studies revealed that pGO-Pt/DOX nanoparticles could be effectively delivered into tumor cells, in which they induced prominent cell apoptosis and necrosis and exhibited higher growth inhibition than the single drug delivery system or free drugs. The pGO-Pt/DOX induced the most prominent cancer cell apoptosis and necrosis rate with 18.6%, which was observed almost 2 times higher than that of pGO-Pt or pGO-DOX groups. in the apoptosis and necrotic quadrants In vivo data confirmed that the pGO-Pt/DOX dual-drug delivery system attenuated the toxicity of Pt and DOX to normal organs compared to free drugs. The tumor inhibition data, histopathology observations, and immunohistochemical staining confirmed that the dual-drug delivery system presented a better anticancer effect than free drugs. These results clearly indicated that the pGO-Pt/DOX dual-drug delivery system provided the means for combination drug delivery in cancer treatment.
Collapse
Affiliation(s)
- Xibo Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhoujie Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xinting Cheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
144
|
Betker JL, Anchordoquy TJ. The Use of Lactose as an Alternative Coating for Nanoparticles. J Pharm Sci 2020; 109:1573-1580. [PMID: 32004536 DOI: 10.1016/j.xphs.2020.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/16/2019] [Accepted: 01/23/2020] [Indexed: 12/22/2022]
Abstract
Nanoparticle-mediated drug delivery has long utilized PEGylation as a mechanism for reducing uptake by the reticuloendothelial system and extending circulation lifetimes. However, studies over the past 2 decades have established that immune responses to PEG can promote clearance on repeat injection and elicit life-threatening anaphylactic reactions in some patients. As a potential alternative to PEGylation, we explored the ability of utilizing lactose, a naturally occurring sugar that is common on the surface of blood cells, as a coating for lipoplexes. Our data indicate that lactose imparts similar effects as PEG in terms of reducing leukocyte uptake, extending circulation half-life, and enhancing delivery to the tumor and other organs. In addition, measurements of blood cytokine levels after repeat injection indicate that reduced levels of inflammatory cytokines (IL-6, IFN-γ, TNFα) are elicited in response to lipoplexes coated with lactose as compared to PEG. These data indicate that a lactose coating on lipoplexes results in slightly improved tumor accumulation as compared to PEGylated formulations while eliciting a reduced innate immune response.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Thomas J Anchordoquy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045.
| |
Collapse
|
145
|
Zhang M, Elango J, Guo X, Fan H, Cui M, Wang M, Liu K. Fe 3+-Coordinated Multifunctional Elastic Nanoplatform for Effective in Vivo Gene Transfection. ACS APPLIED MATERIALS & INTERFACES 2020; 12:3453-3464. [PMID: 31895537 DOI: 10.1021/acsami.9b19585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The common phenomenon that the nonviral vectors have much lower transfection efficiency in vivo than in vitro greatly restricts their further developments and applications. Possible reasons are lacking targeting ability, elimination by the reticuloendothelial system (RES), and insufficient nuclear transport. Here, a novel, flexible, and deformable polymer Fe@PEI-R12 (tLyp-1-NLS) is reported for shortening the gap between in vitro and in vivo gene transfection efficiency. The amorphous network structure Fe@PEI with deformation ability acquired by coordination cross-linking of Fe3+ and low-molecular-weight polyethylenimine (LMW-PEI) constructs the core and serves as the gene reservoir, and it can squeeze out through RES filter holes when trapped in the spleen. The bifunctional peptide R12 provided tumor targeting and enhanced nuclear delivery ability. Additionally, the Fe3+ from Fe@PEI-R12 could trigger endogenous hydrogen peroxide (H2O2) decomposition to produce O2, thereby reducing the adverse effects of tumor hypoxia. It is demonstrated that the Fe@PEI-R12/pDNA complexes could pass through membrane filters, subsequently achieving long circulation time, and Fe@PEI-R12 had a tendency to accumulate in tumor tissue and mediate pGL3-control expression. Therefore, the multifunctional nanoplatform has the potential for effective in vivo gene delivery.
Collapse
Affiliation(s)
- Min Zhang
- College of Food Science and Technology , Shanghai Ocean University , Shanghai 201306 , China
| | - Jeevithan Elango
- College of Food Science and Technology , Shanghai Ocean University , Shanghai 201306 , China
| | - Xinli Guo
- College of Food Science and Technology , Shanghai Ocean University , Shanghai 201306 , China
| | - Hua Fan
- Institut für Laboratoriumsmedizin , Charité-Universitätsmedizin Berlin , Campus Virchow Klinikum , Berlin 13353 , Germany
| | - Mingxiao Cui
- College of Food Science and Technology , Shanghai Ocean University , Shanghai 201306 , China
| | - Mingfu Wang
- College of Food Science and Technology , Shanghai Ocean University , Shanghai 201306 , China
- School of Biological Sciences , University of Hong Kong , Pokfulam Road , Hong Kong 999077 , China
| | - Kehai Liu
- College of Food Science and Technology , Shanghai Ocean University , Shanghai 201306 , China
| |
Collapse
|
146
|
Ibaraki H, Kanazawa T, Owada M, Iwaya K, Takashima Y, Seta Y. Anti-Metastatic Effects on Melanoma via Intravenous Administration of Anti-NF-κB siRNA Complexed with Functional Peptide-Modified Nano-Micelles. Pharmaceutics 2020; 12:pharmaceutics12010064. [PMID: 31952106 PMCID: PMC7022256 DOI: 10.3390/pharmaceutics12010064] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Controlling metastasis is an important strategy in cancer treatment. Nanotechnology and nucleic acids with novel modalities are promising regulators of cancer metastasis. We aimed to develop a small interfering RNA (siRNA) systemic delivery and anti-metastasis system using nanotechnology. We previously reported that polyethylene glycol-polycaprolactone (PEG-PCL) and functional peptide CH2R4H2C nano-micelle (MPEG-PCL-CH2R4H2C) has high siRNA silencing effects, indicated by increased drug accumulation in tumor-bearing mice, and has an anti-tumor effect on solid tumors upon systemic injection. In this study, we aimed to apply our micelles to inhibit metastasis and evaluated the inhibitory effect of anti-RelA siRNA (siRelA), which is a subunit of NF-κB conjugated with MPEG-PCL-CH2R4H2C, via systemic administration. We report that siRelA/MPEG-PCL-CH2R4H2C had a high cellular uptake and suppressed the migration/invasion of cells in B16F10 cells without toxicity. In addition, in a lung metastasis mouse model using intravenous administration of B16F10 cells treated with siRelA/MPEG-PCL-CH2R4H2C, the number of lung nodules in lung tissue significantly decreased compared to naked siRelA and siControl/MPEG-PCL-CH2R4H2C micelle treatments. Hence, we show that RelA expression can reduce cancer metastasis, and MPEG-PCL-CH2R4H2C is an effective siRNA carrier for anti-metastasis cancer therapies.
Collapse
Affiliation(s)
- Hisako Ibaraki
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (H.I.); (M.O.); (K.I.); (Y.T.); (Y.S.)
| | - Takanori Kanazawa
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (H.I.); (M.O.); (K.I.); (Y.T.); (Y.S.)
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
- Correspondence: ; Tel./Fax: +81-47-465-6587
| | - Minami Owada
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (H.I.); (M.O.); (K.I.); (Y.T.); (Y.S.)
| | - Keiko Iwaya
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (H.I.); (M.O.); (K.I.); (Y.T.); (Y.S.)
| | - Yuuki Takashima
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (H.I.); (M.O.); (K.I.); (Y.T.); (Y.S.)
| | - Yasuo Seta
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan; (H.I.); (M.O.); (K.I.); (Y.T.); (Y.S.)
| |
Collapse
|
147
|
Levingstone TJ, Herbaj S, Redmond J, McCarthy HO, Dunne NJ. Calcium Phosphate Nanoparticles-Based Systems for RNAi Delivery: Applications in Bone Tissue Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E146. [PMID: 31947548 PMCID: PMC7023416 DOI: 10.3390/nano10010146] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/16/2019] [Accepted: 12/21/2019] [Indexed: 12/11/2022]
Abstract
Bone-related injury and disease constitute a significant global burden both socially and economically. Current treatments have many limitations and thus the development of new approaches for bone-related conditions is imperative. Gene therapy is an emerging approach for effective bone repair and regeneration, with notable interest in the use of RNA interference (RNAi) systems to regulate gene expression in the bone microenvironment. Calcium phosphate nanoparticles represent promising materials for use as non-viral vectors for gene therapy in bone tissue engineering applications due to their many favorable properties, including biocompatibility, osteoinductivity, osteoconductivity, and strong affinity for binding to nucleic acids. However, low transfection rates present a significant barrier to their clinical use. This article reviews the benefits of calcium phosphate nanoparticles for RNAi delivery and highlights the role of surface functionalization in increasing calcium phosphate nanoparticles stability, improving cellular uptake and increasing transfection efficiency. Currently, the underlying mechanistic principles relating to these systems and their interplay during in vivo bone formation is not wholly understood. Furthermore, the optimal microRNA targets for particular bone tissue regeneration applications are still unclear. Therefore, further research is required in order to achieve the optimal calcium phosphate nanoparticles-based systems for RNAi delivery for bone tissue regeneration.
Collapse
Affiliation(s)
- Tanya J. Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, 9 Dublin, Ireland; (T.J.L.); (S.H.); (J.R.)
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, 9 Dublin, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, 9 Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland
| | - Simona Herbaj
- School of Mechanical and Manufacturing Engineering, Dublin City University, 9 Dublin, Ireland; (T.J.L.); (S.H.); (J.R.)
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, 9 Dublin, Ireland
| | - John Redmond
- School of Mechanical and Manufacturing Engineering, Dublin City University, 9 Dublin, Ireland; (T.J.L.); (S.H.); (J.R.)
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, 9 Dublin, Ireland
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK;
| | - Nicholas J. Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, 9 Dublin, Ireland; (T.J.L.); (S.H.); (J.R.)
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, 9 Dublin, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, 9 Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK;
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, 2 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, 2 Dublin, Ireland
| |
Collapse
|
148
|
Steffes VM, Zhang Z, MacDonald S, Crowe J, Ewert KK, Carragher B, Potter CS, Safinya CR. PEGylation of Paclitaxel-Loaded Cationic Liposomes Drives Steric Stabilization of Bicelles and Vesicles thereby Enhancing Delivery and Cytotoxicity to Human Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2020; 12:151-162. [PMID: 31820904 PMCID: PMC6984750 DOI: 10.1021/acsami.9b16150] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Poly(ethylene glycol) (PEG) is a polymer used widely in drug delivery to create "stealth" nanoparticles (NPs); PEG coatings suppress NP detection and clearance by the immune system and beneficially increase NP circulation time in vivo. However, NP PEGylation typically obstructs cell attachment and uptake in vitro compared to the uncoated equivalent. Here, we report on a cationic liposome (CL) NP system loaded with the hydrophobic cancer drug paclitaxel (PTX) in which PEGylation (i.e., PEG-CLPTX NPs) unexpectedly enhances, rather than diminishes, delivery efficacy and cytotoxicity to human cancer cells. This highly unexpected enhancement occurs even when the PEG-chains coating the NP are in the transition regime between the mushroom and brush conformations. Cryogenic transmission electron microscopy (TEM) of PEG-CLPTX NPs shows that PEG causes the proliferation of a mixture of sterically stabilized nanometer-scale vesicles and anisotropic micelles (e.g., bicelles). Remarkably, the onset of bicelles at sub-monolayer concentrations of the PEG coat has to our knowledge not been previously reported; it was previously thought that PEG-lipid in this composition regime was incorporated into vesicles but did not alter their shape. Confocal microscopy and flow cytometry reveal significantly greater PTX cell uptake from stabilized PEG-CLPTX NPs (vesicles and bicelles) in contrast to bare CLPTX NPs, which can aggregate in cell medium. This underscores the ability of steric stabilization to facilitate NP entry into cells via distinct size-dependent endocytic pathways, some of which cannot transport large NP aggregates into cells. This study highlights the value of understanding how PEGylation alters NP shape and structure, and thus NP efficacy, to design next-generation stealth drug carriers that integrate active cell-targeting strategies into NPs for in vivo delivery.
Collapse
Affiliation(s)
- Victoria M. Steffes
- Materials Department, University of California, Santa Barbara, California 93106, USA
- Chemistry and Biochemistry Department, University of California, Santa Barbara, California 93106, USA
| | - Zhening Zhang
- The National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY 10027, USA
- Present Address: Biochemistry and Molecular Biophysics Department, Columbia University Medical Center, New York, NY 10032, USA
| | - Scott MacDonald
- Physics Department, University of California, Santa Barbara, California 93106, USA
| | - John Crowe
- Physics Department, University of California, Santa Barbara, California 93106, USA
| | - Kai K. Ewert
- Materials Department, University of California, Santa Barbara, California 93106, USA
| | - Bridget Carragher
- The National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY 10027, USA
| | - Clinton S. Potter
- The National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY 10027, USA
| | - Cyrus R. Safinya
- Materials Department, University of California, Santa Barbara, California 93106, USA
- Physics Department, University of California, Santa Barbara, California 93106, USA
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, USA
| |
Collapse
|
149
|
Asayama S. Molecular Design of Polymer-based Carriers for Plasmid DNA Delivery In Vitro and In Vivo. CHEM LETT 2020. [DOI: 10.1246/cl.190696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Shoichiro Asayama
- Department of Applied Chemistry, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| |
Collapse
|
150
|
Kumar S, Singh D, Kumari P, Malik RS, Poonam, Parang K, Tiwari RK. PEGylation and Cell-Penetrating Peptides: Glimpse into the Past and Prospects in the Future. Curr Top Med Chem 2020; 20:337-348. [PMID: 31994461 DOI: 10.2174/1568026620666200128142603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/30/2019] [Accepted: 10/20/2019] [Indexed: 11/22/2022]
Abstract
Several drug molecules have shown low bioavailability and pharmacokinetic profile due to metabolism by enzymes, excretion by the renal system, or due to other physiochemical properties of drug molecules. These problems have resulted in the loss of efficacy and the gain of side effects associated with drug molecules. PEGylation is one of the strategies to overcome these pharmacokinetic issues and has been successful in the clinic. Cell-penetrating Peptides (CPPs) help to deliver molecules across biological membranes and could be used to deliver cargo selectively to the intracellular site or to the drug target. Hence CPPs could be used to improve the efficacy and selectivity of the drug. However, due to the peptidic nature of CPPs, they have a low pharmacokinetic profile. Using PEGylation and CPPs together as a component of a drug delivery system, the and efficacy of drug molecules could be improved. The other important pharmacokinetic properties such as short half-life, solubility, stability, absorption, metabolism, and elimination could be also improved. Here in this review, we summarized PEGylated CPPs or PEGylation based formulations for CPPs used in a drug delivery system for several biomedical applications until August 2019.
Collapse
Affiliation(s)
- Sumit Kumar
- Department of Chemistry, Deenbandhu Chottu Ram University of Science and Technology, Murthal 131039, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohta 124001, India
| | - Pooja Kumari
- Department of Chemistry, Deenbandhu Chottu Ram University of Science and Technology, Murthal 131039, India
| | - Rajender Singh Malik
- Department of Chemistry, Deenbandhu Chottu Ram University of Science and Technology, Murthal 131039, India
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi, Delhi 110007, India
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, United States
| | - Rakesh Kumar Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, United States
| |
Collapse
|