1
|
Shahsavari S, Rad MB, Hajiaghajani A, Rostami M, Hakimian F, Jafarzadeh S, Hasany M, Collingwood JF, Aliakbari F, Fouladiha H, Bardania H, Otzen DE, Morshedi D. Magnetoresponsive liposomes applications in nanomedicine: A comprehensive review. Biomed Pharmacother 2024; 181:117665. [PMID: 39541790 DOI: 10.1016/j.biopha.2024.117665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Safe and effective cancer therapy requires a suitable nanocarrier that can target particular sites, such as cancer cells, in a selective manner. With the tremendous growth in nanotechnology, liposomes, among various competing nanocarriers, have shown promising advances in cancer therapy. Magnetic nanoparticles and metal ions are wide-reaching candidates for conferring magnetic properties and for incorporation into liposomes. Combining liposomes with magnetic structures enables construction of magnetoresponsive liposomes, allowing stimuli-responsiveness to an alternating magnetic field, magnetic targeting, and tracking by magnetic resonance imaging, which could all occur in parallel. This review presents a comprehensive analysis of the practical advances and novel aspects of design, synthesis and engineering magnetoresponsive liposomes, emphasizing their diverse properties for various applications. Our work explores the innovative uses of these structures, extending beyond drug delivery to include smart contrast agents, cell labeling, biosensing, separation, and filtering. By comparing new findings with earlier studies, we showcase significant improvements in efficiency and uncover new potentials, setting a new benchmark for future research in the field of magnetoresponsive liposomes.
Collapse
Affiliation(s)
- Shayan Shahsavari
- Iran Nanotechnology Innovation Council, Nanoclub Elites Association, Tehran, Iran
| | - Mohammad Behnam Rad
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, Tehran, Iran
| | - Amirhossein Hajiaghajani
- School of Electrical Engineering, Iran University of Science and Technology, Tehran 1684613114, Iran
| | | | - Fatemeh Hakimian
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, Tehran, Iran
| | - Sina Jafarzadeh
- Department of Energy Conversion and Storage, Technical University of Denmark, Anker Engelunds Vej, Lyngby 2800 Kgs, Denmark
| | - Masoud Hasany
- Department of Civil and Mechanical Engineering, Technical University of Denmark, Lyngby 2800 Kgs, Denmark
| | | | - Farhang Aliakbari
- National Institute of Genetic Engineering and Biotechnology, Shahrak-e Pajoohesh, km 15 Tehran - Karaj Highway, P.O.Box:14965/161, Tehran, Iran; Molecular Medicine Research Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Hamideh Fouladiha
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Centre (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, Aarhus C DK-8000, Denmark.
| | - Dina Morshedi
- National Institute of Genetic Engineering and Biotechnology, Shahrak-e Pajoohesh, km 15 Tehran - Karaj Highway, P.O.Box:14965/161, Tehran, Iran.
| |
Collapse
|
2
|
Sun X, Tan A, Boyd BJ. Magnetically‐activated lipid nanocarriers in biomedical applications: A review of current status and perspective. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 15:e1863. [PMID: 36428234 DOI: 10.1002/wnan.1863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/27/2022] [Accepted: 09/03/2022] [Indexed: 11/28/2022]
Abstract
Magnetically-activated lipid nanocarriers have become a research hotspot in the field of biomedicine. Liposomes and other lipid-based carriers possess good biocompatibility as well as the ability to carrying therapeutic cargo with a range of physicochemical properties. Previous studies have demonstrated that magnetic materials have potential wide applications in clinical diagnosis and therapy, such as in MRI as contrast agents and in hyperthermic obliteration of cancer tissues. More recently magneto-thermal activation of lipid carriers to stimulate drug release has extended the range of further therapeutic benefits. Here, an overview of the current development of magnetically-activated lipid nanocarriers in the field of biomedicine is provided, including the methods of fabrication of the nanocarriers and their in vitro and in vivo performance. A discussion of the current barriers to translation of these materials as medicines is provided in the context of clinical and regulatory complexities of using magnetically responsive materials in therapeutic applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- Xiaohan Sun
- Drug Delivery, Disposition and Dynamics Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus) Parkville Victoria Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus) Parkville Victoria Australia
| | - Angel Tan
- Drug Delivery, Disposition and Dynamics Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus) Parkville Victoria Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus) Parkville Victoria Australia
| | - Ben J. Boyd
- Drug Delivery, Disposition and Dynamics Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus) Parkville Victoria Australia
- Department of Pharmacy University of Copenhagen Copenhagen Denmark
| |
Collapse
|
3
|
Toro-Córdova A, Llaguno-Munive M, Jurado R, Garcia-Lopez P. The Therapeutic Potential of Chemo/Thermotherapy with Magnetoliposomes for Cancer Treatment. Pharmaceutics 2022; 14:2443. [PMID: 36432634 PMCID: PMC9697689 DOI: 10.3390/pharmaceutics14112443] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer represents a very grave and quickly growing public health problem worldwide. Despite the breakthroughs in treatment and early detection of the disease, an increase is projected in the incidence rate and mortality during the next 30 years. Thus, it is important to develop new treatment strategies and diagnostic tools. One alternative is magnetic hyperthermia, a therapeutic approach that has shown promising results, both as monotherapy and in combination with chemo- and radiotherapy. However, there are still certain limitations and questions with respect to the safety of the systemic administration of magnetic nanoparticles. To deal with these issues, magnetoliposomes were conceived as a new generation of liposomes that incorporate superparamagnetic nanoparticles and oncological pharmaceuticals within their structure. They have the advantage of targeted and selective drug delivery to the diseased organs and tissues. Some of them can avoid the immune response of the host. When exposed to a magnetic field of alternating current, magnetoliposomes produce hyperthermia, which acts synergistically with the released drug. The aim of the present review is to describe the most recent advances in the use of magnetoliposomes and point out what research remains to be done for their application to chemo-thermal therapy in cancer patients.
Collapse
Affiliation(s)
- Alfonso Toro-Córdova
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
- Departamento de Formulación de Vacunas de mRNA, CerTest Biotec S.L., 50840 Zaragoza, Spain
| | - Monserrat Llaguno-Munive
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
- Laboratorio de Física Médica, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| | - Rafael Jurado
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| | - Patricia Garcia-Lopez
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| |
Collapse
|
4
|
Mehta N, Pai R. Amalgamation of Nanoparticles within Drug Carriers: A Synergistic Approach or a Futile Attempt? Pharm Nanotechnol 2022; 10:PNT-EPUB-126127. [PMID: 36056844 DOI: 10.2174/2211738510666220902150449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/25/2022] [Accepted: 05/03/2022] [Indexed: 11/22/2022]
Abstract
In recent years, nanotechnology has gained much attention from scientists and significant advances in therapeutic potential. Nano-delivery systems have emerged as an effective way in order to improve the therapeutic properties of drugs including solubility, stability, prolongation of half-life as well as promoting the accumulation of drug at the target site. The nanoparticles have also been incorporated into various conventional drug delivery systems. This review study aims to introduce the amalgamation of nanoparticles into drug carriers. To overcome the limitations of single nanoparticles such as toxicity, high instability, rapid drug release as well as limited drug loading capacity, a multi-component system is developed. Liposomes, microparticles, nanofibers, dendrimers etc., are promising drug carriers, having some limitations that can be minimized, and the compilation of nanoparticles synergizes the properties. The amalgamated nanocarriers are used for the diagnostic purpose as well as treatment of various chronic diseases. It also increases the solubility of hydrophobic drugs. However, each system has its advantages and disadvantages based on its physicochemical properties, efficacy, and other parameters. This review details the past and present state of development for the fusion of nanoparticles within drug carriers and from which we identify future research works needed for the same.
Collapse
Affiliation(s)
- Nikhil Mehta
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM\\\'s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai- 400056, India
| | - Rohan Pai
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM\\\'s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai- 400056, India
| |
Collapse
|
5
|
Shilova EV, Koltakov IA, Kannykin SV, Artyukhov VG. Inclusion of Magnetite Nanoparticles Stabilized with Cetyltrimethylammonium Bromide in Soy Lecithin-Based Liposomes. Biophysics (Nagoya-shi) 2022. [DOI: 10.1134/s0006350922030216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
6
|
Mahmoud K, Swidan S, El-Nabarawi M, Teaima M. Lipid based nanoparticles as a novel treatment modality for hepatocellular carcinoma: a comprehensive review on targeting and recent advances. J Nanobiotechnology 2022; 20:109. [PMID: 35248080 PMCID: PMC8898455 DOI: 10.1186/s12951-022-01309-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022] Open
Abstract
Liver cancer is considered one of the deadliest diseases with one of the highest disease burdens worldwide. Among the different types of liver cancer, hepatocellular carcinoma is considered to be the most common type. Multiple conventional approaches are being used in treating hepatocellular carcinoma. Focusing on drug treatment, regular agents in conventional forms fail to achieve the intended clinical outcomes. In order to improve the treatment outcomes, utilizing nanoparticles-specifically lipid based nanoparticles-are considered to be one of the most promising approaches being set in motion. Multiple forms of lipid based nanoparticles exist including liposomes, solid lipid nanoparticles, nanostructured lipid carriers, microemulsion, nanoemulsion, phytosomes, lipid coated nanoparticles, and nanoassemblies. Multiple approaches are used to enhance the tumor uptake as well tumor specificity such as intratumoral injection, passive targeting, active targeting, and stimuli responsive nanoparticles. In this review, the effect of utilizing lipidic nanoparticles is being discussed as well as the different tumor uptake enhancement techniques used.
Collapse
Affiliation(s)
- Khaled Mahmoud
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt
| | - Shady Swidan
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt.
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt.
| | - Mohamed El-Nabarawi
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Mahmoud Teaima
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
7
|
Fortes Brollo ME, Domínguez-Bajo A, Tabero A, Domínguez-Arca V, Gisbert V, Prieto G, Johansson C, Garcia R, Villanueva A, Serrano MC, Morales MDP. Combined Magnetoliposome Formation and Drug Loading in One Step for Efficient Alternating Current-Magnetic Field Remote-Controlled Drug Release. ACS APPLIED MATERIALS & INTERFACES 2020; 12:4295-4307. [PMID: 31904927 DOI: 10.1021/acsami.9b20603] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We have developed a reproducible and facile one step strategy for the synthesis of doxorubicin loaded magnetoliposomes by using a thin-layer evaporation method. Liposomes of around 200 nm were made of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and iron oxide nanoparticles (NPs) with negative, positive, and hydrophobic surfaces that were incorporated outside, inside, or between the lipid bilayers, respectively. To characterize how NPs are incorporated in liposomes, advanced cryoTEM and atomic force microscope (AFM) techniques have been used. It was observed that only when the NPs are attached outside the liposomes, the membrane integrity is preserved (lipid melt transition shifts to 38.7 °C with high enthalpy 34.8 J/g) avoiding the leakage of the encapsulated drug while having good colloidal properties and the best heating efficiency under an alternating magnetic field (AMF). These magnetoliposomes were tested with two cancer cell lines, MDA-MB-231 and HeLa cells. First, 100% of cellular uptake was achieved with a high cell survival (above 80%), which is preserved (83%) for doxorubicin-loaded magnetoliposomes. Then, we demonstrate that doxorubicin release can be triggered by remote control, using a noninvasive external AMF for 1 h, leading to a cell survival reduction of 20%. Magnetic field conditions of 202 kHz and 30 mT seem to be enough to produce an effective heating to avoid drug degradation. In conclusion, these drug-loaded magnetoliposomes prepared in one step could be used for drug release on demand at a specific time and place, efficiently using an external AMF to reduce or even eliminate side effects.
Collapse
Affiliation(s)
- Maria Eugenia Fortes Brollo
- Departamento de Energia, Medio Ambiente y Salud , Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Cientı́ficas , Madrid 28049 , Spain
| | - Ana Domínguez-Bajo
- Departamento de Energia, Medio Ambiente y Salud , Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Cientı́ficas , Madrid 28049 , Spain
| | - Andrea Tabero
- Departamento de Biología , Universidad Autónoma de Madrid , Madrid 28049 Spain
| | - Vicente Domínguez-Arca
- Departamento de Física Aplicada , Universidad de Santiago de Compostela , Santiago de Compostela 15782 Spain
| | - Victor Gisbert
- Departamento de Energia, Medio Ambiente y Salud , Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Cientı́ficas , Madrid 28049 , Spain
| | - Gerardo Prieto
- Departamento de Física Aplicada , Universidad de Santiago de Compostela , Santiago de Compostela 15782 Spain
| | | | - Ricardo Garcia
- Departamento de Energia, Medio Ambiente y Salud , Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Cientı́ficas , Madrid 28049 , Spain
| | - Angeles Villanueva
- Departamento de Biología , Universidad Autónoma de Madrid , Madrid 28049 Spain
- IMDEA-Nanociencia , Madrid 28049 Spain
| | - María Concepción Serrano
- Departamento de Energia, Medio Ambiente y Salud , Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Cientı́ficas , Madrid 28049 , Spain
| | - María Del Puerto Morales
- Departamento de Energia, Medio Ambiente y Salud , Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Cientı́ficas , Madrid 28049 , Spain
| |
Collapse
|
8
|
Vlasova KY, Piroyan A, Le-Deygen IM, Vishwasrao HM, Ramsey JD, Klyachko NL, Golovin YI, Rudakovskaya PG, Kireev II, Kabanov AV, Sokolsky-Papkov M. Magnetic liposome design for drug release systems responsive to super-low frequency alternating current magnetic field (AC MF). J Colloid Interface Sci 2019; 552:689-700. [PMID: 31176052 PMCID: PMC7012191 DOI: 10.1016/j.jcis.2019.05.071] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/30/2019] [Accepted: 05/22/2019] [Indexed: 02/02/2023]
Abstract
HYPOTHESIS Magnetic liposomes are shown to release the entrapped dye once modulated by low frequency AC MF. The mechanism and effectiveness of MF application should depend on lipid composition, magnetic nanoparticles (MNPs) properties, temperature and field parameters. EXPERIMENTS The study was performed using liposomes of various lipid composition and embedded hydrophobic MNPs. The liposomes structural changes were studied by the transmission electron microscopy (TEM) and attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopy and the leakage was monitored by the fluorescent dye release. FINDINGS Magnetic liposomes exposure to the AC MF resulted in the clustering of the MNPs in the membranes and disruption of the lipid packaging. Addition of cholesterol diminished the dye release from the saturated lipid-based liposomes. Replacement of the saturated lipid for unsaturated one also decreased the dye release. The dye release depended on the strength, but not the frequency of the field. Thus, the oscillating motion of MNPs in AC MF ruptures the gel phase membranes of saturated lipids. As the temperature increases the disruption also increases. In the liquid crystalline membranes formed by unsaturated lipids the deformations and defects created by mechanical motion of the MNPs are more likely to heal and results in decreased release.
Collapse
Affiliation(s)
- Kseniya Yu Vlasova
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Alexander Piroyan
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Irina M Le-Deygen
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Hemant M Vishwasrao
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Jacob D Ramsey
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Natalia L Klyachko
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, Lomonosov Moscow State University, Moscow 119992, Russia; Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; G.R. Derzhavin Tambov State University, Tambov 392000, Russia.
| | - Yuri I Golovin
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, Lomonosov Moscow State University, Moscow 119992, Russia; G.R. Derzhavin Tambov State University, Tambov 392000, Russia
| | - Polina G Rudakovskaya
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Igor I Kireev
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Alexander V Kabanov
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, Lomonosov Moscow State University, Moscow 119992, Russia; Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA.
| |
Collapse
|
9
|
Thermosensitive hydrogels for sustained-release of sorafenib and selenium nanoparticles for localized synergistic chemoradiotherapy. Biomaterials 2019; 216:119220. [DOI: 10.1016/j.biomaterials.2019.05.031] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/13/2019] [Accepted: 05/18/2019] [Indexed: 12/17/2022]
|
10
|
Liu JF, Jang B, Issadore D, Tsourkas A. Use of magnetic fields and nanoparticles to trigger drug release and improve tumor targeting. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1571. [PMID: 31241251 DOI: 10.1002/wnan.1571] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/29/2019] [Accepted: 05/31/2019] [Indexed: 12/21/2022]
Abstract
Drug delivery strategies aim to maximize a drug's therapeutic index by increasing the concentration of drug at target sites while minimizing delivery to off-target tissues. Because biological tissues are minimally responsive to magnetic fields, there has been a great deal of interest in using magnetic nanoparticles in combination with applied magnetic fields to selectively control the accumulation and release of drug in target tissues while minimizing the impact on surrounding tissue. In particular, spatially variant magnetic fields have been used to encourage accumulation of drug-loaded magnetic nanoparticles at target sites, while time-variant magnetic fields have been used to induce drug release from thermally sensitive nanocarriers. In this review, we discuss nanoparticle formulations and approaches that have been developed for magnetic targeting and/or magnetically induced drug release, as well as ongoing challenges in using magnetism for therapeutic applications. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Jessica F Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bian Jang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Issadore
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Wang Q, Zhong Y, Liu W, Wang Z, Gu L, Li X, Zheng J, Du H, Zhong Z, Xie F. Enhanced chemotherapeutic efficacy of the low-dose doxorubicin in breast cancer via nanoparticle delivery system crosslinked hyaluronic acid. Drug Deliv 2019; 26:12-22. [PMID: 30691317 PMCID: PMC6352940 DOI: 10.1080/10717544.2018.1507057] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite the development of treatment options in breast cancer, many patients die of recurrence and metastasis. Owing to enhanced permeability and retention in solid tumor tissue, nanoparticle (NP) delivery systems have been emerged as novel strategy in cancer chemotherapy. As extracellular matrix, glycosaminoglycan hyaluronan (HA) could bind its surface receptor adhesion molecule CD44 which is strongly expressed on breast cancer. We have previously reported a doxorubicin (DOX)-loaded HA-Lys-LA X-NPs (X-NP-DOX) NP delivery system for breast cancer treatment. In this study, we further investigated the antitumor effect of X-NP-DOX NP delivery system using low-dose DOX in both in vitro and in vivo systems. We demonstrated that low-dose X-NP-DOX possessed the ability for inhibiting MCF-7 breast cancer cell growth, invasion, and migration, and inducing apoptosis in vitro. In in vivo experiments, injection of low-dose X-NP-DOX into tumor-bearing mouse resulted in significant reduction of tumor size. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining further revealed that low-dose X-NP-DOX induced higher percentage of apoptotic cells compared with free DOX or saline. Furthermore, our study demonstrated that low-dose X-NP-DOX inhibited Notch1 and Ras/MAPK pathways, decreased cancer stem cell population, and reduced tumorigenesis compared to free DOX in both in vitro and in vivo settings. Owing to its enhanced efficacy and higher targetability compared to free DOX, low-dose DOX delivered by NP system may be a promising novel strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Qin Wang
- a Department of Pathology, Institutes of Biology and Medical Sciences , Soochow University Medical College, Soochow University , Suzhou P. R. China.,b Department of Immunology, Institutes of Biology and Medical Sciences , Soochow University Medical College, Soochow University , Suzhou , P. R. China
| | - Yinan Zhong
- c Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , Suzhou P. R. China
| | - Wenting Liu
- a Department of Pathology, Institutes of Biology and Medical Sciences , Soochow University Medical College, Soochow University , Suzhou P. R. China.,d Department of Pathology , The Frist Affiliated Hospital of Soochow University , Suzhou , P.R. China
| | - Zemin Wang
- e Investigative Toxicology and Pathology Laboratory, School of Public Health , Indiana University , Bloomington , IN , USA
| | - Liqin Gu
- f Department of Pathology , Taicang Traditional Medicine Hospital of Jiangsu Province , Taicang , P.R. China
| | - Xuejiao Li
- a Department of Pathology, Institutes of Biology and Medical Sciences , Soochow University Medical College, Soochow University , Suzhou P. R. China
| | - Jiqing Zheng
- a Department of Pathology, Institutes of Biology and Medical Sciences , Soochow University Medical College, Soochow University , Suzhou P. R. China
| | - Huan Du
- a Department of Pathology, Institutes of Biology and Medical Sciences , Soochow University Medical College, Soochow University , Suzhou P. R. China
| | - Zhiyuan Zhong
- c Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , Suzhou P. R. China
| | - Fang Xie
- a Department of Pathology, Institutes of Biology and Medical Sciences , Soochow University Medical College, Soochow University , Suzhou P. R. China
| |
Collapse
|
12
|
Boruah JS, Chowdhury D. Hybrid Oleic Acid‐Graphene Quantum Dot Vesicles for Drug Delivery. ChemistrySelect 2019. [DOI: 10.1002/slct.201803619] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Jayanta S. Boruah
- Material Nanochemistry LaboratoryPhysical Sciences DivisionInstitute of Advanced Study in Science and Technology, PaschimBoragaon, Garchuk Guwahati-781035 India
| | - Devasish Chowdhury
- Material Nanochemistry LaboratoryPhysical Sciences DivisionInstitute of Advanced Study in Science and Technology, PaschimBoragaon, Garchuk Guwahati-781035 India
| |
Collapse
|
13
|
Amiri M, Salavati-Niasari M, Akbari A. Magnetic nanocarriers: Evolution of spinel ferrites for medical applications. Adv Colloid Interface Sci 2019; 265:29-44. [PMID: 30711796 DOI: 10.1016/j.cis.2019.01.003] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 01/30/2023]
Abstract
A valuable site-directed application in the field of nanomedicine is targeted drug delivery using magnetic metal oxide nanoparticles by applying an external magnetic field at the target tissue. The magnetic property of these structures allows controlling the orientation and location of particles by changing the direction of the applied external magnetic field. Pharmaceutical design and research in the field of nanotechnology offer novel solutions for diagnosis and therapies. This review summarizes magnetic nanoparticles and magnetic spinel ferrit's properties, remarkable approaches in magnetic liposomes, magnetic polymeric nanoparticles, MRI, hyperthermia and especially magnetic drug delivery systems, which have recently developed in the field of magnetic nanoparticles and their medicinal applications. Here, we discuss spinel ferrite (SF) as magnetic materials that are a significant class of composite metal oxides. They contain ferric ions and have the general structural formula M2+Fe23+O4 (where M = Co,Ni,Zn,etc.). This structure indicates unique multifunctional properties, such as excellent magnetic characteristics, high specific surface area, surface active sites, high chemical stability, tuneable shape and size, and options for functionalization. The review assesses the current efforts on synthesis, properties and medical application of magnetic spinel ferrites nanoparticles based on cobalt, nickel and zinc. Based on this review, it can be concluded that MNPs and SFNPs have unlimited ability in biomedical applications. However, the practical application of SFNPs on a huge scale still needs to be considered and evaluated.
Collapse
|
14
|
Triggering antitumoural drug release and gene expression by magnetic hyperthermia. Adv Drug Deliv Rev 2019; 138:326-343. [PMID: 30339825 DOI: 10.1016/j.addr.2018.10.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 09/06/2018] [Accepted: 10/08/2018] [Indexed: 01/08/2023]
Abstract
Magnetic nanoparticles (MNPs) are promising tools for a wide array of biomedical applications. One of their most outstanding properties is the ability to generate heat when exposed to alternating magnetic fields, usually exploited in magnetic hyperthermia therapy of cancer. In this contribution, we provide a critical review of the use of MNPs and magnetic hyperthermia as drug release and gene expression triggers for cancer therapy. Several strategies for the release of chemotherapeutic drugs from thermo-responsive matrices are discussed, providing representative examples of their application at different levels (from proof of concept to in vivo applications). The potential of magnetic hyperthermia to promote in situ expression of therapeutic genes using vectors that contain heat-responsive promoters is also reviewed in the context of cancer gene therapy.
Collapse
|
15
|
Liu JF, Neel N, Dang P, Lamb M, McKenna J, Rodgers L, Litt B, Cheng Z, Tsourkas A, Issadore D. Radiofrequency-Triggered Drug Release from Nanoliposomes with Millimeter-Scale Resolution Using a Superimposed Static Gating Field. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802563. [PMID: 30286280 PMCID: PMC6397654 DOI: 10.1002/smll.201802563] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/27/2018] [Indexed: 05/17/2023]
Abstract
Drug delivery to a specific site in the body typically relies on the use of targeting agents that recognize a unique biomarker. Unfortunately, it is often difficult to identify unique molecular signatures that exist only at the site of interest. An alternative strategy is to deliver energy (e.g., light) to locally trigger release from a drug carrier; however, the use of this approach is limited because energy delivery to deep tissues is often impractical or invasive. In this work, radiofrequency-responsive superparamagnetic iron oxide nanoparticles (SPIONs) are used to trigger drug release from nanoscale vesicles. Because the body is inherently nonmagnetic, this approach allows for deep tissue targeting. To overcome the unfavorable meter-scale diffraction limit of SPION-compatible radiofrequency (RF) fields, a strong static gating field containing a sharp zero point is superimposed on the RF field. Only drug carriers that are at or near the zero point are susceptible to RF-triggered drug release, thereby localizing drug delivery with millimeter-scale resolution. This approach induces >40% drug release from thermally responsive doxorubicin-loaded liposomes within a 3.2 mm radius of the zero point with <10% release in the surrounding area, leading to a >2.5 therapeutic index in Huh 7 hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Jessica F Liu
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - Nishant Neel
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - Phillip Dang
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - Max Lamb
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - Jaime McKenna
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - Lauren Rodgers
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - Brian Litt
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| | - David Issadore
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St., Philadelphia, PA, 19104, USA
| |
Collapse
|
16
|
Toro-Cordova A, Flores-Cruz M, Santoyo-Salazar J, Carrillo-Nava E, Jurado R, Figueroa-Rodriguez PA, Lopez-Sanchez P, Medina LA, Garcia-Lopez P. Liposomes Loaded with Cisplatin and Magnetic Nanoparticles: Physicochemical Characterization, Pharmacokinetics, and In-Vitro Efficacy. Molecules 2018; 23:molecules23092272. [PMID: 30200551 PMCID: PMC6225157 DOI: 10.3390/molecules23092272] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 02/01/2023] Open
Abstract
With the aim improving drug delivery, liposomes have been employed as carriers for chemotherapeutics achieving promising results; their co-encapsulation with magnetic nanoparticles is evaluated in this work. The objective of this study was to examine the physicochemical characteristics, the pharmacokinetic behaviour, and the efficacy of pegylated liposomes loaded with cisplatin and magnetic nanoparticles (magnetite) (Cis-MLs). Cis-MLs were prepared by a modified reverse-phase evaporation method. To characterize their physicochemical properties, an evaluation was made of particle size, ζ-potential, phospholipid and cholesterol concentration, phase transition temperature (Tm), the encapsulation efficiency of cisplatin and magnetite, and drug release profiles. Additionally, pharmacokinetic studies were conducted on normal Wistar rats, while apoptosis and the cytotoxic effect were assessed with HeLa cells. We present a method for simultaneously encapsulating cisplatin at the core and also embedding magnetite nanoparticles on the membrane of liposomes with a mean vesicular size of 104.4 ± 11.5 nm and a ζ-potential of −40.5 ± 0.8 mV, affording a stable formulation with a safe pharmacokinetic profile. These liposomes elicited a significant effect on cell viability and triggered apoptosis in HeLa cells.
Collapse
Affiliation(s)
- Alfonso Toro-Cordova
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, 14080 CDMX, Mexico.
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, 11340 CDMX, Mexico.
| | - Mario Flores-Cruz
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, 14080 CDMX, Mexico.
| | - Jaime Santoyo-Salazar
- Departamento de Física, Centro de Investigacion y de Estudios Avanzados del Instituto Politécnico Nacional, CINVESTAV-IPN, Zacatenco, 07360 CDMX, Mexico.
| | - Ernesto Carrillo-Nava
- Laboratorio de Biofisicoquímica, Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, 014510 CDMX, Mexico.
| | - Rafael Jurado
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, 14080 CDMX, Mexico.
| | - Pavel A Figueroa-Rodriguez
- Unidad de Investigación Biomédica en Cáncer INCan-UNAM, Instituto Nacional de Cancerología, 14080 CDMX, Mexico.
| | - Pedro Lopez-Sanchez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, 11340 CDMX, Mexico.
| | - Luis A Medina
- Unidad de Investigación Biomédica en Cáncer INCan-UNAM, Instituto Nacional de Cancerología, 14080 CDMX, Mexico.
- Instituto de Física, Universidad Nacional Autónoma de México, 04510 CDMX, Mexico.
| | - Patricia Garcia-Lopez
- Laboratorio de Farmacología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, 14080 CDMX, Mexico.
| |
Collapse
|
17
|
Magnetic field triggered drug release from lipid microcapsule containing lipid-coated magnetic nanoparticles. Chem Phys Lett 2018. [DOI: 10.1016/j.cplett.2018.06.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
18
|
Guisasola E, Baeza A, Vallet M. Magnetically-responsive DDS. STIMULI-RESPONSIVE DRUG DELIVERY SYSTEMS 2018. [DOI: 10.1039/9781788013536-00145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Magnetic-responsive drug delivery systems have received great attention due to the possibility of building theranostic systems. The application of a non-invasive external stimuli as a magnetic field that also allows the imaging and localization of the devices and the release of therapeutic drugs means a great opportunity for the development of new treatments to prevent diseases such as cancer. This chapter will focus on smart materials based on magnetic nanoparticles that have been studied for the formulation of such delivery systems and their synergic effect in combination with drugs for potential applications in the biomedical field. In addition, the possibility of applying hyperthermia at the macro and nanoscale levels and their implications will be discussed.
Collapse
Affiliation(s)
- E. Guisasola
- Dpto. Química en Ciencias Farmacéuticas, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Universidad Complutense de Madrid, Plaza Ramon y Cajal s/n and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Av. Monforte de Lemos, 3-5, Pabellón 11, Planta 0 28029 Madrid Spain
| | - A. Baeza
- Dpto. Química en Ciencias Farmacéuticas, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Universidad Complutense de Madrid, Plaza Ramon y Cajal s/n and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Av. Monforte de Lemos, 3-5, Pabellón 11, Planta 0 28029 Madrid Spain
| | - M. Vallet
- Dpto. Química en Ciencias Farmacéuticas, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Universidad Complutense de Madrid, Plaza Ramon y Cajal s/n and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Av. Monforte de Lemos, 3-5, Pabellón 11, Planta 0 28029 Madrid Spain
| |
Collapse
|
19
|
Xie W, Guo Z, Gao F, Gao Q, Wang D, Liaw BS, Cai Q, Sun X, Wang X, Zhao L. Shape-, size- and structure-controlled synthesis and biocompatibility of iron oxide nanoparticles for magnetic theranostics. Theranostics 2018; 8:3284-3307. [PMID: 29930730 PMCID: PMC6010979 DOI: 10.7150/thno.25220] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/29/2018] [Indexed: 12/23/2022] Open
Abstract
In the past decade, iron oxide nanoparticles (IONPs) have attracted more and more attention for their excellent physicochemical properties and promising biomedical applications. In this review, we summarize and highlight recent progress in the design, synthesis, biocompatibility evaluation and magnetic theranostic applications of IONPs, with a special focus on cancer treatment. Firstly, we provide an overview of the controlling synthesis strategies for fabricating zero-, one- and three-dimensional IONPs with different shapes, sizes and structures. Then, the in vitro and in vivo biocompatibility evaluation and biotranslocation of IONPs are discussed in relation to their chemo-physical properties including particle size, surface properties, shape and structure. Finally, we also highlight significant achievements in magnetic theranostic applications including magnetic resonance imaging (MRI), magnetic hyperthermia and targeted drug delivery. This review provides a background on the controlled synthesis, biocompatibility evaluation and applications of IONPs as cancer theranostic agents and an overview of the most up-to-date developments in this area.
Collapse
Affiliation(s)
- Wensheng Xie
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
- Advanced Materials of Ministry of Education of China, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
| | - Zhenhu Guo
- School of Earth Sciences and Resources, China University of Geosciences, Beijing, 10083, China
| | - Fei Gao
- College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi 710069, China
| | - Qin Gao
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
- Advanced Materials of Ministry of Education of China, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
| | - Dan Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
- Advanced Materials of Ministry of Education of China, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
| | - Bor-shuang Liaw
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
- Advanced Materials of Ministry of Education of China, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
| | - Qiang Cai
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
- Advanced Materials of Ministry of Education of China, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
- Advanced Materials of Ministry of Education of China, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
- Advanced Materials of Ministry of Education of China, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
- Advanced Materials of Ministry of Education of China, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
20
|
Millart E, Lesieur S, Faivre V. Superparamagnetic lipid-based hybrid nanosystems for drug delivery. Expert Opin Drug Deliv 2018. [DOI: 10.1080/17425247.2018.1453804] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- E. Millart
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - S. Lesieur
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - V. Faivre
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
21
|
Zheng XC, Ren W, Zhang S, Zhong T, Duan XC, Yin YF, Xu MQ, Hao YL, Li ZT, Li H, Liu M, Li ZY, Zhang X. The theranostic efficiency of tumor-specific, pH-responsive, peptide-modified, liposome-containing paclitaxel and superparamagnetic iron oxide nanoparticles. Int J Nanomedicine 2018; 13:1495-1504. [PMID: 29559778 PMCID: PMC5856286 DOI: 10.2147/ijn.s157082] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background In the present study, the tumor-specific, pH-responsive peptide H7K(R2)2-modified, theranostic liposome-containing paclitaxel (PTX) and superparamagnetic iron oxide nanoparticles (SPIO NPs), PTX/SPIO-SSL-H7K(R2)2, was prepared by using H7K(R2)2 as the targeting ligand, SPIO NPs as the magnetic resonance imaging (MRI) agent, PTX as antitumor drug. Methods The PTX/SPIO-SSL-H7K(R2)2 was prepared by a thin film hydration method. The characteristics of PTX/SPIO-SSL-H7K(R2)2 were evaluated. The targeting effect, MRI, and antitumor activity of PTX/SPIO-SSL-H7K(R2)2 were investigated detail in vitro and in vivo in human breast carcinoma MDA-MB-231 cell models. Results Our results of in vitro flow cytometry, in vivo imaging, and in vivo MR imaging confirmed the pH-responsive characteristic of H7K(R2)2 in MDA-MB-231 cell line in vitro and in vivo. The results of in vivo MRI and in vivo antitumor activity confirmed the theranostic effect of PTX/SPIO-SSL-H7K(R2)2 in MDA-MB-231 tumor-bearing model. Conclusion Considering all our in vitro and in vivo results, we conclude that we developed targeting modified theranostic liposome which could achieve both role of antitumor and MRI.
Collapse
Affiliation(s)
- Xiu-Chai Zheng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Wei Ren
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Shuang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Ting Zhong
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Xiao-Chuan Duan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Yi-Fan Yin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Mei-Qi Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Yan-Li Hao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Zhan-Tao Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Hui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Man Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Zhuo-Yue Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Xuan Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| |
Collapse
|
22
|
Belyanina I, Kolovskaya O, Zamay S, Gargaun A, Zamay T, Kichkailo A. Targeted Magnetic Nanotheranostics of Cancer. Molecules 2017; 22:E975. [PMID: 28604617 PMCID: PMC6152710 DOI: 10.3390/molecules22060975] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/31/2022] Open
Abstract
Current advances in targeted magnetic nanotheranostics are summarized in this review. Unique structural, optical, electronic and thermal properties of magnetic materials in nanometer scale are attractive in the field of biomedicine. Magnetic nanoparticles functionalized with therapeutic molecules, ligands for targeted delivery, fluorescent and other chemical agents can be used for cancer diagnostic and therapeutic purposes. High selectivity, small size, and low immunogenicity of synthetic nucleic acid aptamers make them attractive delivery agents for therapeutic purposes. Properties, production and functionalization of magnetic nanoparticles and aptamers as ligands for targeted delivery are discussed herein. In recent years, magnetic nanoparticles have been widely used in diagnostic methods, such as scintigraphy, single photon emission computed tomography (SPECT), positron emission tomography (PET), magnetic resonance imaging (MRI), and Raman spectroscopy. Therapeutic purposes of magnetic nanoconstructions are also promising. They are used for effective drug delivery, magnetic mediated hypertermia, and megnetodynamic triggering of apoptosis. Thus, magnetic nanotheranostics opens a new venue for complex differential diagnostics, and therapy of metastatic cancer.
Collapse
Affiliation(s)
- Irina Belyanina
- Krasnoyarsk State Medical University named after prof. V.F. Voino-Yaseneckii, 660022 Krasnoyarsk, Russia.
| | - Olga Kolovskaya
- Krasnoyarsk State Medical University named after prof. V.F. Voino-Yaseneckii, 660022 Krasnoyarsk, Russia.
- Federal Research Center, KSC Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia.
| | - Sergey Zamay
- Federal Research Center, KSC Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia.
| | - Ana Gargaun
- Independent Researcher Vancouver, Vancouver, BC V6K 1C4, Canada.
| | - Tatiana Zamay
- Krasnoyarsk State Medical University named after prof. V.F. Voino-Yaseneckii, 660022 Krasnoyarsk, Russia.
- Federal Research Center, KSC Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia.
| | - Anna Kichkailo
- Krasnoyarsk State Medical University named after prof. V.F. Voino-Yaseneckii, 660022 Krasnoyarsk, Russia.
- Federal Research Center, KSC Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia.
| |
Collapse
|
23
|
Guo Y, Zhang Y, Ma J, Li Q, Li Y, Zhou X, Zhao D, Song H, Chen Q, Zhu X. Light/magnetic hyperthermia triggered drug released from multi-functional thermo-sensitive magnetoliposomes for precise cancer synergetic theranostics. J Control Release 2017; 272:145-158. [PMID: 28442407 DOI: 10.1016/j.jconrel.2017.04.028] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/06/2017] [Accepted: 04/13/2017] [Indexed: 12/13/2022]
Abstract
Precise delivery of antineoplastic drugs to specific tumor region has drawn much attention in recent years. Herein, a light/magnetic hyperthermia triggered drug delivery with multiple functionality is designed based on methotrexate (MTX) modified thermo-sensitive magnetoliposomes (MTX-MagTSLs). In this system, MTX and oleic acid modified magnetic nanoparticles (MNPs) can be applied in biological and magnetic targeting. Meanwhile, lipophilic fluorescent dye Cy5.5 and MNPs are encapsulated into the bilayer of liposomes, which can not only achieve dual-imaging effect to verify the MTX-MagTSLs accumulation in tumor region, but also provide an appropriate laser irradiation region to release Doxorubicin (Dox) under alternating magnetic field (AMF). Both in vitro and in vivo results revealed that MTX-MagTSLs possessed an excellent targeting ability towards HeLa cells and HeLa tumor-bearing mice. Furthermore, the heating effect of MTX-MagTSLs was amplified 4.2-fold upon combination with AMF and local precise near-infrared laser irradiation (808nm) (DUAL-mode) to rapidly reach the phase change temperature (Tm) of MTX-MagTSLs in 5min compared with either AMF or laser stimulation alone, resulting in a significantly enhanced release of Dox at tumor region and precise cancer synergetic theranostics.
Collapse
Affiliation(s)
- Yuxin Guo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Jinyuan Ma
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qi Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yang Li
- College of Materials, Xiamen University, Xiamen, China
| | - Xinyi Zhou
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Dan Zhao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Hua Song
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qing Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
24
|
Chen J, Ratnayaka S, Alford A, Kozlovskaya V, Liu F, Xue B, Hoyt K, Kharlampieva E. Theranostic Multilayer Capsules for Ultrasound Imaging and Guided Drug Delivery. ACS NANO 2017; 11:3135-3146. [PMID: 28263564 PMCID: PMC5682940 DOI: 10.1021/acsnano.7b00151] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Despite the accessibility of ultrasound, the clinical potential of ultrasound-active theranostic agents has not been fully realized because it requires combining sufficient imaging contrast, high encapsulation efficiency, and ultrasound-triggered release in one entity. We report on theranostic polymer microcapsules composed of hydrogen-bonded multilayers of tannic acid and poly(N-vinylpyrrolidone) that produce high imaging contrast and deliver the anticancer drug doxorubicin upon low-power diagnostic or high-power therapeutic ultrasound irradiation. These capsules exhibit excellent imaging contrast in both brightness and harmonic modes and show prolonged contrast over six months, unlike commercially available microbubbles. We also demonstrate low-dose gradual and high-dose fast release of doxorubicin from the capsules by diagnostic (∼100 mW/cm2) and therapeutic (>10 W/cm2) ultrasound irradiation, respectively. We show that the imaging contrast of the capsules can be controlled by varying the number of layers, polymer type (relatively rigid tannic acid versus more flexible poly(methacrylic acid)), and polymer molecular weight. In vitro studies demonstrate that 50% doxorubicin release from ultrasound-treated capsules induces 97% cytotoxicity to MCF-7 human cancer cells, while no cytotoxicity is found without the treatment. Considering the strong ultrasound imaging contrast, high encapsulation efficiency, biocompatibility, and tunable drug release, these microcapsules can be used as theranostic agents for ultrasound-guided chemotherapy.
Collapse
Affiliation(s)
- Jun Chen
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Sithira Ratnayaka
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Aaron Alford
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Veronika Kozlovskaya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Fei Liu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Bing Xue
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Eugenia Kharlampieva
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| |
Collapse
|
25
|
Goins B, Phillips WT, Bao A. Strategies for improving the intratumoral distribution of liposomal drugs in cancer therapy. Expert Opin Drug Deliv 2016; 13:873-89. [PMID: 26981891 DOI: 10.1517/17425247.2016.1167035] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION A major limitation of current liposomal cancer therapies is the inability of liposome therapeutics to penetrate throughout the entire tumor mass. This inhomogeneous distribution of liposome therapeutics within the tumor has been linked to treatment failure and drug resistance. Both liposome particle transport properties and tumor microenvironment characteristics contribute to this challenge in cancer therapy. This limitation is relevant to both intravenously and intratumorally administered liposome therapeutics. AREAS COVERED Strategies to improve the intratumoral distribution of liposome therapeutics are described. Combination therapies of intravenous liposome therapeutics with pharmacologic agents modulating abnormal tumor vasculature, interstitial fluid pressure, extracellular matrix components, and tumor associated macrophages are discussed. Combination therapies using external stimuli (hyperthermia, radiofrequency ablation, magnetic field, radiation, and ultrasound) with intravenous liposome therapeutics are discussed. Intratumoral convection-enhanced delivery (CED) of liposomal therapeutics is reviewed. EXPERT OPINION Optimization of the combination therapies and drug delivery protocols are necessary. Further research should be conducted in appropriate cancer types with consideration of physiochemical features of liposomes and their timing sequence. More investigation of the role of tumor associated macrophages in intratumoral distribution is warranted. Intratumoral infusion of liposomes using CED is a promising approach to improve their distribution within the tumor mass.
Collapse
Affiliation(s)
- Beth Goins
- a Department of Radiology , University of Texas Health Science Center San Antonio , San Antonio , TX , USA
| | - William T Phillips
- a Department of Radiology , University of Texas Health Science Center San Antonio , San Antonio , TX , USA
| | - Ande Bao
- b Department of Radiation Oncology, School of Medicine, Case Western Reserve University/University Hospitals Case Medical Center , Cleveland , OH , USA
| |
Collapse
|
26
|
German SV, Bratashov DN, Navolokin NA, Kozlova AA, Lomova MV, Novoselova MV, Burilova EA, Zyev VV, Khlebtsov BN, Bucharskaya AB, Terentyuk GS, Amirov RR, Maslyakova GN, Sukhorukov GB, Gorin DA. In vitro and in vivo MRI visualization of nanocomposite biodegradable microcapsules with tunable contrast. Phys Chem Chem Phys 2016; 18:32238-32246. [DOI: 10.1039/c6cp03895f] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tunable MRI contrast of microcapsules was obtained.
Collapse
Affiliation(s)
| | | | - Nikita A. Navolokin
- Saratov State University
- Saratov
- Russia
- Saratov State Medical University
- Saratov
| | | | | | | | | | - Victor V. Zyev
- Saratov State University
- Saratov
- Russia
- Saratov State Medical University
- Saratov
| | - Boris N. Khlebtsov
- Saratov State University
- Saratov
- Russia
- Institute of Biochemistry and Physiology of Plants and Microorganisms
- Russia
| | | | | | - Rustem R. Amirov
- Alexander Butlerov Institute of Chemistry
- Kazan Federal University
- Kazan
- Russia
| | | | | | | |
Collapse
|
27
|
Hauser AK, Wydra RJ, Stocke NA, Anderson KW, Hilt JZ. Magnetic nanoparticles and nanocomposites for remote controlled therapies. J Control Release 2015; 219:76-94. [PMID: 26407670 PMCID: PMC4669063 DOI: 10.1016/j.jconrel.2015.09.039] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/19/2015] [Indexed: 12/17/2022]
Abstract
This review highlights the state-of-the-art in the application of magnetic nanoparticles (MNPs) and their composites for remote controlled therapies. Novel macro- to nano-scale systems that utilize remote controlled drug release due to actuation of MNPs by static or alternating magnetic fields and magnetic field guidance of MNPs for drug delivery applications are summarized. Recent advances in controlled energy release for thermal therapy and nanoscale energy therapy are addressed as well. Additionally, studies that utilize MNP-based thermal therapy in combination with other treatments such as chemotherapy or radiation to enhance the efficacy of the conventional treatment are discussed.
Collapse
Affiliation(s)
- Anastasia K Hauser
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - Robert J Wydra
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - Nathanael A Stocke
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - Kimberly W Anderson
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA
| | - J Zach Hilt
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
28
|
Nanoparticle-triggered release from lipid membrane vesicles. N Biotechnol 2015; 32:665-72. [DOI: 10.1016/j.nbt.2014.12.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 11/21/2022]
|
29
|
Malekar SA, Sarode AL, Bach AC, Bose A, Bothun G, Worthen DR. Radio Frequency-Activated Nanoliposomes for Controlled Combination Drug Delivery. AAPS PharmSciTech 2015; 16:1335-43. [PMID: 25899799 DOI: 10.1208/s12249-015-0323-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/12/2015] [Indexed: 11/30/2022] Open
Abstract
This work was conducted in order to design, characterize, and evaluate stable liposomes containing the hydrophobic drug raloxifene HCl (RAL) and hydrophilic doxycycline HCl (DOX), two potentially synergistic agents for treating osteoporosis and other bone lesions, in conjunction with a radio frequency-induced, hydrophobic magnetic nanoparticle-dependent triggering mechanism for drug release. Both drugs were successfully incorporated into liposomes by lipid film hydration, although combination drug loading compromised liposome stability. Liposome stability was improved by reducing the drug load and by including Pluronics® (PL) in the formulations. DOX did not appear to interact with the phospholipid membranes comprising the liposomes, and its release was maximized in the presence of radio frequency (RF) heating. In contrast, differential scanning calorimetry (DSC) and phosphorus-31 nuclear magnetic resonance ((31)P-NMR) analysis revealed that RAL developed strong interactions with the phospholipid membranes, most notably with lipid phosphate head groups, resulting in significant changes in membrane thermodynamics. Likewise, RAL release from liposomes was minimal, even in the presence of RF heating. These studies may offer useful insights into the design and optimization of multidrug containing liposomes. The effects of RAL on liposome characteristics and drug release performance underscore the importance of appropriate physical-chemical analysis in order to identify and characterize drug-lipid interactions that may profoundly affect liposome properties and performance early in the formulation development process.
Collapse
|
30
|
German SV, Navolokin NA, Kuznetsova NR, Zuev VV, Inozemtseva OA, Anis'kov AA, Volkova EK, Bucharskaya AB, Maslyakova GN, Fakhrullin RF, Terentyuk GS, Vodovozova EL, Gorin DA. Liposomes loaded with hydrophilic magnetite nanoparticles: Preparation and application as contrast agents for magnetic resonance imaging. Colloids Surf B Biointerfaces 2015; 135:109-115. [PMID: 26241922 DOI: 10.1016/j.colsurfb.2015.07.042] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 07/15/2015] [Accepted: 07/17/2015] [Indexed: 10/23/2022]
Abstract
Magnetic fluid-loaded liposomes (MFLs) were fabricated using magnetite nanoparticles (MNPs) and natural phospholipids via the thin film hydration method followed by extrusion. The size distribution and composition of MFLs were studied using dynamic light scattering and spectrophotometry. The effective ranges of magnetite concentration in MNPs hydrosol and MFLs for contrasting at both T2 and T1 relaxation were determined. On T2 weighted images, the MFLs effectively increased the contrast if compared with MNPs hydrosol, while on T1 weighted images, MNPs hydrosol contrasting was more efficient than that of MFLs. In vivo magnetic resonance imaging (MRI) contrasting properties of MFLs and their effects on tumor and normal tissues morphology, were investigated in rats with transplanted renal cell carcinoma upon intratumoral administration of MFLs. No significant morphological changes in rat internal organs upon intratumoral injection of MFLs were detected, suggesting that the liposomes are relatively safe and can be used as the potential contrasting agents for MRI.
Collapse
Affiliation(s)
- S V German
- Saratov State University, 410012 Saratov, Russia
| | - N A Navolokin
- Saratov Medical State University, 410012 Saratov, Russia
| | - N R Kuznetsova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - V V Zuev
- Saratov Medical State University, 410012 Saratov, Russia
| | | | - A A Anis'kov
- Saratov State University, 410012 Saratov, Russia
| | - E K Volkova
- Saratov State University, 410012 Saratov, Russia
| | | | - G N Maslyakova
- Saratov Medical State University, 410012 Saratov, Russia
| | - R F Fakhrullin
- Bionanotechnology Lab, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Republic of Tatarstan, Russia
| | - G S Terentyuk
- Saratov State University, 410012 Saratov, Russia; Saratov Medical State University, 410012 Saratov, Russia
| | - E L Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - D A Gorin
- Saratov State University, 410012 Saratov, Russia.
| |
Collapse
|
31
|
Booth A, Pintre IC, Lin Y, Gough JE, Webb SJ. Release of proteins and enzymes from vesicular compartments by alternating magnetic fields. Phys Chem Chem Phys 2015; 17:15579-88. [PMID: 25785572 DOI: 10.1039/c4cp05872k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The magnetic release of catalytically active enzymes from vesicular compartments within aggregated nanomaterials has been demonstrated. These nanomaterials, magnetic nanoparticle-vesicle aggregates (MNPVs), were formed by the self-assembly of biotinylated silica-coated Fe3O4 nanoparticles, biotinylated vesicles and tetrameric avidin. The unique features of nanoscale magnetite allow adhesion between membranes to be combined with magnetically triggered transit of reagents across membranes. Adding short spacers between the adhesive biotin groups and the nanoparticle or vesicle surfaces was found to strengthen binding to avidin, with binding of avidin to biotinylated bilayers and biotinylated nanoparticles monitored by quartz crystal microgravimetry with dissipation (QCM-D). Three different reagents were released from the vesicle compartments of MNPVs by a pulse of alternating magnetic field, with the release of a dye modelling the release of small molecule substrates, and the release of cytochrome c modelling the release of biological polymers, such as enzymes. To confirm that enzymes could be released and maintain activity, trypsin was encapsulated and shown to digest casein after magnetically triggered release.
Collapse
Affiliation(s)
- Andrew Booth
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
| | | | | | | | | |
Collapse
|
32
|
Xu Y, Wang L, Li YK, Wang CQ. Oxidation and pH responsive nanoparticles based on ferrocene-modified chitosan oligosaccharide for 5-fluorouracil delivery. Carbohydr Polym 2014; 114:27-35. [DOI: 10.1016/j.carbpol.2014.08.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/31/2014] [Accepted: 08/01/2014] [Indexed: 01/01/2023]
|