3051
|
Ucisik MH, Küpcü S, Schuster B, Sleytr UB. Characterization of CurcuEmulsomes: nanoformulation for enhanced solubility and delivery of curcumin. J Nanobiotechnology 2013; 11:37. [PMID: 24314310 PMCID: PMC4029586 DOI: 10.1186/1477-3155-11-37] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 10/31/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Curcumin is a polyphenolic compound isolated from the rhizomes of the plant Curcuma longa and shows intrinsic anti-cancer properties. Its medical use remains limited due to its extremely low water solubility and bioavailability. Addressing this problem, drug delivery systems accompanied by nanoparticle technology have emerged. The present study introduces a novel nanocarrier system, so-called CurcuEmulsomes, where curcumin is encapsulated inside the solid core of emulsomes. RESULTS CurcuEmulsomes are spherical solid nanoparticles with an average size of 286 nm and a zeta potential of 37 mV. Encapsulation increases the bioavailability of curcumin by up to 10,000 fold corresponding to a concentration of 0.11 mg/mL. Uptaken by HepG2 human liver carcinoma cell line, CurcuEmulsomes show a significantly prolonged biological activity and demonstrated therapeutic efficacy comparable to free curcumin against HepG2 in vitro - with a delay in response, as assessed by cell viability, apoptosis and cell cycle studies. The delay is attributed to the solid character of the nanocarrier prolonging the release of curcumin inside the HepG2 cells. CONCLUSIONS Incorporation of curcumin into emulsomes results in water-soluble and stable CurcuEmulsome nanoformulations. CurcuEmulsomes do not only successfully facilitate the delivery of curcumin into the cell in vitro, but also enable curcumin to reach its effective concentrations inside the cell. The enhanced solubility of curcumin and the promising in vitro efficacy of CurcuEmulsomes highlight the potential of the system for the delivery of lipophilic drugs. Moreover, high degree of compatibility, prolonged release profile and tailoring properties feature CurcuEmulsomes for further therapeutic applications in vivo.
Collapse
Affiliation(s)
- Mehmet H Ucisik
- Department of Nanobiotechnology, Institute for Synthetic Bioarchitectures,
University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse
11, Vienna 1190, Austria
| | - Seta Küpcü
- Department of Nanobiotechnology, Institute for Synthetic Bioarchitectures,
University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse
11, Vienna 1190, Austria
| | - Bernhard Schuster
- Department of Nanobiotechnology, Institute for Synthetic Bioarchitectures,
University of Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse
11, Vienna 1190, Austria
| | - Uwe B Sleytr
- Department of Nanobiotechnology, Institute for Biophysics, University of
Natural Resources and Life Sciences (BOKU) Vienna, Muthgasse 11, Vienna
1190, Austria
| |
Collapse
|
3052
|
Hauert S, Berman S, Nagpal R, Bhatia SN. A computational framework for identifying design guidelines to increase the penetration of targeted nanoparticles into tumors. NANO TODAY 2013; 8:566-576. [PMID: 25009578 PMCID: PMC4084751 DOI: 10.1016/j.nantod.2013.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Targeted nanoparticles are increasingly being engineered for the treatment of cancer. By design, they can passively accumulate in tumors, selectively bind to targets in their environment, and deliver localized treatments. However, the penetration of targeted nanoparticles deep into tissue can be hindered by their slow diffusion and a high binding affinity. As a result, they often localize to areas around the vessels from which they extravasate, never reaching the deep-seeded tumor cells, thereby limiting their efficacy. To increase tissue penetration and cellular accumulation, we propose generalizable guidelines for nanoparticle design and validate them using two different computer models that capture the potency, motion, binding kinetics, and cellular internalization of targeted nanoparticles in a section of tumor tissue. One strategy that emerged from the models was delaying nanoparticle binding until after the nanoparticles have had time to diffuse deep into the tissue. Results show that nanoparticles that are designed according to these guidelines do not require fine-tuning of their kinetics or size and can be administered in lower doses than classical targeted nanoparticles for a desired tissue penetration in a large variety of tumor scenarios. In the future, similar models could serve as a testbed to explore engineered tissue-distributions that arise when large numbers of nanoparticles interact in a tumor environment.
Collapse
Affiliation(s)
- Sabine Hauert
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Spring Berman
- Mechanical and Aerospace Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Radhika Nagpal
- Computer Science, Harvard University, Cambridge, MA 02138, USA
| | - Sangeeta N. Bhatia
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
3053
|
Gentile E, Cilurzo F, Di Marzio L, Carafa M, Anna Ventura C, Wolfram J, Paolino D, Celia C. Liposomal chemotherapeutics. Future Oncol 2013; 9:1849-59. [DOI: 10.2217/fon.13.146] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Currently, six liposomal chemotherapeutics have received clinical approval and many more are in clinical trials or undergoing preclinical evaluation. Liposomes exhibit low toxicity and improve the biopharmaceutical features and therapeutic index of drugs, thereby increasing efficacy and reducing side effects. In this review we discuss the advantages of using liposomes for the delivery of chemotherapeutics. Gemcitabine and paclitaxel have been chosen as examples to illustrate how the performance of a metabolically unstable or poorly water-soluble drug can be greatly improved by liposomal incorporation. We look at the beneficial effects of liposomes in a variety of solid and blood-borne tumors, including thyroid cancer, pancreatic cancer, breast cancer and multiple myeloma.
Collapse
Affiliation(s)
- Emanuela Gentile
- Department of Health Sciences, University ‘Magna Graecia‘ of Catanzaro, University Campus ‘S. Venuta‘, Building of BioSciences, V.le ‘S. Venuta‘ 88100 Germaneto – Catanzaro, Italy
- Department of Nanomedicine, The Methodist Hospital Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Felisa Cilurzo
- Department of Health Sciences, University ‘Magna Graecia‘ of Catanzaro, University Campus ‘S. Venuta‘, Building of BioSciences, V.le ‘S. Venuta‘ 88100 Germaneto – Catanzaro, Italy
| | - Luisa Di Marzio
- Department of Pharmacy, University ‘G. d‘Annunzio‘ of Chieti - Pescara, Via dei Vestini 31, 66013 Chieti, Italy
| | - Maria Carafa
- Department of Drug Chemistry & Technologies, University ‘La Sapienza‘ of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Cinzia Anna Ventura
- Department of Drug Science & Health Products, University of Messina, Viale Annunziata, 98168 Messina, Italy
| | - Joy Wolfram
- Department of Nanomedicine, The Methodist Hospital Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, Beijing 100190, China
| | - Donatella Paolino
- Department of Health Sciences, University ‘Magna Graecia‘ of Catanzaro, University Campus ‘S. Venuta‘, Building of BioSciences, V.le ‘S. Venuta‘ 88100 Germaneto – Catanzaro, Italy
| | - Christian Celia
- Department of Nanomedicine, The Methodist Hospital Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| |
Collapse
|
3054
|
Parise A, Milelli A, Tumiatti V, Minarini A, Neviani P, Zuccari G. Preparation, characterization and in vitro evaluation of sterically stabilized liposome containing a naphthalenediimide derivative as anticancer agent. Drug Deliv 2013; 22:590-7. [PMID: 24286206 DOI: 10.3109/10717544.2013.861042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to incorporate a new naphthalenediimide derivative (AN169) with a promising anticancer activity into pegylated liposomes to an extent that allows its in vitro and in vivo testing without use of toxic solvent. AN169-loaded liposomes were prepared using the thin-film hydration method and characterized for size, polydispersity index, drug content and drug release. We examined their lyophilization ability in the presence of cryoprotectants (trehalose, sucrose and lysine) and the long-term stability of the lyophilized products stored at 4 °C for 3 and 6 months by particle size changes and drug leakage. AN169 was successfully loaded into liposomes with an entrapment efficiency of 87.3 ± 2.5%. The hydrodynamic diameter of these liposomes after sonication was ∼ 145 nm with a high degree of monodispersity. Trehalose was found to be superior to the other lyoprotectants. In particular, trehalose 1:10 lipid:cryoprotectant molar ratio may provide stable lyophilized liposomes with the conservation of physicochemical properties upon freeze-drying and long-term storage conditions. We also assessed their in vitro antitumor activity in human cancer cell lines (HTLA-230 neuroblastoma, Mel 3.0 melanoma, OVCAR-3 ovarian carcinoma and SV620 prostate cancer cells). However, only after 72 h incubation, loaded liposomes showed almost the same IC50 as free AN169. In conclusion, we developed a stable lyophilized liposomal formulation for intravenous administration of AN169 as anticancer drug, with the advantage of avoiding the use of potentially toxic solubilizing agents for future in vivo experiments.
Collapse
Affiliation(s)
- Amelia Parise
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna , Rimini , Italy and
| | | | | | | | | | | |
Collapse
|
3055
|
Mellal D, Zumbuehl A. Exit-strategies - smart ways to release phospholipid vesicle cargo. J Mater Chem B 2013; 2:247-252. [PMID: 32261503 DOI: 10.1039/c3tb21086c] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
This highlight describes recent trends in fundamental phospholipid research towards possible future drug delivery technology. In particular it focuses on synthetic phospholipids and their vesicular constructs and describes selected "smart" ways to release cargo from liposomes. Various chemical and physical release triggers are discussed such as temperature changes, application of ultrasound, enzyme degradation, changes in pH, redox reactions, photochemical reactions, as well as the effects of shear stress on vesicles.
Collapse
Affiliation(s)
- Denia Mellal
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland.
| | | |
Collapse
|
3056
|
Hua S, Wu SY. The use of lipid-based nanocarriers for targeted pain therapies. Front Pharmacol 2013; 4:143. [PMID: 24319430 PMCID: PMC3836271 DOI: 10.3389/fphar.2013.00143] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/04/2013] [Indexed: 12/29/2022] Open
Abstract
Sustained delivery of analgesic agents at target sites remains a critical issue for effective pain management. The use of nanocarriers has been reported to facilitate effective delivery of these agents to target sites while minimizing systemic toxicity. These include the use of biodegradable liposomal or polymeric carriers. Of these, liposomes present as an attractive delivery system due to their flexible physicochemical properties which allow easy manipulation in order to address different delivery considerations. Their favorable toxicity profiles and ease of large scale production also make their clinical use feasible. In this review, we will discuss the concept of using liposomes as a drug delivery carrier, their in vitro characteristics as well as in vivo behavior. Current advances in the targeted liposomal delivery of analgesic agents and their impacts on the field of pain management will be presented.
Collapse
Affiliation(s)
- Susan Hua
- School of Biomedical Sciences and Pharmacy, The University of Newcastle Callaghan, NSW, Australia
| | | |
Collapse
|
3057
|
Yaroslavov AA, Sybachin AV, Zaborova OV, Pergushov DV, Zezin AB, Melik-Nubarov NS, Plamper FA, Müller AHE, Menger FM. Electrostatically driven complexation of liposomes with a star-shaped polyelectrolyte to low-toxicity multi-liposomal assemblies. Macromol Biosci 2013; 14:491-5. [PMID: 24243764 DOI: 10.1002/mabi.201300436] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 10/10/2013] [Indexed: 11/08/2022]
Abstract
Anionic liposomes are electrostatically complexed to a star-shaped cationic polyelectrolyte. Upon complexation, the liposomes retain their integrity and the resulting liposome-star complexes do not dissociate in a physiological solution with 0.15 M NaCl. This provides a multi-liposomal container for possible use as a high-capacity carrier.
Collapse
Affiliation(s)
- Alexander A Yaroslavov
- Lomonosov Moscow State University, Chemistry Department, Leninskie Gory 1-3, 119991, Moscow, Russia
| | | | | | | | | | | | | | | | | |
Collapse
|
3058
|
Hong BJ, Chipre AJ, Nguyen ST. Acid-degradable polymer-caged lipoplex (PCL) platform for siRNA delivery: facile cellular triggered release of siRNA. J Am Chem Soc 2013; 135:17655-8. [PMID: 24000948 DOI: 10.1021/ja404491r] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
An acid-degradable polymer-caged lipoplex (PCL) platform consisting of a cationic lipoplex core and a biocompatible, pH-responsive polymer shell has been developed for the effective delivery of small interfering RNA (siRNA) through a combination of facile loading, rapid acid-triggered release, cellular internalization, and effective endosomal escape. In vitro testing of this degradable PCL delivery platform reveals ∼45- and ∼2.5-fold enhancement of enhanced green fluorescent protein knockdown in cancer cells in comparison to either free siRNA or siRNA-loaded non-acid-degradable lipoplex formulations, respectively.
Collapse
Affiliation(s)
- Bong Jin Hong
- Department of Chemistry, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| | | | | |
Collapse
|
3059
|
Nam J, Ha YS, Hwang S, Lee W, Song J, Yoo J, Kim S. pH-responsive gold nanoparticles-in-liposome hybrid nanostructures for enhanced systemic tumor delivery. NANOSCALE 2013; 5:10175-10178. [PMID: 24057056 DOI: 10.1039/c3nr03698g] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
We report a pH-responsive gold nanoparticles-in-liposome hybrid nanostructure, which effectively combines the pH-responsive assembly and surface plasmon property changes of 'smart' gold nanoparticles and enhanced systemic circulation and tumor accumulation of the PEG-grafted liposomes.
Collapse
Affiliation(s)
- Jutaek Nam
- Department of Chemistry, Pohang University of Science & Technology (POSTECH), San 31, Hyojadong, Namgu, Pohang 790-784, South Korea.
| | | | | | | | | | | | | |
Collapse
|
3060
|
Noble GT, Stefanick JF, Ashley JD, Kiziltepe T, Bilgicer B. Ligand-targeted liposome design: challenges and fundamental considerations. Trends Biotechnol 2013; 32:32-45. [PMID: 24210498 DOI: 10.1016/j.tibtech.2013.09.007] [Citation(s) in RCA: 352] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 12/18/2022]
Abstract
Nanomedicine, particularly liposomal drug delivery, has expanded considerably over the past few decades, and several liposomal drugs are already providing improved clinical outcomes. Liposomes have now progressed beyond simple, inert drug carriers and can be designed to be highly responsive in vivo, with active targeting, increased stealth, and controlled drug-release properties. Ligand-targeted liposomes (LTLs) have the potential to revolutionize the treatment of cancer. However, these highly engineered liposomes generate new problems, such as accelerated clearance from circulation, compromised targeting owing to non-specific serum protein binding, and hindered tumor penetration. This article highlights recent challenges facing LTL strategies and describes the advanced design elements used to circumvent them.
Collapse
Affiliation(s)
- Gavin T Noble
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jared F Stefanick
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jonathan D Ashley
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Tanyel Kiziltepe
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Mike and Josie Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Advanced Diagnostics and Therapeutics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Basar Bilgicer
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Mike and Josie Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Advanced Diagnostics and Therapeutics, University of Notre Dame, Notre Dame, IN 46556, USA; Center for Rare & Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
3061
|
|
3062
|
Markman JL, Rekechenetskiy A, Holler E, Ljubimova JY. Nanomedicine therapeutic approaches to overcome cancer drug resistance. Adv Drug Deliv Rev 2013; 65:1866-79. [PMID: 24120656 PMCID: PMC5812459 DOI: 10.1016/j.addr.2013.09.019] [Citation(s) in RCA: 503] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 09/29/2013] [Accepted: 09/30/2013] [Indexed: 12/27/2022]
Abstract
Nanomedicine is an emerging form of therapy that focuses on alternative drug delivery and improvement of the treatment efficacy while reducing detrimental side effects to normal tissues. Cancer drug resistance is a complicated process that involves multiple mechanisms. Here we discuss the major forms of drug resistance and the new possibilities that nanomedicines offer to overcome these treatment obstacles. Novel nanomedicines that have a high ability for flexible, fast drug design and production based on tumor genetic profiles can be created making drug selection for personal patient treatment much more intensive and effective. This review aims to demonstrate the advantage of the young medical science field, nanomedicine, for overcoming cancer drug resistance. With the advanced design and alternative mechanisms of drug delivery known for different nanodrugs including liposomes, polymer conjugates, micelles, dendrimers, carbon-based, and metallic nanoparticles, overcoming various forms of multi-drug resistance looks promising and opens new horizons for cancer treatment.
Collapse
Affiliation(s)
- Janet L Markman
- Nanomedicine Research Center, Department of Neurosurgery at Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | | | | | | |
Collapse
|
3063
|
Hirai M, Kimura R, Takeuchi K, Hagiwara Y, Kawai-Hirai R, Ohta N, Igarashi N, Shimuzu N. Structure of liposome encapsulating proteins characterized by X-ray scattering and shell-modeling. JOURNAL OF SYNCHROTRON RADIATION 2013; 20:869-74. [PMID: 24121330 PMCID: PMC3795546 DOI: 10.1107/s0909049513020827] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/25/2013] [Indexed: 05/25/2023]
Abstract
Lipid liposomes are promising drug delivery systems because they have superior curative effects owing to their high adaptability to a living body. Lipid liposomes encapsulating proteins were constructed and the structures examined using synchrotron radiation small- and wide-angle X-ray scattering (SR-SWAXS). The liposomes were prepared by a sequential combination of natural swelling, ultrasonic dispersion, freeze-throw, extrusion and spin-filtration. The liposomes were composed of acidic glycosphingolipid (ganglioside), cholesterol and phospholipids. By using shell-modeling methods, the asymmetric bilayer structure of the liposome and the encapsulation efficiency of proteins were determined. As well as other analytical techniques, SR-SWAXS and shell-modeling methods are shown to be a powerful tool for characterizing in situ structures of lipid liposomes as an important candidate of drug delivery systems.
Collapse
Affiliation(s)
- Mitsuhiro Hirai
- Graduate School of Engineering, Gunma University, 4-2 Aramaki, Maebashi, Gunma 371-8510, Japan
| | - Ryota Kimura
- Graduate School of Engineering, Gunma University, 4-2 Aramaki, Maebashi, Gunma 371-8510, Japan
| | - Kazuki Takeuchi
- Graduate School of Engineering, Gunma University, 4-2 Aramaki, Maebashi, Gunma 371-8510, Japan
| | - Yoshihiko Hagiwara
- Graduate School of Engineering, Gunma University, 4-2 Aramaki, Maebashi, Gunma 371-8510, Japan
| | - Rika Kawai-Hirai
- Institute for Molecular and Celluar Regulation, Gunma University, 3-39-15 Shouwa, Maebashi 371-8512, Japan
| | - Noboru Ohta
- JASRI, 1-1-1 Kuoto, Sayo-cho, Sayo-gun, Hyogo 679-5198, Japan
| | | | | |
Collapse
|
3064
|
Bajoria R, Sooranna S, Chatterjee R. Effect of lipid composition of cationic SUV liposomes on materno-fetal transfer of warfarin across the perfused human term placenta. Placenta 2013; 34:1216-22. [PMID: 24183755 DOI: 10.1016/j.placenta.2013.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/03/2013] [Accepted: 10/07/2013] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Use of drugs that cross the placenta freely are currently avoided during pregnancy. We investigated whether cationic small unilamellar (SUV) liposomes of different lipid compositions could prevent the transfer and uptake of warfarin across human term placenta. METHODS Cationic liposomes encapsulated warfarin was prepared by using lecithin (F-SUV) or sterylamine (S-SUV) with cholesterol and stearylamine. The size distribution, encapsulation efficiency, and stability were determined in blood-based media. The transfer kinetics of free and liposomally encapsulated warfarin were studied in a dually perfused isolated lobule of human term placenta with creatinine. Concentrations of warfarin were measured by fluorimetry. Data are expressed as % of initial dose added and given as mean ± sd. RESULTS Warfarin crossed the placenta freely (14.9 ± 1.1%). Trans placental transfer of warfarin was significantly reduced by F-SUV (6.4 ± 0.6%; P < 0.001) and S-SUV liposomes (5.0 ± 0.8%; P < 0.001). Placental uptake of F-SUV (6.3 ± 1.7%; P < 0.001) was greater than that of S-SUV liposomes (2.2 ± 0.5%; P < 0.001). CONCLUSION Our data suggest that cationic liposomes reduce trans placental transfer of warfarin. If confirmed "in vivo", liposomes might provide an alternative non-invasive method of drug delivery to the mother.
Collapse
Affiliation(s)
- R Bajoria
- Imperial College, School of Medicine, Chelsea and Westminster Hospital, London, UK; University College London, Institute for Women's Health London, UK.
| | | | | |
Collapse
|
3065
|
Nag OK, Awasthi V. Surface engineering of liposomes for stealth behavior. Pharmaceutics 2013; 5:542-69. [PMID: 24300562 PMCID: PMC3873679 DOI: 10.3390/pharmaceutics5040542] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/10/2013] [Accepted: 10/14/2013] [Indexed: 02/08/2023] Open
Abstract
Liposomes are used as a delivery vehicle for drug molecules and imaging agents. The major impetus in their biomedical applications comes from the ability to prolong their circulation half-life after administration. Conventional liposomes are easily recognized by the mononuclear phagocyte system and are rapidly cleared from the blood stream. Modification of the liposomal surface with hydrophilic polymers delays the elimination process by endowing them with stealth properties. In recent times, the development of various materials for surface engineering of liposomes and other nanomaterials has made remarkable progress. Poly(ethylene glycol)-linked phospholipids (PEG-PLs) are the best representatives of such materials. Although PEG-PLs have served the formulation scientists amazingly well, closer scrutiny has uncovered a few shortcomings, especially pertaining to immunogenicity and pharmaceutical characteristics (drug loading, targeting, etc.) of PEG. On the other hand, researchers have also begun questioning the biological behavior of the phospholipid portion in PEG-PLs. Consequently, stealth lipopolymers consisting of non-phospholipids and PEG-alternatives are being developed. These novel lipopolymers offer the potential advantages of structural versatility, reduced complement activation, greater stability, flexible handling and storage procedures and low cost. In this article, we review the materials available as alternatives to PEG and PEG-lipopolymers for effective surface modification of liposomes.
Collapse
Affiliation(s)
- Okhil K Nag
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 North Stonewall Avenue, Oklahoma City, OK 73117, USA.
| | | |
Collapse
|
3066
|
Wolfram J, Suri K, Yang Y, Shen J, Celia C, Fresta M, Zhao Y, Shen H, Ferrari M. Shrinkage of pegylated and non-pegylated liposomes in serum. Colloids Surf B Biointerfaces 2013; 114:294-300. [PMID: 24216620 DOI: 10.1016/j.colsurfb.2013.10.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 09/24/2013] [Accepted: 10/08/2013] [Indexed: 12/15/2022]
Abstract
An essential requisite for the design of nanodelivery systems is the ability to characterize the size, homogeneity and zeta potential of nanoparticles. Such properties can be tailored in order to create the most efficient drug delivery platforms. An important question is whether these characteristics change upon systemic injection. Here, we have studied the behavior of phosphatidylcholine/cholesterol liposomes exposed to serum proteins. The results reveal a serum-induced reduction in the size and homogeneity of both pegylated and non-pegylated liposomes, implicating the possible role of osmotic forces. In addition, changes to zeta-potential were observed upon exposing liposomes to serum. The liposomes with polyethylene glycol expressed different characteristics than their non-polymeric counterparts, suggesting the potential formation of a denser protein corona around the non-pegylated liposomes.
Collapse
Affiliation(s)
- Joy Wolfram
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, Beijing 100190, China; Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Krishna Suri
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yong Yang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jianliang Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Christian Celia
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Pharmacy, University G. d'Annunzio of Chieti, Pescara, 66013 Chieti, Italy
| | - Massimo Fresta
- Department of Health Science, University Magna Græcia of Catanzaro, Germaneto 88100, Italy
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, Beijing 100190, China; Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
3067
|
Falsini S, Ciani L, Ristori S, Fortunato A, Arcangeli A. Advances in Lipid-Based Platforms for RNAi Therapeutics. J Med Chem 2013; 57:1138-46. [DOI: 10.1021/jm400791q] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sara Falsini
- Department
of Chemistry “Ugo Schiff” and CSGI, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino (Fi), Italy
- Department
of Clinical and Experimental Medicine, University of Florence, Viale G.
B. Morgagni 50, 50134, Firenze, Italy
| | - Laura Ciani
- Department
of Chemistry “Ugo Schiff” and CSGI, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino (Fi), Italy
| | - Sandra Ristori
- Department
of Chemistry “Ugo Schiff” and CSGI, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino (Fi), Italy
| | - Angelo Fortunato
- Department
of Clinical and Experimental Medicine, University of Florence, Viale G.
B. Morgagni 50, 50134, Firenze, Italy
| | - Annarosa Arcangeli
- Department
of Clinical and Experimental Medicine, University of Florence, Viale G.
B. Morgagni 50, 50134, Firenze, Italy
| |
Collapse
|
3068
|
Alexander A, Ajazuddin, Khan J, Saraf S, Saraf S. Poly(ethylene glycol)-poly(lactic-co-glycolic acid) based thermosensitive injectable hydrogels for biomedical applications. J Control Release 2013; 172:715-29. [PMID: 24144918 DOI: 10.1016/j.jconrel.2013.10.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 01/17/2023]
Abstract
Stimuli triggered polymers provide a variety of applications related with the biomedical fields. Among various stimuli triggered mechanisms, thermoresponsive mechanisms have been extensively investigated, as they are relatively more convenient and effective stimuli for biomedical applications. In a contemporary approach for achieving the sustained action of proteins, peptides and bioactives, injectable depots and implants have always remained the thrust areas of research. In the same series, Poloxamer based thermogelling copolymers have their own limitations regarding biodegradability. Thus, there is a need to have an alternative biomaterial for the formulation of injectable hydrogel, which must remain biocompatible along with safety and efficacy. In the same context, poly(ethylene glycol) (PEG) based copolymers play a crucial role as a biomedical material for biomedical applications, because of their biocompatibility, biodegradability, thermosensitivity and easy controlled characters. This review stresses on the physicochemical property, stability and composition prospects of smart PEG/poly(lactic-co-glycolic acid) (PLGA) based thermoresponsive injectable hydrogels, recently utilized for biomedical applications. The manuscript also highlights the synthesis scheme and stability characteristics of these copolymers, which will surely help the researchers working in the same area. We have also emphasized the applied use of these smart copolymers along with their formulation problems, which could help in understanding the possible modifications related with these, to overcome their inherent associated limitations.
Collapse
Affiliation(s)
- Amit Alexander
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, C.G. 492010, India.
| | | | | | | | | |
Collapse
|
3069
|
Chan KWY, Bulte JWM, McMahon MT. Diamagnetic chemical exchange saturation transfer (diaCEST) liposomes: physicochemical properties and imaging applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2013; 6:111-24. [PMID: 24339357 DOI: 10.1002/wnan.1246] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Chemical exchange saturation transfer (CEST) is a new type of magnetic resonance imaging (MRI) contrast based on labile spins which rapidly exchange with solvent, resulting in an amplification of signal which allows detection of solute protons at millimolar to micromolar concentrations. An additional feature of these agents is that natural organic and biodegradable compounds can provide strong CEST contrast, allowing the development of diamagnetic CEST (diaCEST) MRI contrast agents. The sensitivity of the CEST approach per unit of agent increases further when diaCEST contrast agents are loaded into liposomes to become diaCEST liposomes. In this review, we will discuss the unique and favorable features of diaCEST liposomes which are well suited for in vivo imaging. diaCEST liposomes are nanocarriers which feature high concentrations of encapsulated contrast material, controlled release of payload, and an adjustable coating for passive or active tumor targeting. These liposomes have water permeable bilayers and both the interior and exterior can be fine-tuned for many biomedical applications. Furthermore, a number of liposome formulations are used in the clinic including Doxil™, which is an approved product for treating patients with cancer for decades, rapid translation of these materials can be envisaged. diaCEST liposomes have shown promise in imaging of cancer, and monitoring of chemotherapy and cell transplants. The unique features of diaCEST liposomes are discussed to provide an overview of the applications currently envisioned for this new technology and to provide an overall insight of their potential.
Collapse
Affiliation(s)
- Kannie W Y Chan
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | | | | |
Collapse
|
3070
|
Raemdonck K, Braeckmans K, Demeester J, De Smedt SC. Merging the best of both worlds: hybrid lipid-enveloped matrix nanocomposites in drug delivery. Chem Soc Rev 2013; 43:444-72. [PMID: 24100581 DOI: 10.1039/c3cs60299k] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The advent of nanotechnology has revolutionized drug delivery in terms of improving drug efficacy and safety. Both polymer-based and lipid-based drug-loaded nanocarriers have demonstrated clinical benefit to date. However, to address the multifaceted drug delivery challenges ahead and further expand the spectrum of therapeutic applications, hybrid lipid-polymer nanocomposites have been designed to merge the beneficial features of both polymeric drug delivery systems and liposomes in a single nanocarrier. This review focuses on different classes of nanohybrids characterized by a drug-loaded polymeric matrix core enclosed in a lipid shell. Various nanoengineering approaches to obtain lipid-polymer nanocomposites with a core-shell nanoarchitecture will be discussed as well as their predominant applications in drug delivery.
Collapse
Affiliation(s)
- Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, B-9000 Ghent, Belgium.
| | | | | | | |
Collapse
|
3071
|
Zhang Q, Tang J, Fu L, Ran R, Liu Y, Yuan M, He Q. A pH-responsive α-helical cell penetrating peptide-mediated liposomal delivery system. Biomaterials 2013; 34:7980-93. [DOI: 10.1016/j.biomaterials.2013.07.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 07/03/2013] [Indexed: 12/11/2022]
|
3072
|
van der Meel R, Vehmeijer LJC, Kok RJ, Storm G, van Gaal EVB. Ligand-targeted particulate nanomedicines undergoing clinical evaluation: current status. Adv Drug Deliv Rev 2013; 65:1284-98. [PMID: 24018362 DOI: 10.1016/j.addr.2013.08.012] [Citation(s) in RCA: 284] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 08/09/2013] [Accepted: 08/29/2013] [Indexed: 12/25/2022]
Abstract
Since the introduction of Doxil® on the market nearly 20years ago, a number of nanomedicines have become part of treatment regimens in the clinic. With the exception of antibody-drug conjugates, these nanomedicines are all devoid of targeting ligands and rely solely on their physicochemical properties and the (patho)physiological processes in the body for their biodistribution and targeting capability. At the same time, many preclinical studies have reported on nanomedicines exposing targeting ligands, or ligand-targeted nanomedicines, yet none of these have been approved at this moment. In the present review, we provide a concise overview of 13 ligand-targeted particulate nanomedicines (ligand-targeted PNMs) that have progressed into clinical trials. The progress of each ligand-targeted PNM is discussed based on available (pre)clinical data. Main conclusions of these analyses are that (a) ligand-targeted PNMs have proven to be safe and efficacious in preclinical models; (b) the vast majority of ligand-targeted PNMs is generated for the treatment of cancer; (c) contribution of targeting ligands to the PNM efficacy is not unambiguously proven; and (d) targeting ligands do not cause localization of the PNM within the target tissue, but rather provide benefits in terms of target cell internalization and target tissue retention once the PNM has arrived at the target site. Increased understanding of the in vivo fate and interactions of the ligand-targeted PNMs with proteins and cells in the human body is mandatory to rationally advance the clinical translation of ligand-targeted PNMs. Future perspectives for ligand-targeted PNM approaches include the delivery of drugs that are unable or inefficient in passing cellular membranes, treatment of drug resistant tumors, targeting of the tumor blood supply, the generation of targeted vaccines and nanomedicines that are able to cross the blood-brain barrier.
Collapse
Affiliation(s)
- Roy van der Meel
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
3073
|
Ernsting MJ, Murakami M, Roy A, Li SD. Factors controlling the pharmacokinetics, biodistribution and intratumoral penetration of nanoparticles. J Control Release 2013; 172:782-94. [PMID: 24075927 DOI: 10.1016/j.jconrel.2013.09.013] [Citation(s) in RCA: 685] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/11/2013] [Accepted: 09/15/2013] [Indexed: 11/30/2022]
Abstract
Nanoparticle drug delivery to the tumor is impacted by multiple factors: nanoparticles must evade clearance by renal filtration and the reticuloendothelial system, extravasate through the enlarged endothelial gaps in tumors, penetrate through dense stroma in the tumor microenvironment to reach the tumor cells, remain in the tumor tissue for a prolonged period of time, and finally release the active agent to induce pharmacological effect. The physicochemical properties of nanoparticles such as size, shape, surface charge, surface chemistry (PEGylation, ligand conjugation) and composition affect the pharmacokinetics, biodistribution, intratumoral penetration and tumor bioavailability. On the other hand, tumor biology (blood flow, perfusion, permeability, interstitial fluid pressure and stroma content) and patient characteristics (age, gender, tumor type, tumor location, body composition and prior treatments) also have impact on drug delivery by nanoparticles. It is now believed that both nanoparticles and the tumor microenvironment have to be optimized or adjusted for optimal delivery. This review provides a comprehensive summary of how these nanoparticle and biological factors impact nanoparticle delivery to tumors, with discussion on how the tumor microenvironment can be adjusted and how patients can be stratified by imaging methods to receive the maximal benefit of nanomedicine. Perspectives and future directions are also provided.
Collapse
Affiliation(s)
- Mark J Ernsting
- Drug Delivery and Formulation, Drug Discovery Program, Ontario Institute for Cancer Research, 101 College Street, Suite 800, Toronto, Ontario M5G 0A3, Canada; Ryerson University, Faculty of Architectural Science and Engineering, Toronto, Ontario M5B 1Z2, Canada
| | | | | | | |
Collapse
|
3074
|
Tam YYC, Chen S, Cullis PR. Advances in Lipid Nanoparticles for siRNA Delivery. Pharmaceutics 2013; 5:498-507. [PMID: 24300520 PMCID: PMC3836621 DOI: 10.3390/pharmaceutics5030498] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/04/2013] [Accepted: 09/12/2013] [Indexed: 01/21/2023] Open
Abstract
Technological advances in both siRNA (small interfering RNA) and whole genome sequencing have demonstrated great potential in translating genetic information into siRNA-based drugs to halt the synthesis of most disease-causing proteins. Despite its powerful promises as a drug, siRNA requires a sophisticated delivery vehicle because of its rapid degradation in the circulation, inefficient accumulation in target tissues and inability to cross cell membranes to access the cytoplasm where it functions. Lipid nanoparticle (LNP) containing ionizable amino lipids is the leading delivery technology for siRNA, with five products in clinical trials and more in the pipeline. Here, we focus on the technological advances behind these potent systems for siRNA-mediated gene silencing.
Collapse
Affiliation(s)
- Yuen Yi C Tam
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C. V6T 1Z3, Canada.
| | | | | |
Collapse
|
3075
|
Platzman I, Janiesch JW, Matić J, Spatz JP. Artificial Antigen-Presenting Interfaces in the Service of Immunology. Isr J Chem 2013. [DOI: 10.1002/ijch.201300060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
3076
|
Shankar R, Samykutty A, Riggin C, Kannan S, Wenzel U, Kolhatkar R. Cathepsin B degradable star-shaped peptidic macromolecules for delivery of 2-methoxyestradiol. Mol Pharm 2013; 10:3776-88. [PMID: 23971990 DOI: 10.1021/mp400261h] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
2-Methoxyestradiol (2ME), a natural metabolite of estradiol, has antiproliferative and antiangiogenic activity. However, its clinical success is limited due to poor water solubility and poor pharmacokinetic parameters suggesting the need for a delivery vehicle. In this study we evaluated cathepsin B degradable star-shaped peptidic macromolecules (SPMs) that can potentially be used to create higher generation and high molecular weight peptidic polymer as delivery vehicle of 2ME. Two peptidic macromolecules having positively charged amine (ASPM) or negatively charged carboxyl surface groups (CSPM) were synthesized and evaluated for their degradation in the presence of cathepsin B and stability in the presence of neutral or acidic buffer and serum. Both ASPM and CSPM degraded rapidly in the presence of cathepsin B. Both were stable in neutral and acidic buffer whereas only CSPM exhibited substantial stability in the presence of serum. Both macromolecules were nontoxic toward breast cancer cells whereas 2ME-containing macromolecules exhibited antiproliferative activity in the micromolar range. Overall, results from the current study indicate that tetrapeptide GFLG can be used to create star-shaped macromolecules that are degraded in the presence of cathepsin B and have the potential to be developed as delivery vehicles of 2ME.
Collapse
Affiliation(s)
- Ravi Shankar
- Department of Biopharmaceutical Sciences, University of Illinois Chicago , Rockford, Illinois 61107, United States
| | | | | | | | | | | |
Collapse
|
3077
|
Nanoparticles targeting mechanisms in cancer therapy: current limitations and emerging solutions. Ther Deliv 2013; 4:1197-209. [DOI: 10.4155/tde.13.75] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
It has been more than one century since Paul Ehrlich spoke about the idea of targeting specific molecules in the cell when he coined the ‘Magic Bullet‘ principle. In most occasions, we seek new pharmacodynamic models for therapy, but nanoparticles provide a chance to modify the already existing pharmacokinetics of drugs to meet needed pharmacodynamic models. In the scope of ‘nanoscale‘, every entity has different characters, and no general rules control pharmacokinetics of nanoparticulate drugs as new physical and physicochemical properties are added to equations. However, such remarkable drug models are still quite far from achieving their potential in clinical application. Among the major obstacles is that most available results in nanoparticles targeting rely upon in vitro and animal models that do not match the tumor environment characteristics in humans. This Review discusses the concept of targeting tumor cells with nanoparticles, the limitations that lead to its incomplete application in clinical practice along with some of the promising solutions to such limitations.
Collapse
|
3078
|
Souza JG, Dias K, Pereira TA, Bernardi DS, Lopez RFV. Topical delivery of ocular therapeutics: carrier systems and physical methods. ACTA ACUST UNITED AC 2013; 66:507-30. [PMID: 24635555 DOI: 10.1111/jphp.12132] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 07/23/2013] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The basic concepts, major mechanisms, technological developments and advantages of the topical application of lipid-based systems (microemulsions, nanoemulsions, liposomes and solid lipid nanoparticles), polymeric systems (hydrogels, contact lenses, polymeric nanoparticles and dendrimers) and physical methods (iontophoresis and sonophoresis) will be reviewed. KEY FINDINGS Although very convenient for patients, topical administration of conventional drug formulations for the treatment of eye diseases requires high drug doses, frequent administration and rarely provides high drug bioavailability. Thus, strategies to improve the efficacy of topical treatments have been extensively investigated. In general, the majority of the successful delivery systems are present on the ocular surface over an extended period of time, and these systems typically improve drug bioavailability in the anterior chamber whereas the physical methods facilitate drug penetration over a very short period of time through ocular barriers, such as the cornea and sclera. SUMMARY Although in the early stages, the combination of these delivery systems with physical methods would appear to be a promising tool to decrease the dose and frequency of administration; thereby, patient compliance and treatment efficacy will be improved.
Collapse
Affiliation(s)
- Joel G Souza
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
3079
|
Karjoo Z, McCarthy HO, Patel P, Nouri FS, Hatefi A. Systematic engineering of uniform, highly efficient, targeted and shielded viral-mimetic nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:2774-2783. [PMID: 23468416 PMCID: PMC5222681 DOI: 10.1002/smll.201300077] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/05/2013] [Indexed: 06/01/2023]
Abstract
In the past decades, numerous types of nanomedicines have been developed for the efficient and safe delivery of nucleic acid-based drugs for cancer therapy. Given that the destination sites for nucleic acid-based drugs are inside cancer cells, delivery systems need to be both targeted and shielded in order to overcome the extracellular and intracellular barriers. One of the major obstacles that has hindered the translation of nanotechnology-based gene-delivery systems into the clinic has been the complexity of the design and assembly processes, resulting in non-uniform nanocarriers with unpredictable surface properties and efficiencies. Consequently, no product has reached the clinic yet. In order to address this shortcoming, a multifunctional targeted biopolymer is genetically engineered in one step, eliminating the need for multiple chemical conjugations. Then, by systematic modulation of the ratios of the targeted recombinant vector to PEGylated peptides of different sizes, a library of targeted-shielded viral-mimetic nanoparticles (VMNs) with diverse surface properties are assembled. Through the use of physicochemical and biological assays, targeted-shielded VMNs with remarkably high transfection efficiencies (>95%) are screened. In addition, the batch-to-batch variability of the assembled targeted-shielded VMNs in terms of uniformity and efficiency is examined and, in both cases, the coefficient of variation is calculated to be below 20%, indicating a highly reproducible and uniform system. These results provide design parameters for engineering uniform, targeted-shielded VMNs with very high cell transfection rates that exhibit the important characteristics for in vivo translation. These design parameters and principles could be used to tailor-make and assemble targeted-shielded VMNs that could deliver any nucleic acid payload to any mammalian cell type.
Collapse
Affiliation(s)
- Zahra Karjoo
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, 08854, USA
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s University, Belfast, BT9 7BL, United Kingdom
| | - Parin Patel
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, 08854, USA
| | | | - Arash Hatefi
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, 08854, USA
| |
Collapse
|
3080
|
Liu D, He C, Wang AZ, Lin W. Application of liposomal technologies for delivery of platinum analogs in oncology. Int J Nanomedicine 2013; 8:3309-19. [PMID: 24023517 PMCID: PMC3767488 DOI: 10.2147/ijn.s38354] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Platinum-based chemotherapy, such as cisplatin, oxaliplatin, and carboplatin, is one of the most widely utilized classes of cancer therapeutics. While highly effective, the clinical applications of platinum-based drugs are limited by their toxicity profiles as well as suboptimal pharmacokinetic properties. Therefore, one of the key research areas in oncology has been to develop novel platinum analog drugs and engineer new platinum drug formulations to improve the therapeutic ratio further. Such efforts have led to the development of platinum analogs including nedaplatin, heptaplatin, and lobaplatin. Moreover, reformulating platinum drugs using liposomes has resulted in the development of L-NDPP (Aroplatin™), SPI-77, Lipoplatin™, Lipoxal™, and LiPlaCis®. Liposomes possess several attractive biological activities, including biocompatibility, high drug loading, and improved pharmacokinetics, that are well suited for platinum drug delivery. In this review, we discuss the various platinum drugs and their delivery using liposome-based drug delivery vehicles. We compare and contrast the different liposome platforms as well as speculate on the future of platinum drug delivery research.
Collapse
Affiliation(s)
- Demin Liu
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
3081
|
Busseron E, Ruff Y, Moulin E, Giuseppone N. Supramolecular self-assemblies as functional nanomaterials. NANOSCALE 2013; 5:7098-140. [PMID: 23832165 DOI: 10.1039/c3nr02176a] [Citation(s) in RCA: 508] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
In this review, we survey the diversity of structures and functions which are encountered in advanced self-assembled nanomaterials. We highlight their flourishing implementations in three active domains of applications: biomedical sciences, information technologies, and environmental sciences. Our main objective is to provide the reader with a concise and straightforward entry to this broad field by selecting the most recent and important research articles, supported by some more comprehensive reviews to introduce each topic. Overall, this compilation illustrates how, based on the rules of supramolecular chemistry, the bottom-up approach to design functional objects at the nanoscale is currently producing highly sophisticated materials oriented towards a growing number of applications with high societal impact.
Collapse
Affiliation(s)
- Eric Busseron
- SAMS Research Group, University of Strasbourg, Institut Charles Sadron, CNRS, 23 rue du Loess, BP 84087, 67034 Strasbourg Cedex 2, France
| | | | | | | |
Collapse
|
3082
|
Yu KF, Zhang WQ, Luo LM, Song P, Li D, Du R, Ren W, Huang D, Lu WL, Zhang X, Zhang Q. The antitumor activity of a doxorubicin loaded, iRGD-modified sterically-stabilized liposome on B16-F10 melanoma cells: in vitro and in vivo evaluation. Int J Nanomedicine 2013; 8:2473-85. [PMID: 23885174 PMCID: PMC3716561 DOI: 10.2147/ijn.s46962] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Considering the fact that iRGD (tumor-homing peptide) demonstrates tumor-targeting and tumor-penetrating activity, and that B16-F10 (murine melanoma) cells overexpress both αv integrin receptor and neuropilin-1 (NRP-1), the purpose of this study was to prepare a novel doxorubicin (DOX)-loaded, iRGD-modified, sterically-stabilized liposome (SSL) (iRGD-SSL-DOX) in order to evaluate its antitumor activity on B16-F10 melanoma cells in vitro and in vivo. The iRGD-SSL-DOX was prepared using a thin-film hydration method. The characteristics of iRGD-SSL-DOX were evaluated. The in vitro leakage of DOX from iRGD-SSL-DOX was tested. The in vitro tumor-targeting and tumor-penetrating characteristics of iRGD-modified liposomes on B16-F10 cells were investigated. The in vivo tumor-targeting and tumor-penetrating activities of iRGD-modified liposomes were performed in B16-F10 tumor-bearing nude mice. The antitumor effect of iRGD-SSL-DOX was evaluated in B16-F10 tumor-bearing C57BL/6 mice in vivo. The average particle size of the iRGD-SSL-DOX was found to be 91 nm with a polydispersity index (PDI) of 0.16. The entrapment efficiency of iRGD-SSL-DOX was 98.36%. The leakage of DOX from iRGD-SSL-DOX at the 24-hour time point was only 7.5%. The results obtained from the in vitro flow cytometry and confocal microscopy, as well as in vivo biodistribution and confocal immunofluorescence microscopy experiments, indicate that the tumor-targeting and tumor-penetrating activity of the iRGD-modified SSL was higher than that of unmodified SSL. In vivo antitumor activity results showed that the antitumor effect of iRGD-SSL-DOX against melanoma tumors was higher than that of SSL-DOX in B16-F10 tumor-bearing mice. In conclusion, the iRGD-SSL-DOX is a tumor-targeting and tumor-penetrating peptide modified liposome which has significant antitumor activity against melanoma tumors.
Collapse
Affiliation(s)
- Ke-Fu Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3083
|
Chitkara D, Kumar N. BSA-PLGA-based core-shell nanoparticles as carrier system for water-soluble drugs. Pharm Res 2013; 30:2396-409. [PMID: 23756758 DOI: 10.1007/s11095-013-1084-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 05/12/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE Preparation, optimization and in vitro evaluation of core-shell nanoparticles comprising of a hydrophilic core of BSA surrounded by a hydrophobic shell of PLGA for loading water-soluble drugs. METHODS A double emulsion method was optimized for preparation of BSA-PLGA based core-shell nanoparticles. Proof of concept for core-shell type structure was established by visual techniques like confocal microscopy and TEM. Characterization was done for particle size, encapsulation efficiency, drug loading and in vitro drug release. Cellular uptake was assessed using confocal microscopy, bio-TEM and HPLC assay, and cytotoxic activity was tested by MTT assay in MG-63 osteosarcoma cells. RESULTS The optimized core-shell nanoparticles showed a particle size of 243 nm (PDI-0.13) and encapsulation efficiency of 40.5% with a drug loading of 8.5% w/w. In vitro drug release studies showed a sustained release for 12 h. Cellular uptake studies indicated a rapid and efficient uptake within 2 h. TEM studies indicated that the core-shell nanoparticles were localized in cytoplasm region of the cells. Gemcitabine loaded core-shell nanoparticles showed enhanced cytotoxicity against MG-63 cells as compared to marketed formulation of gemcitabine (GEMCITE®). CONCLUSION These results indicate that core-shell nanoparticles can be a good carrier system for delivering hydrophilic drugs like gemcitabine successfully to the cells with enhanced efficacy.
Collapse
Affiliation(s)
- Deepak Chitkara
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S Nagar, Punjab, 160 062, India
| | | |
Collapse
|
3084
|
Loi M, Di Paolo D, Soster M, Brignole C, Bartolini A, Emionite L, Sun J, Becherini P, Curnis F, Petretto A, Sani M, Gori A, Milanese M, Gambini C, Longhi R, Cilli M, Allen TM, Bussolino F, Arap W, Pasqualini R, Corti A, Ponzoni M, Marchiò S, Pastorino F. Novel phage display-derived neuroblastoma-targeting peptides potentiate the effect of drug nanocarriers in preclinical settings. J Control Release 2013; 170:233-41. [PMID: 23714122 DOI: 10.1016/j.jconrel.2013.04.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 04/23/2013] [Accepted: 04/29/2013] [Indexed: 01/20/2023]
Abstract
Molecular targeting of drug delivery nanocarriers is expected to improve their therapeutic index while decreasing their toxicity. Here we report the identification and characterization of novel peptide ligands specific for cells present in high-risk neuroblastoma (NB), a childhood tumor mostly refractory to current therapies. To isolate such targeting moieties, we performed combined in vitro/ex-vivo phage display screenings on NB cell lines and on tumors derived from orthotopic mouse models of human NB. By designing proper subtractive protocols, we identified phage clones specific either for the primary tumor, its metastases, or for their respective stromal components. Globally, we isolated 121 phage-displayed NB-binding peptides: 26 bound the primary tumor, 15 the metastatic mass, 57 and 23 their respective microenvironments. Of these, five phage clones were further validated for their specific binding ex-vivo to biopsies from stage IV NB patients and to NB tumors derived from mice. All five clones also targeted tumor cells and vasculature in vivo when injected into NB-bearing mice. Coupling of the corresponding targeting peptides with doxorubicin-loaded liposomes led to a significant inhibition in tumor volume and enhanced survival in preclinical NB models, thereby paving the way to their clinical development.
Collapse
Affiliation(s)
- Monica Loi
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3085
|
Sen K, Mandal M. Second generation liposomal cancer therapeutics: transition from laboratory to clinic. Int J Pharm 2013; 448:28-43. [PMID: 23500602 DOI: 10.1016/j.ijpharm.2013.03.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 03/03/2013] [Accepted: 03/06/2013] [Indexed: 01/10/2023]
Abstract
Recent innovations and developments in nanotechnology have revolutionized cancer therapeutics. Engineered nanomaterials are the current workhorses in the emerging field of cancer nano-therapeutics. Lipid vesicles bearing anti-tumor drugs have turned out to be a clinically feasible and promising nano-therapeutic approach to treat cancer. Efficient entrapment of therapeutics, biocompatibility, biodegradability, low systemic toxicity, low immunogenicity and ability to bypass multidrug resistance mechanisms has made liposomes a versatile drug/gene delivery system in cancer chemotherapy. The present review attempts to explore the recent key advances in liposomal research and the vast arsenal of liposomal formulations currently being utilized in treatment and diagnosis of cancer.
Collapse
Affiliation(s)
- Kacoli Sen
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, India
| | | |
Collapse
|
3086
|
Sapra P, Shor B. Monoclonal antibody-based therapies in cancer: advances and challenges. Pharmacol Ther 2013; 138:452-69. [PMID: 23507041 DOI: 10.1016/j.pharmthera.2013.03.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 02/19/2013] [Indexed: 12/25/2022]
Abstract
Conventional anticancer therapeutics often suffer from lack of specificity, resulting in toxicities to normal healthy tissues and poor therapeutic index. Antibody-mediated delivery of anticancer drugs or toxins to tumor cells through tumor selective or overexpressed antigens is progressively being recognized as an effective strategy for increasing the therapeutic index of anticancer drugs. In this review we focus on three therapeutic modalities in the field of antibody-mediated targeting, including antibody-drug conjugates (ADCs), immunotoxins (ITs) and immunoliposomes (ILs). Design considerations for development of each of the above therapeutic modalities are discussed. Furthermore, an overview of ADCs, ITs or ILs approved for use in clinical oncology and those currently in clinical development is provided. Challenges encountered by the field of antibody-based targeting are discussed and concepts around development of the next generation of antibody therapeutics are presented.
Collapse
Affiliation(s)
- Puja Sapra
- Bioconjugates Discovery and Development, Oncology Research Unit, Pfizer Worldwide Research and Development, 401 North Middletown Road, Pearl River, NY, 10965, USA.
| | | |
Collapse
|
3087
|
Recent trends in multifunctional liposomal nanocarriers for enhanced tumor targeting. JOURNAL OF DRUG DELIVERY 2013; 2013:705265. [PMID: 23533772 PMCID: PMC3606784 DOI: 10.1155/2013/705265] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 02/06/2013] [Indexed: 12/30/2022]
Abstract
Liposomes are delivery systems that have been used to formulate a vast variety of therapeutic and imaging agents for the past several decades. They have significant advantages over their free forms in terms of pharmacokinetics, sensitivity for cancer diagnosis and therapeutic efficacy. The multifactorial nature of cancer and the complex physiology of the tumor microenvironment require the development of multifunctional nanocarriers. Multifunctional liposomal nanocarriers should combine long blood circulation to improve pharmacokinetics of the loaded agent and selective distribution to the tumor lesion relative to healthy tissues, remote-controlled or tumor stimuli-sensitive extravasation from blood at the tumor's vicinity, internalization motifs to move from tumor bounds and/or tumor intercellular space to the cytoplasm of cancer cells for effective tumor cell killing. This review will focus on current strategies used for cancer detection and therapy using liposomes with special attention to combination therapies.
Collapse
|
3088
|
Abstract
This position paper discusses progress made and to be made with so-called advanced drug delivery systems, particularly but not exclusively those in the nanometre domain. The paper has resulted from discussions with a number of international experts in the field who shared their views on aspects of the subject, from the nomenclature used for such systems, the sometimes overwrought claims made in the era of nanotechnology, the complex nature of targeting delivery systems to specific destinations in vivo, the need for setting standards for the choice and characterisation of cell lines used in in vitro studies, to attention to the manufacturability, stability and analytical profiling of systems and more relevant studies on toxicology. The historical background to the development of many systems is emphasised. So too is the stochastic nature of many of the steps to successful access to and action in targets. A lacuna in the field is the lack of availability of data on a variety of carrier systems using the same models in vitro and in vivo using standard controls. The paper asserts that greater emphasis must also be paid to the effective levels of active attained in target organs, for without such crucial data it will be difficult for many experimental systems to enter the clinic. This means the use of diagnostic/imaging technologies to monitor targeted drug delivery and stratify patient groups, identifying patients with optimum chances for successful therapy. Last, but not least, the critical importance of the development of science bases for regulatory policies, scientific platforms overseeing the field and new paradigms of financing are discussed.
Collapse
Affiliation(s)
- Daan J A Crommelin
- Department of Pharmaceutical Sciences, University of Utrecht, 3511 ME Utrecht, The Netherlands.
| | | |
Collapse
|
3089
|
Affiliation(s)
- Rogério Gaspar
- Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
| |
Collapse
|
3090
|
Nogueira E, Loureiro A, Nogueira P, Freitas J, Almeida CR, Härmark J, Hebert H, Moreira A, Carmo AM, Preto A, Gomes AC, Cavaco-Paulo A. Liposome and protein based stealth nanoparticles. Faraday Discuss 2013; 166:417-29. [DOI: 10.1039/c3fd00057e] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|