1
|
Zamora ME, Essien EO, Bhamidipati K, Murthy A, Liu J, Kim H, Patel MN, Nong J, Wang Z, Espy C, Chaudhry FN, Ferguson LT, Tiwari S, Hood ED, Marcos-Contreras OA, Omo-Lamai S, Shuvaeva T, Arguiri E, Wu J, Rauova L, Poncz M, Basil MC, Cantu E, Planer JD, Spiller K, Zepp J, Muzykantov VR, Myerson JW, Brenner JS. Marginated Neutrophils in the Lungs Effectively Compete for Nanoparticles Targeted to the Endothelium, Serving as a Part of the Reticuloendothelial System. ACS NANO 2024; 18:22275-22297. [PMID: 39105696 PMCID: PMC11935960 DOI: 10.1021/acsnano.4c06286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Nanomedicine has long pursued the goal of targeted delivery to specific organs and cell types but has yet to achieve this goal with the vast majority of targets. One rare example of success in this pursuit has been the 25+ years of studies targeting the lung endothelium using nanoparticles conjugated to antibodies against endothelial surface molecules. However, here we show that such "endothelial-targeted" nanocarriers also effectively target the lungs' numerous marginated neutrophils, which reside in the pulmonary capillaries and patrol for pathogens. We show that marginated neutrophils' uptake of many of these "endothelial-targeted" nanocarriers is on par with endothelial uptake. This generalizes across diverse nanomaterials and targeting moieties and was even found with physicochemical lung tropism (i.e., without targeting moieties). Further, we observed this in ex vivo human lungs and in vivo healthy mice, with an increase in marginated neutrophil uptake of nanoparticles caused by local or distant inflammation. These findings have implications for nanomedicine development for lung diseases. These data also suggest that marginated neutrophils, especially in the lungs, should be considered a major part of the reticuloendothelial system (RES), with a special role in clearing nanoparticles that adhere to the lumenal surfaces of blood vessels.
Collapse
Affiliation(s)
- Marco E Zamora
- Drexel University School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Eno-Obong Essien
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Kartik Bhamidipati
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Aditi Murthy
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Jing Liu
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Hyunjun Kim
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Manthan N Patel
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jia Nong
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Zhicheng Wang
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Carolann Espy
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Fatima N Chaudhry
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Laura T Ferguson
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Sachchidanand Tiwari
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Serena Omo-Lamai
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Tea Shuvaeva
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Evguenia Arguiri
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jichuan Wu
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Lubica Rauova
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Mortimer Poncz
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Maria C Basil
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Edward Cantu
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Joseph D Planer
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Kara Spiller
- Drexel University School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
| | - Jarod Zepp
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
2
|
Lin P, Gao R, Fang Z, Yang W, Tang Z, Wang Q, Wu Y, Fang J, Yu W. Precise nanodrug delivery systems with cell-specific targeting for ALI/ARDS treatment. Int J Pharm 2023; 644:123321. [PMID: 37591476 DOI: 10.1016/j.ijpharm.2023.123321] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/22/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common acute and critical diseases in clinics and have no effective treatment to date. With the concept of "precision medicine", research into the precise drug delivery of therapeutic and diagnostic drugs has become a frontier in nanomedicine research and has entered the era of design of precise nanodrug delivery systems (NDDSs) with cell-specific targeting. Owing to the distinctive characteristics of ALI/ARDS, designing NDDSs for specific focal sites is an important strategy for changing drug distribution in the body and specifically increasing drug concentration at target sites while decreasing drug concentration at non-target sites. This strategy enhances drug efficacy, reduces adverse reactions, and ensures accurate nano-targeted treatment. On the basis of the characteristics of pathological ALI/ARDS microenvironments, this paper reviews NDDSs targeting vascular endothelial cells, neutrophils, alveolar macrophages, and alveolar epithelial cells to provide reference for designing accurate NDDSs for ALI/ARDS and novel insights into targeted treatments for ALI/ARDS.
Collapse
Affiliation(s)
- Peihong Lin
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Rui Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Zhengyu Fang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Wenjing Yang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Zhan Tang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Qiao Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Yueguo Wu
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Jie Fang
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou 310013, China.
| | - Wenying Yu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China.
| |
Collapse
|
3
|
Liu GW, Guzman EB, Menon N, Langer RS. Lipid Nanoparticles for Nucleic Acid Delivery to Endothelial Cells. Pharm Res 2023; 40:3-25. [PMID: 36735106 PMCID: PMC9897626 DOI: 10.1007/s11095-023-03471-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
Endothelial cells play critical roles in circulatory homeostasis and are also the gateway to the major organs of the body. Dysfunction, injury, and gene expression profiles of these cells can cause, or are caused by, prevalent chronic diseases such as diabetes, cardiovascular disease, and cancer. Modulation of gene expression within endothelial cells could therefore be therapeutically strategic in treating longstanding disease challenges. Lipid nanoparticles (LNP) have emerged as potent, scalable, and tunable carrier systems for delivering nucleic acids, making them attractive vehicles for gene delivery to endothelial cells. Here, we discuss the functions of endothelial cells and highlight some receptors that are upregulated during health and disease. Examples and applications of DNA, mRNA, circRNA, saRNA, siRNA, shRNA, miRNA, and ASO delivery to endothelial cells and their targets are reviewed, as well as LNP composition and morphology, formulation strategies, target proteins, and biomechanical factors that modulate endothelial cell targeting. Finally, we discuss FDA-approved LNPs as well as LNPs that have been tested in clinical trials and their challenges, and provide some perspectives as to how to surmount those challenges.
Collapse
Affiliation(s)
- Gary W Liu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Edward B Guzman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Nandita Menon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Strand Therapeutics, MA, 02215, Boston, USA
| | - Robert S Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
4
|
Ferguson LT, Hood ED, Shuvaeva T, Shuvaev VV, Basil MC, Wang Z, Nong J, Ma X, Wu J, Myerson JW, Marcos-Contreras OA, Katzen J, Carl JM, Morrisey EE, Cantu E, Villa CH, Mitragotri S, Muzykantov VR, Brenner JS. Dual Affinity to RBCs and Target Cells (DART) Enhances Both Organ- and Cell Type-Targeting of Intravascular Nanocarriers. ACS NANO 2022; 16:4666-4683. [PMID: 35266686 PMCID: PMC9339245 DOI: 10.1021/acsnano.1c11374] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
A long-standing goal of nanomedicine is to improve a drug's benefit by loading it into a nanocarrier that homes solely to a specific target cell and organ. Unfortunately, nanocarriers usually end up with only a small percentage of the injected dose (% ID) in the target organ, due largely to clearance by the liver and spleen. Further, cell-type-specific targeting is rarely achieved without reducing target organ accumulation. To solve these problems, we introduce DART (dual affinity to RBCs and target cells), in which nanocarriers are conjugated to two affinity ligands, one binding red blood cells and one binding a target cell (here, pulmonary endothelial cells). DART nanocarriers first bind red blood cells and then transfer to the target organ's endothelial cells as the bound red blood cells squeeze through capillaries. We show that within minutes after intravascular injection in mice nearly 70% ID of DART nanocarriers accumulate in the target organ (lungs), more than doubling the % ID ceiling achieved by a multitude of prior technologies, finally achieving a majority % ID in a target organ. Humanized DART nanocarriers in ex vivo perfused human lungs recapitulate this phenomenon. Furthermore, DART enhances the selectivity of delivery to target endothelial cells over local phagocytes within the target organ by 6-fold. DART's marked improvement in both organ- and cell-type targeting may thus be helpful in localizing drugs for a multitude of medical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Samir Mitragotri
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts 02138, United States
| | | | | |
Collapse
|
5
|
Cavalcanti IDL, de Fátima Ramos Dos Santos Medeiros SM, Dos Santos Macêdo DC, Ferro Cavalcanti IM, de Britto Lira Nogueira MC. Nanocarriers in the Delivery of Hydroxychloroquine to the Respiratory System: An Alternative to COVID-19. Curr Drug Deliv 2021; 18:583-595. [PMID: 32860358 DOI: 10.2174/1567201817666200827110445] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 11/22/2022]
Abstract
In response to the global outbreak caused by SARS-CoV-2, this article aims to propose the development of nanosystems for the delivery of hydroxychloroquine in the respiratory system to the treatment of COVID-19. A descriptive literature review was conducted, using the descriptors "COVID-19", "Nanotechnology", "Respiratory Syndrome" and "Hydroxychloroquine", in the PubMed, ScienceDirect and SciElo databases. After analyzing the articles according to the inclusion and exclusion criteria, they were divided into 3 sessions: Coronavirus: definitions, classifications and epidemiology, pharmacological aspects of hydroxychloroquine and pharmaceutical nanotechnology in targeting of drugs. We used 131 articles published until July 18, 2020. Hydroxychloroquine seems to promote a reduction in viral load, in vivo studies, preventing the entry of SARS-CoV-2 into lung cells, and the safety of its administration is questioned due to the toxic effects that it can develop, such as retinopathy, hypoglycemia and even cardiotoxicity. Nanosystems for the delivery of drugs in the respiratory system may be a viable alternative for the administration of hydroxychloroquine, which may enhance the therapeutic effect of the drug with a consequent decrease in its toxicity, providing greater safety for implementation in the clinic in the treatment of COVID-19.
Collapse
|
6
|
Di J, Xie F, Xu Y. When liposomes met antibodies: Drug delivery and beyond. Adv Drug Deliv Rev 2020; 154-155:151-162. [PMID: 32926944 DOI: 10.1016/j.addr.2020.09.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
Drug encapsulated liposomes and monoclonal antibodies (Mabs) are two distinctively different classes of therapeutics, but both aim to become the ultimate "magic bullet". While PEGylated liposomes rely on the enhanced permeability and retention (EPR) effect for accumulation in solid tumor tissues, Mabs are designed to bind tightly to specific surface antigens on target cells to exert effector functions. Immunoliposome (IL) refers to the structural combination of liposomes and antibodies, whereas the antibodies are usually decorated on the liposome surface. ILs can therefore take advantage of interactions between antibodies and cancer cells for more efficient endocytosis and intracellular drug delivery. The antibody structure, affinity, density, as well as the liposome surface properties and drug to lipid ratios all contribute to the IL pharmacokinetic(PK) and pharmacodynamic(PD) behaviors. The optimal formulation parameters may vary for different target cells and tissues. Furthermore, besides the delivery of cytotoxic drugs to cancer cells, new ILs are being developed to interact with multiple target receptors, multiple target cells and trigger multiple therapeutic effects. We envision that the IL format can be a great platform for the molecular engineering of multi-valent, multi-specific interactions to achieve complex biological functions for therapeutic benefits, especially in the area of cancer immunotherapy.
Collapse
Affiliation(s)
- Jiaxing Di
- School of Pharmacy, Shanghai Jiao Tong University, China
| | - Fang Xie
- Department of Biomedical Engineering, Johns Hopkins University, United States of America
| | - Yuhong Xu
- College of Pharmacy and Chemistry, Dali University, China.
| |
Collapse
|
7
|
Onuki Y, Obata Y, Kawano K, Sano H, Matsumoto R, Hayashi Y, Takayama K. Membrane Microdomain Structures of Liposomes and Their Contribution to the Cellular Uptake Efficiency into HeLa Cells. Mol Pharm 2016; 13:369-78. [PMID: 26709741 DOI: 10.1021/acs.molpharmaceut.5b00601] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The purpose of this study is to obtain a comprehensive relationship between membrane microdomain structures of liposomes and their cellular uptake efficiency. Model liposomes consisting of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC)/1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC)/cholesterol (Ch) were prepared with various lipid compositions. To detect distinct membrane microdomains in the liposomes, fluorescence-quenching assays were performed at temperatures ranging from 25 to 60 °C using 1,6-diphenyl-1,3,5-hexatriene-labeled liposomes and (2,2,6,6-tetramethylpiperidin-1-yl)oxyl. From the data analysis using the response surface method, we gained a better understanding of the conditions for forming distinct domains (Lo, Ld, and gel phase membranes) as a function of lipid composition. We further performed self-organizing maps (SOM) clustering to simplify the complicated behavior of the domain formation to obtain its essence. As a result, DPPC/DOPC/Ch liposomes in any lipid composition were integrated into five distinct clusters in terms of similarity of the domain structure. In addition, the findings from synchrotron small-angle X-ray scattering analysis offered further insight into the domain structures. As a last phase of this study, an in vitro cellular uptake study using HeLa cells was conducted using SOM clusters' liposomes with/without PEGylation. As a consequence of this study, higher cellular uptake was observed from liposomes having Ch-rich ordered domains.
Collapse
Affiliation(s)
- Yoshinori Onuki
- Department of Pharmaceutical Technology, Graduate School of Medical and Pharmaceutical Science, Unversity of Toyama , Sugitani 2630, Toyama-shi, Toyama 930-0194, Japan
| | - Yasuko Obata
- Department of Pharmaceutics and §Department of Drug Delivery Research, Hoshi University , Ebara 2-4-41, Shinagawa, Tokyo 142-8501, Japan
| | | | - Hiromu Sano
- Department of Pharmaceutics and §Department of Drug Delivery Research, Hoshi University , Ebara 2-4-41, Shinagawa, Tokyo 142-8501, Japan
| | - Reina Matsumoto
- Department of Pharmaceutics and §Department of Drug Delivery Research, Hoshi University , Ebara 2-4-41, Shinagawa, Tokyo 142-8501, Japan
| | - Yoshihiro Hayashi
- Department of Pharmaceutical Technology, Graduate School of Medical and Pharmaceutical Science, Unversity of Toyama , Sugitani 2630, Toyama-shi, Toyama 930-0194, Japan
| | - Kozo Takayama
- Department of Pharmaceutics and §Department of Drug Delivery Research, Hoshi University , Ebara 2-4-41, Shinagawa, Tokyo 142-8501, Japan
| |
Collapse
|
8
|
Suzuki R, Omata D, Oda Y, Unga J, Negishi Y, Maruyama K. Cancer Therapy with Nanotechnology-Based Drug Delivery Systems: Applications and Challenges of Liposome Technologies for Advanced Cancer Therapy. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3121-7_23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
9
|
Bansal D, Gulbake A, Tiwari J, Jain SK. Development of liposomes entrapped in alginate beads for the treatment of colorectal cancer. Int J Biol Macromol 2016; 82:687-95. [DOI: 10.1016/j.ijbiomac.2015.09.052] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/16/2015] [Accepted: 09/27/2015] [Indexed: 11/28/2022]
|
10
|
Icam-1 targeted nanogels loaded with dexamethasone alleviate pulmonary inflammation. PLoS One 2014; 9:e102329. [PMID: 25019304 PMCID: PMC4096597 DOI: 10.1371/journal.pone.0102329] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 06/18/2014] [Indexed: 11/19/2022] Open
Abstract
Lysozyme dextran nanogels (NG) have great potential in vitro as a drug delivery platform, combining simple chemistry with rapid uptake and cargo release in target cells with “stealth” properties and low toxicity. In this work, we study for the first time the potential of targeted NG as a drug delivery platform in vivo to alleviate acute pulmonary inflammation in animal model of LPS-induced lung injury. NG are targeted to the endothelium via conjugation with an antibody (Ab) directed to Intercellular Adhesion Molecule-1(ICAM-NG), whereas IgG conjugated NG (IgG-NG) are used for control formulations. The amount of Ab conjugated to the NG and distribution in the body after intravenous (IV) injection have been quantitatively analyzed using a tracer isotope-labeled [125I]IgG. As a proof of concept, Ab-NG are loaded with dexamethasone, an anti-inflammatory therapeutic, and the drug uptake and release kinetics are measured by HPLC. In vivo studies in mice showed that: i) ICAM-NG accumulates in mouse lungs (∼120% ID/g vs ∼15% ID/g of IgG-NG); and, ii) DEX encapsulated in ICAM-NG, but not in IgG-NG practically blocks LPS-induced overexpression of pro-inflammatory cell adhesion molecules including ICAM-1 in the pulmonary inflammation.
Collapse
|
11
|
Bansal D, Yadav K, Pandey V, Ganeshpurkar A, Agnihotri A, Dubey N. Lactobionic acid coupled liposomes: an innovative strategy for targeting hepatocellular carcinoma. Drug Deliv 2014; 23:140-6. [DOI: 10.3109/10717544.2014.907373] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Divya Bansal
- Pharmaceutics Research Laboratory, Shri Ram Institute of Technology – Pharmacy, Jabalpur, Madhya Pradesh, India and
| | - Kamini Yadav
- Pharmaceutics Research Laboratory, Shri Ram Institute of Technology – Pharmacy, Jabalpur, Madhya Pradesh, India and
| | - Vikas Pandey
- Pharmaceutics Research Laboratory, Shri Ram Institute of Technology – Pharmacy, Jabalpur, Madhya Pradesh, India and
| | - Aditya Ganeshpurkar
- Drug Discovery Laboratory, Shri Ram Institute of Technology – Pharmacy, Jabalpur, Madhya Pradesh, India
| | - Abhishek Agnihotri
- Drug Discovery Laboratory, Shri Ram Institute of Technology – Pharmacy, Jabalpur, Madhya Pradesh, India
| | - Nazneen Dubey
- Drug Discovery Laboratory, Shri Ram Institute of Technology – Pharmacy, Jabalpur, Madhya Pradesh, India
| |
Collapse
|
12
|
Akbulut M, D’Addio SM, Gindy ME, Prud’homme RK. Novel methods of targeted drug delivery: the potential of multifunctional nanoparticles. Expert Rev Clin Pharmacol 2014; 2:265-82. [DOI: 10.1586/ecp.09.4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
13
|
Endothelial targeting of liposomes encapsulating SOD/catalase mimetic EUK-134 alleviates acute pulmonary inflammation. J Control Release 2014; 177:34-41. [PMID: 24412573 DOI: 10.1016/j.jconrel.2013.12.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/26/2013] [Accepted: 12/30/2013] [Indexed: 12/24/2022]
Abstract
Production of excessive levels of reactive oxygen species (ROS) in the vascular endothelium is a common pathogenic pathway in many dangerous conditions, including acute lung injury, ischemia-reperfusion, and inflammation. Ineffective delivery of antioxidants to the endothelium limits their utility for management of these conditions. In this study, we devised a novel translational antioxidant intervention targeted to the vascular endothelium using PEG-liposomes loaded with EUK-134 (EUK), a potent superoxide dismutase/catalase mimetic. EUK loaded into antibody-coated liposomes (size 197.8±4.5 nm diameter, PDI 0.179±0.066) exerted partial activity in the intact carrier, while full activity was recovered upon liposome disruption. For targeting we used antibodies (Abs) to platelet-endothelial cell adhesion molecule (PECAM-1). Both streptavidin-biotin and SATA/SMCC conjugation chemistries provided binding of 125-150 Ab molecules per liposome. Ab/EUK/liposomes, but not IgG/EUK/liposomes: i) bound to endothelial cells and inhibited cytokine-induced inflammatory activation in vitro; and, ii) accumulated in lungs after intravascular injection, providing >60% protection against pulmonary edema in endotoxin-challenged mice (vs <6% protection afforded by IgG/liposome/EUK counterpart). Since the design elements of this drug delivery system are already in clinical use (PEG-liposomes, antibodies, SATA/SMCC conjugation), it is an attractive candidate for translational interventions using antioxidant molecules such as EUK and other clinically acceptable drugs.
Collapse
|
14
|
Baskar S, Muthusamy N. Antibody-based therapeutics for the treatment of human B cell malignancies. Curr Allergy Asthma Rep 2013; 13:33-43. [PMID: 23229130 PMCID: PMC3674564 DOI: 10.1007/s11882-012-0327-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The dynamic expression of various phenotypic markers during B cell development not only defines the particular stage in ontogeny but also provides the necessary growth, differentiation, maturation and survival signals. When a B cell at any given stage becomes cancerous, these cell surface molecules, intracellular signaling molecules, and the over-expressed gene products become favorite targets for potential therapeutic intervention. Various adaptive and adoptive immunotherapeutic approaches induce T cell and antibody responses against cancer cells, and successful remission leading to minimal residual disease has been obtained. Nonetheless, subsequent relapse and development of resistant clones prompted further development and several novel strategies are evolving. Engineered monoclonal antibodies with high affinity and specificity to target antigens have been developed and used either alone or in combination with chemotherapeutic drugs. They are also used as vehicles to deliver cytotoxic drugs, toxins, or radionuclides that are either directly conjugated or encapsulated in liposomal vesicles. Likewise, genetically engineered T cells bearing chimeric antigen receptors are used to redirect cytotoxicity to antigen-positive target cells. This review describes recent advancements in some of these adoptive immunotherapeutic strategies targeting B cell malignancies.
Collapse
Affiliation(s)
- Sivasubramanian Baskar
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10/CRC; Room 3E-3248, Bethesda, MD 20892, USA
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine; Molecular Virology, Immunology and Medical Genetics and Veterinary BioSciences, The OSU Comprehensive Cancer Center, The Ohio State University, 455E, OSUCCC, 410, West 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Adv Drug Deliv Rev 2013; 65:36-48. [PMID: 23036225 DOI: 10.1016/j.addr.2012.09.037] [Citation(s) in RCA: 3097] [Impact Index Per Article: 258.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 09/10/2012] [Accepted: 09/20/2012] [Indexed: 02/06/2023]
Abstract
The first closed bilayer phospholipid systems, called liposomes, were described in 1965 and soon were proposed as drug delivery systems. The pioneering work of countless liposome researchers over almost 5 decades led to the development of important technical advances such as remote drug loading, extrusion for homogeneous size, long-circulating (PEGylated) liposomes, triggered release liposomes, liposomes containing nucleic acid polymers, ligand-targeted liposomes and liposomes containing combinations of drugs. These advances have led to numerous clinical trials in such diverse areas as the delivery of anti-cancer, anti-fungal and antibiotic drugs, the delivery of gene medicines, and the delivery of anesthetics and anti-inflammatory drugs. A number of liposomes (lipidic nanoparticles) are on the market, and many more are in the pipeline. Lipidic nanoparticles are the first nanomedicine delivery system to make the transition from concept to clinical application, and they are now an established technology platform with considerable clinical acceptance. We can look forward to many more clinical products in the future.
Collapse
|
16
|
|
17
|
Suana AJ, Tuffin G, Frey BM, Knudsen L, Mühlfeld C, Rödder S, Marti HP. Single application of low-dose mycophenolate mofetil-OX7-immunoliposomes ameliorates experimental mesangial proliferative glomerulonephritis. J Pharmacol Exp Ther 2011; 337:411-22. [PMID: 21349934 DOI: 10.1124/jpet.110.176222] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
IgA nephropathy, one of the most frequent forms of glomerulonephritis, characterized by mesangial hypercellularity and glomerular extracellular matrix (ECM) expansion, often leads to end-stage renal disease over a prolonged period. We investigated whether antiproliferative treatment in a single low dose specifically targeted to the glomerular mesangium by immunoliposomes (ILs) results in an amelioration of mesangial proliferative glomerulonephritis in rats (anti-Thy1.1 nephritis). Mycophenolate mofetil (MMF) containing ILs was generated that targets the Thy1.1 antigen (OX-7) in rat mesangial cells. Treatment benefit of a single intravenous dose of these ILs given 2 days after disease induction was investigated by stereology, immunohistochemistry, and functional analyses (creatinine, albuminuria) until day +9 and was compared among untreated and free MMF-treated rats using six male Wistar rats per group. MMF-loaded OX7-IL prevented creatinine increase and albuminuria. Stereological analyses of MMF OX7-IL-treated animals yielded 30% reduction of mesangial cells on day +9 and a 40% reduction of glomerular ECM volume on day +5, compared with all of the other nephritic animals. Furthermore, at days +5 and +9 we observed decreased ECM content and decreased glomerular volume (day +5) in the MMF-OX7-IL-treated group compared with the nephritic group treated with free MMF. In conclusion, MMF-OX7-IL-based directed drug delivery represents a novel approach for treating mesangial cell-mediated forms of glomerulonephritis.
Collapse
Affiliation(s)
- A J Suana
- Department of Nephrology and Hypertension, University of Bern Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | | | | | | | | | | | | |
Collapse
|
18
|
Maruyama K. Intracellular targeting delivery of liposomal drugs to solid tumors based on EPR effects. Adv Drug Deliv Rev 2011; 63:161-9. [PMID: 20869415 DOI: 10.1016/j.addr.2010.09.003] [Citation(s) in RCA: 456] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 07/31/2010] [Accepted: 09/01/2010] [Indexed: 11/26/2022]
Abstract
The success of an effective drug delivery system using liposomes for solid tumor targeting based on EPR effects is highly dependent on both size ranging from 100-200 nm in diameter and prolonged circulation half-life in the blood. A major development was the synthesis of PEG-liposomes with a prolonged circulation time in the blood. Active targeting of immunoliposomes to the solid tumor tissue can be achieved by the Fab' fragment which is better than whole IgG in terms of designing PEG-immunoliposomes with prolonged circulation. For intracellular targeting delivery to solid tumors based on EPR effects, transferrin-PEG-liposomes can stay in blood circulation for a long time and extravasate into the extravascular of tumor tissue by the EPR effect as PEG-liposomes. The extravasated transferrin-PEG-liposomes can maintain anti cancer drugs in interstitial space for a longer period, and deliver them into the cytoplasm of tumor cells via transferrin receptor-mediated endocytosis. Transferrin-PEG-liposomes improve the safety and efficacy of anti cancer drug by both passive targeting by prolonged circulation and active targeting by transferrin.
Collapse
|
19
|
Bagari R, Bansal D, Gulbake A, Jain A, Soni V, Jain SK. Chondroitin sulfate functionalized liposomes for solid tumor targeting. J Drug Target 2010; 19:251-7. [DOI: 10.3109/1061186x.2010.492525] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Zhang GH, Liu YF, Hu HY. Preparation and Cytotoxicity Effect of Anti-Hepatocellular Carcinoma Scfv Immunoliposome on Hepatocarcinoma Cell in Vitro. EUR J INFLAMM 2010. [DOI: 10.1177/1721727x1000800204] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The use of PE38 for cancer therapy has attracted considerable attention for a long time. However, the extensive use of PE38 is prohibited by its severe side effects. Even though immunotoxin PE38 has been researched for cancer therapy, it has displayed low antitumor activity. The aim of this study is to compare the killing efficacy on Hepatocellular carcinoma (HCC) SMMC-7721 cell of immunoliposome PE38, immunotoxin PE38 and liposome PE38. In this study, the sterically stabilized liposomal PE38 was prepared using soybean phosphatidylcholine, cholesterol, and Cholesterol-PEG-COOH. The humanized anti-hepatoma disulfide-stabilized Fv (hdsFv25) was coupled to sterically stabilized liposomes using the N-hydroxysuccinimide ester method. The immunoliposome PE38 was prepared in our lab using the above-mentioned single-chain antibody. The hdsFv25-immunoliposomes were immunoreactive as determined by ELISA assay. Immunoliposome PE38 can kill SMMC-7721 cells in vitro with higher efficiency than non-targeted liposomes. These results indicate that immunoliposome PE38 may be potential in the treatment of hepatocarcinoma.
Collapse
Affiliation(s)
| | - Y-F. Liu
- Department of Patholgy, the Fourth Military Medical University, Xi'an, Shaanxi
| | - H-Y. Hu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, PR. China
| |
Collapse
|
21
|
|
22
|
|
23
|
Allen TM, Agrawal AK, Ahmad I, Hansen CB, Zalipsky S. Antibody-Mediated Targeting of Long-Circulating (StealthR) Liposomes. J Liposome Res 2008. [DOI: 10.3109/08982109409037027] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
|
25
|
|
26
|
|
27
|
|
28
|
|
29
|
Literature Alerts. J Microencapsul 2008. [DOI: 10.3109/02652049209021229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
30
|
Longman SA, Cullis PR, Bally MB. A model approach for assessing liposome targetingin vivo. Drug Deliv 2008. [DOI: 10.3109/10717549509031365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
31
|
Literature Alerts. J Microencapsul 2008. [DOI: 10.3109/02652049109069569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Redelmeier TE, Guillet JGÉR, Ballyt MB. High-affinity targeting of biotin-labeled liposomes to streptavidin-conjugated ligands. Drug Deliv 2008. [DOI: 10.3109/10717549509031357] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
33
|
Affiliation(s)
- Vladimir Torchilin
- Northeastern University, Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, 360 Huntington Avenue, Boston, MA 02115, USA ;
| |
Collapse
|
34
|
Targeted gene therapy of LS174 T human colon carcinoma by anti-TAG-72 immunoliposomes. Cancer Gene Ther 2008; 15:331-40. [DOI: 10.1038/cgt.2008.11] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Zhao XB, Muthusamy N, Byrd JC, Lee RJ. Cholesterol as a bilayer anchor for PEGylation and targeting ligand in folate-receptor-targeted liposomes. J Pharm Sci 2007; 96:2424-35. [PMID: 17588260 DOI: 10.1002/jps.20885] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Phospholipids have been extensively evaluated as an anchor for both PEGylation and receptor-targeting in liposomal formulations. However, cholesterol, another important component in biomembranes, has not been fully investigated as an alternative anchor. In this study, the potential role of cholesterol for anchoring PEG and folate was investigated. Cholesterol derivatives were synthesized for PEGylation (mPEG-cholesterol) and folate receptor (FR) targeting (folate-PEG-cholesterol) and incorporated into the bilayer of FR-targeted liposomal doxorubicin. The colloidal stability of these cholesterol derivative-containing liposomes was superior to non-PEGylated liposomes, indicating that steric barrier provided by mPEG-cholesterol can efficiently inhibit aggregation of liposomes. FR-targeting activity of these liposomes was demonstrated by in vitro cell-binding studies on FR-overexpressing KB cells. In addition, in vivo circulation of cholesterol-anchored liposomes was prolonged compared to non-PEGylated liposomes. These studies suggest that cholesterol is a viable bilayer anchor for synthesis of PEGylated and FR-targeted liposomes.
Collapse
Affiliation(s)
- Xiaobin B Zhao
- Division of Pharmaceutics, NCI OSU Comprehensive Cancer Center, NSF Nanoscales Science and Engineering Center (NSEC), College of Pharmacy, The Ohio State University, 542 LM Parks Hall, 500 W. 12th Ave, Columbus, Ohio 43210, USA
| | | | | | | |
Collapse
|
36
|
Suzuki R, Takizawa T, Negishi Y, Utoguchi N, Sawamura K, Tanaka K, Namai E, Oda Y, Matsumura Y, Maruyama K. Tumor specific ultrasound enhanced gene transfer in vivo with novel liposomal bubbles. J Control Release 2007; 125:137-44. [PMID: 18035442 DOI: 10.1016/j.jconrel.2007.08.025] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 08/13/2007] [Accepted: 08/19/2007] [Indexed: 10/22/2022]
Abstract
Bubble liposomes (liposomes which entrap an ultrasound imaging gas) may constitute a unique system for delivering various molecules efficiently into mammalian cells in vitro. In this study, Bubble liposomes were compared with cationic lipid (CL)-DNA complexes as potential gene delivery carriers into tumor in vivo. The delivery of genes by Bubble liposomes depended on the intensity of the applied ultrasound. Transfection efficiency plateaued at 0.7 W/cm(2) ultrasound intensity. Bubble liposomes efficiently transferred genes into cultured cells even when the cells were exposed to ultrasound for only 1 s. In addition, Bubble liposomes could introduce the luciferase gene more effectively than CL-DNA complexes into mouse ascites tumor cells and solid tumor tissue. We conclude that the combination of Bubble liposomes and ultrasound is a minimally-invasive and tumor specific gene transfer method in vivo.
Collapse
Affiliation(s)
- Ryo Suzuki
- Department of Biopharmaceutics, School of Pharmaceutical Sciences, Teikyo University, 1091-1 Suwarashi, Sagamiko, Sagamihara, Kanagawa 229-0195, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kobayashi T, Ishida T, Okada Y, Ise S, Harashima H, Kiwada H. Effect of transferrin receptor-targeted liposomal doxorubicin in P-glycoprotein-mediated drug resistant tumor cells. Int J Pharm 2007; 329:94-102. [PMID: 16997518 DOI: 10.1016/j.ijpharm.2006.08.039] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 07/21/2006] [Accepted: 08/20/2006] [Indexed: 11/28/2022]
Abstract
The over-expression of P-glycoprotein (P-gp) has been associated with the development of multidrug resistance (MDR) in cancer cells. In this study, we examined whether transferrin receptor (Tf-R) targeted liposomes can efficiently deliver encapsulated doxorubicin (DXR) into MDR cells (SBC-3/ADM) via Tf-R-mediated endocytosis thus overcoming MDR by by-passing P-gp-mediated drug efflux. We prepared four types of liposome, i.e. untargeted and Tf-R-targeted, made of either egg-PC/cholesterol or hydrogenated egg PC/cholesterol. Only with the targeted EPC-liposome we achieved significant delivery of encapsulated DXR and increased cytotoxicity of encapsulated DXR on the MDR cells (3.5-fold higher than free DXR). Confocal microscopy and an intracellular drug-accumulation assay indicated that the targeted liposomes efficiently delivered DXR into cells where it readily accumulated in the nucleus, in both drug-sensitive and MDR cells. These findings suggest that the targeted liposomes are rapidly internalized via Tf-R-mediated endocytosis followed by release of their contents into the cytoplasm. The rapid internalization and content release, most likely facilitated by the higher fluidity of the EPC-based liposomes, may explain why only targeted EPC-liposomes were able to prevent drug efflux by P-gp and to consequently circumvent MDR. Our results indicate that in order to achieve MDR circumvention by means of liposomal encapsulation of DXR the liposomes not only need to be targeted, but also to have the proper physicochemical properties for adequate release of the drug. Furthermore, these in vitro results suggest that Tf-R targeted EPC-liposomes are a potentially useful drug delivery system to circumvent P-gp-mediated MDR of tumors.
Collapse
Affiliation(s)
- Tomotaka Kobayashi
- Department of Pharmacokinetics and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, The University of Tokushima, 1-78-1 Sho-machi, Tokushima 770-8505, Tokushima, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Atobe K, Ishida T, Ishida E, Hashimoto K, Kobayashi H, Yasuda J, Aoki T, Obata KI, Kikuchi H, Akita H, Asai T, Harashima H, Oku N, Kiwada H. In Vitro Efficacy of a Sterically Stabilized Immunoliposomes Targeted to Membrane Type 1 Matrix Metalloproteinase (MT1-MMP). Biol Pharm Bull 2007; 30:972-8. [PMID: 17473445 DOI: 10.1248/bpb.30.972] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The poor selective cytotoxicity of anticancer drugs lead to dose-limiting adverse effects which compromise the clinical outcome. Solid tumors recruit new blood vessels to support their growth, and epitopes that are uniquely expressed on tumor cells and tumor endothelial cells (ECs) can function as targets for immunoliposomal anticancer drugs. Membrane type 1 matrix metalloproteinase (MT1-MMP), an important protein related to tumor growth and angiogenesis, is expressed on malignant tumor cells and is activated ECs. Selective delivery could be achieved by targeting MT1-MMP, as well as other angiogenic ECs. In this regard, an anti-MT1-MMP Fab' antibody was used to prepare a MT1-MMP targeted sterically stabilized immunoliposomes (SIL[anti-MT1-MMP(Fab')]). The binding and intracellular distribution of SIL[anti-MT1-MMP(Fab')] and a non-targeted sterically stabilized liposomes (SL) were examined using human fibrosarcoma HT-1080 cells. SIL[anti-MT1-MMP(Fab')] was taken up by the cells in a lipid concentration, temperature, and time dependent manner, ultimately accumulating in the lysosomes. The cytotoxicity of doxorubicin (DXR)-containing SIL[anti-MT1-MMP(Fab')] (DXR-SIL[anti-MT1-MMP(Fab')]) was significantly higher than that of DXR-containing SL. The cellular internalization of SIL[anti-MT1-MMP(Fab')] was inhibited by endocytosis inhibitors, suggesting that their internalization was mediated via clathrin- or caveolae-dependent endocytosis. Furthermore, the efficient binding of SIL[anti-MT1-MMP(Fab')] was observed on human umbilical vein endothelial cells (HUVEC). Based on these results, it would be expected that DXR-SIL[anti-MT1-MMP(Fab')] may achieve direct tumor cell kill and indirect tumor cell kill via the destruction of the tumor endothelium in vivo. This strategy may have the potential for overcoming some major limitations in conventional chemotherapy in vivo.
Collapse
Affiliation(s)
- Kazutaka Atobe
- Department of Pharmacokinetics and Biopharmaceutics, Subdivision of Biopharmaceutical Sciences, Institute of Health Biosciences, The University of Tokushima, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Suzuki R, Takizawa T, Negishi Y, Hagisawa K, Tanaka K, Sawamura K, Utoguchi N, Nishioka T, Maruyama K. Gene delivery by combination of novel liposomal bubbles with perfluoropropane and ultrasound. J Control Release 2006; 117:130-6. [PMID: 17113176 DOI: 10.1016/j.jconrel.2006.09.008] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 08/07/2006] [Accepted: 09/14/2006] [Indexed: 12/20/2022]
Abstract
Microbubbles and ultrasound have recently been investigated with a view to improving the transfection efficiency of non-viral gene delivery systems. However, microbubbles are unstable and their targeting ability is insufficient for clinical use. To circumvent these problems, we developed novel polyethyleneglycol (PEG) modified liposomes (Bubble liposomes) containing perfluoropropane, which is an ultrasound imaging gas. Here, we used ultrasound to induce cavitation in Bubble liposomes and then investigated their ability to deliver genes in vitro and in vivo. Bubble liposomes could deliver plasmid DNA to many cell types without cytotoxicity. Additionally, in vivo gene delivery, Bubble liposomes were more effective delivery into femoral artery than lipofection method. Thus, Bubble liposomes might be efficient and novel non-viral tools for gene delivery.
Collapse
Affiliation(s)
- Ryo Suzuki
- Department of Biopharmaceutics, School of Pharmaceutical Sciences, Teikyo University, 1091-1 Suwarashi, Sagamiko-cho, Sagamihara, Kanagawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hu H, Chen D, Liu Y, Deng Y, Yang S, Qiao M, Zhao J, Zhao X. Target ability and therapy efficacy of immunoliposomes using a humanized antihepatoma disulfide-stabilized Fv fragment on tumor cells. J Pharm Sci 2006; 95:192-9. [PMID: 16315243 DOI: 10.1002/jps.20517] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recently the use of peptides in bee venom (PBV) for cancer therapy has attracted considerable attention. However, PBV's extensive use is prohibited by its intense hemolytic activity. In this study, the sterically stabilized liposomal PBV (PBV-SL) was prepared using soybean phosphatidylcholine, cholesterol, and cholesterol-PEG-COOH. The humanized antihepatoma disulfide-stabilized Fv (hdsFv25) was reengineered, expressed, and coupled to sterically stabilized liposomes using the N-hydroxysuccinimide ester method. The hdsFv25-immunoliposomes (SIL [hdscFv25]) were immunoreactive as determined by ELISA assay. PBV-SIL [hdscFv25] can kill SMMC-7721 cells in vitro with higher efficiency than nontargeted liposomes. PBV-SIL [hdsFv25] displayed high antitumor activity and resulted in a significant reduction in tumor size compared to nontargeted liposomes and PBV. These results indicated that this strategy should be applicable to applicable in the treatment of other cancers.
Collapse
Affiliation(s)
- Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, 110016, RP China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Voinea M, Manduteanu I, Dragomir E, Capraru M, Simionescu M. Immunoliposomes directed toward VCAM-1 interact specifically with activated endothelial cells--a potential tool for specific drug delivery. Pharm Res 2005; 22:1906-17. [PMID: 16088429 DOI: 10.1007/s11095-005-7247-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2005] [Accepted: 07/05/2005] [Indexed: 02/02/2023]
Abstract
PURPOSE Immunoliposomes can be potentially used as carriers for drug delivery to specific cells. The aim of this paper was to exploit the overexpression of vascular cell adhesion molecule-1 (VCAM-1) on activated human endothelial cells (HEC) for targeting of anti-VCAM-1 coupled liposomes with the intent for further use as drug carriers. METHODS TNF-alpha-activated HEC were exposed to liposomes, either plain or coupled with antibodies to VCAM-1 (L-VCAM-1) or to irrelevant IgG (L-IgG); nonactivated HEC subjected to the same conditions were used as control. For binding studies, the cells were incubated with fluorescently labeled liposomes at 4 degrees C, and after 2 h, fluorescence intensity was assessed by flow cytometry; specificity of binding was determined by performing the experiments in the presence of excess anti-VCAM-1. Cellular internalization of liposomes was studied employing radioactively or fluorescently labelled liposomes; to detect the mechanisms of uptake, experiments were performed in the presence of agents that interfere in the endocytotic pathway. Transmigration of liposomes was monitored in a two-chamber culture model. The effect of L-VCAM-1 binding to HEC on intracellular calcium ([Ca(2+)](i)) and distribution of actin was determined by fluorimetry and fluorescence microscopy. RESULTS (1) L-VCAM-1 binds selectively and specifically to TNF-alpha activated HEC. (2) Approximately 50% of L-VCAM-1 is taken up by receptor-mediated endocytosis via clathrin-coated vesicles. (3) Binding of L-VCAM-1 to HEC surface induces a rise in [Ca(2+)](i) and reorganization of actin filaments. (4) A small percentage of liposomes migrates across HEC. CONCLUSION The data indicate that VCAM-1 may be an appropriate target for specific drug delivery to activated HEC using immunoliposomes.
Collapse
Affiliation(s)
- Manuela Voinea
- Institute of Cellular Biology and Pathology, Bucharest, Romania.
| | | | | | | | | |
Collapse
|
42
|
Nobs L, Buchegger F, Gurny R, Allémann E. Current methods for attaching targeting ligands to liposomes and nanoparticles. J Pharm Sci 2005; 93:1980-92. [PMID: 15236448 DOI: 10.1002/jps.20098] [Citation(s) in RCA: 212] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Liposomes and nanoparticles have emerged as versatile carrier systems for delivering active molecules in the organism. These colloidal particles have demonstrated enhanced efficacy compared to conventional drugs. However, the design of liposomes and nanoparticles with a prolonged circulation time and ability to deliver active compounds specifically to target sites remains an ongoing research goal. One interesting way to achieve active targeting is to attach ligands, such as monoclonal antibodies or peptides, to the carrier. These surface-bound ligands recognize and bind specifically to target cells. To this end, various techniques have been described, including covalent and noncovalent approaches. Both in vitro and in vivo studies have proved the efficacy of the concept of active targeting. The present review summarizes the most common coupling techniques developed for binding homing moieties to the surface of liposomes and nanoparticles. Various coupling methods, covalent and noncovalent, will be reviewed, with emphasis on the major differences between the coupling reactions, on their advantages and drawbacks, on the coupling efficiency obtained, and on the importance of combining active targeting with long-circulating particles.
Collapse
Affiliation(s)
- Leila Nobs
- School of Pharmacy, University of Geneva, Quai Ernest-Ansermet 30, 1211 4, Switzerland
| | | | | | | |
Collapse
|
43
|
Andresen TL, Jensen SS, Jørgensen K. Advanced strategies in liposomal cancer therapy: problems and prospects of active and tumor specific drug release. Prog Lipid Res 2005; 44:68-97. [PMID: 15748655 DOI: 10.1016/j.plipres.2004.12.001] [Citation(s) in RCA: 400] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Tumor specific drug delivery has become increasingly interesting in cancer therapy, as the use of chemotherapeutics is often limited due to severe side effects. Conventional drug delivery systems have shown low efficiency and a continuous search for more advanced drug delivery principles is therefore of great importance. In the first part of this review, we present current strategies in the drug delivery field, focusing on site-specific triggered drug release from liposomes in cancerous tissue. Currently marketed drug delivery systems lack the ability to actively release the carried drug and rely on passive diffusion or slow non-specific degradation of the liposomal carrier. To obtain elevated tumor-to-normal tissue drug ratios, it is important to develop drug delivery strategies where the liposomal carriers are actively degraded specifically in the tumor tissue. Many promising strategies have emerged ranging from externally triggered light- and thermosensitive liposomes to receptor targeted, pH- and enzymatically triggered liposomes relying on an endogenous trigger mechanism in the cancerous tissue. However, even though several of these strategies were introduced three decades ago, none of them have yet led to marketed drugs and are still far from achieving this goal. The most advanced and prospective technologies are probably the prodrug strategies where non-toxic drugs are carried and activated specifically in the malignant tissue by overexpressed enzymes. In the second part of this paper, we review our own work, exploiting secretory phospholipase A2 as a site-specific trigger and prodrug activator in cancer therapy. We present novel prodrug lipids together with biophysical investigations of liposome systems, constituted by these new lipids and demonstrate their degradability by secretory phospholipase A2. We furthermore give examples of the biological performance of the enzymatically degradable liposomes as advanced drug delivery systems.
Collapse
Affiliation(s)
- Thomas L Andresen
- Department of Chemistry, Technical University of Denmark, Building 207, DK-2800 Lyngby, Denmark.
| | | | | |
Collapse
|
44
|
Wilson A, He F, Li J, Ma Z, Pitt B, Li S. Targeted delivery of therapeutic oligonucleotides to pulmonary circulation. ADVANCES IN GENETICS 2005; 54:21-41. [PMID: 16096006 DOI: 10.1016/s0065-2660(05)54002-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Functional oligodeoxynucleotides (ODN) such as antisense ODN (AS-ODN) show promise as new therapeutics for the treatment of a number of pulmonary diseases. They also hold potential to serve as a research tool for the study of gene function related to lung physiology. The success of their application is largely dependent on the development of an efficient delivery vehicle. This chapter summarizes work toward the development of lipidic vectors for targeted ODN delivery to pulmonary circulation. Recent advancements in the development of novel ODN are also discussed briefly.
Collapse
Affiliation(s)
- Annette Wilson
- Department of Environmental and Occupational Health Graduate School of Public Health, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | | | |
Collapse
|
45
|
Sapra P, Moase EH, Ma J, Allen TM. Improved Therapeutic Responses in a Xenograft Model of Human B Lymphoma (Namalwa) for Liposomal VincristineversusLiposomal Doxorubicin Targeted via Anti-CD19 IgG2a or Fab′ Fragments. Clin Cancer Res 2004; 10:1100-11. [PMID: 14871990 DOI: 10.1158/1078-0432.ccr-03-0041] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Monoclonal antibody-mediated targeting of liposomal anticancer drugs to surface antigens expressed on malignant B cells can be an effective strategy for treating B-cell malignancies. In a murine model of human B-cell lymphoma, we have made in vitro and in vivo comparisons of long-circulating sterically stabilized (Stealth) immunoliposome (SIL) formulations of two anticancer drugs, vincristine (VCR) and doxorubicin (DXR), with different mechanisms of action and drug release rates. EXPERIMENTAL DESIGN SIL formulations of VCR or DXR were conjugated to the monoclonal antibody anti-CD19 (SIL[alphaCD19]) or its Fab' fragments (SIL[Fab']). Specific binding of SILs to Namalwa cells was studied using radiolabeled liposomes, and cytotoxicities of DXR- or VCR-loaded SILs were quantitated by a tetrazolium assay. Pharmacokinetic and drug leakage experiments were performed in mice using dual-labeled liposomes, and the therapeutic responses of SILs were evaluated in a Namalwa (human B lymphoma) cell xenograft model. RESULTS SIL[alphaCD19] or SIL[Fab'] had higher association with and cytotoxicity against Namalwa cells than nontargeted liposomes. SIL[Fab'] had longer circulation times than SIL[alphaCD19], and VCR had faster release rates from the liposomes than DXR. SIL formulations of either VCR or DXR had significantly better therapeutic outcomes than nontargeted liposomes or free drugs. SILs loaded with VCR were superior to those loaded with DXR. SIL[Fab'] had better therapeutic outcomes than SIL[alphaCD19] for the drug DXR but were equally efficacious for the drug VCR. CONCLUSIONS Treatment of a B lymphoma model with single injections of anti-CD19-targeted liposomal formulations of VCR resulted in high levels of response and long-term survivors. Responses to anti-CD19-targeted liposomal DXR were more modest, although the longer circulation times of SIL[Fab'] versus SIL[alphaCD19] led to superior therapeutics for DXR-loaded immunoliposomes.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/pharmacokinetics
- Antibodies, Monoclonal/chemistry
- Antigens, CD19/biosynthesis
- Antineoplastic Agents, Phytogenic/pharmacokinetics
- Cell Line, Tumor
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Doxorubicin/pharmacokinetics
- Female
- Humans
- Immunoglobulin Fragments/chemistry
- Immunoglobulin G/chemistry
- Inhibitory Concentration 50
- Liposomes/chemistry
- Liposomes/metabolism
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/pathology
- Mice
- Mice, Inbred BALB C
- Mice, SCID
- Neoplasm Transplantation
- Tetrazolium Salts/pharmacology
- Thiazoles/pharmacology
- Time Factors
- Vincristine/pharmacokinetics
Collapse
Affiliation(s)
- Puja Sapra
- Department of Pharmacology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | | | | | | |
Collapse
|
46
|
Schnyder A, Krähenbühl S, Török M, Drewe J, Huwyler J. Targeting of skeletal muscle in vitro using biotinylated immunoliposomes. Biochem J 2004; 377:61-7. [PMID: 14516278 PMCID: PMC1223851 DOI: 10.1042/bj20031034] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2003] [Revised: 09/16/2003] [Accepted: 09/30/2003] [Indexed: 11/17/2022]
Abstract
In the present study, a non-covalent (biotin-streptavidin) coupling procedure for the preparation of pegylated immunoliposomes is presented, which simplifies the attachment of targeting vectors to sterically stabilized liposomes. A biotinylated poly(ethylene glycol) (PEG)-phospholipid [bio-PEG-distearoylphosphatidylethanolamine (DSPE)] was used as a linker between a streptavidin-conjugated monoclonal antibody (mAb) (i.e. the OX26 mAb raised against the rat transferrin receptor) and 150 nm liposomes. OX26-streptavidin had a biotin binding capacity of two to three biotin molecules per OX26-streptavidin conjugate. Immunostaining experiments with the OX26 mAb followed by fluorescent confocal microscopy revealed immunofluorescence labelling of the transferrin receptor on skeletal muscle, as well as in L6 cells, a continuous cell line derived from rat skeletal muscle. Uptake experiments with L6 cells using the OX26 mAb, fluorescence-labelled OX26-streptavidin or fluorescent OX26-immunoliposomes demonstrated cellular uptake and accumulation within an intracellular compartment of the OX26 mAb and its conjugates. Cellular uptake of OX26 conjugates was sensitive to competition with free OX26 antibody. In summary, these studies describe the design of biotinylated immunoliposomes as a universal drug transport vector and their potential for targeting of the transferrin receptor of skeletal muscle.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Biotinylation
- Cell Line
- Drug Delivery Systems
- Liposomes/chemistry
- Microscopy, Confocal
- Microscopy, Fluorescence
- Muscle, Skeletal/chemistry
- Muscle, Skeletal/metabolism
- Phosphatidylethanolamines/chemistry
- Polyethylene Glycols/chemistry
- Rats
- Receptors, Transferrin/analysis
- Receptors, Transferrin/immunology
- Receptors, Transferrin/metabolism
Collapse
Affiliation(s)
- Anita Schnyder
- Department of Research and Division of Clinical Pharmacology, University Hospital Basel, CH-4031 Basel, Switzerland
| | | | | | | | | |
Collapse
|
47
|
Abstract
Antibody or ligand-mediated targeting of liposomal anticancer drugs to antigens expressed selectively or over-expressed on tumor cells is increasingly being recognized as an effective strategy for increasing the therapeutic indices of anticancer drugs. This review summarizes some recent advances in the field of ligand-targeted liposomes (LTLs) for the delivery of anticancer drugs. New approaches used in the design and optimization of LTLs is discussed and the advantages and potential problems associated with their therapeutic applications are described. New technologies are widening the spectrum of ligands available for targeting and are allowing choices to be made regarding affinity, internalization and size. The time is rapidly approaching where we will see translation of anticancer drugs entrapped in LTLs to the clinic.
Collapse
Affiliation(s)
- P Sapra
- Department of Pharmacology, University of Alberta, Edmonton AB, Canada, T6G 2H7
| | | |
Collapse
|
48
|
Voinea M, Dragomir E, Manduteanu I, Simionescu M. Binding and uptake of transferrin-bound liposomes targeted to transferrin receptors of endothelial cells. Vascul Pharmacol 2002; 39:13-20. [PMID: 12616986 DOI: 10.1016/s1537-1891(02)00165-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The use of liposomes as carriers for site-specific delivery is an attractive strategy, especially for the vascular endothelium that by position is an accessible target for drug and gene delivery via the blood circulation. The aim of this study was to detect whether liposomes coupled to transferrin (Tf)-bound and are taken up by aortic endothelial cells (EC) following the pathway of Tf interaction with transferrin receptors, reportedly expressed on their cell membrane. To this purpose, small unilamellar liposomes of different compositions, either classical (C) or sterically stabilized (SS), have been prepared, characterized and coupled with transferrin (Tf-liposomes). To assess the binding and uptake, cultured EC were incubated with fluorescently labelled Tf-liposomes for various times intervals (from 5 min to 24 h) at 4 and 37 degrees C, and further investigated by flow cytometry, fluorimetry and fluorescence microscopy. The results showed that: (i) binding of Tf-liposomes to EC was specific; (ii) the EC binding of SS-Tf-liposomes was lower than that of C-Tf-liposomes; and (iii) after 30 min of incubation, both C- and SS-Tf-liposomes appeared localized in the acidic compartments of the cells. Together, the data indicate that transferrin-bound liposomes are specifically taken up by EC by a receptor-mediated mechanism employing the pathway of surface-exposed Tf receptors.
Collapse
Affiliation(s)
- Manuela Voinea
- Institute of Cellular Biology and Pathology Nicolae Simionescu, 8 BP Hasdeu Street, PO Box 35-14, 79691 Bucharest, Romania
| | | | | | | |
Collapse
|
49
|
Hofland HEJ, Masson C, Iginla S, Osetinsky I, Reddy JA, Leamon CP, Scherman D, Bessodes M, Wils P. Folate-targeted gene transfer in vivo. Mol Ther 2002; 5:739-44. [PMID: 12027558 DOI: 10.1006/mthe.2002.0604] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nonviral systemic delivery is one of the most attractive approaches for cancer gene therapy. To achieve this goal, various laboratories have developed cationic liposomes. However, when injected intravenously, cationic lipid-DNA complexes accumulate mostly into and transfect lung tissue. Here, we describe a method by which these complexes can be targeted to tumors using folic acid. Adding polyethylene glycol (PEG)-lipids to the complexes dramatically reduced both lung accumulation and gene transfer to lungs and tumors after intravenous administration. The presence of folic acid at the distal end of the PEG-lipid did not modify tumor accumulation of the complexes. However, with folate-targeted complexes, gene transfer activity was restored in tumors while the activity in lungs was reduced by 50- to 100-fold compared with nontargeted lipid-DNA complexes. This approach provides a first in vivo proof of concept to achieve targeted tumor gene delivery.
Collapse
Affiliation(s)
- Hans E J Hofland
- Gencell, Aventis Pharma, 3825 Bay Center Place, Hayward, California 94545, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ishida O, Maruyama K, Tanahashi H, Iwatsuru M, Sasaki K, Eriguchi M, Yanagie H. Liposomes bearing polyethyleneglycol-coupled transferrin with intracellular targeting property to the solid tumors in vivo. Pharm Res 2001; 18:1042-8. [PMID: 11496943 DOI: 10.1023/a:1010960900254] [Citation(s) in RCA: 176] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE The purpose of this study was to determine the usefulness of transferrin (TF)-pendant-type polyethyleneglycol (PEG)-liposomes (TF-PEG-liposomes), in which TF was covalently linked to the distal terminal of PEG chains on the external surface of PEG-liposomes as a carrier for in vivo cytoplasmic targeting to tumor cells. METHODS Small unilamellar TF-PEG-liposomes (100-140 nm in diameter) were prepared from DSPC, CH, DSPE-PEG, and DSPE-PEG-COOH (2:1:0.11:0.021, molar ratio), and were conjugated to TF via the carboxyl residue of DSPE-PEG-COOH. The intracellular targeting ability of TF-PEG-liposomes to tumor cells was examined in vitro and in Colon 26 tumor-bearing mice. RESULTS TF-PEG-liposomes, bearing approximately 25 TF molecules per liposome, readily bound to mouse Colon 26 cells in vitro and were internalized by receptor-mediated endocytosis. TF-PEG-liposomes showed a prolonged residence time in the circulation and low RES uptake in Colon 26 tumor-bearing mice, resulting in enhanced extravasation of the liposomes into the solid tumor tissue. Electron microscopic studies in Colon 26 tumor-bearing mice revealed that the extravasated TF-PEG-liposomes were internalized into tumor cells by receptor-mediated endocytosis. CONCLUSION TF-PEG-liposomes had the capabilities of specific receptor binding and receptor-mediated endocytosis to target cells after extravasation into solid tumors in vivo. Such liposomes should be useful for in vivo cytoplasmic targeting of chemotherapeutic agents or plasmid DNAs to target cells.
Collapse
Affiliation(s)
- O Ishida
- Faculty of Pharmaceutical Sciences, Teikyo University, Kanagawa, Japan
| | | | | | | | | | | | | |
Collapse
|