1
|
Sukocheva OA, Liu J, Neganova ME, Beeraka NM, Aleksandrova YR, Manogaran P, Grigorevskikh EM, Chubarev VN, Fan R. Perspectives of using microRNA-loaded nanocarriers for epigenetic reprogramming of drug resistant colorectal cancers. Semin Cancer Biol 2022; 86:358-375. [PMID: 35623562 DOI: 10.1016/j.semcancer.2022.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/07/2023]
Abstract
Epigenetic regulation by microRNAs (miRs) demonstrated a promising therapeutic potential of these molecules to regulate genetic activity in different cancers, including colorectal cancers (CRCs). The RNA-based therapy does not change genetic codes in tumor cells but can silence oncogenes and/or reactivate inhibited tumor suppressor genes. In many cancers, specific miRs were shown to promote or stop tumor progression. Among confirmed and powerful epigenetic regulators of colon carcinogenesis and development of resistance are onco-miRs, which include let-7, miR-21, miR-22, miR-23a, miR-27a, miR-34, miR-92, miR-96, miR-125b, miR-135b, miR-182, miR-200c, miR-203, miR-221, miR-421, miR-451, and others. Moreover, various tumor-suppressor miRs (miR-15b-5b, miR-18a, miR-20b, miR-22, miR-96, miR-139-5p, miR-145, miR-149, miR-197, miR-199b, miR-203, miR-214, miR-218, miR-320, miR-375-3p, miR-409-3p, miR-450b-5p, miR-494, miR-577, miR-874, and others) were found silenced in drug-resistant CRCs. Re-expression of tumor suppressor miR is complicated by the chemical nature of miRs that are not long-lasting compounds and require protection from the enzymatic degradation. Several recent studies explored application of miRs using nanocarrier complexes. This study critically describes the most successfully tested nanoparticle complexes used for intracellular delivery of nuclear acids and miRs, including micelles, liposomes, inorganic and polymeric NPs, dendrimers, and aptamers. Nanocarriers shield incorporated miRs and improve the agent stability in circulation. Attachment of antibodies and/or specific peptide or ligands facilitates cell-targeted miR delivery. Addressing in vivo challenges, a broad spectrum of non-toxic materials has been tested and indicated reliable advantages of lipid-based (lipoplexes) and polymer-based liposomes. Recent cutting-edge developments indicated that lipid-based complexes with multiple cargo, including several miRs, are the most effective approach to eradicate drug-resistant tumors. Focusing on CRC-specific miRs, this review provides a guidance and insights towards the most promising direction to achieve dramatic reduction in tumor growth and metastasis using miR-nanocarrier complexes.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China; The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Queensland, Australia; Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA 5042, Australia.
| | - Junqi Liu
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka 142432, Russia
| | - Narasimha M Beeraka
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China; Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow 119991, Russia; Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical College, Mysuru, Karnataka, India
| | - Yulia R Aleksandrova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka 142432, Russia
| | - Prasath Manogaran
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Ekaterina M Grigorevskikh
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow 119991, Russia
| | - Vladimir N Chubarev
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow 119991, Russia
| | - Ruitai Fan
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China.
| |
Collapse
|
2
|
Zarei H, Malaekeh-Nikouei B, Ramezani M, Soltani F. Multifunctional peptides based on low molecular weight protamine (LMWP) in the structure of polyplexes and lipopolyplexes: Design, preparation and gene delivery characterization. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
3
|
Surface engineering of nanomaterials with phospholipid-polyethylene glycol-derived functional conjugates for molecular imaging and targeted therapy. Biomaterials 2019; 230:119646. [PMID: 31787335 DOI: 10.1016/j.biomaterials.2019.119646] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 11/16/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
Abstract
In recent years, phospholipid-polyethylene glycol-derived functional conjugates have been widely employed to decorate different nanomaterials, due to their excellent biocompatibility, long blood circulation characteristics, and specific targeting capability. Numerous in vivo studies have demonstrated that nanomedicines peripherally engineered with phospholipid-polyethylene glycol-derived functional conjugates show significantly increased selective and efficient internalization by target cells/tissues. Targeting moieties including small-molecule ligands, peptides, proteins, and antibodies are generally conjugated onto PEGylated phospholipids to decorate liposomes, micelles, hybrid nanoparticles, nanocomplexes, and nanoemulsions for targeted delivery of diagnostic and therapeutic agents to diseased sites. In this review, the synthesis methods of phospholipid-polyethylene glycol-derived functional conjugates, biophysicochemical properties of nanomedicines decorated with these conjugates, factors dominating their targeting efficiency, as well as their applications for in vivo molecular imaging and targeted therapy were summarized and discussed.
Collapse
|
4
|
Abstract
Liposomes are one of the most widely investigated carriers for CRISPR/Cas9 delivery. The surface properties of liposomal carriers, including the surface charge, PEGylation, and ligand modification can significantly affect the gene silencing efficiency. Three barriers of systemic CRISPR/Cas9 delivery (long blood circulation, efficient tumor penetration, and efficient cellular uptake/endosomal escape) are analyzed on liposomal carriers with different surface charges, PEGylations, and ligand modifications. Cationic formulations dominate CRISPR/Cas9 delivery and neutral formulations also have good performance while anionic formulations are generally not proper for CRISPR/Cas9 delivery. The PEG dilemma (prolonged blood circulation vs. reduced cellular uptake/endosomal escape) and the side effect of repeated PEGylated formulation (accelerated blood clearance) were discussed. Effects of ligand modification on cationic and neutral formulations were analyzed. Finally, we summarized the achievements in liposomal CRISPR/Cas9 delivery, outlined existing problems, and provided some future perspectives. Liposomes are one of the most widely investigated carriers for CRISPR/Cas9 delivery. The surface properties of liposomal carriers, including the surface charge, PEGylation, and ligand modification can significantly affect the gene silencing efficiency. Three barriers of systemic siRNA delivery (long blood circulation, efficient tumor penetration, and efficient cellular uptake/endosomal escape) are analyzed on liposomal carriers with different surface charges, PEGylations, and ligand modifications. Cationic formulations dominate CRISPR/Cas9 delivery and neutral formulations also have good performance while anionic formulations are generally not proper for CRISPR/Cas9 delivery. The PEG dilemma (prolonged blood circulation vs. reduced cellular uptake/endosomal escape) and the side effect of repeated PEGylated formulation (accelerated blood clearance) were discussed. Effects of ligand modification on cationic and neutral formulations were analyzed. Finally, we summarized the achievements in liposomal CRISPR/Cas9 delivery, outlined existing problems, and provided some future perspectives.
Collapse
|
5
|
Mohammed-Saeid W, Chitanda J, Al-Dulaymi M, Verrall R, Badea I. Design and Evaluation of RGD-Modified Gemini Surfactant-Based Lipoplexes for Targeted Gene Therapy in Melanoma Model. Pharm Res 2017. [PMID: 28643235 DOI: 10.1007/s11095-017-2197-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE We have developed and evaluated novel peptide-targeted gemini surfactant-based lipoplexes designed for melanoma gene therapy. METHODS Integrin receptor targeting peptide, cyclic-arginylglycylaspartic acid (cRGD), was either chemically coupled to a gemini surfactant backbone or physically co-formulated with lipoplexes. Several formulations and transfection techniques were developed. Transfection efficiency and cellular toxicity of the lipoplexes were evaluated in an in vitro human melanoma model. Physicochemical properties were examined using dynamic light scattering, zeta-potential, and small-angle X-ray scattering measurements. RESULTS RGD-modified gemini surfactant based lipoplexes showed significant enhancement in gene transfection activity in A375 cell lines compared to the standard non-targeted formulation, especially when RGD was chemically conjugated to the gemini surfactant (RGD-G). The RGD had no effect on the cell toxicity profile of the lipoplex systems. Targeting specificity was confirmed by using an excess of free RGD and negative control peptide (RAD) and was demonstrated by using normal human epidermal keratinocytes. Physicochemical characterization showed that all nanoparticles were in the optimal size range for cellular uptake and there were no significant differences between RGD-modified and standard lipoplexes. CONCLUSIONS These findings indicate the potential of RGD-modified gemini surfactant-based lipoplexes for use in melanoma gene therapy as an alternative to conventional chemotherapy.
Collapse
Affiliation(s)
- Waleed Mohammed-Saeid
- Drug Design and Discovery Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Room 3D01.5, Saskatoon, Saskatchewan, S7N 5E5, Canada.,College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Jackson Chitanda
- Department of Chemical and Biological Engineering, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Mays Al-Dulaymi
- Drug Design and Discovery Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Room 3D01.5, Saskatoon, Saskatchewan, S7N 5E5, Canada
| | - Ronald Verrall
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ildiko Badea
- Drug Design and Discovery Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Room 3D01.5, Saskatoon, Saskatchewan, S7N 5E5, Canada.
| |
Collapse
|
6
|
Gjetting T, Jølck RI, Andresen TL. Effective nanoparticle-based gene delivery by a protease triggered charge switch. Adv Healthc Mater 2014; 3:1107-18. [PMID: 24652709 DOI: 10.1002/adhm.201300503] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 12/10/2013] [Indexed: 11/06/2022]
Abstract
Gene carriers made from synthetic materials are of interest in relation to gene therapy but suffer from lack of transfection efficiency upon systemic delivery. To address this problem, a novel lipo-peptide-PEG conjugate constituted by a lipid-anchor, a peptide sensitive to proteases and a poly (ethylene glycol) (PEG) chain is investigated. Utilizing ethanol-mediated nucleic acid encapsulation to prepare lipo-nanoparticles (LNPs), LNPs that are stable in serum are obtained. The LNPs constitute a highly effective gene delivery systems in vitro and possess the right features for further investigation in vivo including a PEG layer and a net negative charge that should ensure long-circulating properties before being activated by proteases in diseased tissue. Protease activation leads to detachment of PEG and a charge switching where the LNPs become positive due to the presence of glutamates in the cleaved peptide moiety. The cationic lipid DOTAP is used mainly to complex DNA and proton titratable DODAP is used to increase endosomal escape and enhance transfection efficiency. The idea of using a mixture of permanently charged and titratable cationic lipids shielded by a protease sensitive negatively charged lipo-peptide-PEG coat appears to be a highly efficient solution for achieving effective non-viral gene delivery and the results warrant further investigations.
Collapse
Affiliation(s)
- Torben Gjetting
- Technical University of Denmark, DTU Nanotech Department of Micro- and Nanotechnology; Center for Nanomedicine and Theranostics; Building 423 2800 Lyngby Denmark
| | - Rasmus Irming Jølck
- Technical University of Denmark, DTU Nanotech Department of Micro- and Nanotechnology; Center for Nanomedicine and Theranostics; Building 423 2800 Lyngby Denmark
| | - Thomas Lars Andresen
- Technical University of Denmark, DTU Nanotech Department of Micro- and Nanotechnology; Center for Nanomedicine and Theranostics; Building 423 2800 Lyngby Denmark
| |
Collapse
|
7
|
Levine RM, Pearce TR, Adil M, Kokkoli E. Preparation and characterization of liposome-encapsulated plasmid DNA for gene delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2013; 29:9208-9215. [PMID: 23837701 DOI: 10.1021/la400859e] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The success of common nonviral gene delivery vehicles, lipoplexes and polyplexes, is limited by the toxicity and instability of these charged molecules. Stealth liposomes could provide a stable, safe alternative to cationic DNA complexes for effective gene delivery. DNA encapsulations in three stealth liposomal formulations prepared by thin film, reverse phase evaporation, and asymmetric liposome formation were compared, and the thin film method was found to produce the highest yields of encapsulated DNA. A DNA quantification method appropriate for DNA encapsulated within liposomes was also developed and verified for accuracy. The effect of initial lipid and DNA concentrations on the encapsulation yield and fraction of DNA-filled liposomes was evaluated. Higher encapsulation yields were achieved by higher lipid contents, while a higher fraction of DNA-filled liposomes was produced by either lower lipid content or higher DNA concentration. Control of these parameters allows for the design of gene delivery nanoparticles with high DNA encapsulation yields or higher fraction of DNA-filled liposomes.
Collapse
Affiliation(s)
- Rachel M Levine
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
8
|
Levine RM, Scott CM, Kokkoli E. Peptide functionalized nanoparticles for nonviral gene delivery. SOFT MATTER 2013; 9:985-1004. [DOI: 10.1039/c2sm26633d] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
9
|
Nimesh S. Protamine nanoparticles. Gene Ther 2013. [DOI: 10.1533/9781908818645.237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
10
|
A review of RGD-functionalized nonviral gene delivery vectors for cancer therapy. Cancer Gene Ther 2012; 19:741-8. [PMID: 23018622 DOI: 10.1038/cgt.2012.64] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The development of effective treatments that enable many patients suffering from cancer to be successfully cured is highly demanded. Angiogenesis, which is a process for the formation of new capillary blood vessels, has a crucial role in solid tumor progression and the development of metastasis. Antiangiogenic therapy designed to prevent tumor angiogenesis, thereby arresting the growth or spread of tumors, has emerged as a non-invasive and safe option for cancer treatment. Due to the fact that integrin receptors are overexpressed on the surface of angiogenic endothelial cells, various strategies have been made to develop targeted delivery systems for cancer gene therapy utilizing integrin-targeting peptides with an exposed arginine-glycine-aspartate (RGD) sequence. The aim of this review is to summarize the progress and prospect of RGD-functionalized nonviral vectors toward targeted delivery of genetic materials in order to achieve an efficient therapeutic outcome for cancer gene therapy, including antiangiogenic therapy.
Collapse
|
11
|
Zohra FT, Maitani Y, Akaike T. mRNA delivery through fibronectin associated liposome-apatite particles: a new approach for enhanced mRNA transfection to mammalian cell. Biol Pharm Bull 2012; 35:111-5. [PMID: 22223346 DOI: 10.1248/bpb.35.111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It was believed for a long time that mRNA is very unstable, and can not be used for therapeutic purposes. In the last decade, however, many research groups proved its transfection feasibility along with advantages and applications. Our investigation is aimed at establishing a potent and efficient mRNA delivery system. We previously reported that an inorganic-organic hybrid carrier by exploiting the advantages of inorganic nano apatite particles onto organic carrier DOTAP {N-[1-(2,3-dioleoloxy)propyl]-N,N,N-trimethyl ammonium chloride} and showed potential effect of carbonate apatite particles on each of the mRNA delivery steps in dividing and non-dividing cell. Here, we report on the development of a more efficient mRNA carrier by complexing ECM protein, fibronectin with the DOTAP-apatite carrier. The carrier showed enhanced uptake of luciferase mRNA both qualitatively and quantitatively. Accelerated cellular endocytosis rate was evaluated using labeled endosome. Finally expression of lucifearse mRNA was higher for fibronectin complexed carrier in compared to the uncoated one.
Collapse
Affiliation(s)
- Fatema Tuj Zohra
- Department of Biomolecular Engineering, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226–8501, Japan
| | | | | |
Collapse
|
12
|
Ligands located within a cholesterol domain enhance gene delivery to the target tissue. J Control Release 2012; 160:57-63. [PMID: 22440429 DOI: 10.1016/j.jconrel.2012.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 03/04/2012] [Indexed: 11/23/2022]
Abstract
Targeted gene delivery provides enormous potential for clinical treatment of many incurable diseases. Liposomes formulated with targeting ligands have been tested extensively both in vitro and in vivo, and many studies have strived to identify more efficacious ligands. However, the environment of the ligand within the delivery vehicle is generally not considered, and this study assesses the effect of ligand microenvironment by utilizing a lipoplex possessing a cholesterol domain. Our recent work has shown that the presence of the targeting ligand within the cholesterol domain promotes more productive transfection in cultured cells. In the present study, lipoplexes having the identical lipid composition were formulated with different conjugates of the folate ligand such that the ligand was included in, or excluded from, the cholesterol domain. The effect of locating the ligand within the cholesterol domain was then tested in a xenograft tumor model in mice. Lipoplexes that included the ligand within the cholesterol domain showed significantly higher luciferase expression and plasmid accumulation in tumors as compared to lipoplexes in which the ligand was excluded from the domain. These results demonstrate that the microenvironment of the ligand can affect gene delivery to tumors, and show that ligand-mediated delivery can be enhanced by locating targeting ligands within a cholesterol domain.
Collapse
|
13
|
Hsu CYM, Uludağ H. Nucleic-acid based gene therapeutics: delivery challenges and modular design of nonviral gene carriers and expression cassettes to overcome intracellular barriers for sustained targeted expression. J Drug Target 2012; 20:301-28. [PMID: 22303844 DOI: 10.3109/1061186x.2012.655247] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The delivery of nucleic acid molecules into cells to alter physiological functions at the genetic level is a powerful approach to treat a wide range of inherited and acquired disorders. Biocompatible materials such as cationic polymers, lipids, and peptides are being explored as safer alternatives to viral gene carriers. However, the comparatively low efficiency of nonviral carriers currently hampers their translation into clinical settings. Controlling the size and stability of carrier/nucleic acid complexes is one of the primary hurdles as the physicochemical properties of the complexes can define the uptake pathways, which dictate intracellular routing, endosomal processing, and nucleocytoplasmic transport. In addition to nuclear import, subnuclear trafficking, posttranscriptional events, and immune responses can further limit transfection efficiency. Chemical moieties, reactive linkers or signal peptide have been conjugated to carriers to prevent aggregation, induce membrane destabilization and localize to subcellular compartments. Genetic elements can be inserted into the expression cassette to facilitate nuclear targeting, delimit expression to targeted tissue, and modulate transgene expression. The modular option afforded by both gene carriers and expression cassettes provides a two-tier multicomponent delivery system that can be optimized for targeted gene delivery in a variety of settings.
Collapse
Affiliation(s)
- Charlie Yu Ming Hsu
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Cananda
| | | |
Collapse
|
14
|
Xu L, Anchordoquy T. Drug delivery trends in clinical trials and translational medicine: challenges and opportunities in the delivery of nucleic acid-based therapeutics. J Pharm Sci 2011; 100:38-52. [PMID: 20575003 DOI: 10.1002/jps.22243] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The ability to deliver nucleic acids (e.g., plasmid DNA, antisense oligonucleotides, siRNA) offers the potential to develop potent vaccines and novel therapeutics. However, nucleic acid-based therapeutics are still in their early stages as a new category of biologics. The efficacy of nucleic acids requires that these molecules be delivered to the interior of the target cell, which greatly complicates delivery strategies and compromises efficiency. Due to the safety concerns of viral vectors, synthetic vectors such as liposomes and polymers are preferred for the delivery of nucleic acid-based therapeutics. Yet, delivery efficiencies of synthetic vectors in the clinic are still too low to obtain therapeutic levels of gene expression. In this review, we focus on some key issues in the field of nucleic acid delivery such as PEGylation, encapsulation and targeted delivery and provide some perspectives for consideration in the development of improved synthetic vectors.
Collapse
Affiliation(s)
- Long Xu
- Department of Pharmaceutical Sciences, University of Colorado, 12700 East Nineteenth Avenue, Aurora, Colorado 80045, USA
| | | |
Collapse
|
15
|
Abstract
Integrins have become key targets for molecular imaging and for selective delivery of anti-cancer agents. Here we review recent work concerning the targeted delivery of antisense and siRNA oligonucleotides via integrins. A variety of approaches have been used to link oligonucleotides to ligands capable of binding integrins with high specificity and affinity. This includes direct chemical conjugation, incorporating oligonucleotides into lipoplexes, and use of various polymeric nanocarriers including dendrimers. The ligand-oligonucleotide conjugate or complex associates selectively with the integrin, followed by internalization into endosomes and trafficking through subcellular compartments. Escape of antisense or siRNA from the endosome to the cytosol and nucleus may come about through endogenous trafficking mechanisms, or because of membrane disrupting capabilities built into the conjugate or complex. Thus a variety of useful strategies are available for using integrins to enhance the pharmacological efficacy of therapeutic oligonucleotides.
Collapse
|
16
|
Xu L, Anchordoquy TJ. Effect of cholesterol nanodomains on the targeting of lipid-based gene delivery in cultured cells. Mol Pharm 2010; 7:1311-7. [PMID: 20568694 DOI: 10.1021/mp100097b] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Targeted gene delivery offers immense potential for clinical applications. Liposomes decorated with targeting ligands have been extensively used for both in vitro and in vivo gene delivery. Lipoplexes with high cholesterol content that result in cholesterol domain formation within the complexes have been shown to exhibit enhanced transfection in vitro and resistance to serum-induced aggregation. In the present study, folate was employed as a targeting ligand that was conjugated with either cholesterol or a diacyl lipid (DSPE), and these conjugates were incorporated into lipoplexes formulated with DOTAP/cholesterol (wt/wt: 31/69) that are known to possess cholesterol nanodomains. Cellular uptake and transfection of these lipoplexes in the presence of 50% serum were examined when the ligand was located within or excluded from the cholesterol nanodomain. Lipoplexes with folate-cholesterol exhibited a 50-fold increase in transfection compared to those with folate-DSPE, while the cellular uptake level is only 40% of that with folate-DSPE. These results indicate that the presence of the ligand within the cholesterol domain promotes more productive transfection in cultured cells, and intracellular trafficking of the lipoplexes after entry into cells plays a crucial role in gene delivery.
Collapse
Affiliation(s)
- Long Xu
- School of Pharmacy, University of Colorado Denver, 12700 East 19th Avenue, Aurora, Colorado 80045, USA
| | | |
Collapse
|
17
|
Brown KC. Peptidic tumor targeting agents: the road from phage display peptide selections to clinical applications. Curr Pharm Des 2010; 16:1040-54. [PMID: 20030617 DOI: 10.2174/138161210790963788] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 09/25/2009] [Indexed: 11/22/2022]
Abstract
Cancer has become the number one cause of death amongst Americans, killing approximately 1,600 people per day. Novel methods for early detection and the development of effective treatments are an eminent priority in medicine. For this reason, isolation of tumor-specific ligands is a growing area of research. Tumor-specific binding agents can be used to probe the tumor cell surface phenotype and to customize treatment accordingly by conjugating the appropriate cell-targeting ligand to an anticancer drug. This refines the molecular diagnosis of the tumor and creates guided drugs that can target the tumor while sparing healthy tissues. Additionally, these targeting agents can be used as in vivo imaging agents that allow for earlier detection of tumors and micrometastasis. Phage display is a powerful technique for the isolation of peptides that bind to a particular target with high affinity and specificity. The biopanning of intact cancer cells or tumors in animals can be used as the bait to isolate peptides that bind to cancer-specific cell surface biomarkers. Over the past 10 years, unbiased biopanning of phage-displayed peptide libraries has generated a suite of cancer targeting peptidic ligands. This review discusses the recent advances in the isolation of cancer-targeting peptides by unbiased biopanning methods and highlights the use of the isolated peptides in clinical applications.
Collapse
Affiliation(s)
- Kathlynn C Brown
- Division of Translational Medicine Departments of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9185, USA.
| |
Collapse
|
18
|
Abstract
IMPORTANCE OF THE FIELD Recently, there has been substantial progress in the development of integrin targeted pharmaceuticals and drug delivery systems. Integrin is an important member in the cell adhesion molecule family, which is involved in regulation of complex biological conditions, from keeping normal physiological activities to causing cellular dysfunction in diseased cells. Hence, it is timely to summarize the recent developments in integrin targeted drug and gene delivery systems to understand better their advantages and limitations. AREAS COVERED IN THIS REVIEW In this review, advances in the discovery and clinical trials of these integrin antagonists against different integrin subunits are summarized and discussed. Besides using integrin inhibitor as a single therapeutic agent, integrin antagonists that were conjugated to cytotoxic drugs by synthetic chemistry or coupled to biomacromolecules by either DNA recombination technology or fusion protein technology for integrin targeted therapy have been explored. Furthermore, nanoparticles with integrin targeting ligands for both drug and gene delivery, typically for antiangiogenesis and anticancer therapy, are highlighted and evaluated. WHAT THE READER WILL GAIN This review sheds light on the future development of integrin targeted drug and/or gene delivery systems. TAKE HOME MESSAGE Although thus far there are still limitations, integrin targeted delivery systems have already shown their potential as important pharmaceuticals in the near future.
Collapse
Affiliation(s)
- Zhe Wang
- National University of Singapore, Department of Pharmacy, 18 Science Drive 4, Singapore 117543, Singapore
| | | | | |
Collapse
|
19
|
ZHOU QINGHUI, YOU YEZI, WU CHAO, HUANG YI, OUPICKÝ DAVID. Cyclic RGD-targeting of reversibly stabilized DNA nanoparticles enhances cell uptake and transfection in vitro. J Drug Target 2009; 17:364-73. [PMID: 19263264 PMCID: PMC4655816 DOI: 10.1080/10611860902807046] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Reversibly stabilized DNA nanoparticles (rSDN) were prepared by coating reducible polycation/DNA complexes with multivalent N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers. RGD-targeted rSDN were formulated by linking cyclic c(RGDyK) to the surface layer of rSDN. Cellular uptake in B16F10 mouse melanoma cells, human umbilical vein endothelial cells (HUVEC), and THLE immortalized hepatic cells was quantified by real-time PCR. RGD-targeted rSDN exhibited approximately twofold higher cell uptake in integrin-positive cells: B16F10 and HUVEC compared to THLE cells with low integrin content. RGD-targeting mediated increased transfection activity in B16F10 cells but not in THLE cells. Overall, the studies show that rSDN can be effectively targeted with RGD while exhibiting reduced nonspecific cell interactions and favorable stability. As such, these gene delivery vectors have the potential to permit targeting therapeutic genes to tumors by systemic delivery. In addition, the study shows that real-time PCR could be used effectively for the quantification of cellular uptake of gene delivery vectors.
Collapse
Affiliation(s)
- QING-HUI ZHOU
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, Tel: 313-577-6511; Fax: 313-577-2033
| | - YE-ZI YOU
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, Tel: 313-577-6511; Fax: 313-577-2033
| | - CHAO WU
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, Tel: 313-577-6511; Fax: 313-577-2033
| | - YI HUANG
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, Tel: 313-577-6511; Fax: 313-577-2033
| | - DAVID OUPICKÝ
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, Tel: 313-577-6511; Fax: 313-577-2033
| |
Collapse
|
20
|
Abstract
This article provides an overview of principles and barriers relevant to intracellular drug and gene transport, accumulation and retention (collectively called as drug delivery) by means of nanovehicles (NV). The aim is to deliver a cargo to a particular intracellular site, if possible, to exert a local action. Some of the principles discussed in this article apply to noncolloidal drugs that are not permeable to the plasma membrane or to the blood-brain barrier. NV are defined as a wide range of nanosized particles leading to colloidal objects which are capable of entering cells and tissues and delivering a cargo intracelullarly. Different localization and targeting means are discussed. Limited discussion on pharmacokinetics and pharmacodynamics is also presented. NVs are contrasted to micro-delivery and current nanotechnologies which are already in commercial use. Newer developments in NV technologies are outlined and future applications are stressed. We also briefly review the existing modeling tools and approaches to quantitatively describe the behavior of targeted NV within the vascular and tumor compartments, an area of particular importance. While we list "elementary" phenomena related to different level of complexity of delivery to cancer, we also stress importance of multi-scale modeling and bottom-up systems biology approach.
Collapse
Affiliation(s)
- Ales Prokop
- Department of Chemical Engineering, 24th Avenue & Garland Avenues, 107 Olin Hall, Vanderbilt University, Nashville, Tennessee 37235, USA.
| | | |
Collapse
|
21
|
Li W, Szoka FC. Lipid-based nanoparticles for nucleic acid delivery. Pharm Res 2007; 24:438-49. [PMID: 17252188 DOI: 10.1007/s11095-006-9180-5] [Citation(s) in RCA: 447] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Accepted: 10/16/2006] [Indexed: 01/13/2023]
Abstract
Lipid-based colloidal particles have been extensively studied as systemic gene delivery carriers. The topic that we would like to emphasize is the formulation/assembly of lipid-based nanoparticles (NP) with diameter under 100 nm for delivering nucleic acid in vivo. NP are different from cationic lipid-nucleic acid complexes (lipoplexes) and are vesicles composed of lipids and encapsulated nucleic acids with a diameter less than 100 nm. The diameter of the NP is an important attribute to enable NP to overcome the various in vivo barriers for systemic gene delivery such as: the blood components, reticuloendothelial system (RES) uptake, tumor access, extracellular matrix components, and intracellular barriers. The major formulation factors that impact the diameter and encapsulation efficiency of DNA-containing NP include the lipid composition, nucleic acid to lipid ratio and formulation method. The particle assembly step is a critical one to make NP suitable for in vivo gene delivery. NP are often prepared using a dialysis method either from an aqueous-detergent or aqueous-organic solvent mixture. The resulting particles have diameters about 100 nm and nucleic acid encapsulation ratios are >80%. Additional components can then be added to the particle after it is formed. This ordered assembly strategy enables one to optimize the particle physico-chemical attributes to devise a biocompatible particle with increased gene transfer efficacy in vivo. The components included in the sequentially assembled NP include: poly(ethylene glycol) (PEG)-shielding to improve the particle pharmacokinetic behavior, a targeting ligand to facilitate the particle-cell recognition and in some case a bioresponsive lipid or pH-triggered polymer to enhance nucleic acid release and intracellular trafficking. A number of groups have observed that a PEG-shielded NP is a robust and modestly effective system for systemic gene or small interfering RNA (siRNA) delivery.
Collapse
Affiliation(s)
- Weijun Li
- Departament of Biopharmaceutidal Sciences, School of Pharmacy, University of California at San Francisco, San Francisco, California 94143-0046, USA
| | | |
Collapse
|
22
|
Hattori Y, Maitani Y. DNA/Lipid complex incorporated with fibronectin to cell adhesion enhances transfection efficiency in prostate cancer cells and xenografts. Biol Pharm Bull 2007; 30:603-7. [PMID: 17329867 DOI: 10.1248/bpb.30.603] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previously we have described the development and applications of lipid-based nanoparticles for gene delivery vector. In an attempt to improve transfection efficiency using the cell adhesion of extracellular matrix (ECM) to DNA/lipid complex (nanoplex), the mRNA expression of integrin alpha2beta1 and CD44 in prostate cancer cells was detected as adhesion molecules for fibronectin (Fn), collagen I (Col) and laminin (Lam) using a commercially available cDNA array (GEArray) system. These ECM proteins could enhance DNA transfection activity in cells when coated on the nanoplex. Among the ECM proteins, Fn-coating nanoplexes significantly increased transfection activity 2-fold in prostate cancer PC-3 cells, and exhibited higher DNA transfection activities to PC-3 xenografts, compared with commercially available cationic polymer in vivo jetPEI. These results indicated that Fn-coating nanoplexes could facilitate efficient transfection of prostate tumor cells.
Collapse
Affiliation(s)
- Yoshiyuki Hattori
- Institute of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | | |
Collapse
|
23
|
Shenoy DB, Amiji MM. An overview of condensing and noncondensing polymeric systems for gene delivery. ACTA ACUST UNITED AC 2007; 2007:pdb.top9. [PMID: 21357090 DOI: 10.1101/pdb.top9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
INTRODUCTIONSelf-assembling synthetic vectors for DNA delivery are designed to fulfill several biological functions. They must be able to deliver their genetic payload specifically to the target tissue/cells in a site-specific manner, while protecting the genetic material from degradation by metabolic or immune pathways. Furthermore, they must exhibit minimal toxicity and be proven safe enough for therapeutic use. Ultimately, they must have the capability to express a therapeutic gene for a finite period of time in an appropriate, regulated fashion. The DNA encapsulated in these vectors may be in a condensed or noncondensed form, depending on the nature of the polymer and the technique used for formulating the vector system. The whole process presents many barriers at both tissue and cellular levels. Overcoming these hurdles is the principal objective for efficient polymer-based DNA therapeutics.
Collapse
|
24
|
Xiong XB, Mahmud A, Uludağ H, Lavasanifar A. Conjugation of Arginine-Glycine-Aspartic Acid Peptides to Poly(ethylene oxide)-b-poly(ε-caprolactone) Micelles for Enhanced Intracellular Drug Delivery to Metastatic Tumor Cells. Biomacromolecules 2007; 8:874-84. [PMID: 17315946 DOI: 10.1021/bm060967g] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An arginine-glycine-aspartic acid (RGD) containing model peptide was conjugated to the surface of poly(ethylene oxide)-block-poly(epsilon-caprolactone) (PEO-b-PCL) micelles as a ligand that can recognize adhesion molecules overexpressed on the surface of metastatic cancer cells, that is, integrins, and that can enhance the micellar delivery of encapsulated hydrophobic drug into a tumor cell. Toward this goal, PEO-b-PCL copolymers bearing acetal groups on the PEO end were synthesized, characterized, and assembled to polymeric micelles. The acetal group on the surface of the PEO-b-PCL micelles was converted to reactive aldehyde under acidic condition at room temperature. An RGD-containing linear peptide, GRGDS, was conjugated on the surface of the aldehyde-decorated PEO-b-PCL micelles by incubation at room temperature. A hydrophobic fluorescent probe, that is, DiI, was physically loaded in prepared polymeric micelles to imitate hydrophobic drugs loaded in micellar carrier. The cellular uptake of DiI loaded GRGDS-modified micelles by melanoma B16-F10 cells was investigated at 4 and 37 degrees C by fluorescent spectroscopy and confocal microscopy techniques and was compared to the uptake of DiI loaded valine-PEO-b-PCL micelles (as the irrelevant ligand decorated micelles) and free DiI. GRGDS conjugation to polymeric micelles significantly facilitated the cellular uptake of encapsulated hydrophobic DiI most probably by intergrin-mediated cell attachment and endocytosis. The results indicate that acetal-terminated PEO-b-PCL micelles are amenable for introducing targeting moieties on the surface of polymeric micelles and that RGD-peptide conjugated PEO-b-PCL micelles are promising ligand-targeted carriers for enhanced drug delivery to metastatic tumor cells.
Collapse
Affiliation(s)
- Xiao-Bing Xiong
- Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2N8, Canada
| | | | | | | |
Collapse
|
25
|
Suk JS, Suh J, Choy K, Lai SK, Fu J, Hanes J. Gene delivery to differentiated neurotypic cells with RGD and HIV Tat peptide functionalized polymeric nanoparticles. Biomaterials 2006; 27:5143-50. [PMID: 16769110 PMCID: PMC5737930 DOI: 10.1016/j.biomaterials.2006.05.013] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Accepted: 05/10/2006] [Indexed: 10/24/2022]
Abstract
A number of neurodegenerative disorders may potentially be treated by the delivery of therapeutic genes to neurons. Nonviral gene delivery systems, however, typically provide low transfection efficiency in post-mitotic differentiated neurons. To uncover mechanistic reasons for this observation, we compared gene transfer to undifferentiated and differentiated SH-SY5Y cells using polyethylenimine (PEI)/DNA nanocomplexes. Differentiated cells exhibited substantially lower uptake of gene vectors. To overcome this bottleneck, RGD or HIV-1 Tat peptides were attached to PEI/DNA nanocomplexes via poly(ethylene glycol) (PEG) spacer molecules. Both RGD and Tat improved the cellular uptake of gene vectors and enhanced gene transfection efficiency of primary neurons up to 14-fold. RGD functionalization resulted in a statistically significant increase in vector escape from endosomes, suggesting it may improve gene delivery by more than one mechanism.
Collapse
Affiliation(s)
- Jung Soo Suk
- Department of Biomedical Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Junghae Suh
- Department of Biomedical Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Kokleong Choy
- Department of Biomedical Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Samuel K. Lai
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Jie Fu
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Justin Hanes
- Department of Biomedical Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
- Corresponding author. Department of Chemical & Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA. Tel.: +1 410 516 3484; fax: +1410 516 5510. (J. Hanes)
| |
Collapse
|
26
|
Dunehoo AL, Anderson M, Majumdar S, Kobayashi N, Berkland C, Siahaan TJ. Cell Adhesion Molecules for Targeted Drug Delivery. J Pharm Sci 2006; 95:1856-72. [PMID: 16850395 DOI: 10.1002/jps.20676] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rapid advancement of the understanding of the structure and function of cell adhesion molecules (i.e., integrins, cadherins) has impacted the design and development of drugs (i.e., peptide, proteins) with the potential to treat cancer and heart and autoimmune diseases. For example, RGD peptides/peptidomimetics have been marketed as anti-thrombic agents and are being investigated for inhibiting tumor angiogenesis. Other cell adhesion peptides derived from ICAM-1 and LFA-1 sequences were found to block T-cell adhesion to vascular endothelial cells and epithelial cells; these peptides are being investigated for treating autoimmune diseases. Recent findings suggest that cell adhesion receptors such as integrins can internalize their peptide ligands into the intracellular space. Thus, many cell adhesion peptides (i.e., RGD peptide) were used to target drugs, particles, and diagnostic agents to a specific cell that has increased expression of cell adhesion receptors. This review is focused on the utilization of cell adhesion peptides and receptors in specific targeted drug delivery, diagnostics, and tissue engineering. In the future, more information on the mechanism of internalization and intracellular trafficking of cell adhesion molecules will be exploited for delivering drug molecules to a specific type of cell or for diagnosis of cancer and heart and autoimmune diseases.
Collapse
Affiliation(s)
- Alison L Dunehoo
- Department of Pharmaceutical Chemistry, The University of Kansas, Simons Research Laboratories, 2095 Constant Avenue, Lawrence, Kansas 66047, USA
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Nonviral vectors continue to be attractive alternatives to viruses due to their low toxicity and immunogenicity, lack of pathogenicity, and ease of pharmacologic production. However, nonviral vectors also continue to suffer from relatively low levels of gene transfer compared to viruses, thus the drive to improve these vectors continues. Many studies on vector-cell interactions have reported that nonviral vectors bind and enter cells efficiently, but yield low gene expression, thus directing our attention to the intracellular trafficking of these vectors to understand where the obstacles occur. Here, we will review nonviral vector trafficking pathways, which will be considered here as the steps from cell binding to nuclear delivery. Studies on the intracellular trafficking of nonviral vectors has given us valuable insights into the barriers these vectors must overcome to mediate efficient gene transfer. Importantly, we will highlight the different approaches used by researchers to overcome certain trafficking barriers to gene transfer, many of which incorporate components from biological systems that have naturally evolved the capacity to overcome such obstacles. The tools used to study trafficking pathways will also be discussed.
Collapse
Affiliation(s)
- L K Medina-Kauwe
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
28
|
Cao A, Briane D, Coudert R. Chapter 5: Cationic Liposomes as Transmembrane Carriers of Nucleic Acids. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/s1554-4516(06)04005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
29
|
Temming K, Schiffelers RM, Molema G, Kok RJ. RGD-based strategies for selective delivery of therapeutics and imaging agents to the tumour vasculature. Drug Resist Updat 2005; 8:381-402. [PMID: 16309948 DOI: 10.1016/j.drup.2005.10.002] [Citation(s) in RCA: 361] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Revised: 10/27/2005] [Accepted: 10/28/2005] [Indexed: 12/24/2022]
Abstract
During the past decade, RGD-peptides have become a popular tool for the targeting of drugs and imaging agents to alphavbeta3-integrin expressing tumour vasculature. RGD-peptides have been introduced by recombinant means into therapeutic proteins and viruses. Chemical means have been applied to couple RGD-peptides and RGD-mimetics to liposomes, polymers, peptides, small molecule drugs and radiotracers. Some of these products show impressive results in preclinical animal models and a RGD targeted radiotracer has already successfully been tested in humans for the visualization of alphavbeta3-integrin, which demonstrates the feasibility of this approach. This review will summarize the structural requirements for RGD-peptides and RGD-mimetics as ligands for alphavbeta3. We will show how they have been introduced in the various types of constructs by chemical and recombinant techniques. The importance of multivalent RGD-constructs for high affinity binding and internalization will be highlighted. Furthermore the in vitro and in vivo efficacy of RGD-targeted therapeutics and diagnostics reported in recent years will be reviewed.
Collapse
Affiliation(s)
- Kai Temming
- Department of Pharmacokinetics and Drug Delivery, Groningen University Institute for Drug Exploration (GUIDE), Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | | | | | | |
Collapse
|
30
|
Zhang Z, Harada H, Tanabe K, Hatta H, Hiraoka M, Nishimoto SI. Aminopeptidase N/CD13 targeting fluorescent probes: synthesis and application to tumor cell imaging. Peptides 2005; 26:2182-7. [PMID: 15885853 DOI: 10.1016/j.peptides.2005.03.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Revised: 03/22/2005] [Accepted: 03/22/2005] [Indexed: 11/28/2022]
Abstract
A family of fluorescein-peptide conjugates (CNP1-3) for aminopeptidase N (APN/CD13) targeting fluorescent probes were designed and synthesized. Among the three conjugates, CNP1 bearing tumor-homing cyclic peptide CNGRC, could selectively label APN/CD13 over-expressing on the surface of tumor cells of HT-1080, as identified by means of fluorescent microscopic cell imaging. CNP1 was shown to be a promising fluorescent probe applicable to tumor-targeting molecular imaging.
Collapse
Affiliation(s)
- Zhouen Zhang
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura Campus, Kyoto 615-8510, Japan.
| | | | | | | | | | | |
Collapse
|
31
|
Xiong XB, Huang Y, Lu WL, Zhang X, Zhang H, Nagai T, Zhang Q. Enhanced intracellular delivery and improved antitumor efficacy of doxorubicin by sterically stabilized liposomes modified with a synthetic RGD mimetic. J Control Release 2005; 107:262-75. [PMID: 16125816 DOI: 10.1016/j.jconrel.2005.03.030] [Citation(s) in RCA: 176] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2004] [Revised: 02/22/2005] [Accepted: 03/07/2005] [Indexed: 11/22/2022]
Abstract
While sterically stabilized liposomes (SSL) can passively accumulate into tumor tissue due to the effect of enhanced permeability and retention (EPR), the intracellular uptake of the entrapped anticancer drugs by the tumor cells should be a determinant step for their antitumor activities. Therefore, strategies that can enhance the intracellular uptake of SSL into tumor cells could lead to an improved therapeutic efficacy for the drugs. To check this possibility, RGD-mimetic-modified SSL (RGDm-SSL) were constructed aimed to achieve tumor accumulation as well as enhanced intracellular delivery, and were loaded with doxorubicin (DOX), an anticancer drug. Flow cytometry and confocal microscopy reveal that RGDm-SSL facilitated the DOX uptake into the melanoma cells via integrin-mediated endocytosis. DOX-loaded RGDm-SSL (RGDm-SSL-DOX) displayed higher cytotoxicity on melanoma cells than DOX-loaded SSL (SSL-DOX). Tissue distribution and therapeutic experiments were examined in C57BL/6 mice carrying melanoma B16 tumors. RGDm-SSL-DOX displayed similar DOX accumulation in tumor tissue to that of SSL-DOX but showed significantly lower DOX level in blood and remarkably higher DOX level in spleen than SSL-DOX. Administration of RGDm-SSL-DOX at a dose of 5 mg DOX/kg resulted in effective retardation of tumor growth and prolonged survival times compared with SSL-DOX. These results suggest that RGDm-modified SSL may be a promising intracellular targeting carrier for efficient delivery of chemotherapeutic agents into tumor cells.
Collapse
Affiliation(s)
- Xiao-Bing Xiong
- School of Pharmaceutical Sciences, Peking University, Beijing, 100083, PR China
| | | | | | | | | | | | | |
Collapse
|
32
|
Xiong XB, Huang Y, Lu WL, Zhang X, Zhang H, Nagai T, Zhang Q. Intracellular delivery of doxorubicin with RGD-modified sterically stabilized liposomes for an improved antitumor efficacy: In vitro and in vivo. J Pharm Sci 2005; 94:1782-93. [PMID: 15986461 DOI: 10.1002/jps.20397] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Passive targeting by sterically stabilized liposomes (SSL), once combined with efficient intracellular delivery, may be a very useful strategy to improve the antitumor efficacy for the anticancer agents. The arginine-glycine-aspartic acid tripeptide (RGD) is known to serve as a recognition motif for several different integrins located on cell surface. In this study, the RGD tripeptide was coupled to the distal end of the poly (ethylene glycol)-coated liposomes (RGD-SSL) aimed to achieve increased tumor accumulation and enhanced intracellular uptake. DOX-loaded RGD-SSL (RGD-SSL-DOX), DOX-loaded SSL (SSL-DOX), and free DOX were compared with respect to their in vitro uptake and cytotoxicity and their in vivo biodistribution and therapeutic efficacy in tumor-bearing mice. Flow cytometry and confocal microscopy studies revealed that RGD-SSL could facilitate the DOX uptake into melanoma cells by integrin-mediated endocytosis. RGD-SSL-DOX displayed higher cytotoxicity on melanoma cells than SSL-DOX. While RGD-SSL-DOX demonstrated prolonged circulation time and increased tumor accumulation as SSL-DOX did, it showed remarkably higher splenic uptake than SSL-DOX. Mice receiving RGD-SSL-DOX (5 mg DOX/kg) showed effective retardation in tumor growth compared with those receiving same dose of SSL-DOX, free DOX solution, or saline. These results suggest that RGD-modified SSL may be a feasible intracellular targeting carrier for efficient delivery of chemotherapeutic agents into tumor cells.
Collapse
Affiliation(s)
- Xiao-Bing Xiong
- School of Pharmaceutical Sciences, Peking University, Beijing 100083, P.R. China
| | | | | | | | | | | | | |
Collapse
|
33
|
Medberry P, Dennis S, Van Hecke T, DeLong RK. pDNA bioparticles: comparative heterogeneity, surface, binding, and activity analyses. Biochem Biophys Res Commun 2004; 319:426-32. [PMID: 15178424 DOI: 10.1016/j.bbrc.2004.04.188] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Indexed: 10/26/2022]
Abstract
New applications for nucleic acid-bound micro/nanoparticles are emerging in drug delivery, biocatalysis, diagnostics, and toxicology. Bioactivity of viral or liposomal based technologies is limited by heterogeneity, partitioning, aggregation, and protein binding in physiological fluids, underlying immunotoxicity, and poor in vitro and cell-culture corollaries. Here we have systematically investigated novel pDNA bioparticles formed through complexation to model non-viral/non-lipid materials, peptides, aminoglycans, and small molecules (polybrene, chitosan, butirosin, protamine, Lys10, RGDS, bupivacaine, and chlorpromazine). On the basis of characterization by heterogeneity, kinetics, partitioning in physiological fluid and serum protein-binding, surface, size and electrophoretic behavior, transfection, and immunotoxicity, notably protamine, and chitosan DNA particles gave a long lifetime (12-18h), low protein-binding (<10microg/ml), good transfection activity (10(2)-10(4)RLU/mg cell protein), and low immunotoxicity. Our results support further evaluation of these materials as potential alternatives to viral or liposomal approaches, in combination with pDNA as binding, expression or therapeutic agents.
Collapse
|