201
|
Zhang L, Zhou H, Belzile O, Thorpe P, Zhao D. Phosphatidylserine-targeted bimodal liposomal nanoparticles for in vivo imaging of breast cancer in mice. J Control Release 2014; 183:114-23. [PMID: 24698945 DOI: 10.1016/j.jconrel.2014.03.043] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/24/2014] [Indexed: 11/30/2022]
Abstract
Phosphatidylserine (PS) that is normally constrained to the inner plasma membrane becomes exposed on the surface of endothelial cells (ECs) in tumor vasculature. In the present study, we report the development of a novel tumor vasculature-targeted liposomal nanoprobe by conjugating a human monoclonal antibody, PGN635 that specifically targets PS to polyethylene glycol-coated liposomes. MR contrast, superparamagnetic iron oxide nanoparticles (SPIO) were packed into the core of liposomes, while near-infrared dye, DiR was incorporated into the lipophilic bilayer. The liposomal nanoprobe PGN-L-IO/DiR was fully characterized, and its binding specificity and subsequent internalization into PS-exposed vascular ECs was confirmed by in vitro MRI and histological staining. In vivo longitudinal MRI and optical imaging were performed after i.v. injection of the liposomal nanoprobes into mice bearing breast MDA-MB231 tumors. At 9.4T, T2-weighted MRI detected drastic reduction on signal intensity and T2 values of tumors at 24h. Ionizing radiation significantly increased PS exposure on tumor vascular ECs, resulting in a further MRI signal loss of tumors. Concurrent with MRI, optical imaging revealed a clear tumor contrast at 24h. Intriguingly, PGN-L-IO/DiR exhibited distinct pharmacokinetics and biodistribution with significantly reduced accumulations in liver or spleen. Localization of PGN-L-IO/DiR to tumor was antigen specific, since a control probe of irrelevant specificity showed minimal accumulation in the tumors. Our studies indicate that PS-targeted liposomes may provide a useful platform for tumor-targeted delivery of imaging contrast agents or potentially anti-cancer drugs for cancer theranostics.
Collapse
Affiliation(s)
- Liang Zhang
- Radiology, UT Southwestern Medical Center, Dallas, USA
| | - Heling Zhou
- Radiology, UT Southwestern Medical Center, Dallas, USA
| | | | - Philip Thorpe
- Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Dawen Zhao
- Radiology, UT Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
202
|
Tavano L, Aiello R, Ioele G, Picci N, Muzzalupo R. Niosomes from glucuronic acid-based surfactant as new carriers for cancer therapy: preparation, characterization and biological properties. Colloids Surf B Biointerfaces 2014; 118:7-13. [PMID: 24709252 DOI: 10.1016/j.colsurfb.2014.03.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 10/25/2022]
Abstract
Niosomes are vesicular systems composed of surfactant molecules, claimed to be used as drug delivery carriers thanks to their physico-chemical and biological properties. The aim of this work was to design niosomes obtained with a surfactant synthesized from glucuronic acid. Doxorubicin and 5FU were used as model drugs. Niosomes were prepared with different ratios between surfactant and cholesterol, and characterized in terms of size, morphology, drugs entrapment efficiency and in vitro releases, to identify the optimal formulation to be used in pharmaceutical fields. In addition, the hemolytic activity of all formulations have been also evaluated. Results showed that dodecylglucuronamide surfactant was able to produce vesicular systems with or without the presence of cholesterol. Niosomes resulted regular in size and shape and they have been found to encapsulate and release in a controlled manner both doxorubicin and 5-fluorouracil. Hemolytic tests showed that the capability of disrupting erythrocyte only depend on the size of colloidal aggregates. Finally, our formulations could be potentially used as antitumoral delivery systems in anticancer therapy.
Collapse
Affiliation(s)
- Lorena Tavano
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Edificio Polifunzionale, Arcavacata di Rende, 87036 Cosenza, Italy; Dipartimento di Ingegneria Informatica, Modellistica, Elettronica e Sistemistica, Università della Calabria, Via P. Bucci Cubo 39/C, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Rossana Aiello
- Dipartimento di Scienze della Salute, Università Magna Graecia, Via T. Campanella 115, 88100 Catanzaro, Italy
| | - Giuseppina Ioele
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Edificio Polifunzionale, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Nevio Picci
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Edificio Polifunzionale, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Rita Muzzalupo
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Edificio Polifunzionale, Arcavacata di Rende, 87036 Cosenza, Italy.
| |
Collapse
|
203
|
Yang KN, Zhang CQ, Wang W, Wang PC, Zhou JP, Liang XJ. pH-responsive mesoporous silica nanoparticles employed in controlled drug delivery systems for cancer treatment. Cancer Biol Med 2014; 11:34-43. [PMID: 24738037 PMCID: PMC3969802 DOI: 10.7497/j.issn.2095-3941.2014.01.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/10/2014] [Indexed: 01/09/2023] Open
Abstract
In the fight against cancer, controlled drug delivery systems have emerged to enhance the therapeutic efficacy and safety of anti-cancer drugs. Among these systems, mesoporous silica nanoparticles (MSNs) with a functional surface possess obvious advantages and were thus rapidly developed for cancer treatment. Many stimuli-responsive materials, such as nanoparticles, polymers, and inorganic materials, have been applied as caps and gatekeepers to control drug release from MSNs. This review presents an overview of the recent progress in the production of pH-responsive MSNs based on the pH gradient between normal tissues and the tumor microenvironment. Four main categories of gatekeepers can respond to acidic conditions. These categories will be described in detail.
Collapse
Affiliation(s)
- Ke-Ni Yang
- 1 State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China ; 2 CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, China ; 3 Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington DC 20060, USA
| | - Chun-Qiu Zhang
- 1 State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China ; 2 CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, China ; 3 Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington DC 20060, USA
| | - Wei Wang
- 1 State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China ; 2 CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, China ; 3 Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington DC 20060, USA
| | - Paul C Wang
- 1 State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China ; 2 CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, China ; 3 Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington DC 20060, USA
| | - Jian-Ping Zhou
- 1 State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China ; 2 CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, China ; 3 Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington DC 20060, USA
| | - Xing-Jie Liang
- 1 State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China ; 2 CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, China ; 3 Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington DC 20060, USA
| |
Collapse
|
204
|
Paliwal SR, Paliwal R, Vyas SP. A review of mechanistic insight and application of pH-sensitive liposomes in drug delivery. Drug Deliv 2014; 22:231-42. [PMID: 24524308 DOI: 10.3109/10717544.2014.882469] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The pH-sensitive liposomes have been extensively used as an alternative to conventional liposomes in effective intracellular delivery of therapeutics/antigen/DNA/diagnostics to various compartments of the target cell. Such liposomes are destabilized under acidic conditions of the endocytotic pathway as they usually contain pH-sensitive lipid components. Therefore, the encapsulated content is delivered into the intracellular bio-environment through destabilization or its fusion with the endosomal membrane. The therapeutic efficacy of pH-sensitive liposomes enables them as biomaterial with commercial utility especially in cancer treatment. In addition, targeting ligands including antibodies can be anchored on the surface of pH-sensitive liposomes to target specific cell surface receptors/antigen present on tumor cells. These vesicles have also been widely explored for antigen delivery and serve as immunological adjuvant to enhance the immune response to antigens. The present review deals with recent research updates on application of pH-sensitive liposomes in chemotherapy/diagnostics/antigen/gene delivery etc.
Collapse
Affiliation(s)
- Shivani Rai Paliwal
- Department of Pharmaceutics, SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya Bilaspur , Chhattisgarh , India
| | | | | |
Collapse
|
205
|
Offerman SC, Kamra Verma AV, Telfer BA, Berk DA, Clarke DJ, Aojula HS. Ability of co-administered peptide liposome nanoparticles to exploit tumour acidity for drug delivery. RSC Adv 2014. [DOI: 10.1039/c3ra44746d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
206
|
Ninomiya K, Kawabata S, Tashita H, Shimizu N. Ultrasound-mediated drug delivery using liposomes modified with a thermosensitive polymer. ULTRASONICS SONOCHEMISTRY 2014; 21:310-316. [PMID: 23948493 DOI: 10.1016/j.ultsonch.2013.07.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 07/21/2013] [Accepted: 07/24/2013] [Indexed: 06/02/2023]
Abstract
Ultrasound-mediated drug delivery was established using liposomes that were modified with the thermosensitive polymer (TSP) poly(NIPMAM-co-NIPAM), which sensitized the liposomes to high temperatures. TSP-modified liposomes (TSP liposomes) released encapsulated calcein under 1 MHz ultrasound irradiation at 0.5 W/cm(2) for 120 s as well as the case under incubation at 42 °C for 15 min. In addition, uptake of the drug released from TSP liposomes by cancer cells was enhanced by ultrasound irradiation. In a cell injury assay using doxorubicin (DOX)-loaded TSP liposomes and ultrasound irradiation, cell viability of HepG2 cells at 6 h after ultrasound irradiation (1 MHz, 0.5 W/cm(2) for 30 s) with DOX-loaded TSP liposomes (TSP/lipid ratio=1) was 60%, which was significantly lower than that of the control conditions such as DOX-loaded TSP liposomes alone and DOX-loaded intact liposomes under ultrasound irradiation.
Collapse
Affiliation(s)
- Kazuaki Ninomiya
- Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa 920-1192, Japan
| | | | | | | |
Collapse
|
207
|
Liposomal encapsulation enhances in vivo near infrared imaging of exposed phosphatidylserine in a mouse glioma model. Molecules 2013; 18:14613-28. [PMID: 24287994 PMCID: PMC6269843 DOI: 10.3390/molecules181214613] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/18/2013] [Accepted: 11/21/2013] [Indexed: 11/17/2022] Open
Abstract
We have previously demonstrated that exposed phosphatidylserine (PS) on tumor vascular endothelial cells is highly tumor specific, and development of the PS targeted near infrared (NIR) optical probe enables successful in vivo optical imaging of U87 gliomas in a mouse model. Liposomes have been widely used as a nanovector for delivery of chemotherapeutics and imaging contrast agents due to their high payload and longer circulation time. In the current study, we have fabricated PS-targeted liposomal nanoprobes encapsulating a NIR dye, IRDye® 800CW, aiming to enhance PS-targeted tumor imaging. Hydrophilic 800CW dye was packed into the core of polyethylene glycol (PEG)-coated liposomes functionalized with F(ab’)2 fragments of PGN635, a fully human monoclonal antibody that binds PS. As expected, in vivo dynamic NIR imaging revealed significantly improved tumor/normal contrast (TNR = 20 ± 3; p < 0.01) of subcutaneous U87 gliomas in mice after injection of the liposomal nanoprobes. Markedly enhanced TNR was observed after the tumors were irradiated to increase PS exposure (TNR = 48 ± 6; p < 0.05). Intriguingly, the liposomal nanoprobes, PGN-L-800CW showed distinct biodistribution and pharmacokinetics compared to the 800CW-PGN probes used in our previous study. Our data further suggest the usefulness of PS-targeted imaging probes for sensitive tumor detection and the potential of utilizing liposomal platform for glioma theranostics.
Collapse
|
208
|
Gao Y, Xie J, Chen H, Gu S, Zhao R, Shao J, Jia L. Nanotechnology-based intelligent drug design for cancer metastasis treatment. Biotechnol Adv 2013; 32:761-77. [PMID: 24211475 DOI: 10.1016/j.biotechadv.2013.10.013] [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: 04/26/2013] [Revised: 10/19/2013] [Accepted: 10/29/2013] [Indexed: 12/21/2022]
Abstract
Traditional chemotherapy used today at clinics is mainly inherited from the thinking and designs made four decades ago when the Cancer War was declared. The potency of those chemotherapy drugs on in-vitro cancer cells is clearly demonstrated at even nanomolar levels. However, due to their non-specific effects in the body on normal tissues, these drugs cause toxicity, deteriorate patient's life quality, weaken the host immunosurveillance system, and result in an irreversible damage to human's own recovery power. Owing to their unique physical and biological properties, nanotechnology-based chemotherapies seem to have an ability to specifically and safely reach tumor foci with enhanced efficacy and low toxicity. Herein, we comprehensively examine the current nanotechnology-based pharmaceutical platforms and strategies for intelligent design of new nanomedicines based on targeted drug delivery system (TDDS) for cancer metastasis treatment, analyze the pros and cons of nanomedicines versus traditional chemotherapy, and evaluate the importance that nanomaterials can bring in to significantly improve cancer metastasis treatment.
Collapse
Affiliation(s)
- Yu Gao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Jingjing Xie
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Haijun Chen
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China; Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Fuzhou University, Fujian 350108, China
| | - Songen Gu
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Rongli Zhao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Jingwei Shao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China.
| |
Collapse
|
209
|
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: 343] [Impact Index Per Article: 28.6] [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
|
210
|
Pourbaghi-Masouleh M, Hosseini V. Amorphous calcium phosphate nanoparticles could function as a novel cancer therapeutic agent by employing a suitable targeted drug delivery platform. NANOSCALE RESEARCH LETTERS 2013; 8:449. [PMID: 24172080 PMCID: PMC3816303 DOI: 10.1186/1556-276x-8-449] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 10/09/2013] [Indexed: 05/08/2023]
Abstract
Employment of nanovehicular system for delivering apoptogenic agent to cancer cells for inducing apoptosis has widely been investigated. Loading efficacy and controlled release of the agents are of the inseparable obstacles that hamper the efforts in reaching an efficacious targeted cancer therapy method. When the carrier itself is apoptogenic, then there is no need to load the carrier with apoptogenic agent and just delivering of the particle to the specific location matters. Hence, we hypothesize that amorphous calcium phosphate nanoparticle (ACPN) is a potent candidate for apoptosis induction, although encapsulation in liposome shell, and surface decoration with targeting ligand (TL), and cell-penetrating peptide (CPP) plays a pivotal role in the employment of this agent. It is well understood that elevation in cytosolic Ca2+ ([Ca2+]c) would result in the induction of apoptosis. ACPN has the potential to cause imbalance in this medium by elevating [Ca2+]c. Owning to the fact that the nanoparticles should be delivered into cytosol, it is necessary to trap them in a liposomal shell for evading endocytosis. It was demonstrated that employment of the trans-activator of transcription (TAT) as CPP eminently enhances the efficacy of endosomal escape; therefore, the platform is designed in a way that TAT is positioned on the surface of the liposome. Due to the fact that the apoptosis should be induced in sole cancer cells, Folate as TL is also attached on the surface of the liposome. This hypothesis heralds the new generation of chemotherapeutic agents and platforms which could have less side effect than the most common ones, in addition to other advantages they have.
Collapse
Affiliation(s)
- Milad Pourbaghi-Masouleh
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj, P.O. Box: 31787/316, Iran
| | - Vahid Hosseini
- Department of Health Science and Technology, Laboratory of Applied Mechanobiology, ETH, Zürich 8093, Switzerland
| |
Collapse
|
211
|
Park K. Programmed sickle cells for targeted delivery to hypoxic tumors. J Control Release 2013; 171:258. [PMID: 24016418 DOI: 10.1016/j.jconrel.2013.08.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 11/26/2012] [Indexed: 10/26/2022]
Affiliation(s)
- Kinam Park
- Purdue University, Departments of Biomedical Engineering and Pharmaceutics, West Lafayette, IN, USA.
| |
Collapse
|
212
|
Efficient hepatic delivery of drugs: novel strategies and their significance. BIOMED RESEARCH INTERNATIONAL 2013; 2013:382184. [PMID: 24286077 PMCID: PMC3826320 DOI: 10.1155/2013/382184] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 08/14/2013] [Accepted: 08/25/2013] [Indexed: 02/06/2023]
Abstract
Liver is a vital organ responsible for plethora of functions including detoxification, protein synthesis, and the production of biochemicals necessary for the sustenance of life. Therefore, patients with chronic liver diseases such as viral hepatitis, liver cirrhosis, and hepatocellular carcinoma need immediate attention to sustain life and as a result are often exposed to the prolonged treatment with drugs/herbal medications. Lack of site-specific delivery of these medications to the hepatocytes/nonparenchymal cells and adverse effects associated with their off-target interactions limit their continuous use. This calls for the development and fabrication of targeted delivery systems which can deliver the drug payload at the desired site of action for defined period of time. The primary aim of drug targeting is to manipulate the whole body distribution of drugs, that is, to prevent distribution to non-target cells and concomitantly increase the drug concentration at the targeted site. Carrier molecules are designed for their selective cellular uptake, taking advantage of specific receptors or binding sites present on the surface membrane of the target cell. In this review, various aspects of liver targeting of drug molecules and herbal medications have been discussed which elucidate the importance of delivering the drugs/herbal medications at their desired site of action.
Collapse
|
213
|
Arouri A, Hansen AH, Rasmussen TE, Mouritsen OG. Lipases, liposomes and lipid-prodrugs. Curr Opin Colloid Interface Sci 2013. [DOI: 10.1016/j.cocis.2013.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
214
|
Chen YC, Chang CH, Yang YM, Maa JR, Lin JL, Wu CH. Interaction of Dipalmitoyl Phosphatidylcholine with n-Hexadecanol in Monolayer and Liposome. J CHIN CHEM SOC-TAIP 2013. [DOI: 10.1002/jccs.200700046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
215
|
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: 659] [Impact Index Per Article: 54.9] [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
|
216
|
Lee J, Lee MG, Jung C, Park YH, Song C, Choi MC, Park HG, Park JK. High-throughput nanoscale lipid vesicle synthesis in a semicircular contraction-expansion array microchannel. BIOCHIP JOURNAL 2013. [DOI: 10.1007/s13206-013-7303-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
217
|
Perera RH, Solorio L, Wu H, Gangolli M, Silverman E, Hernandez C, Peiris PM, Broome AM, Exner AA. Nanobubble ultrasound contrast agents for enhanced delivery of thermal sensitizer to tumors undergoing radiofrequency ablation. Pharm Res 2013; 31:1407-17. [PMID: 23943542 DOI: 10.1007/s11095-013-1100-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 06/04/2013] [Indexed: 12/20/2022]
Abstract
PURPOSE Pluronic has been shown to sensitize various tumor cell lines to chemotherapy and hyperthermia by altering the membrane fluidity, depleting ATP, and modulating the heat shock protein 70 expression. In our prior work, Pluronic was also used to formulate nanosized ultrasound contrast agents. In the current study we evaluate the use of these contrast agents as vehicles for image-guided delivery of Pluronic to improve outcomes of tumor radiofrequency (RF) ablation. METHODS Lipid-shelled Pluronic nanobubbles were prepared and examined for size distribution, zeta potential, stability, biodistribution, accumulation of nanobubbles in the tumor, and treatment efficacy. LS174-T xenograft tumor-bearing mice were used to evaluate tumor growth suppression and measure treatment efficacy after RF ablation. RESULTS The average diameter of Pluronic bubbles was 230 nm, and initial bubble echogenicity was 16 dB. In vitro, cells exposed to Pluronic nanobubbles exhibited low cytotoxicity in the absence of ultrasound, even if heat (43 ºC) was applied. When the cells were exposed to Pluronic nanobubbles, heat, and ultrasound; viability was significantly reduced. In vivo, tumors treated with ultrasound-modulated nanobubbles prior to RF ablation showed a significant reduction in growth compared to the RF alone (P<0.05). CONCLUSION Lipid and Pluronic-shelled, echogenic nanobubbles combined with ultrasound modulation can serve as an effective theranostic method for sensitization of tumors to RF ablation.
Collapse
Affiliation(s)
- Reshani H Perera
- Case Center for Imaging Research, Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, Ohio, 44106-5056, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Choe SW, Terman DS, Rivers AE, Rivera J, Lottenberg R, Sorg BS. Drug-loaded sickle cells programmed ex vivo for delayed hemolysis target hypoxic tumor microvessels and augment tumor drug delivery. J Control Release 2013; 171:184-92. [PMID: 23871960 DOI: 10.1016/j.jconrel.2013.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 06/27/2013] [Accepted: 07/02/2013] [Indexed: 12/22/2022]
Abstract
Selective drug delivery to hypoxic tumor niches remains a significant therapeutic challenge that calls for new conceptual approaches. Sickle red blood cells (SSRBCs) have shown an ability to target such hypoxic niches and induce tumoricidal effects when used together with exogenous pro-oxidants. Here we determine whether the delivery of a model therapeutic encapsulated in murine SSRBCs can be enhanced by ex vivo photosensitization under conditions that delay autohemolysis to a time that coincides with maximal localization of SSRBCs in a hypoxic tumor. Hyperspectral imaging of 4T1 carcinomas shows oxygen saturation levels <10% in a large fraction (commonly 50% or more) of the tumor. Using video microscopy of dorsal skin window chambers implanted with 4T1 tumors, we demonstrate that allogeneic SSRBCs, but not normal RBCs (nRBCs), selectively accumulate in hypoxic 4T1 tumors between 12 and 24h after systemic administration. We further show that ex vivo photo-oxidation can program SSRBCs to postpone hemolysis/release of a model therapeutic to a point that coincides with their maximum sequestration in hypoxic tumor microvessels. Under these conditions, drug-loaded photosensitized SSRBCs show a 3-4 fold greater drug delivery to tumors compared to non-photosensitized SSRBCs, drug-loaded photosensitized nRBCs, and free drug. These results demonstrate that photo-oxidized SSRBCs, but not photo-oxidized nRBCs, sequester and hemolyze in hypoxic tumors and release substantially more drug than photo-oxidized nRBCs and non-photo-oxidized SSRBCs. Photo-oxidation of drug-loaded SSRBCs thus appears to exploit the unique tumor targeting and carrier properties of SSRBCs to optimize drug delivery to hypoxic tumors. Such programmed and drug-loaded SSRBCs therefore represent a novel and useful tool for augmenting drug delivery to hypoxic solid tumors.
Collapse
Affiliation(s)
- Se-woon Choe
- Gumi Electronics & Information Technology Research Institute, Gumi, Republic of Korea
| | | | | | | | | | | |
Collapse
|
219
|
Lorenzato C, Cernicanu A, Meyre ME, Germain M, Pottier A, Levy L, de Senneville BD, Bos C, Moonen C, Smirnov P. MRI contrast variation of thermosensitive magnetoliposomes triggered by focused ultrasound: a tool for image-guided local drug delivery. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:185-92. [PMID: 23281291 DOI: 10.1002/cmmi.1515] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/28/2012] [Accepted: 10/10/2012] [Indexed: 11/10/2022]
Abstract
Improved drug delivery control during chemotherapy has the potential to increase the therapeutic index. MRI contrast agent such as iron oxide nanoparticles can be co-encapsulated with drugs in nanocarrier liposomes allowing their tracking and/or visualization by MRI. Furthermore, the combination of a thermosensitive liposomal formulation with an external source of heat such as high intensity focused ultrasound guided by MR temperature mapping allows the controlled local release of the content of the liposome. MRI-guided high-intensity focused ultrasound (HIFU), in combination represents a noninvasive technique to generate local hyperthermia for drug release. In this study we used ultrasmall superparamagnetic iron oxide nanoparticles (USPIO) encapsulated in thermosensitive liposomes to obtain thermosensitive magnetoliposomes (TSM). The transverse and longitudinal relaxivities of this MRI contrast agent were measured upon TSM membrane phase transition in vitro using a water bath or HIFU. The results showed significant differences for MRI signal enhancement and relaxivities before and after heating, which were absent for nonthermosensitive liposomes and free nanoparticles used as controls. Thus, incorporation of USPIO as MRI contrast agents into thermosensitive liposomes should, besides TSM tumor accumulation monitoring, allow the visualization of TSM membrane phase transition upon temperature elevation. In conclusion, HIFU under MR image guidance in combination with USPIO-loaded thermosensitive liposomes as drug delivery system has the potential for a better control of drug delivery and to increase the drug therapeutic index.
Collapse
Affiliation(s)
- Cyril Lorenzato
- Imaging Division, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Abstract
BACKGROUND Nanoparticles are increasingly being incorporated into the design of diagnostic imaging agents. Significant research efforts have been conducted with one class of lipid nanoparticle (liposomes) radiolabeled with gamma-emitting radionuclides as radiopharmaceuticals for scintigraphic imaging of cancer, inflammation/infection and sentinel lymph node detection. OBJECTIVE This article reviews the current literature with special emphasis on the clinical studies performed with liposome radiopharmaceuticals for detection of tumors, infectious/inflammatory sites or metastatic lymph nodes. Future uses of liposome radiopharmaceuticals are also described. METHODS Characteristics required of the radionuclide, liposome formulation and radiolabeling method for an effective radiopharmaceutical are discussed. A description of the procedures and instrumentation for conducting an imaging study with liposome radiopharmaceutical is included. Clinical studies using liposome radiopharmaceuticals are summarized. Future imaging applications of first- and second-generation radiolabeled liposomes for chemodosimetry and the specific targeting of a disease process are also described. RESULTS/CONCLUSION The choice of radionuclide, liposome formulation and radiolabeling method must be carefully considered during the design of a liposome radiopharmaceutical for a given application. After-loading and surface chelation methods are the most efficient and practical. Clinical studies with liposome radiopharmaceuticals demonstrated that a wide variety of tumors could be detected with good sensitivity and specificity. Liposome radiopharmaceuticals could also clearly detect various soft tissue and bone inflammatory/infectious lesions, and performed equal to or better than infection imaging agents that are approved at present. Yet, despite these favorable results, no liposome radiopharmaceutical has been approved for any indication. Some of the reasons for this can be attributed to reports of an unexpected infusion-related adverse reaction in two studies, the requirement of more complex liposome manufacturing procedures, and the adoption of other competing imaging procedures. Continued research of liposome radiopharmaceutical design based on a better understanding of liposome biology, improved radiolabeling methodologies and advances in gamma camera technology is warranted.
Collapse
Affiliation(s)
- Beth A Goins
- The University of Texas Health Science Center at San Antonio, TX Department of Radiology, Mail Code 7800, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA +1 210 567 5575 ; +1 210 567 5549 ;
| |
Collapse
|
221
|
Joshi N, Saha R, Shanmugam T, Balakrishnan B, More P, Banerjee R. Carboxymethyl-chitosan-tethered lipid vesicles: hybrid nanoblanket for oral delivery of paclitaxel. Biomacromolecules 2013; 14:2272-82. [PMID: 23721348 DOI: 10.1021/bm400406x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We describe the development and evaluation of a hybrid lipopolymeric system comprising carboxymethyl chitosan (CMC), covalently tethered to phosphatidylethanolamine units on the surface of lipid nanovesicles, for oral delivery of paclitaxel. The bioploymer is intended to act as a blanket, thereby shielding the drug from harsh gastrointestinal conditions, whereas the lipid nanovesicle ensures high encapsulation efficiency of paclitaxel and its passive targeting to tumor. CMC-tethered nanovesicles (LN-C-PTX) in the size range of 200-300 nm improved the gastrointestinal resistance and mucoadhesion properties as compared with unmodified lipid nanovesicles (LN-PTX). Conjugation of CMC did not compromise the cytotoxic potential of paclitaxel yet facilitated the interaction and uptake of the nanovesicles by murine melanoma (B16F10) cells through an ATP-dependent process. CMC-conjugated nanovesicles, upon oral administration in rats, improved the plasma concentration profile of paclitaxel, with 1.5 fold increase in its bioavailability and 5.5 folds increase in elimination half life in comparison with Taxol. We also found that CMC in addition to providing a gastric resistant coating also imparted stealth character to the nanovesicles, thereby reducing their reticuloendothelial system (RES)-mediated uptake by liver and spleen and bypassing the need for PEGylation. In vivo efficacy in subcutaneous model of B16F10 showed significantly improved tumor growth inhibition and survival with CMC-tethered nanovesicles as compared with unmodified nanovesicles, both administered orally. LN-C-PTX exhibited therapeutic efficacy comparable to Taxol and Abraxane and also showed reduced toxicity and improved survival. Overall, these results suggest the therapeutic potential of CMC tethered nanovesicles as a platform for oral administration of paclitaxel and also unravel the ability of CMC to impart stealth character to the nanoparticles, thereby preventing their RES clearance.
Collapse
Affiliation(s)
- Nitin Joshi
- WRCBB, Department of Biosciences and Bioengineeering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | | | | | | | | | | |
Collapse
|
222
|
Zhu X, Fryd M, Wayland BB. Kinetic-mechanistic studies of lipase-polymer micelle binding and catalytic degradation: Enzyme interfacial activation. Polym Degrad Stab 2013. [DOI: 10.1016/j.polymdegradstab.2013.03.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
223
|
Rai Paliwal S, Paliwal R, Vyas SP. pH-sensitive Liposomes in Drug Delivery. SMART MATERIALS FOR DRUG DELIVERY 2013. [DOI: 10.1039/9781849736800-00080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The pH-sensitive liposomes have been extensively studied in recent years as an advantageous alternative to conventional liposomes in effective targeting and accumulation of anticancer drugs in tumors. pH-sensitive liposomes usually contain phosphatidylethanolamine and stabilizing amphiphiles and can destabilize under acidic conditions of the endocytotic pathway. The drug loaded is thought to be delivered into the cytoplasm, probably through destabilization of or fusion with the endosome membrane. This fusogenic property makes the pH-sensitive liposomes more efficient in delivering anticancer drugs than conventional liposomes. The intra-cellular release of drug/gene/diagnostic agents can be achieved without altering their therapeutic efficacy by means of the endosomal escape phenomenon. Cell surface targeting ligands, including antibodies, can be appended on the surface of pH-sensitive liposomes to target specific receptors on tumor cells. This chapter provides an introduction to pH-sensitive liposomes and examples of their therapeutic interest as smart drug-delivery systems.
Collapse
Affiliation(s)
- Shivani Rai Paliwal
- Drug Delivery Research Laboratory Department of Pharmaceutical Sciences, Dr H. S. Gour Vishwavidyalaya (A Central University), Sagar M.P. India, 470003
- Department of Pharmaceutics SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G. India, 495009
| | - Rishi Paliwal
- Drug Delivery Research Laboratory Department of Pharmaceutical Sciences, Dr H. S. Gour Vishwavidyalaya (A Central University), Sagar M.P. India, 470003
| | - Suresh P Vyas
- Drug Delivery Research Laboratory Department of Pharmaceutical Sciences, Dr H. S. Gour Vishwavidyalaya (A Central University), Sagar M.P. India, 470003
| |
Collapse
|
224
|
Enhanced therapeutic efficacy of iRGD-conjugated crosslinked multilayer liposomes for drug delivery. BIOMED RESEARCH INTERNATIONAL 2013; 2013:378380. [PMID: 23691500 PMCID: PMC3652104 DOI: 10.1155/2013/378380] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 03/06/2013] [Accepted: 03/18/2013] [Indexed: 12/27/2022]
Abstract
Targeting nanoparticles by conjugating various specific ligands has shown potential therapeutic efficacy in nanomedicine. However, poor penetration of antitumor drugs into solid tumors remains a major obstacle. Here, we describe a targeting strategy for antitumor drug delivery by conjugating a crosslinked multilamellar liposomal vesicle (cMLV) formulation with a tumor-penetrating peptide, iRGD. The results showed that iRGD peptides could facilitate the binding and cellular uptake of drug-loaded cMLVs and consequently enhance the antitumor efficacy in breast tumor cells, including multidrug-resistant cells. Moreover, colocalization data revealed that iRGD-conjugated cMLVs (iRGD-cMLVs) entered cells via the clathrin-mediated pathway, followed by endosome-lysosome transport for efficient drug delivery. Finally, in vivo study indicated that iRGD-cMLVs could deliver anticancer drugs efficiently to mediate significant tumor suppression.
Collapse
|
225
|
Joo KI, Xiao L, Liu S, Liu Y, Lee CL, Conti PS, Wong MK, Li Z, Wang P. Crosslinked multilamellar liposomes for controlled delivery of anticancer drugs. Biomaterials 2013; 34:3098-109. [PMID: 23375392 PMCID: PMC3995748 DOI: 10.1016/j.biomaterials.2013.01.039] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/05/2013] [Indexed: 02/07/2023]
Abstract
Liposomes constitute one of the most popular nanocarriers for the delivery of cancer therapeutics. However, since their potency is limited by incomplete drug release and inherent instability in the presence of serum components, their poor delivery occurs in certain circumstances. In this study, we address these shortcomings and demonstrate an alternative liposomal formulation, termed crosslinked multilamellar liposome (CML). With its properties of improved sustainable drug release kinetics and enhanced vesicle stability, CML can achieve controlled delivery of cancer therapeutics. CML stably encapsulated the anticancer drug doxorubicin (Dox) in the vesicle and exhibited a remarkably controlled rate of release compared to that of the unilamellar liposome (UL) with the same lipid composition or Doxil-like liposome (DLL). Our imaging study demonstrated that the CMLs were mainly internalized through a caveolin-dependent pathway and were further trafficked through the endosome-lysosome compartments. Furthermore, in vivo experiments showed that the CML-Dox formulation reduced systemic toxicity and significantly improved therapeutic activity in inhibiting tumor growth compared to that of UL-Dox or DLL-Dox. This drug packaging technology may therefore provide a new treatment option to better manage cancer and other diseases.
Collapse
Affiliation(s)
- Kye-Il Joo
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089
| | - Liang Xiao
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089
| | - Shuanglong Liu
- Molecular Imaging Center, Department of Radiology, University of Southern California, Los Angeles, CA 90033
| | - Yarong Liu
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089
| | - Chi-Lin Lee
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089
| | - Peter S. Conti
- Molecular Imaging Center, Department of Radiology, University of Southern California, Los Angeles, CA 90033
| | - Michael K. Wong
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Zibo Li
- Molecular Imaging Center, Department of Radiology, University of Southern California, Los Angeles, CA 90033
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
226
|
Malaekeh-Nikouei B, Mousavi SH, Shahsavand S, Mehri S, Nassirli H, Moallem SA. Assessment of cytotoxic properties of safranal and nanoliposomal safranal in various cancer cell lines. Phytother Res 2013; 27:1868-73. [PMID: 23494763 DOI: 10.1002/ptr.4945] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Revised: 11/30/2012] [Accepted: 01/16/2013] [Indexed: 11/09/2022]
Abstract
Saffron (Crocus sativus) is a widely used food additive used for its color and taste. It has been reported that saffron possesses significant in vivo and in vitro anti-tumor activity. In the present study, anti-tumor effects of safranal, the major aromatic compound in saffron, and its liposomal form were investigated. The role of apoptosis has also been explored in this toxicity. HeLa, MCF7 and L929 cell lines were cultured and exposed to safranal (0.01-3 mM) or liposomal safranal (0.04-0.32 mM). 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium (MTT) assay was performed to assess cytotoxicity. Apoptosis was evaluated by staining cells with propidium iodide and quantifying sub-Gl peak by flow cytometry. MTT assay revealed a significant and concentration-dependent cytotoxic effect of safranal on HeLa and MCF7 cell lines. Liposomal safranal showed enhanced effect compared to the safranal solution, as compared by their IC50 concentrations. Flow cytometry results revealed induction of apoptosis by safranal. It might be concluded that safranal could be involved in saffron-induced cell death in HeLa and MCF7 cells. Liposome encapsulation improved anti-tumor effect of safranal. Safranal and particularly its liposomal form could be investigated as promising chemotherapeutic agents in cancer treatment.
Collapse
Affiliation(s)
- Bizhan Malaekeh-Nikouei
- Nanotechnology Research Centre, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | | | | | | |
Collapse
|
227
|
Smits EA, Smits CJ, Vromans H. The development of a method to quantify encapsulated and free prednisolone phosphate in liposomal formulations. J Pharm Biomed Anal 2013; 75:47-54. [DOI: 10.1016/j.jpba.2012.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/19/2012] [Accepted: 11/05/2012] [Indexed: 11/17/2022]
|
228
|
Zhu C, Yang Q, Lv F, Liu L, Wang S. Conjugated polymer-coated bacteria for multimodal intracellular and extracellular anticancer activity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2013; 25:1203-8. [PMID: 23280674 DOI: 10.1002/adma.201204550] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Indexed: 05/27/2023]
Affiliation(s)
- Chunlei Zhu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | | | | | | | | |
Collapse
|
229
|
Schully KL, Sharma S, Peine KJ, Pesce J, Elberson MA, Fonseca ME, Prouty AM, Bell MG, Borteh H, Gallovic M, Bachelder EM, Keane-Myers A, Ainslie KM. Rapid vaccination using an acetalated dextran microparticulate subunit vaccine confers protection against triplicate challenge by bacillus anthracis. Pharm Res 2013; 30:1349-61. [PMID: 23354770 DOI: 10.1007/s11095-013-0975-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/04/2013] [Indexed: 01/20/2023]
Abstract
PURPOSE A rapid immune response is required to prevent death from Anthrax, caused by Bacillus anthracis. METHOD We formulated a vaccine carrier comprised of acetalated dextran microparticles encapsulating recombinant protective antigen (rPA) and resiquimod (a toll-like receptor 7/8 agonist). RESULTS We were able to protect against triplicate lethal challenge by vaccinating twice (Days 0, 7) and then aggressively challenging on Days 14, 21, 28. A significantly higher level of antibodies was generated by day 14 with the encapsulated group compared to the conventional rPA and alum group. Antibodies produced by the co-encapsulated group were only weakly-neutralizing in toxin neutralization; however, survival was not dependent on toxin neutralization, as all vaccine formulations survived all challenges except control groups. Post-mortem culture swabs taken from the hearts of vaccinated groups that did not produce significant neutralizing titers failed to grow B. anthracis. CONCLUSIONS Results indicate that protective antibodies are not required for rapid protection; indeed, cytokine results indicate that T cell protection may play a role in protection from anthrax. We report the first instance of use of a particulate carrier to generate a rapid protective immunity against anthrax.
Collapse
Affiliation(s)
- Kevin L Schully
- Vaccine and Medical Countermeasures Department Biological Defense Research Directorate Naval Medical Research Center, Silver Spring, Maryland 20910, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Mussi SV, Torchilin VP. Recent trends in the use of lipidic nanoparticles as pharmaceutical carriers for cancer therapy and diagnostics. J Mater Chem B 2013; 1:5201-5209. [DOI: 10.1039/c3tb20990c] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
231
|
Petrichenko O, Erglis K, Cēbers A, Plotniece A, Pajuste K, Béalle G, Ménager C, Dubois E, Perzynski R. Bilayer properties of giant magnetic liposomes formed by cationic pyridine amphiphile and probed by active deformation under magnetic forces. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2013; 36:9. [PMID: 23359032 DOI: 10.1140/epje/i2013-13009-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 01/16/2013] [Indexed: 06/01/2023]
Abstract
We synthesize giant magnetic liposomes by a reverse-phase evaporation method (REV) using a new self-assembling Cationic Pyridine Amphiphile (CPA) derived from 1,4-dihydropyridine as liposome-forming agent and a magnetic ferrofluid based on γ-Fe(2)O(3) nanoparticles. Having in view the potential interest of CPA in targeted transport by magnetic forces, the mechanical elastic properties of such bilayers are here directly investigated in vesicles loaded with magnetic nanoparticles. Bending elastic modulus K(b) ∼ 0.2 to 5k(B)T and pre-stress τ ∼ 3.2 to 12.10(-6) erg/cm(2) are deduced from the under-field deformations of the giant magnetic liposomes. The obtained K(b) values are discussed in terms of A. Wurgers's theory.
Collapse
Affiliation(s)
- O Petrichenko
- University of Latvia, Zeļļu-8, LV-1002, Rıga, Latvia
| | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Niezgoda N, Mituła P, Kempińska K, Wietrzyk J, Wawrzeńczyk C. Synthesis of Phosphatidylcholine with Conjugated Linoleic Acid and Studies on Its Cytotoxic Activity. Aust J Chem 2013. [DOI: 10.1071/ch12404] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Phospholipids with conjugated linoleic acid (CLA), which are potential lipid prodrugs, were synthesised. CLA was obtained by the alkali-isomerisation of linoleic acid and was subsequently used in the synthesis of 1,2-di(conjugated)linoleoyl-sn-glycero-3-phosphocholine in good (82 %) yield. 1-Palmitoyl-2-(conjugated)linoleoyl-sn-glycero-3-phosphocholine was obtained by a two-step synthesis in 87 % yield. All the compounds were tested in an in vitro cytotoxicity assay against two human cancer cell lines, HL-60 and MCF-7, and a mouse fibroblast cell line, Balb/3T3. The free form of CLA exhibited the highest activity against all cancer cell lines. Results obtained for the Balb/3T3 line proved that phosphatidylcholine derivatives decreased the cytotoxic effect of CLA against healthy cell lines.
Collapse
|
233
|
Membrane-perturbing effect of fatty acids and lysolipids. Prog Lipid Res 2013; 52:130-40. [DOI: 10.1016/j.plipres.2012.09.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 08/20/2012] [Accepted: 09/13/2012] [Indexed: 12/13/2022]
|
234
|
Lafon C, Somaglino L, Bouchoux G, Mari JM, Chesnais S, Ngo J, Mestas JL, Fossheim SL, Nilssen EA, Chapelon JY. Feasibility study of cavitation-induced liposomal doxorubicin release in an AT2 Dunning rat tumor model. J Drug Target 2012; 20:691-702. [PMID: 22845841 DOI: 10.3109/1061186x.2012.712129] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Targeted and triggered release of liposomal drug using heat or ultrasound represents a promising treatment modality able to increase the therapeutic-totoxicity ratio of encapsulated drugs. PURPOSE To study the ability for high-intensity focused ultrasound to induce liposomal drug release mainly by focused inertial cavitation in vitro and in an animal model. METHODS A 1 MHz ultrasound setup has been developed for in vitro and in vivo drug release from a specific liposomal doxorubicin formulation at a target cavitation dose. RESULTS Controlled cavitation at 1 MHz was applied within the tumors 48 hours after liposome injection according to preliminary pharmacokinetic study. A small non-significant therapeutic effect of US-liposomal treatment was observed compared to liposomes alone suggesting no beneficial effect of ultrasound in the current setup. CONCLUSION The in vitro study provided a suitable ultrasound setup for delivering a cavitation dose appropriate for safe liposomal drug release. However, when converting to an in vivo model, no therapeutic benefit was observed. This may be due to a number of reasons, one of which may be the difficulty in converting in vitro findings to an in vivo model. In light of these findings, we discuss important design features for future studies.
Collapse
Affiliation(s)
- Cyril Lafon
- INSERM U1032, Université de Lyon, 151 Cours Albert Thomas, Lyon, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Franzen U, Østergaard J. Physico-chemical characterization of liposomes and drug substance–liposome interactions in pharmaceutics using capillary electrophoresis and electrokinetic chromatography. J Chromatogr A 2012; 1267:32-44. [DOI: 10.1016/j.chroma.2012.07.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/02/2012] [Accepted: 07/06/2012] [Indexed: 01/19/2023]
|
236
|
Liu C, Faller R. Conformational, dynamical. and tensional study of tethered bilayer lipid membranes in coarse-grained molecular simulations. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2012; 28:15907-15915. [PMID: 23092246 DOI: 10.1021/la303511p] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Tethered bilayer lipid membranes (tBLMs) have attracted great interest recently due to their crucial roles in elucidating fundamental membrane characteristics and the implications in biochemical sensors and pharmaceutical drug carriers. Nevertheless, they have not yet been investigated computationally on the molecular scale. Here, we study tBLMs consisting of DOPCs (1,2-dioleoyl-sn-glycero-3-phosphocholine) as free lipids and pegylated DOPCs (on phosphate group) as tethers in water by a variation of the MARTINI model. By varying grafting densities and tether lengths, distinct conformational changes from planar to undulated bilayers are observed. Lateral diffusivities and lateral pressure profiles show that the dynamical and tensional states are specific to the system configurations. These results suggest that the conformations, fluidity, and elasticity of the tBLMs can be tuned and manipulated to conform to various requirements in theoretical investigations and technological applications.
Collapse
Affiliation(s)
- Chueh Liu
- Department of Chemical Engineering and Materials Science, University of California-Davis, Davis, California 95616, United States
| | | |
Collapse
|
237
|
Sun H, Benjaminsen RV, Almdal K, Andresen TL. Hyaluronic Acid Immobilized Polyacrylamide Nanoparticle Sensors for CD44 Receptor Targeting and pH Measurement in Cells. Bioconjug Chem 2012; 23:2247-55. [DOI: 10.1021/bc300349n] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Honghao Sun
- Department
of Pharmacy, College
of Bioengineering, Hubei University of Technology, Wuhan 430068, China
| | | | | | | |
Collapse
|
238
|
Leung SJ, Romanowski M. Light-activated content release from liposomes. Am J Cancer Res 2012; 2:1020-36. [PMID: 23139729 PMCID: PMC3493200 DOI: 10.7150/thno.4847] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 10/01/2012] [Indexed: 12/31/2022] Open
Abstract
Successful integration of diagnostic and therapeutic actions at the level of individual cells requires new materials that combine biological compatibility with functional versatility. This review focuses on the development of liposome-based functional materials, where payload release is activated by light. Methods of sensitizing liposomes to light have progressed from the use of organic molecular moieties to the use of metallic plasmon resonant structures. This development has facilitated application of near infrared light for activation, which is preferred for its deep penetration and low phototoxicity in biological tissues. Presented mechanisms of light-activated liposomal content release enable precise in vitro manipulation of minute amounts of reagents, but their use in clinical diagnostic and therapeutic applications will require demonstration of safety and efficacy.
Collapse
|
239
|
Comparison of conventional chemotherapy, stealth liposomes and temperature-sensitive liposomes in a mathematical model. PLoS One 2012; 7:e47453. [PMID: 23082168 PMCID: PMC3474827 DOI: 10.1371/journal.pone.0047453] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 09/17/2012] [Indexed: 12/26/2022] Open
Abstract
Various liposomal drug carriers have been developed to overcome short plasma half-life and toxicity related side effects of chemotherapeutic agents. We developed a mathematical model to compare different liposome formulations of doxorubicin (DOX): conventional chemotherapy (Free-DOX), Stealth liposomes (Stealth-DOX), temperature sensitive liposomes (TSL) with intra-vascular triggered release (TSL-i), and TSL with extra-vascular triggered release (TSL-e). All formulations were administered as bolus at a dose of 9 mg/kg. For TSL, we assumed locally triggered release due to hyperthermia for 30 min. Drug concentrations were determined in systemic plasma, aggregate body tissue, cardiac tissue, tumor plasma, tumor interstitial space, and tumor cells. All compartments were assumed perfectly mixed, and represented by ordinary differential equations. Contribution of liposomal extravasation was negligible in the case of TSL-i, but was the major delivery mechanism for Stealth-DOX and for TSL-e. The dominant delivery mechanism for TSL-i was release within the tumor plasma compartment with subsequent tissue- and cell uptake of released DOX. Maximum intracellular tumor drug concentrations for Free-DOX, Stealth-DOX, TSL-i, and TSL-e were 3.4, 0.4, 100.6, and 15.9 µg/g, respectively. TSL-i and TSL-e allowed for high local tumor drug concentrations with reduced systemic exposure compared to Free-DOX. While Stealth-DOX resulted in high tumor tissue concentrations compared to Free-DOX, only a small fraction was bioavailable, resulting in little cellular uptake. Consistent with clinical data, Stealth-DOX resulted in similar tumor intracellular concentrations as Free-DOX, but with reduced systemic exposure. Optimal release time constants for maximum cellular uptake for Stealth-DOX, TSL-e, and TSL-i were 45 min, 11 min, and <3 s, respectively. Optimal release time constants were shorter for MDR cells, with ∼4 min for Stealth-DOX and for TSL-e. Tissue concentrations correlated well quantitatively with a prior in-vivo study. Mathematical models may thus allow optimization of drug delivery systems to achieve a better therapeutic index.
Collapse
|
240
|
Song H, Wang R, Xiao H, Cai H, Zhang W, Xie Z, Huang Y, Jing X, Liu T. A cross-linked polymeric micellar delivery system for cisplatin(IV) complex. Eur J Pharm Biopharm 2012; 83:63-75. [PMID: 23046872 DOI: 10.1016/j.ejpb.2012.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 09/09/2012] [Accepted: 09/10/2012] [Indexed: 10/27/2022]
Abstract
A polymeric cisplatin(IV) prodrug in the form of cross-linked micelles (M(Pt(IV)) was prepared by first constructing MPEG-b-PCL-b-PLL micelles and then attaching a cisplatin(IV) complex with two axial succinic moieties to the lysine residues of the carrier polymer in aqueous medium. The micelles obtained were characterized by TEM, DLS, and zeta potential measurement. Their in vitro release experiments were carried out at pH 7.4 and 5.0 or in the presence of 5mM sodium ascorbate (NaAsc). Results showed that the micelles were sensitive to both acidic hydrolysis and mild reducing agents; in the presence of 5mM NaAsc, cisplatin(II) was directly released and the released cisplatin(II) could chelate with nucleobases; the micelles displayed comparable cytotoxicities to cisplatin; and the micelles were much more efficiently internalized by the cells than cisplatin(II) and cisplatin(IV) counterparts. Moreover, in vivo study showed accumulation of more Pt species in the tumor site and lower systematic toxicity compared to free cisplatin(II) and cisplatin(IV). This polymeric prodrug of cisplatin is expected to be used more for future study and applications.
Collapse
Affiliation(s)
- Haiqin Song
- Department of Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Petersen AL, Hansen AE, Gabizon A, Andresen TL. Liposome imaging agents in personalized medicine. Adv Drug Deliv Rev 2012; 64:1417-35. [PMID: 22982406 DOI: 10.1016/j.addr.2012.09.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 09/02/2012] [Accepted: 09/03/2012] [Indexed: 12/20/2022]
Abstract
In recent years the importance of molecular and diagnostic imaging has increased dramatically in the treatment planning of many diseases and in particular in cancer therapy. Within nanomedicine there are particularly interesting possibilities for combining imaging and therapy. Engineered liposomes that selectively localize in tumor tissue can transport both drugs and imaging agents, which allows for a theranostic approach with great potential in personalized medicine. Radiolabeling of liposomes have for many years been used in preclinical studies for evaluating liposome in vivo performance and has been an important tool in the development of liposomal drugs. However, advanced imaging systems now provide new possibilities for non-invasive monitoring of liposome biodistribution in humans. Thus, advances in imaging and developments in liposome radiolabeling techniques allow us to enter a new arena where we start to consider how to use imaging for patient selection and treatment monitoring in connection to nanocarrier based medicines. Nanocarrier imaging agents could furthermore have interesting properties for disease diagnostics and staging. Here, we review the major advances in the development of radiolabeled liposomes for imaging as a tool in personalized medicine.
Collapse
Affiliation(s)
- Anncatrine L Petersen
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet 423, 2800 Lyngby, Denmark
| | | | | | | |
Collapse
|
242
|
Nguyen TTTN, Østergaard J, Stürup S, Gammelgaard B. Metallomics in drug development: characterization of a liposomal cisplatin drug formulation in human plasma by CE–ICP–MS. Anal Bioanal Chem 2012; 405:1845-54. [DOI: 10.1007/s00216-012-6355-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/06/2012] [Accepted: 08/13/2012] [Indexed: 10/27/2022]
|
243
|
Mendoza MF, Hollabaugh NM, Hettiarachchi SU, McCarley RL. Human NAD(P)H:quinone oxidoreductase type I (hNQO1) activation of quinone propionic acid trigger groups. Biochemistry 2012; 51:8014-26. [PMID: 22989153 DOI: 10.1021/bi300760u] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
NAD(P)H:quinone oxidoreductase type I (NQO1) is a target enzyme for triggered delivery of drugs at inflamed tissue and tumor sites, particularly those that challenge traditional therapies. Prodrugs, macromolecules, and molecular assemblies possessing trigger groups that can be cleaved by environmental stimuli are vehicles with the potential to yield active drug only at prescribed sites. Furthermore, quinone propionic acids (QPAs) covalently attached to prodrugs or liposome surfaces can be removed by application of a reductive trigger stimulus, such as that from NQO1; their rates of reductive activation should be tunable via QPA structure. We explored in detail the recombinant human NAD(P)H:quinone oxidoreductase type I (rhNQO1)-catalyzed NADH reduction of a family of substituted QPAs and obtained high precision kinetic parameters. It is found that small changes in QPA structure-in particular, single atom and function group substitutions on the quinone ring at R(1)-lead to significant impacts on the Michaelis constant (K(m)), maximum velocity (V(max)), catalytic constant (k(cat)), and catalytic efficiency (k(cat)/K(m)). Molecular docking simulations demonstrate that alterations in QPA structure result in large changes in QPA alignment and placement with respect to the flavin isoalloxazine ring in the active site of rhNQO1; a qualitative relationship exists between the kinetic parameters and the depth of QPA penetration into the rhNQO1 active site. From a quantitative perspective, a very good correlation is observed between log(k(cat)/K(m)) and the molecular-docking-derived distance between the flavin hydride donor site and quinone hydride acceptor site in the QPAs, an observation that is in agreement with developing theories. The comprehensive kinetic and molecular modeling knowledge obtained for the interaction of recombinant human NQO1 with the quinone propionic acid analogues provides insight into the design and implementation of the QPA trigger groups for drug delivery applications.
Collapse
Affiliation(s)
- Maria F Mendoza
- Department of Chemistry, Louisiana State University, 232 Choppin Hall, Baton Rouge, Louisiana 70803-1804, USA
| | | | | | | |
Collapse
|
244
|
Enhanced growth inhibition of osteosarcoma by cytotoxic polymerized liposomal nanoparticles targeting the alcam cell surface receptor. Sarcoma 2012; 2012:126906. [PMID: 23024593 PMCID: PMC3447386 DOI: 10.1155/2012/126906] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 07/03/2012] [Indexed: 11/17/2022] Open
Abstract
Osteosarcoma is the most common primary malignancy of bone in children, adolescents, and adults. Despite extensive surgery and adjuvant aggressive high-dose systemic chemotherapy with potentially severe bystander side effects, cure is attainable in about 70% of patients with localized disease and only 20%-30% of those patients with metastatic disease. Targeted therapies clearly are warranted in improving our treatment of this adolescent killer. However, a lack of osteosarcoma-associated/specific markers has hindered development of targeted therapeutics. We describe a novel osteosarcoma-associated cell surface antigen, ALCAM. We, then, create an engineered anti-ALCAM-hybrid polymerized liposomal nanoparticle immunoconjugate (α-AL-HPLN) to specifically target osteosarcoma cells and deliver a cytotoxic chemotherapeutic agent, doxorubicin. We have demonstrated that α-AL-HPLNs have significantly enhanced cytotoxicity over untargeted HPLNs and over a conventional liposomal doxorubicin formulation. In this way, α-AL-HPLNs are a promising new strategy to specifically deliver cytotoxic agents in osteosarcoma.
Collapse
|
245
|
de la Rica R, Aili D, Stevens MM. Enzyme-responsive nanoparticles for drug release and diagnostics. Adv Drug Deliv Rev 2012; 64:967-78. [PMID: 22266127 DOI: 10.1016/j.addr.2012.01.002] [Citation(s) in RCA: 474] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 01/05/2012] [Accepted: 01/09/2012] [Indexed: 12/20/2022]
Abstract
Enzymes are key components of the bionanotechnology toolbox that possess exceptional biorecognition capabilities and outstanding catalytic properties. When combined with the unique physical properties of nanomaterials, the resulting enzyme-responsive nanoparticles can be designed to perform functions efficiently and with high specificity for the triggering stimulus. This powerful concept has been successfully applied to the fabrication of drug delivery schemes where the tissue of interest is targeted via release of cargo triggered by the biocatalytic action of an enzyme. Moreover, the chemical transformation of the carrier by the enzyme can also generate therapeutic molecules, therefore paving the way to design multimodal nanomedicines with synergistic effects. Dysregulation of enzymatic activity has been observed in a number of severe pathological conditions, and this observation is useful not only to program drug delivery in vivo but also to fabricate ultrasensitive sensors for diagnosing these diseases. In this review, several enzyme-responsive nanomaterials such as polymer-based nanoparticles, liposomes, gold nanoparticles and quantum dots are introduced, and the modulation of their physicochemical properties by enzymatic activity emphasized. When known, toxicological issues related to the utilization nanomaterials are highlighted. Key examples of enzyme-responsive nanomaterials for drug delivery and diagnostics are presented, classified by the type of effector biomolecule, including hydrolases such as proteases, lipases and glycosidases, and oxidoreductases.
Collapse
|
246
|
Ikeuchi R, Iwasaki Y. High mineral affinity of polyphosphoester ionomer-phospholipid vesicles. J Biomed Mater Res A 2012; 101:318-25. [PMID: 22829566 DOI: 10.1002/jbm.a.34321] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 05/07/2012] [Accepted: 06/20/2012] [Indexed: 12/19/2022]
Abstract
Bone-specific drug delivery is important for the treatment of osteoporosis and osseous metastases. However, there have been limitations in the design of drug carriers having bone affinity. We synthesized amphiphilic polyphosphoester ionomers (CH-PHE) and modified them to 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) vesicles. The ζ-potential of the vesicles was decreased by immobilization of CH-PHE; the amount was influenced by the structure and fraction of CH-PHE. The release rate of 5-carboxyfluorescein from the vesicles could be controlled by changing the fraction of DOPC and CH-PHE. In particular, the release of CF from DOPC vesicles containing 3% CH-PHE was most reduced. In addition, the enzymatic degradation of DOPC was reduced by immobilization with polyphosphoester ionomers; enzyme tolerance was increased with an increase in the molar fraction of polyphosphoester ionomers. Hemolytic activity of the phospholipid vesicles bearing CH-PHE was infrequently observed and was similar to that of the DOPC vesicles. Although a decrease in the viability of mouse osteoblastic cells (MC3T3-E1) in contact with the vesicles bearing CH-PHE was observed when the DOPC concentration of the vesicles bearing 20 mol % CH-PHE with highly ionized units was greater than 200 μM, the cytotoxicity was diminished by sodium salt formation of the CH-PHE. The affinity of the vesicles to calcium deposits generated by MC3T3-E1 cells was significantly improved by the immobilization polyphosphoesters.
Collapse
Affiliation(s)
- Ryota Ikeuchi
- Department of Chemistry and Materials Engineering, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Osaka, Japan
| | | |
Collapse
|
247
|
pH-sensitive degradable chimaeric polymersomes for the intracellular release of doxorubicin hydrochloride. Biomaterials 2012; 33:7291-9. [PMID: 22795540 DOI: 10.1016/j.biomaterials.2012.06.034] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 06/22/2012] [Indexed: 11/21/2022]
Abstract
pH-sensitive degradable chimaeric polymersomes were developed based on asymmetric poly(ethylene glycol)-b-poly(2,4,6-trimethoxybenzylidene-1,1,1-tris(hydroxymethyl)ethane methacrylate)-b-poly(acrylic acid) (PEG-PTTMA-PAA) triblock copolymers for active loading as well as triggered intracellular release of hydrophilic doxorubicin hydrochloride (DOX·HCl). PEG-PTTMA-PAA copolymers were readily prepared with M(n PAA) ranging from 1.5, 2.1 to 2.7 kg/mol by sequential reversible addition-fragmentation chain transfer (RAFT) copolymerization of 2,4,6-trimethoxybenzylidene-1,1,1-tris(hydroxymethyl)ethane methacrylate (TTMA) and acrylic acid (AA) using PEG-CPADN (M(n PEG) = 5.0 kg/mol; CPADN: 4-cyanopentanoic acid dithionaphthalenoate) as a macro-RAFT agent. PEG-PTTMA-PAA copolymers formed mono-disperse polymersomes with average sizes of 63.9-112.1 nm, which decreased with increasing M(n PAA). The polymersomal structure was confirmed by transmission electron microscopy (TEM) and confocal laser scanning microscopy (CLSM). Notably, the acetals in polymersomes while sufficiently stable at pH 7.4 were prone to rapid hydrolysis at mildly acidic pHs of 4.0 and 5.0, which resulted in swelling and eventually disassembly of polymersomes. These chimaeric polymersomes could actively load DOX·HCl resulting in remarkably high drug loading contents (up to 15.9 wt.%) and loading efficiencies (up to 88.8%). The in vitro release studies showed that DOX·HCl was released from chimaeric polymersomes in a controlled and pH-dependent manner. CLSM observations revealed that these chimaeric polymersomes could efficiently deliver and release DOX·HCl into the nuclei of HeLa cells. MTT assays in HeLa cells demonstrated that DOX·HCl-loaded PEG-PTTMA-PAA polymersomes exhibited high anti-tumor activity with IC(50) (inhibitory concentration to produce 50% cell death) of 1.48-1.67 μg/mL, close to that of free DOX·HCl, while blank polymersomes were practically non-toxic up to a tested concentration of 2.0 mg/mL. These pH-sensitive degradable chimaeric polymersomes have appeared to be a promising alternative to liposomes for tumor-targeted delivery of DOX·HCl.
Collapse
|
248
|
Interaction of antitumoral fluorescent heteroaromatic compounds, a benzothienopyrrole and two thienoindoles, with DNA and lipid membranes. J Photochem Photobiol A Chem 2012. [DOI: 10.1016/j.jphotochem.2012.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
249
|
Pedersen PJ, Viart HMF, Melander F, Andresen TL, Madsen R, Clausen MH. Synthesis of tocopheryl succinate phospholipid conjugates and monitoring of phospholipase A2 activity. Bioorg Med Chem 2012; 20:3972-8. [DOI: 10.1016/j.bmc.2012.05.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Revised: 04/30/2012] [Accepted: 05/11/2012] [Indexed: 01/20/2023]
|
250
|
Chekhun VF, Demash DV, Nalieskina LA. Evaluation of biological effects and possible mechanisms of action of static magnetic field. ACTA ACUST UNITED AC 2012. [DOI: 10.15407/fz58.03.085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|