101
|
Kong ZL, Kuo HP, Johnson A, Wu LC, Chang KLB. Curcumin-Loaded Mesoporous Silica Nanoparticles Markedly Enhanced Cytotoxicity in Hepatocellular Carcinoma Cells. Int J Mol Sci 2019; 20:ijms20122918. [PMID: 31207976 PMCID: PMC6628080 DOI: 10.3390/ijms20122918] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/03/2019] [Accepted: 06/13/2019] [Indexed: 02/07/2023] Open
Abstract
Curcumin, a natural polyphenol extracted from a perennial herb Curcuma longa has been verified for many physiological activities such as anti-oxidant, anti-inflammatory, and anti-tumor properties. The direct use of curcumin cytotoxicity studies are limited due to its unstable chemical structure, low bioavailability, easy oxidation, and degradation by ultraviolet (UV) light etc. Trying to overcome this problem, silica-encapsulated curcumin nanoparticles (SCNP) and chitosan with silica co-encapsulated curcumin nanoparticles (CSCNP) were prepared by silicification and biosilicification methods, respectively, and encapsulated curcumin within it. We investigated the antitumor properties of SCNP and CSCNP on different tumor cell lines. Scanning electron microscopy (SEM) analysis revealed that both SCNP and CSCNP were almost spherical in shape and the average particle size of CSCNP was 75.0 ± 14.62 nm, and SCNP was 61.7 ± 23.04 nm. The results show that CSCNP has more anti-oxidant activity as compared to curcumin and SCNP. The higher cytotoxicity towards different cancerous cell lines was also observed in CSCNP treated tumor cells. It was noted that the SCNP and CSCNP has a high percentage of IC50 values in Hep G2 cells. The encapsulation of curcumin improved instability, antioxidant activity, and antitumor activity. Our results demonstrated that nanoencapsulation of curcumin with silica and chitosan not only increase curcumin stability but also enhance its cytotoxic activity on hepatocellular carcinoma cells. On the basis of these primary studies, the curcumin-loaded nanoparticles appear to be promising as an innovative therapeutic material for the treatment of tumors.
Collapse
Affiliation(s)
- Zwe-Ling Kong
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan.
| | - Hsiang-Ping Kuo
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan.
| | - Athira Johnson
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan.
| | - Li-Cyuan Wu
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan.
| | - Ke Liang B Chang
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan.
| |
Collapse
|
102
|
Mahmoudi A, Jaafari MR, Ramezanian N, Gholami L, Malaekeh-Nikouei B. BR2 and CyLoP1 enhance in-vivo SN38 delivery using pegylated PAMAM dendrimers. Int J Pharm 2019; 564:77-89. [DOI: 10.1016/j.ijpharm.2019.04.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/14/2022]
|
103
|
Bratovš A, Kramer L, Mikhaylov G, Vasiljeva O, Turk B. Stefin A-functionalized liposomes as a system for cathepsins S and L-targeted drug delivery. Biochimie 2019; 166:94-102. [PMID: 31163196 DOI: 10.1016/j.biochi.2019.05.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 05/30/2019] [Indexed: 01/26/2023]
Abstract
Proteolytic activity in the tumor microenvironment is one of the key elements supporting tumor development and metastasis. One of the key families of proteases that are overexpressed in various types of cancer and implicated in different stages of tumor progression are cysteine cathepsins. Among them, cathepsins S and L can be secreted into the tumor microenvironment by tumor and/or immune cells, making them promising drug delivery targets. Here we present a new system for cathepsin S/L targeting using a liposomal drug carrier system functionalized with the endogenous cysteine cathepsin inhibitor, stefin A. The selective targeting of cathepsins by stefin A-conjugated liposomes was confirmed in vitro and in vivo, demonstrating the potential of this approach for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Andreja Bratovš
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; Jozef Stefan International Postgraduate School, Jamova 39, Sl-1000 Ljubljana, Slovenia
| | - Lovro Kramer
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Georgy Mikhaylov
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Olga Vasiljeva
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, Večna Pot 113, University of Ljubljana, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
104
|
de Kruijff RM, Raavé R, Kip A, Molkenboer-Kuenen J, Roobol SJ, Essers J, Heskamp S, Denkova AG. Elucidating the Influence of Tumor Presence on the Polymersome Circulation Time in Mice. Pharmaceutics 2019; 11:E241. [PMID: 31137479 PMCID: PMC6572275 DOI: 10.3390/pharmaceutics11050241] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 11/16/2022] Open
Abstract
The use of nanoparticles as tumor-targeting agents is steadily increasing, and the influence of nanoparticle characteristics such as size and stealthiness have been established for a large number of nanocarrier systems. However, not much is known about the impact of tumor presence on nanocarrier circulation times. This paper reports on the influence of tumor presence on the in vivo circulation time and biodistribution of polybutadiene-polyethylene oxide (PBd-PEO) polymersomes. For this purpose, polymersomes were loaded with the gamma-emitter 111In and administered intravenously, followed by timed ex vivo biodistribution. A large reduction in circulation time was observed for tumor-bearing mice, with a circulation half-life of merely 5 min (R2 = 0.98) vs 117 min (R2 = 0.95) in healthy mice. To determine whether the rapid polymersome clearance observed in tumor-bearing mice was mediated by macrophages, chlodronate liposomes were administered to both healthy and tumor-bearing mice prior to the intravenous injection of radiolabeled polymersomes to deplete their macrophages. Pretreatment with chlodronate liposomes depleted macrophages in the spleen and liver and restored the circulation time of the polymersomes with no significant difference in circulation time between healthy mice and tumor-bearing mice pretreated with clodronate liposomes (15.2 ± 1.2% ID/g and 13.6 ± 2.7% ID/g, respectively, at 4 h p.i. with p = 0.3). This indicates that activation of macrophages due to tumor presence indeed affected polymersome clearance rate. Thus, next to particle design, the presence of a tumor can also greatly impact circulation times and should be taken into account when designing studies to evaluate the distribution of polymersomes.
Collapse
Affiliation(s)
- Robin M de Kruijff
- Radiation Science and Technology, Delft University of Technology, 2629 JB Delft, The Netherlands.
| | - René Raavé
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Annemarie Kip
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Janneke Molkenboer-Kuenen
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Stefan J Roobol
- Molecular Genetics, Oncode Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
- Radiology and Nuclear Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Jeroen Essers
- Molecular Genetics, Oncode Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Sandra Heskamp
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Antonia G Denkova
- Radiation Science and Technology, Delft University of Technology, 2629 JB Delft, The Netherlands.
| |
Collapse
|
105
|
Sutariya V, Kelly SJ, Weigel RG, Tur J, Halasz K, Sharma NS, Tipparaju SM. Nanoparticle drug delivery characterization for fluticasone propionate and in vitro testing 1. Can J Physiol Pharmacol 2019; 97:675-684. [PMID: 31100204 DOI: 10.1139/cjpp-2018-0569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucocorticoids, such as fluticasone propionate (FP), are used for the treatment of inflammation and alleviation of nasal symptoms and allergies, and as an antipruritic. However, both short- and long-term therapeutic use of glucocorticoids can lead to muscle weakness and atrophy. In the present study, we evaluated the feasibility of the nanodelivery of FP with poly(dl-lactide-co-glycolide) (PLGA) and tested in vitro function. FP-loaded PLGA nanoparticles were prepared via nanoprecipitation and morphological characteristics were studied via scanning electron microscopy. FP-loaded nanoparticles demonstrated an encapsulation efficiency of 68.6% ± 0.5% with a drug loading capacity of 4.6% ± 0.04%, were 128.8 ± 0.6 nm in diameter with a polydispersity index of 0.07 ± 0.008, and displayed a zeta potential of -19.4 ± 0.7. A sustained in vitro drug release pattern was observed for up to 7 days. The use of fluticasone nanoparticle decreased lipopolysaccharide (LPS)-induced lactate dehydrogenase release compared with LPS alone in C2C12 treated cells. FP also decreased expression of LPS-induced inflammatory genes in C2C12 treated cells as compared with LPS alone. Taken together, the present study demonstrates in vitro feasibility of PLGA-FP nanoparticle delivery to the skeletal muscle cells, which may be beneficial for treating inflammation.
Collapse
Affiliation(s)
- Vijaykumar Sutariya
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Shannon J Kelly
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Robert G Weigel
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Jared Tur
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Kathleen Halasz
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Nirmal S Sharma
- b Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Srinivas M Tipparaju
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
106
|
Ma X, Özliseli E, Zhang Y, Pan G, Wang D, Zhang H. Fabrication of redox-responsive doxorubicin and paclitaxel prodrug nanoparticles with microfluidics for selective cancer therapy. Biomater Sci 2019; 7:634-644. [PMID: 30534690 DOI: 10.1039/c8bm01333k] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cancer is an exceptionally confounding disease that demands the development of powerful drug/drugs, without inducing heavy adverse side effects. Thus, different approaches have been applied to improve the targeted delivery of cancer drugs: for example by using nanocarriers. However, nanocarriers are foreign materials, which need further validation for their biocompatibility and biodegradability. In this study, we have chemically conjugated the hydrophilic anticancer drug doxorubicin (DOX) with the hydrophobic drug paclitaxel (PTX) through a redox-sensitive disulfide bond, abbreviated to DOX-S-S-PTX. Subsequently, due to its amphiphilic characterization, the prodrug can self-assemble into nanoparticles under microfluidic nanoprecipitation. These novel prodrug nanoparticles have a super-high drug loading degree of 89%, which is impossible to achieve by any nanocarrier systems, and can be tailored to 180 nm to deliver themselves to the target, and release DOX and PTX under redox conditions, which are often found in cancer cells. By evaluating cell viability in MDA-MB-231, MDA-MB-231/ADR and MEF cell lines, we observed that the prodrug nanoparticles effectively killed the cancer cells, and selectively conquered the MDA-MB-231/ADR. Meanwhile, MEF cells were spared due to their lack of a redox condition. The cell interaction results show that the reduced intermediate of the prodrug can also bind to parent drug biological targets. The hemolysis results show that the nanoparticles are biocompatible in blood. Computer modelling suggested that the prodrug is unlikely to bind to biological targets that parent drugs still strongly interact with. Finally, we confirm that the prodrug nanoparticles have no therapeutic effect in blood or healthy cells, but can selectively eliminate the cancer cells that meet the redox conditions to cleave the disulfide bond and release the drugs DOX and PTX.
Collapse
Affiliation(s)
- Xiaodong Ma
- Department of Radiology affiliated Hospital of Jiangsu University Jiangsu University, 212001 Zhenjiang, P.R. China
| | | | | | | | | | | |
Collapse
|
107
|
Mignani S, Rodrigues J, Roy R, Shi X, Ceña V, El Kazzouli S, Majoral JP. Exploration of biomedical dendrimer space based on in-vivo physicochemical parameters: Key factor analysis (Part 2). Drug Discov Today 2019. [DOI: https://doi.org/10.1016/j.drudis.2019.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
108
|
Mignani S, Rodrigues J, Roy R, Shi X, Ceña V, El Kazzouli S, Majoral JP. Exploration of biomedical dendrimer space based on in-vivo physicochemical parameters: Key factor analysis (Part 2). Drug Discov Today 2019; 24:1184-1192. [PMID: 30904723 DOI: 10.1016/j.drudis.2019.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/01/2019] [Accepted: 03/01/2019] [Indexed: 02/08/2023]
Abstract
In nanomedicine, the widespread concern of nanoparticles in general, and dendrimers, in particular, is the analysis of key in-vivo physicochemical parameters to ensure the preclinical and clinical development of 'safe' bioactive nanomaterials. It is clear that for biomedical applications, biocompatible dendrimers, used as nanocarriers or active per se, should be devoid of toxicity and immunogenicity, and have adequate PK/PD behaviors (adequate exposure) in order to diffuse in different tissues. Functionalization of dendrimers has a dramatic effect on in-vivo physicochemical parameters. In this review, we highlighted key in-vivo physicochemical properties, based on data from biochemical, cellular and animal models, to provide biocompatible dendrimers. Up-to-date, only scarce studies have been described on this topic.
Collapse
Affiliation(s)
- Serge Mignani
- Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, 45, rue des Saints Peres, 75006 Paris, France; CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; Glycovax Pharma, 424 Guy Street, Suite 202, Montreal, Quebec H3J 1S6, Canada; Department of Pharmacy, Zhengzhou Railway Vocational & Technical College, Zhengzhou 450018, China.
| | - João Rodrigues
- CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; School of Materials Science and Engineering/Center for Nano Energy Materials, Northwestern Polytechnical University, Xi'an 710072, China.
| | - René Roy
- Glycovax Pharma, 424 Guy Street, Suite 202, Montreal, Quebec H3J 1S6, Canada.
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, PR China
| | - Valentin Ceña
- Unidad Asociada Neurodeath, Universidad de Castilla-La Mancha, 02006 Albacete, Spain; Centro de Investigación Biomédica en Red para Enfermedades Neurodegenerativas, ISCIII, 28031 Madrid, Spain
| | - Saïd El Kazzouli
- Euromed Research Center, Euromed Faculty of Engineering, Euromed University of Fes (UEMF), Route de Meknès, 30000 Fès, Morocco
| | - Jean-Pierre Majoral
- Department of Pharmacy, Zhengzhou Railway Vocational & Technical College, Zhengzhou 450018, China; Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France; Université Toulouse 118 route de Narbonne, 31077 Toulouse Cedex 4, France.
| |
Collapse
|
109
|
Abstract
Radiation therapy has made tremendous progress in oncology over the last decades due to advances in engineering and physical sciences in combination with better biochemical, genetic and molecular understanding of this disease. Local delivery of optimal radiation dose to a tumor, while sparing healthy surrounding tissues, remains a great challenge, especially in the proximity of vital organs. Therefore, imaging plays a key role in tumor staging, accurate target volume delineation, assessment of individual radiation resistance and even personalized dose prescription. From this point of view, radiotherapy might be one of the few therapeutic modalities that relies entirely on high-resolution imaging. Magnetic resonance imaging (MRI) with its superior soft-tissue resolution is already used in radiotherapy treatment planning complementing conventional computed tomography (CT). Development of systems integrating MRI and linear accelerators opens possibilities for simultaneous imaging and therapy, which in turn, generates the need for imaging probes with therapeutic components. In this review, we discuss the role of MRI in both external and internal radiotherapy focusing on the most important examples of contrast agents with combined therapeutic potential.
Collapse
|
110
|
Kafshgari MH, Mazare A, Distaso M, Goldmann WH, Peukert W, Fabry B, Schmuki P. Intracellular Drug Delivery with Anodic Titanium Dioxide Nanotubes and Nanocylinders. ACS APPLIED MATERIALS & INTERFACES 2019; 11:14980-14985. [PMID: 30916543 DOI: 10.1021/acsami.9b01211] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Titanium dioxide (TiO2) holds remarkable promises for developing current theranostic strategies. Anodic TiO2 nanostructures as a porous scaffold have offered a broad range of useful theranostic properties; however, previous attempts to generate single and uniform TiO2 one-dimensional nanocarriers from anodic nanotube arrays have resulted in a broad cluster size distribution of arbitrarily broken tubes that are unsuitable for therapeutic delivery systems due to poor biodistribution and the risk of introducing tissue inflammation. Here, we achieve well-separated, uniformly shaped anodic TiO2 nanotubes and nanocylinders through a time-varying electrochemical anodization protocol that leads to the generation of planar sheets of weakly connected nanotubes with a defined fracture point near the base. Subsequent sonication cleanly detaches the nanotubes from the base. Depending on the position of the fracture point, we can fabricate single-anodic nanocylinders that are open on both ends and nanotubes that are closed on one end. We proceed to show that anodic nanotubes and nanocylinders are nontoxic at therapeutic concentrations. When conjugated with the anticancer drug doxorubicin using a pH-responsive linker, they are readily internalized by cells and subsequently release their drug cargo into acidic intracellular compartments. Our results demonstrate that uniformly sized anodic TiO2 nanotubes and nanocylinders are suitable for subcellular delivery of therapeutic agents in cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Wolfgang H Goldmann
- Department of Physics, Biophysics Group , University of Erlangen-Nuremberg , Erlangen 91052 , Germany
| | | | - Ben Fabry
- Department of Physics, Biophysics Group , University of Erlangen-Nuremberg , Erlangen 91052 , Germany
| | | |
Collapse
|
111
|
Lobaz V, Konefał R, Pánek J, Vlk M, Kozempel J, Petřík M, Novy Z, Gurská S, Znojek P, Štěpánek P, Hrubý M. In Situ In Vivo radiolabeling of polymer-coated hydroxyapatite nanoparticles to track their biodistribution in mice. Colloids Surf B Biointerfaces 2019; 179:143-152. [PMID: 30954015 DOI: 10.1016/j.colsurfb.2019.03.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 11/26/2022]
Abstract
The imaging of healthy tissues and solid tumors benefits from the application of nanoparticle probes with altered pharmacokinetics, not available to low molecular weight compounds. However, the distribution and accumulation of nanoprobes in vivo typically take at least tens of hours to be efficient. For nanoprobes bearing a radioactive label, this is contradictory to the requirement of minimizing the radiation dose for patients by using as-short-as-feasible half-life radionuclides in diagnostics. Thus, we developed a two-stage diagnostic concept for monitoring long-lasting targeting effects with short-lived radioactive labels using bone-mimicking biocompatible polymer-coated and colloidally fully stabilized hydroxyapatite nanoparticles (HAP NPs) and bone-seeking radiopharmaceuticals. Within the pretargeting stage, the nonlabeled nanoparticles are allowed to circulate in the blood. Afterward, 99mTc-1-hydroxyethylidene-1.1-diphosphonate (99mTc-HEDP) is administered intravenously for in situ labeling of the nanoparticles and subsequent single-photon emission computed tomography/computed tomography (SPECT/CT) visualization. The HAP NPs, stabilized with tailored hydrophilic polymers, are not cytotoxic in vitro, as shown by several cell lines. The polymer coating prolongs the circulation of HAP NPs in the blood. The nanoparticles were successfully labeled in vivo with 99mTc-HEDP, 1 and 24 h after injection, and they were visualized by SPECT/CT over time in healthy mice.
Collapse
Affiliation(s)
- Volodymyr Lobaz
- Institute of Macromolecular Chemistry AS CR, Heyrovsky Sq. 1888/2, 162 06, Prague 6, Czech Republic.
| | - Rafał Konefał
- Institute of Macromolecular Chemistry AS CR, Heyrovsky Sq. 1888/2, 162 06, Prague 6, Czech Republic
| | - Jiří Pánek
- Institute of Macromolecular Chemistry AS CR, Heyrovsky Sq. 1888/2, 162 06, Prague 6, Czech Republic
| | - Martin Vlk
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University, Břehová 7, 115 19, Prague 1, Czech Republic
| | - Ján Kozempel
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University, Břehová 7, 115 19, Prague 1, Czech Republic
| | - Miloš Petřík
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 5, 779 00, Olomouc, Czech Republic
| | - Zbyněk Novy
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 5, 779 00, Olomouc, Czech Republic
| | - Soňa Gurská
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 5, 779 00, Olomouc, Czech Republic
| | - Pawel Znojek
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 5, 779 00, Olomouc, Czech Republic
| | - Petr Štěpánek
- Institute of Macromolecular Chemistry AS CR, Heyrovsky Sq. 1888/2, 162 06, Prague 6, Czech Republic
| | - Martin Hrubý
- Institute of Macromolecular Chemistry AS CR, Heyrovsky Sq. 1888/2, 162 06, Prague 6, Czech Republic
| |
Collapse
|
112
|
Andersen MN, Etzerodt A, Graversen JH, Holthof LC, Moestrup SK, Hokland M, Møller HJ. STAT3 inhibition specifically in human monocytes and macrophages by CD163-targeted corosolic acid-containing liposomes. Cancer Immunol Immunother 2019; 68:489-502. [PMID: 30637473 PMCID: PMC11028169 DOI: 10.1007/s00262-019-02301-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/06/2019] [Indexed: 01/05/2023]
Abstract
Tumor-associated macrophages (TAMs) are of major importance in cancer-related immune suppression, and tumor infiltration by CD163pos TAMs is associated with poor outcome in most human cancers. Therefore, therapeutic strategies for reprogramming TAMs from a tumor-supporting (M2-like) phenotype towards a tumoricidal (M1-like) phenotype are of great interest. Activation of the transcription factor STAT3 within the tumor microenvironment is associated with worse prognosis, and STAT3 activation promotes the immunosuppressive phenotype of TAMs. Therefore, we aimed to develop a drug for inhibition of STAT3 specifically within human TAMs by targeting the endocytic CD163 scavenger receptor, which is highly expressed on TAMs. Here, we report the first data on a CD163-targeted STAT3-inhibitory drug consisting of corosolic acid (CA) packaged within long-circulating liposomes (LCLs), which are CD163-targeted by modification with monoclonal anti-CD163 antibodies (αCD163)-CA-LCL-αCD163. We show, that activation of STAT3 (by phosphorylation) was inhibited by CA-LCL-αCD163 specifically within CD163pos cells, with minor effect on CD163neg cells. Furthermore, CA-LCL-αCD163 inhibited STAT3-regulated gene expression of IL-10, and increased expression of TNFα, thus indicating a pro-inflammatory effect of the drug on human macrophages. This M1-like reprogramming at the mRNA level was confirmed by significantly elevated levels of pro-inflammatory cytokines (IFNγ, IL-12, TNFα, IL-2) in the culture medium. Since liposomes are attractive vehicles for novel anti-cancer drugs, and since direct TAM-targeting may decrease adverse effects of systemic inhibition of STAT3, the present results encourage future investigation of CA-LCL-αCD163 in the in vivo setting.
Collapse
Affiliation(s)
- Morten Nørgaard Andersen
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensen Boulevard 99, 8200, Aarhus N, Denmark.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark.
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jonas H Graversen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lisa C Holthof
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Søren K Moestrup
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensen Boulevard 99, 8200, Aarhus N, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Holger J Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensen Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
113
|
Kim MW, Jeong HY, Kang SJ, Jeong IH, Choi MJ, You YM, Im CS, Song IH, Lee TS, Lee JS, Lee A, Park YS. Anti-EGF Receptor Aptamer-Guided Co-Delivery of Anti-Cancer siRNAs and Quantum Dots for Theranostics of Triple-Negative Breast Cancer. Am J Cancer Res 2019; 9:837-852. [PMID: 30809312 PMCID: PMC6376474 DOI: 10.7150/thno.30228] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/28/2018] [Indexed: 12/18/2022] Open
Abstract
Many aptamers have been evaluated for their ability as drug delivery vehicles to target ligands, and a variety of small interfering RNAs (siRNAs) have been tested for their anti-cancer properties. However, since these two types of molecules have similar physicochemical properties, it has so far been difficult to formulate siRNA-encapsulating carriers guided by aptamers. Here, we propose aptamer-coupled lipid nanocarriers encapsulating quantum dots (QDs) and siRNAs for theragnosis of triple-negative breast cancer (TNBC). Methods: Hydrophobic QDs were effectively incorporated into lipid bilayers, and then therapeutic siRNAs were complexed with QD-lipid nanocarriers (QLs). Finally, anti-EGFR aptamer-lipid conjugates were inserted into the QLs for TNBC targeting (aptamo-QLs). TNBC-targeting aptamo-QLs were directly compared to anti-EGFR antibody-coupled immuno-QLs. The in vitro delivery of therapeutic siRNAs and QDs to target cells was assessed by flow cytometry and confocal microscopy. The in vivo targeting of siRNAs to tumors and their therapeutic efficacy were evaluated in mice carrying MDA-MB-231 tumors. Results: Both types of EGFR-targeting QLs showed enhanced delivery to target cancer cells, resulting in more effective gene silencing and enhanced tumor imaging compared to non-targeting control QLs. Moreover, combinatorial therapy with Bcl-2 and PKC-ι siRNAs loaded into the anti-EGFR QLs was remarkably effective in inhibiting tumor growth and metastasis. Conclusion: In general, the aptamo-QLs showed competitive in vivo delivery and therapeutic efficacy compared to immuno-QLs under the same experimental conditions. Our results show that the anti-EGFR aptamer-guided lipid carriers may be a potential theranostic delivery vehicle for RNA interference and fluorescence imaging of TNBCs.
Collapse
|
114
|
Ueda M, Seo S, Nair BG, Müller S, Takahashi E, Arai T, Iyoda T, Fujii SI, Tsuneda S, Ito Y. End-Sealed High Aspect Ratio Hollow Nanotubes Encapsulating an Anticancer Drug: Torpedo-Shaped Peptidic Nanocapsules. ACS NANO 2019; 13:305-312. [PMID: 30606006 DOI: 10.1021/acsnano.8b06189] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanomaterial morphology is important for the targeted delivery of drugs to tissues as well as subsequent cellular uptake. Hollow nanotubes composed of peptides, with a diameter of 80 nm and various lengths (100, 200, 300, 600 nm), were successfully capped and sealed with a peptide hemisphere to encapsulate the anticancer drug, cisplatin. The torpedo-shaped nanocapsules with an aspect ratio (length/diameter) of 2.4 showed more rapid cellular uptake and accumulation at the tumor site compared with spherical analogues. Successful delivery of cisplatin to tumors was achieved in a mouse model and tumor growth was efficiently suppressed.
Collapse
Affiliation(s)
- Motoki Ueda
- Emergent Bioengineering Materials Research Team , RIKEN Center for Emergent Matter Science (CEMS) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
- Nano Medical Engineering Laboratory , RIKEN Cluster for Pioneering Research (CPR) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
| | - Siyoong Seo
- Emergent Bioengineering Materials Research Team , RIKEN Center for Emergent Matter Science (CEMS) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
- Nano Medical Engineering Laboratory , RIKEN Cluster for Pioneering Research (CPR) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
- Department of Life Science and Medical Bioscience , Waseda University , 2-2 Wakamatsu-cho , Shinjuku-ku, Tokyo 162-8480 , Japan
| | - Baiju G Nair
- Nano Medical Engineering Laboratory , RIKEN Cluster for Pioneering Research (CPR) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
| | - Stefan Müller
- Emergent Bioengineering Materials Research Team , RIKEN Center for Emergent Matter Science (CEMS) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
- Research Resources Division , RIKEN Center for Brain Science (CBS) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
| | - Eiki Takahashi
- Research Resources Division , RIKEN Center for Brain Science (CBS) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
| | - Takashi Arai
- Research Resources Division , RIKEN Center for Brain Science (CBS) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
| | - Tomonori Iyoda
- Laboratory for Immunotherapy , RIKEN Center for Integrative Medical Science (IMS) , 1-7-22 Suehiro-cho , Tsurumi-ku, Yokohama , Kanagawa 230-0045 , Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy , RIKEN Center for Integrative Medical Science (IMS) , 1-7-22 Suehiro-cho , Tsurumi-ku, Yokohama , Kanagawa 230-0045 , Japan
| | - Satoshi Tsuneda
- Department of Life Science and Medical Bioscience , Waseda University , 2-2 Wakamatsu-cho , Shinjuku-ku, Tokyo 162-8480 , Japan
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research Team , RIKEN Center for Emergent Matter Science (CEMS) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
- Nano Medical Engineering Laboratory , RIKEN Cluster for Pioneering Research (CPR) , 2-1 Hirosawa , Wako, Saitama 351-0198 , Japan
| |
Collapse
|
115
|
Liu J, Lécuyer T, Seguin J, Mignet N, Scherman D, Viana B, Richard C. Imaging and therapeutic applications of persistent luminescence nanomaterials. Adv Drug Deliv Rev 2019; 138:193-210. [PMID: 30414492 DOI: 10.1016/j.addr.2018.10.015] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/26/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022]
Abstract
The development of probes for biomolecular imaging and diagnostics is a very active research area. Among the different imaging modalities, optics emerged since it is a noninvasive and cheap imaging technique allowing real time imaging. In vitro, this technique is very useful however in vivo, fluorescence suffers from low signal-to-noise ratio due to tissue autofluorescence under constant excitation. To address this limitation, novel types of optical nanoprobes are actually being developed and among them, persistent luminescence nanoparticles (PLNPs), with long lasting near-infrared (NIR) luminescence capability, allows doing optical imaging without constant excitation and so without autofluorescence. This review will begin by introducing the physical phenomenon associated to the long luminescence decay of such nanoprobes, from minutes to hours after ceasing the excitation. Then we will show how this property can be used to develop in vivo imaging probes and also more recently nanotheranostic agents. Finally, preliminary data on their biocompatibility will be mentioned and we will conclude by envisioning on the future applications and improvements of such nanomaterials.
Collapse
|
116
|
Ngoune R, Contini C, Hoffmann MM, von Elverfeldt D, Winkler K, Putz G. Optimizing Antitumor Efficacy and Adverse Effects of Pegylated Liposomal Doxorubicin by Scheduled Plasmapheresis: Impact of Timing and Dosing. Curr Drug Deliv 2018; 15:1261-1270. [PMID: 29779479 PMCID: PMC6327121 DOI: 10.2174/1567201815666180518125839] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/11/2018] [Accepted: 05/11/2018] [Indexed: 12/25/2022]
Abstract
Background: Nanoscale drug delivery systems accumulate in solid tumors preferentially by the enhanced permeation and retention effect (EPR-effect). Nevertheless, only a miniscule fraction of a given dosage reaches the tumor, while >90% of the given drug ends up in otherwise healthy tissues, lead-ing to the severe toxic reactions observed during chemotherapy. Once accumulation in the tumor has reached its maximum, extracorporeal elimination of circulating nanoparticles by plasmapheresis can dimin-ish toxicities. Objective: In this study, we investigated the effect of dosing and plasmapheresis timing on adverse events and antitumor efficacy in a syngeneic rat tumor model. Methods: MAT-B-III cells transfected with a luciferase reporter plasmid were inoculated into female Fisher rats, and pegylated liposomal doxorubicin (PLD) was used for treatment. Plasmapheresis was performed in a discontinuous manner via centrifugation and subsequent filtration of isolated plasma. Results: Bioluminescence measurements of tumor growth could not substitute caliper measurements of tumor size. In the control group, raising the dosage above 9 mg PLD/kg body weight did not increase therapeutic efficacy in our fully immunocompetent animal model. Plasmapheresis was best done 36 h after injecting PLD, leading to similar antitumor efficacy with significantly less toxicity. Plasmapheresis 24 h after injection interfered with therapeutic efficacy, while plasmapheresis after 48 h led to fewer side effects but also to increased weight loss. Conclusion: Long-circulating nanoparticles offer the unique possibility to eliminate the excess of circulat-ing particles after successful accumulation in tumors by EPR, thereby reducing toxicities and likely toxici-ty-related therapeutic limitations
Collapse
Affiliation(s)
- Romeo Ngoune
- Medical Center - University of Freiburg, Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Freiburg, Germany
| | - Christine Contini
- Medical Center - University of Freiburg, Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Freiburg, Germany
| | - Michael M Hoffmann
- Medical Center - University of Freiburg, Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Freiburg, Germany
| | - Dominik von Elverfeldt
- Medical Center - University of Freiburg, Faculty of Medicine, Department of Diagnostic Radiology Medical Physics, Freiburg, Germany
| | - Karl Winkler
- Medical Center - University of Freiburg, Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Freiburg, Germany
| | - Gerhard Putz
- Medical Center - University of Freiburg, Faculty of Medicine, Institute for Clinical Chemistry and Laboratory Medicine, Freiburg, Germany
| |
Collapse
|
117
|
The availability of drug by liposomal drug delivery : Individual kinetics and tissue distribution of encapsulated and released drug in mice after administration of PEGylated liposomal prednisolone phosphate. Invest New Drugs 2018; 37:890-901. [PMID: 30547315 PMCID: PMC6736927 DOI: 10.1007/s10637-018-0708-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022]
Abstract
Lately, the usefulness of liposomal drug delivery systems has been debated. To better understand the underlying pharmacokinetics of the targeted drug delivery by liposomes, individual encapsulated and non-encapsulated drug concentrations in blood, tumor, liver, spleen and kidneys were quantified after i.v. administration of liposomal prednisolone phosphate in mice. Kinetic analysis shows that the tumor influx of encapsulated drug is not dominant compared to the uptake by the other tissues. Further, from a quantitative point of view, the availability of non-encapsulated drug in the tumor tissue after liposomal delivery is not pronounced as compared to the other tissues studied. However, drug release in the tumor seems more extended than in the other tissues and the non-encapsulated drug concentration decreases more slowly in the tumor than in the liver and spleen. The spleen shows a high affinity for the uptake of encapsulated drug as well as the release of drug from the liposomes. Subsequently, released drug in the spleen, and possibly also in other tissues, is probably quickly redistributed towards the blood and other tissues. This also impairs the drug delivery effect of the liposomes. In contrast to the released drug in the central circulation, liver and spleen, the released drug concentration in the tumor remains at a fairly constant level likely due to the extended release kinetics from the liposomes. These extended release characteristics in the tumor most probably contribute to the beneficial effect. Nevertheless, it should be noted that larger released drug concentrations are formed in healthy tissues.
Collapse
|
118
|
The impact of protein corona on the behavior and targeting capability of nanoparticle-based delivery system. Int J Pharm 2018; 552:328-339. [DOI: 10.1016/j.ijpharm.2018.10.011] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/04/2018] [Accepted: 10/06/2018] [Indexed: 01/04/2023]
|
119
|
Braham MV, Deshantri AK, Minnema MC, Öner FC, Schiffelers RM, Fens MH, Alblas J. Liposomal drug delivery in an in vitro 3D bone marrow model for multiple myeloma. Int J Nanomedicine 2018; 13:8105-8118. [PMID: 30555229 PMCID: PMC6278842 DOI: 10.2147/ijn.s184262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose Liposomal drug delivery can improve the therapeutic index of treatments for multiple myeloma. However, an appropriate 3D model for the in vitro evaluation of liposomal drug delivery is lacking. In this study, we applied a previously developed 3D bone marrow (BM) myeloma model to examine liposomal drug therapy. Material and methods Liposomes of different sizes (~75-200 nm) were tested in a 3D BM myeloma model, based on multipotent mesenchymal stromal cells, endothelial progenitor cells, and myeloma cells cocultured in hydrogel. The behavior and efficacy of liposomal drug therapy was investigated, evaluating the feasibility of testing liposomal drug delivery in 3D in vitro. Intracellular uptake of untargeted and integrin α4β1 (very late antigen-4) targeted liposomes was compared in myeloma and supporting cells, as well as the effectivity of free and liposome-encapsulated chemotherapy (bortezomib, doxorubicin). Either cocultured myeloma cell lines or primary CD138+ myeloma cells received the treatments. Results Liposomes (~75-110 nm) passively diffused throughout the heterogeneously porous (~80-850 nm) 3D hydrogel model after insertion. Cellular uptake of liposomes was observed and was increased by targeting very late antigen-4. Liposomal bortezomib and doxorubicin showed increased cytotoxic effects toward myeloma cells compared with the free drugs, using either a cell line or primary myeloma cells. Cytotoxicity toward supporting BM cells was reduced using liposomes. Conclusion The 3D model allows the study of liposome-encapsulated molecules on multiple myeloma and supporting BM cells, looking at cellular targeting, and general efficacy of the given therapy. The advantages of liposomal drug delivery were demonstrated in a primary myeloma model, enabling the study of patient-to-patient responses to potential drugs and treatment regimes.
Collapse
Affiliation(s)
- Maaike Vj Braham
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, the Netherlands,
| | - Anil K Deshantri
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Pharmacology, Sun Pharma Advanced Research Company Limited, Vadodara, Gujarat, India
| | - Monique C Minnema
- Department of Hematology, University Medical Center Utrecht Cancer Center, Utrecht, the Netherlands
| | - F Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, the Netherlands,
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marcel Ham Fens
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, the Netherlands,
| |
Collapse
|
120
|
Enhanced nanoparticle delivery exploiting tumour-responsive formulations. Cancer Nanotechnol 2018; 9:10. [PMID: 30595759 PMCID: PMC6276285 DOI: 10.1186/s12645-018-0044-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/01/2018] [Indexed: 12/27/2022] Open
Abstract
Nanoparticles can be used as drug carriers, contrast
agents and radiosensitisers for the treatment of cancer. Nanoparticles can either passively accumulate within tumour sites, or be conjugated with targeting ligands to actively enable tumour deposition. With respect to passive accumulation, particles < 150 nm accumulate with higher efficiency within the tumour microenvironment, a consequence of the enhanced permeability and retention effect. Despite these favourable properties, clinical translation of nano-therapeutics is inhibited due to poor in vivo stability, biodistribution and target cell internalisation. Nano-therapeutics can be modified to exploit features of the tumour microenvironment such as elevated hypoxia, increased pH and a compromised extracellular matrix. This is in contrast to cytotoxic chemotherapies which generally do not exploit the characteristic pathological features of the tumour microenvironment, and as such are prone to debilitating systemic toxicities. This review examines strategies for tumour microenvironment targeting to improve nanoparticle delivery, with particular focus on the delivery of nucleic acids and gold nanoparticles. Evidence for key research areas and future technologies are presented and critically evaluated. Among the most promising technologies are the development of next-generation cell penetrating peptides and the incorporation of micro-environment responsive stealth molecules.
Collapse
|
121
|
Troendle EP, Khan A, Searson PC, Ulmschneider MB. Predicting drug delivery efficiency into tumor tissues through molecular simulation of transport in complex vascular networks. J Control Release 2018; 292:221-234. [PMID: 30415016 PMCID: PMC10131895 DOI: 10.1016/j.jconrel.2018.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022]
Abstract
Efficient delivery of anticancer drugs into tumor tissues at maximally effective and minimally toxic concentrations is vital for therapeutic success. At present, no method exists that can predict the spatial and temporal distribution of drugs into a target tissue after administration of a specific dose. This prevents accurate estimation of optimal dosage regimens for cancer therapy. Here we present a new method that predicts quantitatively the time-dependent spatial distribution of drugs in tumor tissues at sub-micrometer resolution. This is achieved by modeling the diffusive flow of individual drug molecules through the three-dimensional network of blood-vessels that vascularize the tumor, and into surrounding tissues, using molecular mechanics techniques. By evaluating delivery into tumors supplied by a series of blood-vessel networks with varying degrees of complexity, we show that the optimal dose depends critically on the precise vascular structure. Finally, we apply our method to calculate the optimal dosage of the cancer drug doxil into a section of a mouse ovarian tumor, and demonstrate the enhanced delivery of liposomally administered doxorubicin when compared to free doxorubicin. Comparison with experimental data and a multiple-compartment model show that the model accurately recapitulates known pharmacokinetics and drug-load predictions. In addition, it provides, for the first time, a detailed picture of the spatial dependence of drug uptake into tissues surrounding tumor vasculatures. This approach is fundamentally different to current continuum models, and reveals that the target tumor vascular topology is as important for therapeutic success as the transport properties of the drug delivery platform itself. This sets the stage for revisiting drug dosage calculations.
Collapse
Affiliation(s)
- Evan P Troendle
- Department of Chemistry, King's College London, London, UK; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ayesha Khan
- University of Exeter Medical School, Exeter, UK
| | - Peter C Searson
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Martin B Ulmschneider
- Department of Chemistry, King's College London, London, UK; University of Exeter Medical School, Exeter, UK; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
122
|
Amirshaghaghi A, Altun B, Nwe K, Yan L, Stein JM, Cheng Z, Tsourkas A. Site-Specific Labeling of Cyanine and Porphyrin Dye-Stabilized Nanoemulsions with Affibodies for Cellular Targeting. J Am Chem Soc 2018; 140:13550-13553. [PMID: 30351141 DOI: 10.1021/jacs.8b07866] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recently, it has been shown that amphiphilic dyes such as Indocyanine Green (ICG) and Protoporphyrin IX (PpIX) can solubilize hydrophobic colloids and/or drugs by driving the formation of stable nanoemulsions. These nanoemulsions are unique in that they can be composed entirely of functional and clinically used materials; however, they lack bio-orthogonal chemical handles for the facile attachment of targeting ligands. The ability to target nanoparticles is desirable because it can lead to improved specificity and reduced side effects. Here, we describe variants of ICG and PpIX with azide handles that can be readily incorporated into dye-stabilized nanoemulsions and facilitate the attachment of targeting ligands via click-chemistry in a simple, scalable, and reproducible reaction. As a model system, an anti-Her2 affibody was site-specifically attached to both ICG and PpIX-stabilized nanoemulsions with encapsulated superparamagnetic iron oxide nanoparticles.
Collapse
Affiliation(s)
- Ahmad Amirshaghaghi
- Department of Bioengineering, School of Engineering and Applied Sciences , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Burcin Altun
- Department of Bioengineering, School of Engineering and Applied Sciences , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Kido Nwe
- Chemical and Nanoparticle Synthesis Core , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Lesan Yan
- Chemical and Nanoparticle Synthesis Core , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Joel M Stein
- Department of Bioengineering, School of Engineering and Applied Sciences , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States.,Department of Radiology, Division of Neuroradiology , Hospital of the University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Zhiliang Cheng
- Department of Bioengineering, School of Engineering and Applied Sciences , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Andrew Tsourkas
- Department of Bioengineering, School of Engineering and Applied Sciences , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| |
Collapse
|
123
|
Raut S, Mooberry L, Sabnis N, Garud A, Dossou AS, Lacko A. Reconstituted HDL: Drug Delivery Platform for Overcoming Biological Barriers to Cancer Therapy. Front Pharmacol 2018; 9:1154. [PMID: 30374303 PMCID: PMC6196266 DOI: 10.3389/fphar.2018.01154] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Drug delivery to malignant tumors is limited by several factors, including off-target toxicities and suboptimal benefits to cancer patient. Major research efforts have been directed toward developing novel technologies involving nanoparticles (NPs) to overcome these challenges. Major obstacles, however, including, opsonization, transport across cancer cell membranes, multidrug-resistant proteins, and endosomal sequestration of the therapeutic agent continue to limit the efficiency of cancer chemotherapy. Lipoprotein-based drug delivery technology, "nature's drug delivery system," while exhibits highly desirable characteristics, it still needs substantial investment from private/government stakeholders to promote its eventual advance to the bedside. Consequently, this review focuses specifically on the synthetic (reconstituted) high-density lipoprotein rHDL NPs, evaluating their potential to overcome specific biological barriers and the challenges of translation toward clinical utilization and commercialization. This highly robust drug transport system provides site-specific, tumor-selective delivery of anti-cancer agents while reducing harmful off-target effects. Utilizing rHDL NPs for anti-cancer therapeutics and tumor imaging revolutionizes the future strategy for the management of a broad range of cancers and other diseases.
Collapse
Affiliation(s)
- Sangram Raut
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Linda Mooberry
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Nirupama Sabnis
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Ashwini Garud
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Akpedje Serena Dossou
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Andras Lacko
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
124
|
Abstract
Imaging plays a key role in the preclinical evaluation of nanomedicine-based drug delivery systems and it has provided important insights into their mechanism of action and therapeutic effect. Its role in supporting the clinical development of nanomedicine products, however, has been less explored. In this review, we summarize clinical studies in which imaging has provided valuable information on the pharmacokinetics, biodistribution, and target site accumulation of nanomedicine-based drug delivery systems. Importantly, these studies provide convincing evidence on the uptake of nanomedicines in tumors, confirming that the enhanced permeability and retention (EPR) effect is a real phenomenon in patients, albeit with fairly high levels of inter- and intraindividual variability. It is gradually becoming clear that imaging is critically important to help address this high heterogeneity. In support of this notion, a decent correlation between nanomedicine uptake in tumors and antitumor efficacy has recently been obtained in two independent studies in patients, exemplifying that image-guided drug delivery can help to pave the way towards individualized and improved nanomedicine therapies.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK.
| |
Collapse
|
125
|
Wang L, Hu Y, Hao Y, Li L, Zheng C, Zhao H, Niu M, Yin Y, Zhang Z, Zhang Y. Tumor-targeting core-shell structured nanoparticles for drug procedural controlled release and cancer sonodynamic combined therapy. J Control Release 2018; 286:74-84. [PMID: 30026078 DOI: 10.1016/j.jconrel.2018.07.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/13/2018] [Accepted: 07/15/2018] [Indexed: 01/10/2023]
Abstract
Combination therapy with multiple drugs or/and multiple assistant treatments has become a hot spot in cancer therapy. In this study, a new type of core-shell structured dual-drug delivery system based on poly (lactic-co-glycolic acid) (PLGA, inner cores) and hyaluronic acid (HA, outer shells) was constructed. Firstly, HA was conjugated to PLGA for preparation of HA-PLGA block copolymer. Secondly, 5-amino levulinic acid (ALA) was connected to PLGA through a pH-sensitive hydrazone bond for synthesization of PLGA-HBA-ALA. Finally, the core-shell structured nanoparticles (HA-PLGA@ART/ALA NPs) were constructed by self-assembled method for artemisinin (ART) loading in PLGA cores. In this co-delivery system, ALA and ART can be released in a manner of procedural controlled release. ALA was released from the NPs at first though the pH sensitive hydrazone bond cleavage in order to generate protoporphyrin IX (PpIX) for heme formation. And the increase of heme can effectively improve the curative effect of the subsequent released ART. Furthermore, this system has also shown obvious sonodynaimc activity which can be used for cancer sonodynamic combination therapy. The in vitro and in vivo anticancer results demonstrate that HA-PLGA@ART/ALA delivery system could provide a prospective comprehensive treatment strategy for cancer therapy.
Collapse
Affiliation(s)
- Lei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Yujie Hu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China; The 7(th) People's Hospital of Zhengzhou, 450006, PR China
| | - Yongwei Hao
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Li Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Cuixia Zheng
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Hongjuan Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Mengya Niu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Yanyan Yin
- College of Basic Medicine, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China.
| | - Yun Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, PR China; Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, 100 Kexue Avenue, Zhengzhou 450001, PR China.
| |
Collapse
|
126
|
Shi NQ, Li Y, Zhang Y, Li ZQ, Qi XR. Deepened cellular/subcellular interface penetration and enhanced antitumor efficacy of cyclic peptidic ligand-decorated accelerating active targeted nanomedicines. Int J Nanomedicine 2018; 13:5537-5559. [PMID: 30271146 PMCID: PMC6154709 DOI: 10.2147/ijn.s172556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Acceleration and improvement of penetration across cell-membrane interfaces of active targeted nanotherapeutics into tumor cells would improve tumor-therapy efficacy by overcoming the issue of poor drug penetration. Cell-penetrating peptides, especially synthetic polyarginine, have shown promise in facilitating cargo delivery. However, it is unknown whether polyarginine can work to overcome the membrane interface in an inserted pattern for cyclic peptide ligand-mediated active targeting drug delivery. Here, we conducted a study to test the hypothesis that tandem-insert nona-arginine (tiR9) can act as an accelerating component for intracellular internalization, enhance cellular penetration, and promote antitumor efficacy of active targeted cyclic asparagine–glycine–arginine (cNGR)-decorated nanoliposomes. Methods Polyarginine was coupled with the polyethylene glycol (PEG) chain and the cNGR moiety, yielding a cNGR–tiR9–PEG2,000–distearoylphosphatidylethanolamine conjugate. Results The accelerating active targeted liposome (Lip) nanocarrier (cNGR-tiR9-Lip–doxorubicin [Dox]) constructed in this study held suitable physiochemical features, such as appropriate particle size of ~150 nm and sustained-release profiles. Subsequently, tiR9 was shown to enhance cellular drug delivery of Dox-loaded active targeted systems (cNGR-Lip-Dox) significantly. Layer-by-layer confocal microscopy indicated that the tandem-insert polyarginine accelerated active targeted system entry into deeper intracellular regions based on observations at marginal and center locations. tiR9 enhanced the penetration depth of cNGR-Lip–coumarin 6 through subcellular membrane barriers and caused its specific accumulation in mitochondria, endoplasmic reticulum, and Golgi apparatus. It was also obvious that cNGR-tiR9-Lip-Dox induced enhanced apoptosis and activated caspase 3/7. Moreover, compared with cNGR-Lip-Dox, cNGR-tiR9-Lip-Dox induced a significantly higher antiproliferative effect and markedly suppressed tumor growth in HT1080-bearing nude mice. Conclusion This active tumor-targeting nanocarrier incorporating a tandem-insert polyarginine (tiR9) as an accelerating motif shows promise as an effective drug-delivery system to accelerate translocation of drugs across tumor-cell/subcellular membrane barriers to achieve improved specific tumor therapy.
Collapse
Affiliation(s)
- Nian-Qiu Shi
- School of Pharmacy, Jilin Medical University, Jilin, Jilin, 132013, China,
| | - Yan Li
- Immunology Department, Laboratory Medical College, Jilin Medical University, Jilin, Jilin, 132013, China
| | - Yong Zhang
- College of Life Science, Jilin University, Changchun, Jilin, 130012, China
| | - Zheng-Qiang Li
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, College of Life Science, Jilin University, Changchun, Jilin, 130012, China,
| | - Xian-Rong Qi
- Department of Pharmaceutics, School of Pharmaceutical Science, Peking University, Beijing, 100191, China
| |
Collapse
|
127
|
van Elk M, van den Dikkenberg JB, Storm G, Hennink WE, Vermonden T, Heger M. Preclinical evaluation of thermosensitive poly(N-(2-hydroxypropyl) methacrylamide mono/dilactate)-grafted liposomes for cancer thermochemotherapy. Int J Pharm 2018; 550:190-199. [PMID: 30130606 DOI: 10.1016/j.ijpharm.2018.08.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 01/15/2023]
Abstract
Thermosensitive liposomes grafted with cholesterol-conjugated poly(N-(2-hydroxypropyl) methacrylamide mono/dilactate) (chol-pHPMAlac) have been developed for heat-induced release of doxorubicin (DOX). These liposomes release DOX completely during mild hyperthermia, but their interaction with blood cells and cancer cells has not been studied. Following intravenous administration, liposomes may interact with plasma proteins and various types of cells (e.g., endothelial cells, platelets, and macrophages), which would reduce their disposition in the tumor stroma. Interaction between liposomes and platelets may further cause platelet activation and thrombosis, which could lead to vascular occlusion and thromboembolic complications. The aim was to investigate DOX release kinetics in the presence of serum, stability, in vitro uptake by and toxicity to cancer cells and somatic cells, and platelet activating potential of the chol-pHPMAlac liposomes. DOX release was determined spectrofluorometrically. Liposome stability was determined in buffer and serum by dynamic light scattering and nanoparticle tracking analysis. Association with/uptake by and toxicity of empty liposomes to AML-12, HepG2 (both hepatocyte-derived cancer cells), RAW 264.7 (macrophages), and HUVEC (endothelial) cells was assayed in vitro. Platelet activation was determined by analysis of P-selectin expression and fibrinogen binding. DOPE:EPC liposomes (diameter = 135 nm) grafted with 5% chol-pHPMAlac (cloud point (CP) = 16 °C; Mn = 8.5 kDa) released less than 10% DOX at 37 °C in 30 min, whereas complete release took place at 47 °C or higher within 10 min. The size of these liposomes remained stable in buffer and serum during 24 h at 37 °C. Fluorescently labeled but DOX-lacking chol-pHPMAlac-liposomes exhibited poor association with/uptake by all cells under investigation, were not cytotoxic, and did not activate platelets in both buffered solution and whole blood. In conclusion, thermosensitive chol-pHPMAlac-grafted liposomes rapidly release DOX during mild hyperthermia. The liposomes are stable in a physiological milieu, are not taken up by cells that are encountered in an in vivo setting, and are non-antagonistic towards platelets. Chol-pHPMAlac-grafted liposomes are therefore good candidates for DOX delivery to tumors and temperature-triggered release in tumor stroma.
Collapse
Affiliation(s)
- Merel van Elk
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Joep B van den Dikkenberg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| | - Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Membrane Biochemistry and Biophysics, Bijvoet Center for Biomolecular Research, Institute of Biomembranes, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
128
|
Guo Q, Chang Z, Khan NU, Miao T, Ju X, Feng H, Zhang L, Sun Z, Li H, Han L. Nanosizing Noncrystalline and Porous Silica Material-Naturally Occurring Opal Shale for Systemic Tumor Targeting Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:25994-26004. [PMID: 30020771 DOI: 10.1021/acsami.8b06275] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Opal shale, as a naturally occurring and noncrystalline silica material with porous structure, has the potential to be a drug delivery carrier. In this study, we obtained opal shale nanoparticles (OS NPs) through the techniques of ultrasonic emulsion and differential centrifugation. The OS NPs exhibited markedly lower cytotoxicity than crystalline mesoporous silica nanoparticles. The highly porous structure and the strong adsorbability endowed OS NPs with the ability of loading and sustained release of doxorubicin (DOX). DOX-loaded OS NPs improved tumor cellular uptake and antiproliferation compared with free drug. Interestingly, OS NPs possessed strong binding with the nuclear envelope, which can be beneficial to the nucleus localization and apoptosis inducing of loaded DOX. We further demonstrated the tumor passive targeting ability, prolonged blood circulation, and enhanced antitumor effect with limited in vivo toxicity. Our results suggest that OS NPs can be applied for tumor targeting drug delivery, which may have a significant influence on the development of silica-based drug delivery system.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhenglong Sun
- Suzhou Institute of Biomedical Engineering and Technology , Chinese Academy of Sciences , Suzhou 215163 , P. R. China
| | - Hui Li
- Suzhou Institute of Biomedical Engineering and Technology , Chinese Academy of Sciences , Suzhou 215163 , P. R. China
| | - Liang Han
- Key Laboratory of Smart Drug Delivery (Fudan University) , Ministry of Education , Shanghai 201203 , P. R. China
| |
Collapse
|
129
|
Lv T, Li Z, Xu L, Zhang Y, Chen H, Gao Y. Chloroquine in combination with aptamer-modified nanocomplexes for tumor vessel normalization and efficient erlotinib/Survivin shRNA co-delivery to overcome drug resistance in EGFR-mutated non-small cell lung cancer. Acta Biomater 2018; 76:257-274. [PMID: 29960010 DOI: 10.1016/j.actbio.2018.06.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/23/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023]
Abstract
Although novel molecular targeted drugs have been recognized as an effective therapy for non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) activating mutations, their efficacy fails to meet the expectation due to the acquired resistance in tumors. Up-regulation of the anti-apoptotic protein Survivin was shown to contribute to the resistance to EGFR tyrosine kinase inhibitors (TKI) in EGFR mutation-positive NSCLC. However, the unorganized tumor blood vessels impeded drug penetration into tumor tissue. The resulting insufficient intracellular drug/gene delivery in drug-resistant cancer cells remarkably weakened the drug efficacy in NSCLC. In this work, a multi-functional drug delivery system AP/ES was developed by using anti-EGFR aptamer (Apt)-modified polyamidoamine to co-deliver erlotinib and Survivin-shRNA. Chloroquine (CQ) was used in combination with AP/ES to normalize tumor vessels for sufficient drug/gene delivery to overcome drug resistance in NSCLC cells. The obtained AP/ES possessed desired physicochemical properties, good biostability, controlled drug release profiles, and strong selectivity to EGFR-mutated NSCLC mediated by Apt. CQ not only enhanced endosomal escape ability of AP/ES for efficient gene transfection to inhibit Survivin, but also showed strong vessel-normalization ability to improve tumor microcirculation, which further promoted drug delivery and enhanced drug efficacy in erlotinib-resistant NSCLC cells. Our innovative gene/drug co-delivery system in combination with CQ showed a promising outcome in fighting against erlotinib resistance both in vitro and in vivo. This work indicates that normalization of tumor vessels could help intracellular erlotinib/Survivin-shRNA delivery and the down-regulation of Survivin could act synergistically with erlotinib for reversal of erlotinib resistance in EGFR mutation-positive NSCLC. STATEMENT OF SIGNIFICANCE NSCLC patients who benefited from EGFR-TKIs inevitably developed acquired resistance. Previous research focused on synthesis of new generation of molecular targeted drugs that could irreversibly inhibit EGFR with a particular gene mutation to overcome drug resistance. However, they failed to inhibit EGFR with other gene mutations. Activation of bypass signaling pathway and the changes of tumor microenvironment are identified as two of the mechanisms of acquired resistance to EGFR-TKIs. We therefore constructed multifunctional gene/drug co-delivery nanocomplexes AP/ES co-formulated with chloroquine that could target the both two mechanisms. We found that chloroquine not only enhanced endosomal escape ability of AP/ES for efficient gene transfection to inhibit Survivin, but also showed strong vessel-normalization ability to improve tumor microcirculation, which further promoted drug delivery into tumor tissue and enhanced drug efficacy in erlotinib-resistant NSCLC.
Collapse
|
130
|
He H, Jiang S, Xie Y, Lu Y, Qi J, Dong X, Zhao W, Yin Z, Wu W. Reassessment of long circulation via monitoring of integral polymeric nanoparticles justifies a more accurate understanding. NANOSCALE HORIZONS 2018; 3:397-407. [PMID: 32254127 DOI: 10.1039/c8nh00010g] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Monitoring of payloads results in a biased perception of long circulation of nanoparticles. Instead, herein, the long-circulation effect was re-confirmed by monitoring integral nanoparticles, but circulation was not found to be as long as generally perceived. In contrast, disparate pharmacokinetics were obtained by monitoring a model drug, paclitaxel, highlighting the bias of the conventional protocol.
Collapse
Affiliation(s)
- Haisheng He
- Key Laboratory of Smart Drug Delivery of MOE and PLA, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Wei Y, Quan L, Zhou C, Zhan Q. Factors relating to the biodistribution & clearance of nanoparticles & their effects on in vivo application. Nanomedicine (Lond) 2018; 13:1495-1512. [DOI: 10.2217/nnm-2018-0040] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles have promising biomedical applications for drug delivery, tumor imaging and tumor treatment. Pharmacokinetics are important for the in vivo application of nanoparticles. Biodistribution and clearance are largely defined as the key points of pharmacokinetics to maximize therapeutic efficacy and to minimize side effects. Different engineered nanoparticles have different biodistribution and clearance processes. The interactions of organs with nanoparticles, which are determined by the characteristics of the organs and the biochemical/physical properties of the nanoparticles, are a major factor influencing biodistribution and clearance. In this review, the clearance functions of organs and the properties related to pharmacokinetics, including nanoparticle size, shape, biodegradation and surface modifications are discussed.
Collapse
Affiliation(s)
- Yanchun Wei
- Provincial Key Laboratory for Interventional Medical Devices, Huaiyin Institute of Technology, Huai'an, Jiangsu 223001, PR China
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
| | - Li Quan
- Provincial Key Laboratory for Interventional Medical Devices, Huaiyin Institute of Technology, Huai'an, Jiangsu 223001, PR China
| | - Chao Zhou
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
| | - Qiuqiang Zhan
- Centre for Optical & Electromagnetic Research, Guangdong Provincial Key Laboratory of Optical Information Materials & Technology, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou 510006, PR China
- Key Laboratory of Optoelectronic Devices & Systems of Ministry of Education & Guangdong Province, Shenzhen University, Shenzhen 518052, PR China
| |
Collapse
|
132
|
Enzyme-triggered size shrink and laser-enhanced NO release nanoparticles for deep tumor penetration and combination therapy. Biomaterials 2018; 168:64-75. [DOI: 10.1016/j.biomaterials.2018.03.046] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/21/2018] [Accepted: 03/27/2018] [Indexed: 11/23/2022]
|
133
|
Rodallec A, Fanciullino R, Lacarelle B, Ciccolini J. Seek and destroy: improving PK/PD profiles of anticancer agents with nanoparticles. Expert Rev Clin Pharmacol 2018; 11:599-610. [PMID: 29768060 DOI: 10.1080/17512433.2018.1477586] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The Pharmacokinetics/pharmacodynamics (PK/PD) relationships with cytotoxics are usually based on a steepening concentration-effect relationship; the greater the drug amount, the greater the effect. The Maximum Tolerated Dose paradigm, finding the balance between efficacy, while keeping toxicities at their manageable level, has been the rule of thumb for the last 50-years. Developing nanodrugs is an appealing strategy to help broaden this therapeutic window. The fact that efficacy and toxicity with cytotoxics are intricately linked is primarily due to the complete lack of specificity toward the tumor tissue during their distribution phase. Because nanoparticles are expected to better target tumor tissue while sparing healthy cells, accumulating large amounts of cytotoxics in tumors could be achieved in a safer way. Areas covered: This review aims at presenting how nanodrugs present unique features leading to reconsidering PK/PD relationships of anticancer agents. Expert commentary: The constant interplay between carrier PK, interactions with cancer cells, payload release, payload PK, target expression and target engagement, makes picturing the exact PK/PD relationships of nanodrugs particularly challenging. However, those improved PK/PD relationships now make the once contradictory higher efficacy and lower toxicities requirement an achievable goal in cancer patients.
Collapse
Affiliation(s)
- Anne Rodallec
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Raphaelle Fanciullino
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Bruno Lacarelle
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| | - Joseph Ciccolini
- a SMARTc Unit, Pharmacokinetics Laboratory, Inserm UMR U1068 Centre de Recherche en Cancérologie de Marseille , Aix-Marseille Universite , Marseille , France
| |
Collapse
|
134
|
Skupin-Mrugalska P, Sobotta L, Warowicka A, Wereszczynska B, Zalewski T, Gierlich P, Jarek M, Nowaczyk G, Kempka M, Gapinski J, Jurga S, Mielcarek J. Theranostic liposomes as a bimodal carrier for magnetic resonance imaging contrast agent and photosensitizer. J Inorg Biochem 2018; 180:1-14. [DOI: 10.1016/j.jinorgbio.2017.11.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/03/2017] [Accepted: 11/27/2017] [Indexed: 01/17/2023]
|
135
|
Ghorbani M, Hamishehkar H. Decoration of gold nanoparticles with thiolated pH-responsive polymeric (PEG-b-p(2-dimethylamio ethyl methacrylate-co-itaconic acid) shell: A novel platform for targeting of anticancer agent. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 81:561-570. [DOI: 10.1016/j.msec.2017.08.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/24/2017] [Accepted: 08/02/2017] [Indexed: 10/19/2022]
|
136
|
Kang K, Ma J, Yi Q, Gu Z. Localized drug release and effective chemotherapy by hyperthermia-governed bubble-generating hybrid nanocapsule system. Nanomedicine (Lond) 2017; 12:2763-2783. [DOI: 10.2217/nnm-2017-0265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: To build up a remote triggering drug delivery system with hyperthermia-responsive ammonium bicarbonate salt and investigate its effects on tumor therapy. Materials & methods: This hybrid nanocapsule system was prepared by a different strategy, doxorubicin (DOX) was encapsulated in the heparin shell first and then ammonium bicarbonate was diffused into the nanocapsules to generate DOX-bicarbonate salt, its characterizations and effects on tumor therapy were investigated. Results: Upon exposure to mild external thermal treatment (42°C), DOX-bicarbonate salt began to decompose with the recovery of DOX fluorescence, carbon dioxide generation and rapid DOX release out of the nanocapsules, exhibiting great abilities to accumulate at tumor site rapidly and inhibit tumor cell growth. Conclusion: These hybrid nanocapsules demonstrate great potential in clinical applications triggering by external thermal treatment.
Collapse
Affiliation(s)
- Ke Kang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Jin Ma
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Zhongwei Gu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
- College of Materials Science & Engineering, Nanjing Tech University, 30 South Puzhu Road, Nanjing 211816, China
| |
Collapse
|
137
|
Pedrosa P, Heuer-Jungemann A, Kanaras AG, Fernandes AR, Baptista PV. Potentiating angiogenesis arrest in vivo via laser irradiation of peptide functionalised gold nanoparticles. J Nanobiotechnology 2017; 15:85. [PMID: 29162137 PMCID: PMC5697398 DOI: 10.1186/s12951-017-0321-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/14/2017] [Indexed: 02/21/2023] Open
Abstract
BACKGROUND Anti-angiogenic therapy has great potential for cancer therapy with several FDA approved formulations but there are considerable side effects upon the normal blood vessels that decrease the potential application of such therapeutics. Chicken chorioallantoic membrane (CAM) has been used as a model to study angiogenesis in vivo. Using a CAM model, it had been previously shown that spherical gold nanoparticles functionalised with an anti-angiogenic peptide can humper neo-angiogenesis. RESULTS Our results show that gold nanoparticles conjugated with an anti-angiogenic peptide can be combined with visible laser irradiation to enhance angiogenesis arrest in vivo. We show that a green laser coupled to gold nanoparticles can achieve high localized temperatures able to precisely cauterize blood vessels. This combined therapy acts via VEGFR pathway inhibition, leading to a fourfold reduction in FLT-1 expression. CONCLUSIONS The proposed phototherapy extends the use of visible lasers in clinics, combining it with chemotherapy to potentiate cancer treatment. This approach allows the reduction of dose of anti-angiogenic peptide, thus reducing possible side effects, while destroying blood vessels supply critical for tumour progression.
Collapse
Affiliation(s)
- Pedro Pedrosa
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Amelie Heuer-Jungemann
- Institute for Life Sciences, Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, SO17 1BJ, UK
| | - Antonios G Kanaras
- Institute for Life Sciences, Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, SO17 1BJ, UK
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal.
| |
Collapse
|
138
|
Xiao W, Ruan S, Yu W, Wang R, Hu C, Liu R, Gao H. Normalizing Tumor Vessels To Increase the Enzyme-Induced Retention and Targeting of Gold Nanoparticle for Breast Cancer Imaging and Treatment. Mol Pharm 2017; 14:3489-3498. [PMID: 28845990 DOI: 10.1021/acs.molpharmaceut.7b00475] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Wei Xiao
- Key Laboratory of Drug Targeting
and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shaobo Ruan
- Key Laboratory of Drug Targeting
and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Wenqi Yu
- Key Laboratory of Drug Targeting
and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ranran Wang
- Key Laboratory of Drug Targeting
and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chuan Hu
- Key Laboratory of Drug Targeting
and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rui Liu
- Key Laboratory of Drug Targeting
and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Huile Gao
- Key Laboratory of Drug Targeting
and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
139
|
Kim MW, Jeong HY, Kang SJ, Choi MJ, You YM, Im CS, Lee TS, Song IH, Lee CG, Rhee KJ, Lee YK, Park YS. Cancer-targeted Nucleic Acid Delivery and Quantum Dot Imaging Using EGF Receptor Aptamer-conjugated Lipid Nanoparticles. Sci Rep 2017; 7:9474. [PMID: 28842588 PMCID: PMC5573382 DOI: 10.1038/s41598-017-09555-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 07/18/2017] [Indexed: 12/11/2022] Open
Abstract
Co-application of fluorescent quantum dot nanocrystals and therapeutics has recently become a promising theranostic methodology for cancer treatment. We developed a tumor-targeted lipid nanocarrier that demonstrates notable efficacy in gene delivery as well as tumor bio-imaging. Coupling of aptamer molecules against the EGF receptor (EGFR) to the distal termini of lipid nanoparticles provided the carrier with tumor-specific recognition capability. The cationic lipid component, referred to as O,O’-dimyristyl-N-lysyl glutamate (DMKE), was able to effectively complex with anionic small-interfering RNA (siRNA). The hydrophobic quantum dots (Q-dots) were effectively incorporated in hydrophobic lipid bilayers at an appropriate Q-dot to lipid ratio. In this study, we optimized the liposomal formula of aptamer-conjugated liposomes containing Q-dots and siRNA molecules (Apt-QLs). The anti-EGFR Apt-QLs exhibited remarkable EGFR-dependent siRNA delivery as well as fluorescence imaging, which were analyzed in cultured cancer cells and tumor xenografts in mice. These results imply that the formulation of Apt-QLs could be widely utilized as a carrier for tumor-directed gene delivery and bio-imaging.
Collapse
Affiliation(s)
- Min Woo Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Hwa Yeon Jeong
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Seong Jae Kang
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Moon Jung Choi
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Young Myoung You
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Chan Su Im
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Tae Sup Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - In Ho Song
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Chang Gun Lee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea
| | - Yeon Kyung Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea.
| |
Collapse
|
140
|
Abstract
Chemoradiotherapy (CRT) plays a key role in the curative treatment of many human cancers. The full utility of this paradigm is often restricted by limitations of conventional drug delivery. Nanotherapeutics have demonstrated great potential to overcome many of these issues. The potential benefits of nanotherapeutics in CRT include improved therapeutic efficacy, decreased toxicity, enhanced real-time in vivo tumor imaging, and the translation of novel small molecule drugs or nucleic acid therapies. Additionally, nanomedicines and radiation therapy exert distinct and complementary effects on the tumor microenvironment which can further enhance therapeutic efficacy compared with either modality alone. In this review, we highlight specific clinical and preclinical examples which demonstrate the potential benefits of nanotherapeutics in CRT.
Collapse
|
141
|
Huang S, Li C, Wang W, Li H, Sun Z, Song C, Li B, Duan S, Hu Y. A54 peptide-mediated functionalized gold nanocages for targeted delivery of DOX as a combinational photothermal-chemotherapy for liver cancer. Int J Nanomedicine 2017; 12:5163-5176. [PMID: 28790823 PMCID: PMC5529379 DOI: 10.2147/ijn.s131089] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The combination of photothermal therapy and chemotherapy (photothermal–chemotherapy) is a promising strategy for cancer therapy. Gold nanocages (AuNCs), with hollow and porous structures and unique optical properties, have become a rising star in the field of drug delivery. Here, we designed a novel targeted drug delivery system based on functionalized AuNCs and evaluated their therapeutic effects in vitro and in vivo. We then loaded doxorubicin into this promising system, designated as DHTPAuNCs consisting of hyaluronic acid-grafted and A54 peptide-targeted PEGylated AuNCs. Its formation was corroborated by ultraviolet–visible spectroscopy, transmission electron microscopy and dynamic light scattering. This delivery platform needed hyaluronidase to release encapsulated drugs, meanwhile the acidic pH and near-infrared irradiation could accelerate the release. In addition, the results of cellular uptake demonstrate that this system could bind specifically with BEL-7402 cells. In vitro, we evaluated therapeutic effects of the DHTPAuNCs in BEL-7402 cells by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-tetrazolium bromide assay. Moreover, in BEL-7402 tumor-bearing nude mice, its therapy effect in vivo was also evaluated. As expected, DHTPAuNCs exhibited excellent therapeutic effect by photothermal–chemotherapy, both in vitro and in vivo. In short, DHTPAuNCs with low toxicity showed great potential as a drug delivery system for cancer therapy.
Collapse
Affiliation(s)
- Shengnan Huang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Chunming Li
- Department of Pharmacy, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing, People's Republic of China
| | - Weiping Wang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Huanjie Li
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Zhi Sun
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Chengzhi Song
- School of Physical Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Benyi Li
- Department of Urology and Cancer Center, the University of Kansas Medical Center, Kansas City, KS, USA
| | - Shaofeng Duan
- College of Pharmacy, Henan University, Kaifeng, People's Republic of China.,Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yurong Hu
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China.,Key Laboratory of Key Technology of Drug Preparation, Ministry of Education, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, People's Republic of China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, People's Republic of China
| |
Collapse
|
142
|
Zhu Z, Li Y, Yang X, Pan W, Pan H. The reversion of anti-cancer drug antagonism of tamoxifen and docetaxel by the hyaluronic acid-decorated polymeric nanoparticles. Pharmacol Res 2017; 126:84-96. [PMID: 28734999 DOI: 10.1016/j.phrs.2017.07.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/24/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022]
Abstract
Docetaxel (DTX) and tamoxifen (TMX) are first-line drugs used to treat breast cancer. However when used in combination, they produce antagonism because of their differential metabolic pathways. In order to prevent this antagonism, an amphiphilic copolymer, cholesterol modified hyaruronic acid (HA-CHOL), was synthesized for investigating the co-delivery of TMX and DTX. In vitro drug release experiment of the Co-encapsulated (encapsulated DTX+TMX) nanoparticles (Co-NPs) revealed that NPs with unique release mechanism can markedly reduce the release of these drugs in the circulatory system. However, when reaching in cell, TMX can release rapidly to prevent DTX from coming into contact with metabolizing enzymes. In vitro cytotoxicity experiment revealed that the Co-NPs exhibited a significant synergistic effect for inhibiting the proliferation of the cancer cell lines A549, MCF7 and S180. NPs carrying Coumarin-6(Cou6) exhibited increased cellular uptake compared with Cou6 solution at similar drug concentrations. As an in vivo treatment of xenograft tumors involving 180 cells, the Co-NPs displayed a clear tumor-inhibiting effect. This led us to conclude that the reversion of drug antagonism by NPs was attributed to the increased stability of the nanoparticles in the blood circulation, the efficient cellular uptake, the hierarchical drug metabolism in the tumor and the good and orderly delivery of the drugs to the tumor tissue.
Collapse
Affiliation(s)
- Zhihong Zhu
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Yuenan Li
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Xinggang Yang
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Weisan Pan
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China.
| | - Hao Pan
- College of Pharmacy, Liaoning University, Shenyang, 110036, China, China.
| |
Collapse
|
143
|
Dai W, Wang X, Song G, Liu T, He B, Zhang H, Wang X, Zhang Q. Combination antitumor therapy with targeted dual-nanomedicines. Adv Drug Deliv Rev 2017; 115:23-45. [PMID: 28285944 DOI: 10.1016/j.addr.2017.03.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 01/01/2023]
Abstract
Combination therapy is one of the important treatment strategies for cancer at present. However, the outcome of current combination therapy based on the co-administration of conventional dosage forms is suboptimal, due to the short half-lives of chemodrugs, their deficient tumor selectivity and so forth. Nanotechnology-based targeted delivery systems show great promise in addressing the associated problems and providing superior therapeutic benefits. In this review, we focus on the combination of therapeutic strategies between different nanomedicines or drug-loaded nanocarriers, rather than the co-delivery of different drugs via a single nanocarrier. We introduce the general concept of various targeting strategies of nanomedicines, present the principles of combination antitumor therapy with dual-nanomedicines, analyze their advantages and limitations compared with co-delivery strategies, and overview the recent advances of combination therapy based on targeted nanomedicines. Finally, we reviewed the challenges and future perspectives regarding the selection of therapeutic agents, targeting efficiency and the gap between the preclinical and clinical outcome.
Collapse
Affiliation(s)
- Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaoyou Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Ge Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Tongzhou Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China.
| |
Collapse
|
144
|
Alibolandi M, Taghdisi SM, Ramezani P, Hosseini Shamili F, Farzad SA, Abnous K, Ramezani M. Smart AS1411-aptamer conjugated pegylated PAMAM dendrimer for the superior delivery of camptothecin to colon adenocarcinoma in vitro and in vivo. Int J Pharm 2017; 519:352-364. [PMID: 28126548 DOI: 10.1016/j.ijpharm.2017.01.044] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/17/2017] [Accepted: 01/21/2017] [Indexed: 12/17/2022]
Abstract
In the current study camptothecin-loaded pegylated PAMAM dendrimer were synthesized and were functionalized with AS1411 anti-nucleolin aptamers for site-specific targeting against colorectal cancer cells which over expresses nucleolin receptors. The morphological properties and size dispersity of the prepared nanoparticles were evaluated using transmission electron microscope (TEM) and DLS. The drug-loading content and encapsulation efficiency were obtained 8.1% and 93.67% respectively. The in vitro release of camptothecin from the formulation was provided the sustained release of encapsulated camptothecin during 4days. Comparative in vitro cytotoxicity experiments demonstrated that the targeted camptothecin loaded-pegylated dendrimers had higher antiproliferation activity, towards nucleolin-positive HT29 and C26 colorectal cancer cells than nucleolin-negative CHO cell line. Fluorscence microscopy and flow cytometry also confirmed the enhanced cellular uptake of AS1411 targeted pegylated-dendrimer. In vivo study in C26 tumor-bearing BALB/C mice revealed that the AS1411-functionalized camptothecin loaded pegylated dendrimers improved antitumor activity and survival rate of the encapsulated camptothecin. Conjugation of AS1411 aptamer to the camptothecin loaded-pegylated dendrimer surface provides site-specific delivery of camptothecin, inhibit C26 tumor growth in vivo and significantly decrease systemic toxicity. These results suggested that the new nucleolin-targeted pegylated PAMAM dendrimer as a delivery system for camptothecin have the potential for the treatment of nucleolin-overexpressed colorectal cancer.
Collapse
Affiliation(s)
- Mona Alibolandi
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouria Ramezani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fazileh Hosseini Shamili
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Amel Farzad
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
145
|
Liu S, Song XL, Wang YH, Wang XM, Xiao Y, Wang X, Cheng L, Li XT. The efficacy of WGA modified daunorubicin anti-resistant liposomes in treatment of drug-resistant MCF-7 breast cancer. J Drug Target 2017; 25:541-553. [PMID: 28277825 DOI: 10.1080/1061186x.2017.1298602] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Breast cancer is the most common malignancy and remains a leading cause of cancer-related deaths in female. Chemotherapy failure of breast cancer is mainly associated with multidrug resistance of cancer cells. PURPOSE The WGA modified daunorubicin anti-resistant liposomes were developed for circumventing the multidrug resistance and eliminating cancer cells. METHODS WGA was modified on liposomal surface for increasing the intracellular uptake. Tetrandrine was inserted into the phospholipid bilayer for reversing cancer drug-resistance, and daunorubicin was encapsulated in liposomal aqueous core as an anticancer agent. Evaluations were performed on MCF-7 cells, MCF-7/ADR cells and xenografts of MCF-7/ADR cells. RESULTS In vitro results showed that WGA modified daunorubicin anti-resistant liposomes exhibited suitable physicochemical properties, significantly increased intracellular uptake in both MCF-7 cells and MCF-7/ADR cells, and circumvented the multidrug resistance via inhibiting P-gp. In vivo results demonstrated that the targeting liposomes showed a long-circulatory effect in blood system, and could remarkably accumulate at the tumor location. The involved action mechanisms for the enhanced anticancer efficacy were activation of pro-apoptotic proteins (Bax and Bok), apoptotic enzymes (caspase 8, caspase 9 and caspase 3). CONCLUSION The established WGA modified daunorubicin anti-resistant liposomes could provide a potential strategy for treating resistant MCF-7 breast cancer.
Collapse
Affiliation(s)
- Shuang Liu
- a School of Pharmacy , Liaoning University of Traditional Chinese Medicine , Dalian , China
| | - Xiao-Li Song
- a School of Pharmacy , Liaoning University of Traditional Chinese Medicine , Dalian , China
| | - Yan-Hong Wang
- a School of Pharmacy , Liaoning University of Traditional Chinese Medicine , Dalian , China
| | - Xiao-Min Wang
- a School of Pharmacy , Liaoning University of Traditional Chinese Medicine , Dalian , China
| | - Yao Xiao
- a School of Pharmacy , Liaoning University of Traditional Chinese Medicine , Dalian , China
| | - Xin Wang
- a School of Pharmacy , Liaoning University of Traditional Chinese Medicine , Dalian , China
| | - Lan Cheng
- a School of Pharmacy , Liaoning University of Traditional Chinese Medicine , Dalian , China
| | - Xue-Tao Li
- a School of Pharmacy , Liaoning University of Traditional Chinese Medicine , Dalian , China
| |
Collapse
|
146
|
Su Y, Hu J, Huang Z, Huang Y, Peng B, Xie N, Liu H. Paclitaxel-loaded star-shaped copolymer nanoparticles for enhanced malignant melanoma chemotherapy against multidrug resistance. Drug Des Devel Ther 2017; 11:659-668. [PMID: 28293102 PMCID: PMC5345981 DOI: 10.2147/dddt.s127328] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Malignant melanoma (MM) is the most dangerous type of skin cancer with annually increasing incidence and death rates. However, chemotherapy for MM is restricted by low topical drug concentration and multidrug resistance. In order to surmount the limitation and to enhance the therapeutic effect on MM, a new nanoformulation of paclitaxel (PTX)-loaded cholic acid (CA)-functionalized star-shaped poly(lactide-co-glycolide) (PLGA)-D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) nanoparticles (NPs) (shortly PTX-loaded CA-PLGA-TPGS NPs) was fabricated by a modified method of nanoprecipitation. The particle size, zeta potential, morphology, drug release profile, drug encapsulation efficiency, and loading content of PTX-loaded NPs were detected. As shown by confocal laser scanning, NPs loaded with coumarin-6 were internalized by human melanoma cell line A875. The cellular uptake efficiency of CA-PLGA-TPGS NPs was higher than those of PLGA NPs and PLGA-TPGS NPs. The antitumor effects of PTX-loaded NPs were evaluated by the MTT assay in vitro and by a xenograft tumor model in vivo, demonstrating that star-shaped PTX-loaded CA-PLGA-TPGS NPs were significantly superior to commercial PTX formulation Taxol®. Such drug delivery nanocarriers are potentially applicable to the improvement of clinical MM therapy.
Collapse
Affiliation(s)
- Yongsheng Su
- Department of Burn and Plastic Surgery, The People’s Hospital of Baoan Shenzhen Affiliated to Southern Medical University
| | - Jian Hu
- Department of Burn and Plastic Surgery, The People’s Hospital of Baoan Shenzhen Affiliated to Southern Medical University
| | - Zhibin Huang
- Department of Burn and Plastic Surgery, The People’s Hospital of Baoan Shenzhen Affiliated to Southern Medical University
| | - Yubin Huang
- Department of Burn and Plastic Surgery, The People’s Hospital of Baoan Shenzhen Affiliated to Southern Medical University
| | - Bingsheng Peng
- Department of Burn and Plastic Surgery, The People’s Hospital of Baoan Shenzhen Affiliated to Southern Medical University
| | - Ni Xie
- Core Laboratory, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, People’s Republic of China
| | - Hui Liu
- Department of Burn and Plastic Surgery, The People’s Hospital of Baoan Shenzhen Affiliated to Southern Medical University
| |
Collapse
|
147
|
Wang Y, Tian Y, Zhu P, Ma Y, He J, Lei J. Self-assembled nanoparticles based on poly(ethylene glycol)–oleanolic acid conjugates for co-delivery of anticancer drugs. RSC Adv 2017. [DOI: 10.1039/c7ra04366j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Oleanolic acid (OA) has shown promising antitumor activity.
Collapse
Affiliation(s)
- Yingsa Wang
- MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy
- Beijing Forestry University
- Beijing
- China
| | - Yajie Tian
- MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy
- Beijing Forestry University
- Beijing
- China
| | - Pengbo Zhu
- MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy
- Beijing Forestry University
- Beijing
- China
| | - Yunyun Ma
- MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy
- Beijing Forestry University
- Beijing
- China
| | - Jing He
- MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy
- Beijing Forestry University
- Beijing
- China
- Beijing Key Laboratory of Lignocellulosic Chemistry
| | - Jiandu Lei
- MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy
- Beijing Forestry University
- Beijing
- China
- Beijing Key Laboratory of Lignocellulosic Chemistry
| |
Collapse
|
148
|
Chen W, Ji S, Qian X, Zhang Y, Li C, Wu W, Wang F, Jiang X. Phenylboronic acid-incorporated elastin-like polypeptide nanoparticle drug delivery systems. Polym Chem 2017. [DOI: 10.1039/c7py00330g] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Packaging hydrophobic drugs into nanoparticles can improve their aqueous solubility, tumor-specific accumulation and therapeutic effect.
Collapse
Affiliation(s)
- Weizhi Chen
- Department of Polymer Science & Engineering
- College of Chemistry & Chemical Engineering
- and Jiangsu Key Laboratory for Nanotechnology
- Nanjing University
- Nanjing
| | - Shilu Ji
- Department of Polymer Science & Engineering
- College of Chemistry & Chemical Engineering
- and Jiangsu Key Laboratory for Nanotechnology
- Nanjing University
- Nanjing
| | - Xiaoping Qian
- Department of Polymer Science & Engineering
- College of Chemistry & Chemical Engineering
- and Jiangsu Key Laboratory for Nanotechnology
- Nanjing University
- Nanjing
| | - Yajun Zhang
- Department of Polymer Science & Engineering
- College of Chemistry & Chemical Engineering
- and Jiangsu Key Laboratory for Nanotechnology
- Nanjing University
- Nanjing
| | - Cheng Li
- Department of Polymer Science & Engineering
- College of Chemistry & Chemical Engineering
- and Jiangsu Key Laboratory for Nanotechnology
- Nanjing University
- Nanjing
| | - Wei Wu
- Department of Polymer Science & Engineering
- College of Chemistry & Chemical Engineering
- and Jiangsu Key Laboratory for Nanotechnology
- Nanjing University
- Nanjing
| | - Fei Wang
- College of Chemical Engineering
- Nanjing Forestry University
- Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals
- Nanjing
- P.R. China
| | - Xiqun Jiang
- Department of Polymer Science & Engineering
- College of Chemistry & Chemical Engineering
- and Jiangsu Key Laboratory for Nanotechnology
- Nanjing University
- Nanjing
| |
Collapse
|
149
|
Yhee JY, Im J, Nho RS. Advanced Therapeutic Strategies for Chronic Lung Disease Using Nanoparticle-Based Drug Delivery. J Clin Med 2016; 5:jcm5090082. [PMID: 27657144 PMCID: PMC5039485 DOI: 10.3390/jcm5090082] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 09/15/2016] [Accepted: 09/17/2016] [Indexed: 12/12/2022] Open
Abstract
Chronic lung diseases include a variety of obstinate and fatal diseases, including asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), and lung cancers. Pharmacotherapy is important for the treatment of chronic lung diseases, and current progress in nanoparticles offers great potential as an advanced strategy for drug delivery. Based on their biophysical properties, nanoparticles have shown improved pharmacokinetics of therapeutics and controlled drug delivery, gaining great attention. Herein, we will review the nanoparticle-based drug delivery system for the treatment of chronic lung diseases. Various types of nanoparticles will be introduced, and recent innovative efforts to utilize the nanoparticles as novel drug carriers for the effective treatment of chronic lung diseases will also be discussed.
Collapse
Affiliation(s)
- Ji Young Yhee
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jintaek Im
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | | |
Collapse
|