951
|
Saeed M, van Brakel M, Zalba S, Schooten E, Rens JAP, Koning GA, Debets R, Ten Hagen TLM. Targeting melanoma with immunoliposomes coupled to anti-MAGE A1 TCR-like single-chain antibody. Int J Nanomedicine 2016; 11:955-75. [PMID: 27022262 PMCID: PMC4792179 DOI: 10.2147/ijn.s96123] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Therapy of melanoma using T-cells with genetically introduced T-cell receptors (TCRs) directed against a tumor-selective cancer testis antigen (CTA) NY-ESO1 demonstrated clear antitumor responses in patients without side effects. Here, we exploited the concept of TCR-mediated targeting through introduction of single-chain variable fragment (scFv) antibodies that mimic TCRs in binding major histocompatibility complex-restricted CTA. We produced scFv antibodies directed against Melanoma AntiGEn A1 (MAGE A1) presented by human leukocyte antigen A1 (HLA-A1), in short M1/A1, and coupled these TCR-like antibodies to liposomes to achieve specific melanoma targeting. Two anti-M1/A1 antibodies with different ligand-binding affinities were derived from a phage-display library and reformatted into scFvs with an added cysteine at their carboxyl termini. Protein production conditions, ie, bacterial strain, temperature, time, and compartments, were optimized, and following production, scFv proteins were purified by immobilized metal ion affinity chromatography. Batches of pure scFvs were validated for specific binding to M1/A1-positive B-cells by flow cytometry. Coupling of scFvs to liposomes was conducted by employing different conditions, and an optimized procedure was achieved. In vitro experiments with immunoliposomes demonstrated binding of M1/A1-positive B-cells as well as M1/A1-positive melanoma cells and internalization by these cells using flow cytometry and confocal microscopy. Notably, the scFv with nonenhanced affinity of M1/A1, but not the one with enhanced affinity, was exclusively bound to and internalized by melanoma tumor cells expressing M1/A1. Taken together, antigen-mediated targeting of tumor cells as well as promoting internalization of nanoparticles by these tumor cells is mediated by TCR-like scFv and can contribute to melanoma-specific targeting.
Collapse
Affiliation(s)
- Mesha Saeed
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Mandy van Brakel
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Sara Zalba
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Erik Schooten
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Joost A P Rens
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Gerben A Koning
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Timo L M Ten Hagen
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
952
|
Karimi M, Ghasemi A, Sahandi Zangabad P, Rahighi R, Moosavi Basri SM, Mirshekari H, Amiri M, Shafaei Pishabad Z, Aslani A, Bozorgomid M, Ghosh D, Beyzavi A, Vaseghi A, Aref AR, Haghani L, Bahrami S, Hamblin MR. Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem Soc Rev 2016; 45:1457-501. [PMID: 26776487 PMCID: PMC4775468 DOI: 10.1039/c5cs00798d] [Citation(s) in RCA: 912] [Impact Index Per Article: 101.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
New achievements in the realm of nanoscience and innovative techniques of nanomedicine have moved micro/nanoparticles (MNPs) to the point of becoming actually useful for practical applications in the near future. Various differences between the extracellular and intracellular environments of cancerous and normal cells and the particular characteristics of tumors such as physicochemical properties, neovasculature, elasticity, surface electrical charge, and pH have motivated the design and fabrication of inventive "smart" MNPs for stimulus-responsive controlled drug release. These novel MNPs can be tailored to be responsive to pH variations, redox potential, enzymatic activation, thermal gradients, magnetic fields, light, and ultrasound (US), or can even be responsive to dual or multi-combinations of different stimuli. This unparalleled capability has increased their importance as site-specific controlled drug delivery systems (DDSs) and has encouraged their rapid development in recent years. An in-depth understanding of the underlying mechanisms of these DDS approaches is expected to further contribute to this groundbreaking field of nanomedicine. Smart nanocarriers in the form of MNPs that can be triggered by internal or external stimulus are summarized and discussed in the present review, including pH-sensitive peptides and polymers, redox-responsive micelles and nanogels, thermo- or magnetic-responsive nanoparticles (NPs), mechanical- or electrical-responsive MNPs, light or ultrasound-sensitive particles, and multi-responsive MNPs including dual stimuli-sensitive nanosheets of graphene. This review highlights the recent advances of smart MNPs categorized according to their activation stimulus (physical, chemical, or biological) and looks forward to future pharmaceutical applications.
Collapse
Affiliation(s)
- Mahdi Karimi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Ghasemi
- Department of Materials Science and Engineering, Sharif University of Technology, 11365-9466, Tehran, Iran
| | - Parham Sahandi Zangabad
- Department of Materials Science and Engineering, Sharif University of Technology, 11365-9466, Tehran, Iran
| | - Reza Rahighi
- Department of Research and Development, Sharif Ultrahigh Nanotechnologists (SUN) Company, P.O. Box: 13488-96394, Tehran, Iran and Nanotechnology Research Center, Research Institute of Petroleum Industry (RIPI), West Entrance Blvd., Olympic Village, P.O. Box: 14857-33111, Tehran, Iran
| | - S Masoud Moosavi Basri
- Bioenvironmental Research Center, Sharif University of Technology, Tehran, Iran and Civil & Environmental Engineering Department, Shahid Beheshti University, Tehran, Iran
| | - H Mirshekari
- Department of Biotechnology, University of Kerala, Trivandrum, India
| | - M Amiri
- Department of Materials Science and Engineering, Sharif University of Technology, 11365-9466, Tehran, Iran
| | - Z Shafaei Pishabad
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - A Aslani
- Department of Materials Science and Engineering, Sharif University of Technology, 11365-9466, Tehran, Iran
| | - M Bozorgomid
- Department of Applied Chemistry, Central Branch of Islamic Azad University of Tehran, Tehran, Iran
| | - D Ghosh
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, Tehran, Iran
| | - A Beyzavi
- School of Mechanical Engineering, Boston University, Boston, MA, USA
| | - A Vaseghi
- Department of Biotechnology, Faculty of Advanced Science and Technologies of Isfahan, Isfahan, Iran
| | - A R Aref
- Department of Cancer Biology, Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Department of Genetics, Harvard Medical School, Boston, MA 02215, USA
| | - L Haghani
- School of Medicine, International Campus of Tehran University of Medical Science, Tehran, Iran
| | - S Bahrami
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. and Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| |
Collapse
|
953
|
Affiliation(s)
- Huachao Chen
- State Key Laboratory of Coordination Chemistry, Coordination Chemistry Institute, School of Chemistry and Chemical Engineering, Nanjing University
| | - Danyang Liu
- State Key Laboratory of Coordination Chemistry, Coordination Chemistry Institute, School of Chemistry and Chemical Engineering, Nanjing University
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, Coordination Chemistry Institute, School of Chemistry and Chemical Engineering, Nanjing University
| |
Collapse
|
954
|
Al-Ahmady Z, Kostarelos K. Chemical Components for the Design of Temperature-Responsive Vesicles as Cancer Therapeutics. Chem Rev 2016; 116:3883-918. [DOI: 10.1021/acs.chemrev.5b00578] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zahraa Al-Ahmady
- Nanomedicine Lab, Faculty of Medical & Human Sciences, University of Manchester, AV Hill Building, Manchester M13 9PT, United Kingdom
- UCL
School of Pharmacy, Faculty of Life Science, University College London, Brunswick Square, London WC1N 1AX, United Kingdom
- Manchester
Pharmacy School, University of Manchester, Stopford Building, Manchester M13 9PT, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine Lab, Faculty of Medical & Human Sciences, University of Manchester, AV Hill Building, Manchester M13 9PT, United Kingdom
- UCL
School of Pharmacy, Faculty of Life Science, University College London, Brunswick Square, London WC1N 1AX, United Kingdom
| |
Collapse
|
955
|
Sigg SJ, Postupalenko V, Duskey JT, Palivan CG, Meier W. Stimuli-Responsive Codelivery of Oligonucleotides and Drugs by Self-Assembled Peptide Nanoparticles. Biomacromolecules 2016; 17:935-45. [PMID: 26871486 DOI: 10.1021/acs.biomac.5b01614] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ever more emerging combined treatments exploiting synergistic effects of drug combinations demand smart, responsive codelivery carriers to reveal their full potential. In this study, a multifunctional stimuli-responsive amphiphilic peptide was designed and synthesized to self-assemble into nanoparticles capable of co-bearing and -releasing hydrophobic drugs and antisense oligonucleotides for combined therapies. The rational design was based on a hydrophobic l-tryptophan-d-leucine repeating unit derived from a truncated sequence of gramicidin A (gT), to entrap hydrophobic cargo, which is combined with a hydrophilic moiety of histidines to provide electrostatic affinity to nucleotides. Stimuli-responsiveness was implemented by linking the hydrophobic and hydrophilic sequence through an artificial amino acid bearing a disulfide functional group (H3SSgT). Stimuli-responsive peptides self-assembled in spherical nanoparticles in sizes (100-200 nm) generally considered as preferable for drug delivery applications. Responsive peptide nanoparticles revealed notable nucleotide condensing abilities while maintaining the ability to load hydrophobic cargo. The disulfide cleavage site introduced in the peptide sequence induced responsiveness to physiological concentrations of reducing agent, serving to release the incorporated molecules. Furthermore, the peptide nanoparticles, singly loaded or coloaded with boron-dipyrromethene (BODIPY) and/or antisense oligonucleotides, were efficiently taken up by cells. Such amphiphilic peptides that led to noncytotoxic, reduction-responsive nanoparticles capable of codelivering hydrophobic and nucleic acid payloads simultaneously provide potential toward combined treatment strategies to exploit synergistic effects.
Collapse
Affiliation(s)
- Severin J Sigg
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Viktoriia Postupalenko
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Jason T Duskey
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Wolfgang Meier
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| |
Collapse
|
956
|
Hu JJ, Liu LH, Li ZY, Zhuo RX, Zhang XZ. MMP-responsive theranostic nanoplatform based on mesoporous silica nanoparticles for tumor imaging and targeted drug delivery. J Mater Chem B 2016; 4:1932-1940. [PMID: 32263070 DOI: 10.1039/c5tb02490k] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this paper, we report on an intelligent mesoporous silica-based multifunctional theranostic nanoplatform (designated as MMTNP) for tumor imaging as well as controlled drug release. This theranostic nanoplatform consists of MCM-41 typical mesoporous silica nanoparticles (MSNs) as a hydrophobic drug carrier, matrix metalloprotease-2 (MMP-2) activated fluorescence imaging peptides on the surface of MSNs served as diagnostic probes as well as enzyme-responsive nanovalves blocking the pores, and cRGD peptides further functionalized on the surface of MSNs for tumor targeting. In the absence of MMP-2 conditions, the proximity between the fluorescent dye 5(6)-carboxytetramethylrhodamine hydrochloride (TAMRA) and the quencher 4,4-dimethylamino-azobenzene-4'-carboxylic acid (Dabcyl) on the surface of MSNs resulted in no fluorescence. When the drug loaded nanoplatform arrived at tumor tissue with overexpressed MMP-2, the fluorescence of TAMRA became recovered efficiently due to the hydrolysis of the MMP-2 sensitive peptide substrate, realizing tumor imaging and triggering drug release. In addition, the further introduced cRGD peptide significantly enhanced the targeting efficiency through receptor-mediated endocytosis in tumor cells.
Collapse
Affiliation(s)
- Jing-Jing Hu
- Key Laboratory of Biomedical Polymers of the Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | | | | | | | | |
Collapse
|
957
|
Hersey JS, LaManna CM, Lusic H, Grinstaff MW. Stimuli responsive charge-switchable lipids: Capture and release of nucleic acids. Chem Phys Lipids 2016; 196:52-60. [PMID: 26896839 DOI: 10.1016/j.chemphyslip.2016.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 01/12/2023]
Abstract
Stimuli responsive lipids, which enable control over the formation, transformation, and disruption of supramolecular assemblies, are of interest for biosensing, diagnostics, drug delivery, and basic transmembrane protein studies. In particular, spatiotemporal control over a supramolecular structure can be achieved using light activated compounds to induce significant supramolecular rearrangements. As such, a family of cationic lipids are described which undergo a permanent switch in charge upon exposure to 365 nm ultraviolet (UV) light to enable the capture of negatively charged nucleic acids within the self-assembled supramolecular structure of the lipids and subsequent release of these macromolecules upon exposure to UV light and disruption of the assemblies. The lipids are composed of either two different tripeptide head groups, Lysine-Glycine-Glycine (KGG) and Glycine-Glycine-Glycine (GGG) and three different hydrocarbon chain lengths (C6, C10, or C14) terminated by a UV light responsive 1-(2-nitrophenyl)ethanol (NPE) protected carboxylic acid. The photolysis of the NPE protected lipid is measured as a function of time, and the resulting changes in net molecular charge are observed using zeta potential analysis for each head group and chain length combination. A proof of concept study for the capture and release of both linear DNA (calf thymus) and siRNA is presented using an ethidium bromide quenching assay where a balance between binding affinity and supramolecular stability are found to be the key to optimal nucleic acid capture and release.
Collapse
Affiliation(s)
- Joseph S Hersey
- Boston University, Biomedical Engineering Department, Boston, MA 02215, USA
| | - Caroline M LaManna
- Boston University, Biomedical Engineering Department, Boston, MA 02215, USA
| | - Hrvoje Lusic
- Boston University, Chemistry Department, Boston, MA 02215, USA
| | - Mark W Grinstaff
- Boston University, Biomedical Engineering Department, Boston, MA 02215, USA; Boston University, Chemistry Department, Boston, MA 02215, USA.
| |
Collapse
|
958
|
Marianecci C, Di Marzio L, Del Favero E, Cantù L, Brocca P, Rondelli V, Rinaldi F, Dini L, Serra A, Decuzzi P, Celia C, Paolino D, Fresta M, Carafa M. Niosomes as Drug Nanovectors: Multiscale pH-Dependent Structural Response. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:1241-9. [PMID: 26740247 DOI: 10.1021/acs.langmuir.5b04111] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The use of nanocarriers, which respond to different stimuli controlling their physicochemical properties and biological responsivness, shows a growing interest in pharmaceutical science. The stimuli are activated by targeting tissues and biological compartments, e.g., pH modification, temperature, redox condition, enzymatic activity, or can be physically applied, e.g., a magnetic field and ultrasound. pH modification represents the easiest method of passive targeting, which is actually used to accumulate nanocarriers in cells and tissues. The aim of this paper was to physicochemically characterize pH-sensitive niosomes using different experimental conditions and demonstrate the effect of surfactant composition on the supramolecular structure of niosomes. In this attempt, niosomes, made from commercial (Tween21) and synthetic surfactants (Tween20 derivatives), were physicochemically characterized by using different techniques, e.g., transmission electron microscopy, Raman spectroscopy, and small-angle X-ray scattering. The changes of niosome structure at different pHs depend on surfactants, which can affect the supramolecular structure of colloidal nanocarriers and their potential use both in vitro and in vivo. At pH 7.4, the shape and structure of niosomes have been maintained; however, niosomes show some differences in terms of bilayer thicknesses, water penetration, membrane coupling, and cholesterol dispersion. The acid pH (5.5) can increase the bilayer fluidity, and affect the cholesterol depletion. In fact, Tween21 niosomes form large vesicles with lower curvature radius at acid pH; while Tween20-derivative niosomes increase the intrachain mobility within a more interchain correlated membrane. These results demonstrate that the use of multiple physicochemical procedures provides more information about supramolecular structures of niosomes and improves the opportunity to deeply investigate the effect of stimuli responsiveness on the niosome structure.
Collapse
Affiliation(s)
- Carlotta Marianecci
- Department of Drug Chemistry and Technology, University of Rome "Sapienza" , 00185 Rome, Italy
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti - Pescara "G d'Annunzio" , 66100 Chieti - Pescara, Italy
| | - Elena Del Favero
- Department of Medical Biotechnologies and Traslational Medicine, University of Milan , LITA, 20122 Milan, Italy
| | - Laura Cantù
- Department of Medical Biotechnologies and Traslational Medicine, University of Milan , LITA, 20122 Milan, Italy
| | - Paola Brocca
- Department of Medical Biotechnologies and Traslational Medicine, University of Milan , LITA, 20122 Milan, Italy
| | - Valeria Rondelli
- Department of Medical Biotechnologies and Traslational Medicine, University of Milan , LITA, 20122 Milan, Italy
| | - Federica Rinaldi
- Center for Life Nano Science@Sapienza, Fondazione Istituto Italiano di Tecnologia , 00197 Rome, Italy
| | - Luciana Dini
- Department of Biological and Environmental Sciences and Technologies, University of Salento , 73100 Lecce, Italy
| | - Antonio Serra
- Department of Physics Applied to Materials Science Laboratory (PAMS-Lab), University of Salento , 73100 Lecce, Italy
| | - Paolo Decuzzi
- Department of Translational Imaging, Houston Methodist Research Institute , Houston, Texas 77030, United States
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , 16163 Genoa, Italy
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia" , 88100 Catanzaro, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G d'Annunzio" , 66100 Chieti - Pescara, Italy
- Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas 77030, United States
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia" , 88100 Catanzaro, Italy
- IRC FSH-Interregional Research Center for Food Safety & Health, University of Catanzaro "Magna Græcia" , 88100 Catanzaro, Italy
| | - Massimo Fresta
- IRC FSH-Interregional Research Center for Food Safety & Health, University of Catanzaro "Magna Græcia" , 88100 Catanzaro, Italy
- Department of Health Sciences, University of Catanzaro "Magna Græcia" , 88100 Catanzaro, Italy
| | - Maria Carafa
- Department of Drug Chemistry and Technology, University of Rome "Sapienza" , 00185 Rome, Italy
| |
Collapse
|
959
|
Sabhachandani P, Motwani V, Cohen N, Sarkar S, Torchilin V, Konry T. Generation and functional assessment of 3D multicellular spheroids in droplet based microfluidics platform. LAB ON A CHIP 2016; 16:497-505. [PMID: 26686985 PMCID: PMC4834071 DOI: 10.1039/c5lc01139f] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Here we describe a robust, microfluidic technique to generate and analyze 3D tumor spheroids, which resembles tumor microenvironment and can be used as a more effective preclinical drug testing and screening model. Monodisperse cell-laden alginate droplets were generated in polydimethylsiloxane (PDMS) microfluidic devices that combine T-junction droplet generation and external gelation for spheroid formation. The proposed approach has the capability to incorporate multiple cell types. For the purposes of our study, we generated spheroids with breast cancer cell lines (MCF-7 drug sensitive and resistant) and co-culture spheroids of MCF-7 together with a fibroblast cell line (HS-5). The device has the capability to house 1000 spheroids on chip for drug screening and other functional analysis. Cellular viability of spheroids in the array part of the device was maintained for two weeks by continuous perfusion of complete media into the device. The functional performance of our 3D tumor models and a dose dependent response of standard chemotherapeutic drug, doxorubicin (Dox) and standard drug combination Dox and paclitaxel (PCT) was analyzed on our chip-based platform. Altogether, our work provides a simple and novel, in vitro platform to generate, image and analyze uniform, 3D monodisperse alginate hydrogel tumors for various omic studies and therapeutic efficiency screening, an important translational step before in vivo studies.
Collapse
Affiliation(s)
- P Sabhachandani
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway, Boston, MA 02115, USA.
| | - V Motwani
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway, Boston, MA 02115, USA.
| | - N Cohen
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway, Boston, MA 02115, USA.
| | - S Sarkar
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway, Boston, MA 02115, USA.
| | - V Torchilin
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway, Boston, MA 02115, USA. and Center for Pharmaceutical Biotechnology & Nanomedicine, Northeastern University, 360 Huntington Avenue, 140 The Fenway, Boston, MA 02115, USA
| | - T Konry
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, 140 The Fenway, Boston, MA 02115, USA.
| |
Collapse
|
960
|
Ewert KK, Kotamraju VR, Majzoub RN, Steffes VM, Wonder EA, Teesalu T, Ruoslahti E, Safinya CR. Synthesis of linear and cyclic peptide-PEG-lipids for stabilization and targeting of cationic liposome-DNA complexes. Bioorg Med Chem Lett 2016; 26:1618-1623. [PMID: 26874401 DOI: 10.1016/j.bmcl.2016.01.079] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 01/27/2016] [Accepted: 01/29/2016] [Indexed: 11/29/2022]
Abstract
Because nucleic acids (NAs) have immense potential value as therapeutics, the development of safe and effective synthetic NA vectors continues to attract much attention. In vivo applications of NA vectors require stabilized, nanometer-scale particles, but the commonly used approaches of steric stabilization with a polymer coat (e.g., PEGylation; PEG=poly(ethylene glycol)) interfere with attachment to cells, uptake, and endosomal escape. Conjugation of peptides to PEG-lipids can improve cell attachment and uptake for cationic liposome-DNA (CL-DNA) complexes. We present several synthetic approaches to peptide-PEG-lipids and discuss their merits and drawbacks. A lipid-PEG-amine building block served as the common key intermediate in all synthetic routes. Assembling the entire peptide-PEG-lipid by manual solid phase peptide synthesis (employing a lipid-PEG-carboxylic acid) allowed gram-scale synthesis but is mostly applicable to linear peptides connected via their N-terminus. Conjugation via thiol-maleimide or strain-promoted (copper-free) azide-alkyne cycloaddition chemistry is highly amenable to on-demand preparation of peptide-PEG-lipids, and the appropriate PEG-lipid precursors are available in a single chemical step from the lipid-PEG-amine building block. Azide-alkyne cycloaddition is especially suitable for disulfide-bridged peptides such as iRGD (cyclic CRGDKGPDC). Added at 10 mol% of a cationic/neutral lipid mixture, the peptide-PEG-lipids stabilize the size of CL-DNA complexes. They also affect cell attachment and uptake of nanoparticles in a peptide-dependent manner, thereby providing a platform for preparing stabilized, affinity-targeted CL-DNA nanoparticles.
Collapse
Affiliation(s)
- Kai K Ewert
- Department of Materials, University of California, Santa Barbara, CA 93106, United States; Department of Physics, University of California, Santa Barbara, CA 93106, United States; Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106, United States.
| | - Venkata Ramana Kotamraju
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States
| | - Ramsey N Majzoub
- Department of Materials, University of California, Santa Barbara, CA 93106, United States; Department of Physics, University of California, Santa Barbara, CA 93106, United States; Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106, United States
| | - Victoria M Steffes
- Department of Materials, University of California, Santa Barbara, CA 93106, United States; Department of Physics, University of California, Santa Barbara, CA 93106, United States; Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106, United States; Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, United States
| | - Emily A Wonder
- Department of Materials, University of California, Santa Barbara, CA 93106, United States; Department of Physics, University of California, Santa Barbara, CA 93106, United States; Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106, United States
| | - Tambet Teesalu
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States; Center for Nanomedicine and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106, United States; Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Erkki Ruoslahti
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States; Center for Nanomedicine and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106, United States
| | - Cyrus R Safinya
- Department of Materials, University of California, Santa Barbara, CA 93106, United States; Department of Physics, University of California, Santa Barbara, CA 93106, United States; Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106, United States.
| |
Collapse
|
961
|
Gener P, Rafael DFDS, Fernández Y, Ortega JS, Arango D, Abasolo I, Videira M, Schwartz S. Cancer stem cells and personalized cancer nanomedicine. Nanomedicine (Lond) 2016; 11:307-20. [DOI: 10.2217/nnm.15.200] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite the progress in cancer treatment over the past years advanced cancer is still an incurable disease. Special attention is pointed toward cancer stem cell (CSC)-targeted therapies, because this minor cell population is responsible for the treatment resistance, metastatic growth and tumor recurrence. The recently described CSC dynamic phenotype and interconversion model of cancer growth hamper even more the possible success of current cancer treatments in advanced cancer stages. Accordingly, CSCs can be generated through dedifferentiation processes from non-CSCs, in particular, when CSC populations are depleted after treatment. In this context, the use of targeted CSC nanomedicines should be considered as a promising tool to increase CSC sensitivity and efficacy of specific anti-CSC therapies.
Collapse
Affiliation(s)
- Petra Gener
- Drug Delivery & Targeting Group; CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Diana Fernandes de Sousa Rafael
- Drug Delivery & Targeting Group; CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- iMed.ULisboa, Research Institute for Medicines. Faculdade de Farmácia da Universidade de Lisboa, Av Prof Gama Pinto, 1649–003 Lisboa, Portugal
| | - Yolanda Fernández
- Drug Delivery & Targeting Group; CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- Functional Validation & Preclinical Studies (FVPR); CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Joan Sayós Ortega
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- Inmunobiology Group; CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Diego Arango
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- Molecular Oncology Group; CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Ibane Abasolo
- Drug Delivery & Targeting Group; CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- Functional Validation & Preclinical Studies (FVPR); CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Mafalda Videira
- iMed.ULisboa, Research Institute for Medicines. Faculdade de Farmácia da Universidade de Lisboa, Av Prof Gama Pinto, 1649–003 Lisboa, Portugal
| | - Simo Schwartz
- Drug Delivery & Targeting Group; CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| |
Collapse
|
962
|
Arosio D, Casagrande C. Advancement in integrin facilitated drug delivery. Adv Drug Deliv Rev 2016; 97:111-43. [PMID: 26686830 DOI: 10.1016/j.addr.2015.12.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/27/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023]
Abstract
The research of integrin-targeted anticancer agents has recorded important advancements in ingenious design of delivery systems, based either on the prodrug approach, or on nanoparticle carriers, but for now, none of these has reached a clinical stage of development. Past work in this area has been extensively reviewed by us and others. Thus, the purpose and scope of the present review is to survey the advancement reported in the last 3years, with focus on innovative delivery systems that appear to afford openings for future developments. These systems exploit the labelling with conventional and novel integrin ligands for targeting the interface of cancer cells and of endothelial cells involved in cancer angiogenesis, with the proteins of the extracellular matrix, in the circulation, in tissues, and in tumour stroma, as the site of progression and metastatic evolution of the disease. Furthermore, these systems implement the expertise in the development of nanomedicines to the purpose of achieving preferential biodistribution and uptake in cancer tissues, internalisation in cancer cells, and release of the transported drugs at intracellular sites. The assessment of the value of controlling these factors, and their combination, for future developments requires support of biological testing in appropriate mechanistic models, but also imperatively demand confirmation in therapeutically relevant in vivo models for biodistribution, efficacy, and lack of off-target effects. Thus, among many studies, we have tried to point out the results supported by relevant in vivo studies, and we have emphasised in specific sections those addressing the medical needs of drug delivery to brain tumours, as well as the delivery of oligonucleotides modulating gene-dependent pathological mechanism. The latter could constitute the basis of a promising third branch in the therapeutic armamentarium against cancer, in addition to antibody-based agents and to cytotoxic agents.
Collapse
Affiliation(s)
- Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari (ISTM), CNR, Via C. Golgi 19, I-20133 Milan, Italy.
| | - Cesare Casagrande
- Università degli Studi di Milano, Dipartimento di Chimica, Via C. Golgi 19, I-20133 Milan, Italy.
| |
Collapse
|
963
|
Casadó A, Giuffrida MC, Sagristá ML, Castelli F, Pujol M, Alsina MA, Mora M. Langmuir monolayers and Differential Scanning Calorimetry for the study of the interactions between camptothecin drugs and biomembrane models. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:422-33. [DOI: 10.1016/j.bbamem.2015.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/20/2015] [Accepted: 12/03/2015] [Indexed: 10/22/2022]
|
964
|
Kamei N, Bech Nielsen EJ, Nakakubo T, Aoyama Y, Rahbek UL, Pedersen BL, Takeda-Morishita M. Applicability and Limitations of Cell-Penetrating Peptides in Noncovalent Mucosal Drug or Carrier Delivery Systems. J Pharm Sci 2016; 105:747-753. [DOI: 10.1016/j.xphs.2015.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/30/2015] [Accepted: 11/09/2015] [Indexed: 12/15/2022]
|
965
|
Dan Z, Cao H, He X, Zhang Z, Zou L, Zeng L, Xu Y, Yin Q, Xu M, Zhong D, Yu H, Shen Q, Zhang P, Li Y. A pH-Responsive Host-guest Nanosystem Loading Succinobucol Suppresses Lung Metastasis of Breast Cancer. Am J Cancer Res 2016; 6:435-45. [PMID: 26909117 PMCID: PMC4737729 DOI: 10.7150/thno.13896] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/28/2015] [Indexed: 12/19/2022] Open
Abstract
Cancer metastasis is the leading reason for the high mortality of breast cancer. Herein, we report on a pH-responsive host-guest nanosystem of succinobucol (PHN) with pH-stimuli controlled drug release behavior to improve the therapeutic efficacy on lung metastasis of breast cancer. PHN was composed of the host polymer of β-cyclodextrin linked with multiple arms of N,N-diisopropylethylenediamine (βCD-DPA), the guest polymer of adamantyl end-capped methoxy poly(ethylene glycol) (mPEG-Ad), and the active agent of succinobucol. PHN comprises nanometer-sized homogenous spherical particles, and exhibits specific and rapid drug release in response to the intracellular acidic pH-stimuli. Then, the anti-metastatic efficacy of PHN is measured in metastatic 4T1 breast cancer cells, which effectively confirms the superior inhibitory effects on cell migration and invasion activities, VCAM-1 expression and cell-cell binding of RAW 264.7 to 4T1 cells. Moreover, PHN can be specifically delivered to the sites of metastatic nodules in lungs, and result in an obviously improved therapeutic efficacy on lung metastasis of breast cancer. Thereby, the pH-responsive host-guest nanosystem can be a promising drug delivery platform for effective treatment of cancer metastasis.
Collapse
|
966
|
Landesman-Milo D, Peer D. Transforming Nanomedicines From Lab Scale Production to Novel Clinical Modality. Bioconjug Chem 2016; 27:855-62. [PMID: 26734836 DOI: 10.1021/acs.bioconjchem.5b00607] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of nanoparticles as anticancer drug carriers has been studied for over 50 years. These nanoparticles that can carry drugs are now termed "nanomedicines". Since the approval of the first FDA "nanodrug", DOXIL in 1995, tremendous efforts have been made to develop hundreds of nanomedicines based on different materials. The development of drug nanocarriers (NCs) for cancer therapy is especially challenging and requires multidisciplinary approach. Not only is the translation from a lab scale production of the NCs to clinical scale a challenge, but tumor biology and its unique physiology also possess challenges that need to be overcome with cleverer approaches. Yet, with all the efforts made to develop new strategies to deliver drugs (including small molecules and biologics) for cancer therapy, the number of new NCs that are reaching clinical trials is extremely low. Here we discuss the reasons most of the NCs loaded with anticancer drugs are not likely to reach the clinic and emphasize the importance of understanding tumor physiology and heterogeneity, the use of predictive animal models, and the importance of sharing data as key denominators for potential successful translation of NCs from a bench scale into clinical modality for cancer care.
Collapse
Affiliation(s)
- Dalit Landesman-Milo
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv 69978, Israel
| |
Collapse
|
967
|
Vizirianakis IS, Mystridis GA, Avgoustakis K, Fatouros DG, Spanakis M. Enabling personalized cancer medicine decisions: The challenging pharmacological approach of PBPK models for nanomedicine and pharmacogenomics (Review). Oncol Rep 2016; 35:1891-904. [PMID: 26781205 DOI: 10.3892/or.2016.4575] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/27/2015] [Indexed: 11/05/2022] Open
Abstract
The existing tumor heterogeneity and the complexity of cancer cell biology critically demand powerful translational tools with which to support interdisciplinary efforts aiming to advance personalized cancer medicine decisions in drug development and clinical practice. The development of physiologically based pharmacokinetic (PBPK) models to predict the effects of drugs in the body facilitates the clinical translation of genomic knowledge and the implementation of in vivo pharmacology experience with pharmacogenomics. Such a direction unequivocally empowers our capacity to also make personalized drug dosage scheme decisions for drugs, including molecularly targeted agents and innovative nanoformulations, i.e. in establishing pharmacotyping in prescription. In this way, the applicability of PBPK models to guide individualized cancer therapeutic decisions of broad clinical utility in nanomedicine in real-time and in a cost-affordable manner will be discussed. The latter will be presented by emphasizing the need for combined efforts within the scientific borderlines of genomics with nanotechnology to ensure major benefits and productivity for nanomedicine and personalized medicine interventions.
Collapse
Affiliation(s)
- Ioannis S Vizirianakis
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR‑54124, Greece
| | - George A Mystridis
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR‑54124, Greece
| | - Konstantinos Avgoustakis
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Patras, Patras GR-26504, Greece
| | - Dimitrios G Fatouros
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Marios Spanakis
- Computational BioMedicine Laboratory, Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion GR-71110, Crete, Greece
| |
Collapse
|
968
|
Sulheim E, Baghirov H, von Haartman E, Bøe A, Åslund AKO, Mørch Y, Davies CDL. Cellular uptake and intracellular degradation of poly(alkyl cyanoacrylate) nanoparticles. J Nanobiotechnology 2016; 14:1. [PMID: 26743777 PMCID: PMC4705582 DOI: 10.1186/s12951-015-0156-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/29/2015] [Indexed: 11/10/2022] Open
Abstract
Background
Poly(alkyl cyanoacrylate) (PACA) nanoparticles have shown promise as drug carriers both to solid tumors and across the blood–brain barrier. Efficient drug delivery requires both high cellular uptake of the nanoparticles and release of the drug from the nanoparticles. Release of hydrophobic drugs from PACA nanoparticles is primarily governed by nanoparticle degradation, and this process has been poorly studied at the cellular level. Here we use the hydrophobic model drug Nile Red 668 (NR668) to investigate intracellular degradation of PACA nanoparticles by measuring changes in NR668 fluorescence emission and lifetime, as the spectral properties of NR668 depend on the hydrophobicity of the dye environment. We also assess the potential of poly(butyl cyanoacrylate) (PBCA) and poly(octyl cyanoacrylate) (POCA) nanoparticles for intracellular drug delivery in the prostate cancer cell line PC3 and rat brain endothelial cell line RBE4 and the role of endocytosis pathways in PACA nanoparticle uptake in those cell lines. Results Fluorescence lifetime imaging, emission spectra analysis and Förster resonance energy transfer indicated that the intracellular degradation was in line with the degradation found by direct methods such as gas chromatography and scanning electron microscopy, showing that PBCA has a faster degradation rate compared to POCA. The combined P(BCA/OCA) nanoparticles had an intermediate degradation rate. The uptake of POCA and PBCA nanoparticles was much higher in RBE4 than in PC3 cells. Endocytosis inhibition studies showed that both clathrin- and caveolin-mediated endocytosis were involved in PACA nanoparticle uptake, and that the former played a predominant role, particularly in PC3 cells. Conclusions In the present study, we used three different optical techniques to show that within a 24-hour period PBCA nanoparticles degraded significantly inside cells, releasing their payload into the cytosol, while POCA nanoparticles remained intact. This indicates that it is possible to tune the intracellular drug release rate by choosing appropriate monomers from the PACA family or by using hybrid PACA nanoparticles containing different monomers. In addition, we showed that the uptake of PACA nanoparticles depends not only on the monomer material, but also on the cell type, and that different cell lines can use different internalization pathways. Electronic supplementary material The online version of this article (doi:10.1186/s12951-015-0156-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Einar Sulheim
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| | - Habib Baghirov
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| | - Eva von Haartman
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway. .,Pharmaceutical Sciences Laboratory, Faculty of Natural Sciences and Technology, Åbo Akademi University, Turku, Finland.
| | - Andreas Bøe
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| | - Andreas K O Åslund
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| | - Yrr Mørch
- SINTEF Materials and Chemistry, Trondheim, Norway.
| | - Catharina de Lange Davies
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| |
Collapse
|
969
|
Thambi T, Son S, Lee DS, Park JH. Poly(ethylene glycol)-b-poly(lysine) copolymer bearing nitroaromatics for hypoxia-sensitive drug delivery. Acta Biomater 2016; 29:261-270. [PMID: 26472611 DOI: 10.1016/j.actbio.2015.10.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 10/07/2015] [Accepted: 10/09/2015] [Indexed: 12/27/2022]
Abstract
Hypoxia occurs in a variety of pathological conditions including stroke, rheumatoid arthritis, atherosclerosis, and tumors. In this study, an amphiphilic block copolymer, composed of poly(ethylene glycol) as the hydrophilic block and poly(ε-(4-nitro)benzyloxycarbonyl-L-lysine) as the hydrophobic block, was prepared for hypoxia-sensitive drug delivery. Owing to its amphiphilic nature, the block copolymer formed micelles and encapsulated doxorubicin (DOX) in an aqueous condition. The DOX-loaded micelles exhibited rapid intracellular release of DOX under the hypoxic condition, implying high potential as a drug carrier for cancer therapy. STATEMENT OF SIGNIFICANCE Hypoxia occurs in a variety of pathological conditions including stroke, rheumatoid arthritis, atherosclerosis, and tumors. In this study, we developed a novel type of hypoxia-sensitive polymeric micelles (HS-PMs) that can specifically release the drug under the hypoxic conditions. HS-PMs were prepared using poly(ethylene glycol) as the hydrophilic block and poly(ε-(4-nitro)benzyloxycarbonyl-L-lysine) as the hydrophobic block. Owing to its amphiphilic nature, the block copolymer formed micelles and encapsulated doxorubicin (DOX) in an aqueous condition. The DOX-loaded micelles exhibited rapid intracellular release of DOX under the hypoxic condition. Overall, it is evident that the HS-PMs prepared in this study have the potential to effectively deliver hydrophobic drugs into the hypoxic cells involved in various intractable diseases.
Collapse
Affiliation(s)
- Thavasyappan Thambi
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Soyoung Son
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Doo Sung Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon 440-746, Republic of Korea.
| |
Collapse
|
970
|
Abstract
In chemotherapy a fine balance between therapeutic and toxic effects needs to be found for each patient, adapting standard combination protocols each time. Nanotherapeutics has been introduced into clinical practice for treating tumors with the aim of improving the therapeutic outcome of conventional therapies and of alleviating their toxicity and overcoming multidrug resistance. Photodynamic therapy (PDT) is a clinically approved, minimally invasive procedure emerging in cancer treatment. It involves the administration of a photosensitizer (PS) which, under light irradiation and in the presence of molecular oxygen, produces cytotoxic species. Unfortunately, most PSs lack specificity for tumor cells and are poorly soluble in aqueous media, where they can form aggregates with low photoactivity. Nanotechnological approaches in PDT (nanoPDT) can offer a valid option to deliver PSs in the body and to solve at least some of these issues. Currently, polymeric nanoparticles (NPs) are emerging as nanoPDT system because their features (size, surface properties, and release rate) can be readily manipulated by selecting appropriate materials in a vast range of possible candidates commercially available and by synthesizing novel tailor-made materials. Delivery of PSs through NPs offers a great opportunity to overcome PDT drawbacks based on the concept that a nanocarrier can drive therapeutic concentrations of PS to the tumor cells without generating any harmful effect in non-target tissues. Furthermore, carriers for nanoPDT can surmount solubility issues and the tendency of PS to aggregate, which can severely affect photophysical, chemical, and biological properties. Finally, multimodal NPs carrying different drugs/bioactive species with complementary mechanisms of cancer cell killing and incorporating an imaging agent can be developed. In the following, we describe the principles of PDT use in cancer and the pillars of rational design of nanoPDT carriers dictated by tumor and PS features. Then we illustrate the main nanoPDT systems demonstrating potential in preclinical models together with emerging concepts for their advanced design.
Collapse
|
971
|
Spiridonov VV, Panova IG, Topchieva IN, Zakharov AN. Metal-free thermally-responsive pseudohybrid nanoparticles based on 2-hydroxypropyl-β-cyclodextrin. RSC Adv 2016. [DOI: 10.1039/c6ra03702j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Partial decomposition of pseudohybrid nanoparticles based on 2-hydroxypropyl-β-cyclodextrin by heat treatment at 37 °C.
Collapse
Affiliation(s)
- Vasily V. Spiridonov
- Department of Chemistry
- M.V.Lomonosov Moscow State University
- 119991 Moscow
- Russian Federation
| | - Irina G. Panova
- Department of Chemistry
- M.V.Lomonosov Moscow State University
- 119991 Moscow
- Russian Federation
| | - Irina N. Topchieva
- Department of Chemistry
- M.V.Lomonosov Moscow State University
- 119991 Moscow
- Russian Federation
| | - Alexandre N. Zakharov
- Department of Chemistry
- M.V.Lomonosov Moscow State University
- 119991 Moscow
- Russian Federation
- Department of Fundamental Sciences
| |
Collapse
|
972
|
Dai L, Zhang Q, Shen X, Sun Q, Mu C, Gu H, Cai K. A pH-responsive nanocontainer based on hydrazone-bearing hollow silica nanoparticles for targeted tumor therapy. J Mater Chem B 2016; 4:4594-4604. [DOI: 10.1039/c6tb01050d] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
pH-responsive hollow silica nanoparticles blocked with hyaluronic acid molecules for targeted tumor therapy with high efficiency and good biocompatibility.
Collapse
Affiliation(s)
- Liangliang Dai
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Qingfeng Zhang
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Xinkun Shen
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Qiang Sun
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Caiyun Mu
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Hao Gu
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| |
Collapse
|
973
|
Wu X, Zhou L, Su Y, Dong CM. An autoreduction method to prepare plasmonic gold-embedded polypeptide micelles for synergistic chemo-photothermal therapy. J Mater Chem B 2016; 4:2142-2152. [DOI: 10.1039/c6tb00198j] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
An autoreduction method was introduced to prepare plasmonic gold-embedded polypeptide micelles, opening up a new avenue for the development of anticancer nanotherapeutics with synergistic chemo-photothermal effects.
Collapse
Affiliation(s)
- Xingjie Wu
- Department of Polymer Science & Engineering
- School of Chemistry and Chemical Engineering
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Linzhu Zhou
- Department of Polymer Science & Engineering
- School of Chemistry and Chemical Engineering
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Yue Su
- Department of Polymer Science & Engineering
- School of Chemistry and Chemical Engineering
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Chang-Ming Dong
- Department of Polymer Science & Engineering
- School of Chemistry and Chemical Engineering
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| |
Collapse
|
974
|
Muniz-Miranda M, Muniz-Miranda F, Pedone A. Raman and DFT study of methimazole chemisorbed on gold colloidal nanoparticles. Phys Chem Chem Phys 2016; 18:5974-80. [DOI: 10.1039/c5cp07597a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The SERS/DFT study of methimazole chemisorbed on Au nanoparticles paves the way for the use of these nanohybrids in biomedicine.
Collapse
Affiliation(s)
| | - Francesco Muniz-Miranda
- Department of Chemical and Geological Sciences (DSCG)
- University of Modena and Reggio Emilia (UniMORE)
- 41125 Modena
- Italy
| | - Alfonso Pedone
- Department of Chemical and Geological Sciences (DSCG)
- University of Modena and Reggio Emilia (UniMORE)
- 41125 Modena
- Italy
| |
Collapse
|
975
|
Corbo C, Molinaro R, Parodi A, Toledano Furman NE, Salvatore F, Tasciotti E. The impact of nanoparticle protein corona on cytotoxicity, immunotoxicity and target drug delivery. Nanomedicine (Lond) 2016; 11:81-100. [PMID: 26653875 PMCID: PMC4910943 DOI: 10.2217/nnm.15.188] [Citation(s) in RCA: 431] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022] Open
Abstract
In a perfect sequence of events, nanoparticles (NPs) are injected into the bloodstream where they circulate until they reach the target tissue. The ligand on the NP surface recognizes its specific receptor expressed on the target tissue and the drug is released in a controlled manner. However, once injected in a physiological environment, NPs interact with biological components and are surrounded by a protein corona (PC). This can trigger an immune response and affect NP toxicity and targeting capabilities. In this review, we provide a survey of recent findings on the NP-PC interactions and discuss how the PC can be used to modulate both cytotoxicity and the immune response as well as to improve the efficacy of targeted delivery of nanocarriers.
Collapse
Affiliation(s)
- Claudia Corbo
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
- Fondazione SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Roberto Molinaro
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
| | - Alessandro Parodi
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
- Fondazione SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Naama E Toledano Furman
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
| | - Francesco Salvatore
- CEINGE, Advanced Biotechnology s.c.a.r.l., Via G. Salvatore 486, 80145 Naples, Italy
| | - Ennio Tasciotti
- Department of Regenerative Medicine, Houston Methodist Research Institute, 6670 Bertner Avenue, 77030 Houston, TX, USA
| |
Collapse
|
976
|
Adnan NNM, Cheng YY, Ong NMN, Kamaruddin TT, Rozlan E, Schmidt TW, Duong HTT, Boyer C. Effect of gold nanoparticle shapes for phototherapy and drug delivery. Polym Chem 2016. [DOI: 10.1039/c6py00465b] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this study, we compared three different hybrid gold nanoparticle shapes (spherical, rod and star) for photothermal therapy and the delivery of doxorubicin.
Collapse
Affiliation(s)
- Nik N. M. Adnan
- Australian Centre for Nanomedicine
- School of Chemical Engineering
- University of New South Wales
- Sydney
- Australia
| | - Y. Y. Cheng
- School of Chemistry
- University of New South Wales
- Sydney
- Australia
| | - Nur M. N. Ong
- Centre for Advanced Macromolecular Design (CAMD)
- School of Chemical Engineering
- University of New South Wales
- Sydney
- Australia
| | - Tuan T. Kamaruddin
- Centre for Advanced Macromolecular Design (CAMD)
- School of Chemical Engineering
- University of New South Wales
- Sydney
- Australia
| | - Eliza Rozlan
- Centre for Advanced Macromolecular Design (CAMD)
- School of Chemical Engineering
- University of New South Wales
- Sydney
- Australia
| | | | - Hien T. T. Duong
- Australian Centre for Nanomedicine
- School of Chemical Engineering
- University of New South Wales
- Sydney
- Australia
| | - Cyrille Boyer
- Australian Centre for Nanomedicine
- School of Chemical Engineering
- University of New South Wales
- Sydney
- Australia
| |
Collapse
|
977
|
Blas-Garcia A, Baldoví HG, Polo M, Victor VM, Garcia H, Herance JR. Toxicological properties of two fluorescent carbon quantum dots with onion ring morphology and their usefulness as bioimaging agents. RSC Adv 2016. [DOI: 10.1039/c5ra27662d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Two carbon quantum dots can be used as bioimaging agents. To check this usefulness, a complete in vitro toxicological study has been performed in human cells. Nanoparticles did not show toxicity at low concentration after a punctual or continuous exposition in these cells.
Collapse
Affiliation(s)
- Ana Blas-Garcia
- Departamento de Farmacología
- Facultad de Medicina
- Universitat de Valencia
- FISABIO-Hospital Universitario Dr Peset
- Valencia
| | - Herme G. Baldoví
- Instituto Universitario de Tecnología Química CSIC-UPV
- Univ. Politécnica de Valencia
- Valencia
- Spain
| | - Miriam Polo
- Departamento de Farmacología
- Facultad de Medicina
- Universitat de Valencia
- FISABIO-Hospital Universitario Dr Peset
- Valencia
| | - Victor M. Victor
- CIBERehd
- Department of Physiology-University of Valencia
- Valencia
- Spain
- Service of Endocrinology
| | - Hermenegildo Garcia
- Instituto Universitario de Tecnología Química CSIC-UPV
- Univ. Politécnica de Valencia
- Valencia
- Spain
| | - Jose Raul Herance
- Service of Endocrinology
- University Hospital Dr. Peset
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO)
- Spain
- Vall d'Hebron Institut de Recerca (VHIR)
| |
Collapse
|
978
|
Chen Q, Lin W, Wang H, Wang J, Zhang L. PDEAEMA-based pH-sensitive amphiphilic pentablock copolymers for controlled anticancer drug delivery. RSC Adv 2016. [DOI: 10.1039/c6ra10757e] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The synthesis of a series of PDEAEMA-based pH-sensitive amphiphilic pentablock copolymers PEG-b-(PDEAEMA-b-PMMA)2 with different compositions proceeded via the combination of a bromination reaction andARGET ATRP.
Collapse
Affiliation(s)
- Quan Chen
- School of Chemistry and Chemical Engineering
- South China University of Technology
- Guangzhou 510640
- PR China
| | - Wenjing Lin
- School of Chemistry and Chemical Engineering
- South China University of Technology
- Guangzhou 510640
- PR China
| | - Haiying Wang
- School of Bioscience & Bioengineering
- South China University of Technology
- Guangzhou 510640
- PR China
| | - Jufang Wang
- School of Bioscience & Bioengineering
- South China University of Technology
- Guangzhou 510640
- PR China
| | - Lijuan Zhang
- School of Chemistry and Chemical Engineering
- South China University of Technology
- Guangzhou 510640
- PR China
| |
Collapse
|
979
|
Abstract
This review focuses on stimuli-responsive polymersomes for cancer therapy, which can be disintegrated by recognizing the specific environments of cancer (e.g., low pH, bioreductive environment, over-expressed enzymes,etc.).
Collapse
Affiliation(s)
- Thavasyappan Thambi
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon 440-746
- Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon 440-746
- Republic of Korea
| | - Doo Sung Lee
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon 440-746
- Republic of Korea
| |
Collapse
|
980
|
Multifunctional all-in-one drug delivery systems for tumor targeting and sequential release of three different anti-tumor drugs. Biomaterials 2016; 76:399-407. [DOI: 10.1016/j.biomaterials.2015.10.069] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/20/2015] [Accepted: 10/26/2015] [Indexed: 11/20/2022]
|
981
|
Cheng CC, Lee DJ, Liao ZS, Huang JJ. Stimuli-responsive single-chain polymeric nanoparticles towards the development of efficient drug delivery systems. Polym Chem 2016. [DOI: 10.1039/c6py01623e] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Novel dynamic single-chain polymeric nanoparticles not only significantly improve drug transport efficiency in vitro but can also reside stably and facilitate precisely triggered drug-release in tumor-like microenvironments.
Collapse
Affiliation(s)
- Chih-Chia Cheng
- Graduate Institute of Applied Science and Technology
- National Taiwan University of Science and Technology
- Taipei 10607
- Taiwan
| | - Duu-Jong Lee
- Department of Chemical Engineering
- National Taiwan University
- Taipei 10617
- Taiwan
- Department of Chemical Engineering
| | - Zhi-Sheng Liao
- Graduate Institute of Applied Science and Technology
- National Taiwan University of Science and Technology
- Taipei 10607
- Taiwan
| | - Jyun-Jie Huang
- Graduate Institute of Applied Science and Technology
- National Taiwan University of Science and Technology
- Taipei 10607
- Taiwan
| |
Collapse
|
982
|
Korolovych VF, Grishina OA, Inozemtseva OA, Selifonov AV, Bratashov DN, Suchkov SG, Bulavin LA, Glukhova OE, Sukhorukov GB, Gorin DA. Impact of high-frequency ultrasound on nanocomposite microcapsules: in silico and in situ visualization. Phys Chem Chem Phys 2016; 18:2389-97. [DOI: 10.1039/c5cp05465f] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Simulation and direct visualization of high-frequency (1.2 MHz) ultrasound impact on microcapsules with ZnO nanoparticles embedded in the shell.
Collapse
Affiliation(s)
- V. F. Korolovych
- Saratov State University
- 410012 Saratov
- Russia
- Taras Shevchenko National University of Kyiv
- Physics Faculty
| | | | | | | | | | | | - L. A. Bulavin
- Taras Shevchenko National University of Kyiv
- Physics Faculty
- Department of Molecular Physics
- Kyiv
- Ukraine
| | | | - G. B. Sukhorukov
- School of Engineering & Materials Science
- Queen Mary University of London
- London
- UK
| | - D. A. Gorin
- Saratov State University
- 410012 Saratov
- Russia
| |
Collapse
|
983
|
Herrmann IK, Rösslein M. Personalized medicine: the enabling role of nanotechnology. Nanomedicine (Lond) 2016; 11:1-3. [DOI: 10.2217/nnm.15.152] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Inge K Herrmann
- Department Materials Meet Life, Swiss Federal Laboratories for Materials Science & Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Matthias Rösslein
- Department Materials Meet Life, Swiss Federal Laboratories for Materials Science & Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| |
Collapse
|
984
|
Johnsen KB, Moos T. Revisiting nanoparticle technology for blood–brain barrier transport: Unfolding at the endothelial gate improves the fate of transferrin receptor-targeted liposomes. J Control Release 2016; 222:32-46. [DOI: 10.1016/j.jconrel.2015.11.032] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/28/2015] [Accepted: 11/30/2015] [Indexed: 12/25/2022]
|
985
|
Grimaldi N, Andrade F, Segovia N, Ferrer-Tasies L, Sala S, Veciana J, Ventosa N. Lipid-based nanovesicles for nanomedicine. Chem Soc Rev 2016; 45:6520-6545. [DOI: 10.1039/c6cs00409a] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multifunctional lipid-based nanovesicles (L-NVs) prepared by molecular self-assembly of membrane components together with (bio)-active molecules, by means of compressed CO2-media or other non-conventional methods lead to highly homogeneous, tailor-made nanovesicles that are used for advanced nanomedicine. Confocal microscopy image of siRNA transfection using L-NVs, reprinted with permission from de Jonge,et al.,Gene Therapy, 2006,13, 400–411.
Collapse
Affiliation(s)
- N. Grimaldi
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Nanomol Technologies SA
| | - F. Andrade
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| | - N. Segovia
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| | - L. Ferrer-Tasies
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Nanomol Technologies SA
| | - S. Sala
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| | - J. Veciana
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| | - N. Ventosa
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| |
Collapse
|
986
|
Conjugate-SELEX: A High-throughput Screening of Thioaptamer-liposomal Nanoparticle Conjugates for Targeted Intracellular Delivery of Anticancer Drugs. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e382. [DOI: 10.1038/mtna.2016.81] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/17/2016] [Indexed: 01/03/2023]
|
987
|
Yin S, Chang L, Li T, Wang G, Gu X, Li J. Construction of novel pH-sensitive hybrid micelles for enhanced extracellular stability and rapid intracellular drug release. RSC Adv 2016. [DOI: 10.1039/c6ra23050d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Novel pH-sensitive hybrid micelles with high entrapment efficiency were constructed to realize rapid intracellular drug release without premature release.
Collapse
Affiliation(s)
- Shaoping Yin
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Liang Chang
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Tie Li
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Guangji Wang
- Center of Pharmacokinetics
- Key Laboratory of Drug Metabolism and Pharmacokinetics
- China Pharmaceutical University
- Nanjing
- China
| | - Xiaochen Gu
- College of Pharmacy
- University of Manitoba
- Winnipeg
- Canada R3E 0T5
| | - Juan Li
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| |
Collapse
|
988
|
Chu D, Gao J, Wang Z. Neutrophil-Mediated Delivery of Therapeutic Nanoparticles across Blood Vessel Barrier for Treatment of Inflammation and Infection. ACS NANO 2015; 9:11800-11. [PMID: 26516654 PMCID: PMC4699556 DOI: 10.1021/acsnano.5b05583] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Endothelial cells form a monolayer in lumen of blood vessels presenting a great barrier for delivery of therapeutic nanoparticles (NPs) into extravascular tissues where most diseases occur, such as inflammation disorders and infection. Here, we report a strategy for delivering therapeutic NPs across this blood vessel barrier by nanoparticle in situ hitchhiking activated neutrophils. Using intravital microscopy of TNF-α-induced inflammation of mouse cremaster venules and a mouse model of acute lung inflammation, we demonstrated that intravenously (iv) infused NPs made from denatured bovine serum albumin (BSA) were specifically internalized by activated neutrophils, and subsequently, the neutrophils containing NPs migrated across blood vessels into inflammatory tissues. When neutrophils were depleted using anti-Gr-1 in a mouse, the transport of albumin NPs across blood vessel walls was robustly abolished. Furthermore, it was found that albumin nanoparticle internalization did not affect neutrophil mobility and functions. Administration of drug-loaded albumin NPs markedly mitigated the lung inflammation induced by LPS (lipopolysaccharide) or infection by Pseudomonas aeruginosa. These results demonstrate the use of an albumin nanoparticle platform for in situ targeting of activated neutrophils for delivery of therapeutics across the blood vessel barriers into diseased sites. This study demonstrates our ability to hijack neutrophils to deliver nanoparticles to targeted diseased sites.
Collapse
|
989
|
Abstract
INTRODUCTION Paclitaxel and docetaxel were two epoch-making anticancer drugs and have been successfully used in chemotherapy for a variety of cancer types. In the year 2010, a new taxane, cabazitaxel, was approved by FDA for use in combination with prednisone for the treatment of metastatic hormone-refractory prostate cancer. Albumin-bound paclitaxel (nab™-paclitaxel; abraxane) nanodroplet formulation was another notable invention (FDA approval 2005 for refractory, metastatic, or relapsed breast cancer). Abraxane in combination with gemcitabine for the treatment of pancreatic cancer was approved by FDA in 2013. Accordingly, there have been a huge number of patent applications dealing with taxane anticancer agents in the last 5 years. Thus, it is a good time to review the progress in this area and find the next wave for new developments. AREA COVERED This review covers the patent literature from the year 2010 to early 2015 on various aspects of taxane-based chemotherapies and drug developments. EXPERT OPINION Three FDA-approved taxane anticancer drugs will continue to expand their therapeutic applications, especially through drug combinations and new formulations. Inspired by the success of abraxane, new nano-formulations are emerging. Highly potent new-generation taxanes will play a key role in the development of efficacious tumor-targeted drug delivery systems.
Collapse
Affiliation(s)
- Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, U. S. A
- Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY 11794-3400, U. S. A
| | - Brendan Lichtenthal
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, U. S. A
| | - Siyeon Lee
- Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY 11794-3400, U. S. A
| | - Changwei Wang
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, U. S. A
| | - Xin Wang
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, U. S. A
| |
Collapse
|
990
|
Lu S, Bennett WFD, Ding Y, Zhang L, Fan HY, Zhao D, Zheng T, Ouyang PK, Li J, Wu Y, Xu W, Chu D, Yuan Y, Heerklotz H, Karttunen M, Chen P. Design and Characterization of a Multifunctional pH-Triggered Peptide C8 for Selective Anticancer Activity. Adv Healthc Mater 2015; 4:2709-18. [PMID: 26474414 DOI: 10.1002/adhm.201500636] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/15/2015] [Indexed: 11/12/2022]
Abstract
Most drug delivery systems have been developed for efficient delivery to tumor sites via targeting and on-demand strategies, but the carriers rarely execute synergistic therapeutic actions. In this work, C8, a cationic, pH-triggered anticancer peptide, is developed by incorporating histidine-mediated pH-sensitivity, amphipathic helix, and amino acid pairing self-assembly design. We designed C8 to function as a pH-responsive nanostructure whose cytotoxicity can be switched on and off by its self-assembly: Noncytotoxic β-sheet fibers at high pH with neutral histidines, and positively charged monomers with membrane lytic activity at low pH. The selective activity of C8, tested for three different cancer cell lines and two noncancerous cell lines, is shown. Based on liposome leakage assays and multiscale computer simulations, its physical mechanisms of pore-forming action and selectivity are proposed, which originate from differences in the lipid composition of the cellular membrane and changes in hydrogen bonding. C8 is then investigated for its potential as a drug carrier. C8 forms a nanocomplex with ellipticine, a nonselective model anticancer drug. It selectively targets cancer cells in a pH-responsive manner, demonstrating enhanced efficacy and selectivity. This study provides a novel powerful strategy for the design and development of multifunctional self-assembling peptides for therapeutic and drug delivery applications.
Collapse
Affiliation(s)
- Sheng Lu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology; University of Waterloo; 200 University Avenue West Waterloo Ontario N2L 3G1 Canada
| | - W. F. Drew Bennett
- Department of Chemistry and Waterloo Institute for Nanotechnology; University of Waterloo; Waterloo, 200 University Avenue West Ontario N2L 3G1 Canada
| | - Yong Ding
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology; University of Waterloo; 200 University Avenue West Waterloo Ontario N2L 3G1 Canada
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology; University of Waterloo; 200 University Avenue West Waterloo Ontario N2L 3G1 Canada
- College of Biological and Pharmaceutical Engineering; Nanjing University of Technology; 30 Puchu Road South Nanjing Jiangsu 211816 China
| | - Helen Y. Fan
- Leslie Dan Faculty of Pharmacy; University of Toronto; 144 College Street Toronto Ontario M5S 3M2 Canada
| | - Danyang Zhao
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology; University of Waterloo; 200 University Avenue West Waterloo Ontario N2L 3G1 Canada
| | - Tao Zheng
- College of Biological and Pharmaceutical Engineering; Nanjing University of Technology; 30 Puchu Road South Nanjing Jiangsu 211816 China
| | - Ping-Kai Ouyang
- College of Biological and Pharmaceutical Engineering; Nanjing University of Technology; 30 Puchu Road South Nanjing Jiangsu 211816 China
| | - Jason Li
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology; University of Waterloo; 200 University Avenue West Waterloo Ontario N2L 3G1 Canada
| | - Yan Wu
- Department of Pharmacy; Shanghai 9th People's Hospital; Shanghai Jiao Tong University School of Medicine; 227 Chongqing Road South Shanghai 201999 China
| | - Wen Xu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology; University of Waterloo; 200 University Avenue West Waterloo Ontario N2L 3G1 Canada
| | - Dafeng Chu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology; University of Waterloo; 200 University Avenue West Waterloo Ontario N2L 3G1 Canada
| | - Yongfang Yuan
- Department of Pharmacy; Shanghai 9th People's Hospital; Shanghai Jiao Tong University School of Medicine; 227 Chongqing Road South Shanghai 201999 China
| | - Heiko Heerklotz
- Leslie Dan Faculty of Pharmacy; University of Toronto; 144 College Street Toronto Ontario M5S 3M2 Canada
- Institute for Pharmaceutical Sciences; Albert-Ludwigs University of Freiburg; Hermann-Herder Str. 9 79104 Freiburg Germany
| | - Mikko Karttunen
- Department of Chemistry and Waterloo Institute for Nanotechnology; University of Waterloo; Waterloo, 200 University Avenue West Ontario N2L 3G1 Canada
- Department of Mathematics and Computer Science and Institute for Complex Molecular Systems; Eindhoven University of Technology; P.O. Box 513, MetaForum 5600 MB Eindhoven The Netherlands
| | - P. Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology; University of Waterloo; 200 University Avenue West Waterloo Ontario N2L 3G1 Canada
| |
Collapse
|
991
|
Srinivas R, Satterlee A, Wang Y, Zhang Y, Wang Y, Huang L. Theranostic etoposide phosphate/indium nanoparticles for cancer therapy and imaging. NANOSCALE 2015; 7:18542-18551. [PMID: 26489694 PMCID: PMC4670036 DOI: 10.1039/c5nr04509f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Etoposide phosphate (EP), a water-soluble anticancer prodrug, is widely used for treatment of many cancers. After administration it is rapidly converted to etoposide, its parent compound, which exhibits anticancer activity. Difficulty in parenteral administration necessitates the development of a suitable nanoparticle delivery system for EP. Here we have used indium both as a carrier to deliver etoposide phosphate to tumor cells and as a SPECT imaging agent through incorporation of (111)In. Etoposide phosphate was successfully encapsulated together with indium in nanoparticles, and exhibited dose dependent cytotoxicity and induction of apoptosis in cultured H460 cancer cells via G2/M cell cycle arrest. In a mouse xenograft lung cancer model, etoposide phosphate/indium nanoparticles induce tumor cell apoptosis, leading to significant enhancement of tumor growth inhibition compared to the free drug.
Collapse
Affiliation(s)
- Ramishetti Srinivas
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrew Satterlee
- UNC and NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC 27599
| | - Yuhua Wang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuan Zhang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yongjun Wang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC and NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC 27599
| |
Collapse
|
992
|
Tu Y, Peng F, Adawy A, Men Y, Abdelmohsen LKEA, Wilson DA. Mimicking the Cell: Bio-Inspired Functions of Supramolecular Assemblies. Chem Rev 2015; 116:2023-78. [DOI: 10.1021/acs.chemrev.5b00344] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yingfeng Tu
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Fei Peng
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Alaa Adawy
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Yongjun Men
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Loai K. E. A. Abdelmohsen
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Daniela A. Wilson
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| |
Collapse
|
993
|
Stewart MP, Lorenz A, Dahlman J, Sahay G. Challenges in carrier-mediated intracellular delivery: moving beyond endosomal barriers. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:465-78. [PMID: 26542891 DOI: 10.1002/wnan.1377] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/07/2015] [Accepted: 09/15/2015] [Indexed: 01/08/2023]
Abstract
The deployment of molecular to microscale carriers for intracellular delivery has tremendous potential for biology and medicine, especially for in vivo therapies. The field remains limited, however, by a poor understanding of how carriers gain access to the cell interior. In this review, we provide an overview of the different types of carriers, their speculated modes of entry, putative pathways of vesicular transport, and sites of endosomal escape. We compare this alongside pertinent examples from the cell biology of how viruses, bacteria, and their effectors enter cells and escape endosomal confinement. We anticipate insights into the mechanisms of cellular entry and endosomal escape will benefit future research efforts on effective carrier-mediated intracellular delivery. WIREs Nanomed Nanobiotechnol 2016, 8:465-478. doi: 10.1002/wnan.1377 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Martin P Stewart
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna Lorenz
- Department of Pharmaceutical Sciences, OSU/OHSU College of Pharmacy, Portland, OR, USA
| | - James Dahlman
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gaurav Sahay
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Pharmaceutical Sciences, OSU/OHSU College of Pharmacy, Portland, OR, USA
| |
Collapse
|
994
|
Guo X, Wei X, Jing Y, Zhou S. Size Changeable Nanocarriers with Nuclear Targeting for Effectively Overcoming Multidrug Resistance in Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:6450-6456. [PMID: 26401989 DOI: 10.1002/adma.201502865] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 08/10/2015] [Indexed: 06/05/2023]
Abstract
A size changeable polymer micelle system with a dual shell, which increases in size under acidic pH conditions and is altered to smaller micelles, triggered by intracellular glutathione (GSH), is successfully developed. It is capable of direct delivering anticancer drugs to the nucleus of multidrug resistance (MDR) tumor cells for highly effective combating of drug resistant breast cancer.
Collapse
Affiliation(s)
- Xing Guo
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P.R. China
| | - Xiao Wei
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P.R. China
| | - Yuting Jing
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P.R. China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P.R. China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P.R. China
| |
Collapse
|
995
|
Azevedo HS, Pashkuleva I. Biomimetic supramolecular designs for the controlled release of growth factors in bone regeneration. Adv Drug Deliv Rev 2015; 94:63-76. [PMID: 26325686 DOI: 10.1016/j.addr.2015.08.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 08/17/2015] [Accepted: 08/25/2015] [Indexed: 12/13/2022]
Abstract
The extracellular matrix (ECM) of tissues is an assembly of insoluble macromolecules that specifically interact with soluble bioactive molecules and regulate their distribution and availability to cells. Recapitulating this ability has been an important target in controlled growth factor delivery strategies for tissue regeneration and requires the design of multifunctional carriers. This review describes the integration of supramolecular interactions on the design of delivery strategies that encompass self-assembling and engineered affinity components to construct advanced biomimetic carriers for growth factor delivery. Several glycan- and peptide-based self-assemblies reported in the literature are highlighted and commented upon. These examples demonstrate how molecular design and chemistry are successfully employed to create versatile multifunctional molecules which self-assemble/disassemble in a precisely predicted manner, thus controlling compartmentalization, transport and delivery. Finally, we discuss whether recent advances in the design and preparation of supramolecular delivery systems have been sufficient to drive real translation towards a clinical impact.
Collapse
Affiliation(s)
- Helena S Azevedo
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK.
| | - Iva Pashkuleva
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
996
|
Stylianopoulos T, Jain RK. Design considerations for nanotherapeutics in oncology. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2015; 11:1893-907. [PMID: 26282377 PMCID: PMC4628869 DOI: 10.1016/j.nano.2015.07.015] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/20/2015] [Accepted: 07/22/2015] [Indexed: 12/24/2022]
Abstract
Nanotherapeutics have improved the quality of life of cancer patients, primarily by reducing the adverse effects of chemotherapeutic agents, but improvements in overall survival are modest. This is in large part due to the fact that the enhanced permeability and retention effect, which is the basis for the use of nanoparticles in cancer, can be also a barrier to the delivery of nanomedicines. A careful design of nanoparticle formulations can overcome barriers posed by the tumor microenvironment and result in better treatments. In this review, we first discuss strengths and limitations of clinically-approved nanoparticles. Then, we evaluate design parameters that can be modulated to optimize delivery. The benefits of active tumor targeting and drug release rate on intratumoral delivery and treatment efficacy are also discussed. Finally, we suggest specific design strategies that should optimize delivery to most solid tumors and discuss under what conditions active targeting would be beneficial. FROM THE CLINICAL EDITOR Advances in nanotechnology have seen the introduction of new treatment modalities for cancer. The principle of action using nanocarriers for drug delivery is based mostly on the Enhanced Permeability and Retention effect. This phenomenon however, can also be a hindrance. In this article, the authors performed an in-depth review on various nanoparticle platforms in cancer therapeutics. They also suggested options to improve drug delivery, in terms of carrier design.
Collapse
Affiliation(s)
- Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
997
|
Shipunova VO, Nikitin MP, Zelepukin IV, Nikitin PI, Deyev SM, Petrov RV. A comprehensive study of interactions between lectins and glycoproteins for the development of effective theranostic nanoagents. DOKL BIOCHEM BIOPHYS 2015; 464:315-8. [PMID: 26518557 DOI: 10.1134/s1607672915050117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Indexed: 02/03/2023]
Abstract
A comprehensive study of the interactions between lectins and glycoproteins possessing different glycosylation profiles in the composition of nanoparticles was carried out in order to find specifically interacting protein pairs for the creation of novel classes of multifunctional nanoagets that based on protein-assisted selfassembly. We obtained information about specific interactions of certain lectins with selected glycoproteins as well as about the ability of certain monosaccharides to competitively inhibit binding of glycoproteins with lectins. These protein-mediated interactions may be involved in the formulation of self-assembled nanoparticles for therapy and diagnostics of various diseases.
Collapse
Affiliation(s)
- V O Shipunova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, Moscow, 117997, Russia.
- Nizhny Novgorod State University, pr. Gagarina 23, Nizhny Novgorod, 603600, Russia.
| | - M P Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, Moscow, 117997, Russia
- Moscow Institute of Physics and Technology (State University), Institutskii per. 9, Dolgoprudnyi, Moscow oblast, 141700, Russia
- Prokhorov Institute of General Physics, Russian Academy of Sciences, ul. Vavilova 38, Moscow, 119991, Russia
| | - I V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, Moscow, 117997, Russia
- Moscow Institute of Physics and Technology (State University), Institutskii per. 9, Dolgoprudnyi, Moscow oblast, 141700, Russia
| | - P I Nikitin
- Prokhorov Institute of General Physics, Russian Academy of Sciences, ul. Vavilova 38, Moscow, 119991, Russia
- National Research Nuclear University "Moscow Engineering Physics Institute,", Moscow, Russia
| | - S M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, Moscow, 117997, Russia
- Nizhny Novgorod State University, pr. Gagarina 23, Nizhny Novgorod, 603600, Russia
| | - R V Petrov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, Moscow, 117997, Russia
| |
Collapse
|
998
|
Zhang S, Xiao Q, Sherman SE, Muncan A, Ramos Vicente ADM, Wang Z, Hammer DA, Williams D, Chen Y, Pochan DJ, Vértesy S, André S, Klein ML, Gabius HJ, Percec V. Glycodendrimersomes from Sequence-Defined Janus Glycodendrimers Reveal High Activity and Sensor Capacity for the Agglutination by Natural Variants of Human Lectins. J Am Chem Soc 2015; 137:13334-44. [DOI: 10.1021/jacs.5b08844] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Shaodong Zhang
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Qi Xiao
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Samuel E. Sherman
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Adam Muncan
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Andrea D. M. Ramos Vicente
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Zhichun Wang
- Department
of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6391, United States
| | - Daniel A. Hammer
- Department
of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6391, United States
| | - Dewight Williams
- Electron
Microscopy Resource Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6082, United States
| | - Yingchao Chen
- Department of Materials Science & Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Darrin J. Pochan
- Department of Materials Science & Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Sabine Vértesy
- Institute
of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinärstrasse 13, 80539 Munich, Germany
| | - Sabine André
- Institute
of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinärstrasse 13, 80539 Munich, Germany
| | - Michael L. Klein
- Institute
of Computational Molecular Science, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Hans-Joachim Gabius
- Institute
of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinärstrasse 13, 80539 Munich, Germany
| | - Virgil Percec
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| |
Collapse
|
999
|
Chen K, Xu X, Guo J, Zhang X, Han S, Wang R, Li X, Zhang J. Enhanced Intracellular Delivery and Tissue Retention of Nanoparticles by Mussel-Inspired Surface Chemistry. Biomacromolecules 2015; 16:3574-83. [DOI: 10.1021/acs.biomac.5b01056] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | | | | | - Ruibing Wang
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | | | | |
Collapse
|
1000
|
Shi Q, Zhang L, Liu M, Zhang X, Zhang X, Xu X, Chen S, Li X, Zhang J. Reversion of multidrug resistance by a pH-responsive cyclodextrin-derived nanomedicine in drug resistant cancer cells. Biomaterials 2015. [DOI: 10.1016/j.biomaterials.2015.07.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|