Nonviral Approaches for Targeted Delivery of Plasmid DNA and Oligonucleotide SHIGERU KAWAKAMI, YURIKO HIGUCHI, MITSURU HASHIDA Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
Received 6 December 2006; revised 22 March 2007; accepted 22 March 2007 Published online in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/jps.21024
ABSTRACT: Successful gene therapy depends on the development of efficient delivery systems. Although pDNA and ODN are novel candidates for nonviral gene therapy, their clinical applications are generally limited owing to their rapid degradation by nucleases in serum and rapid clearance. A great deal of effort had been devoted to developing gene delivery systems, including physical methods and carrier-mediated methods. Both methods could improve transfection efficacy and achieve high gene expression in vitro and in vivo. As for carrier-mediated delivery in vivo, since gene expression depends on the particle size, charge ratio, and interaction with blood components, these factors must be optimized. Furthermore, a lack of cell-selectivity limits the wide application to gene therapy; therefore, the use of ligand-modified carriers is a promising strategy to achieve well-controlled gene expression in target cells. In this review, we will focus on the in vivo targeted delivery of pDNA and ODN using nonviral carriers. ß 2007 Wiley-Liss, Inc. and the American Pharmacists Association J Pharm Sci 97:726–745, 2008
Keywords: DNA/oligonucleotide delivery; nonviral gene delivery; targeted drug delivery; biomaterials; nanotechnology
INTRODUCTION The basic concept underlying gene therapy is that human disease may be treated by the transfer of genetic material into specific cells of a patient in order to enhance gene expression or inhibit production of a target protein. In the past few decades, several systems including viral and nonviral carriers that can be used to transfer foreign genetic material into cells have been developed with the aim of enhancing gene transfer in vitro and in vivo. In theory, virals carrier could provide a high transfection rate and a rapid
Correspondence to: Mitsuru Hashida (Telephone: þ81-75753-4545, Fax: þ81-75-753-4575; E-mail:
[email protected]) Journal of Pharmaceutical Sciences, Vol. 97, 726–745 (2008) ß 2007 Wiley-Liss, Inc. and the American Pharmacist Association
726
transcription of the foreign material inserted in the viral genome. However, viral carriers present potential problems for patients, such as the immunogenicity of the viral proteins, lack of desired tissue selectivity, potential for oncogenesis due to chromosomal integration, and generation of infectious viruses due to recombination. On the other side, nonviral vectors are potentially less immunogenic, relatively easy to produce in clinically relevant quantities, and associated with fewer safety concerns. Furthermore, synthetic nonviral vectors provide flexibility in formulation design and can be tailored to interact efficiency with DNA cargo and the specific route of vector administration, and can be enhanced delivery to specific tissues or cells through incorporation of a targeting ligand. Therefore, the evaluation and development of targeted delivery system by nonviral carriers should overcome low gene transfer efficacy compared with viral vectors.
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
Broadly speaking, in the gene therapy, recombinant plasmid DNA (pDNA) encoding therapeutic proteins and oligonucleotides, relatively small fragments of synthetic DNA or RNA, designed to block gene expression relating to a particular disease have been used like a ‘drug’. Since pDNA and oligonucleotide are not extensively taken up by cells, efficient gene transfer relies on the development of carriers. Moreover, there are many obstacles to in vivo gene delivery, such as degradation by enzymes in blood, interaction with blood components, and nonspecific uptake by the cells, which govern biodistribution in the body. In this review, targeted gene delivery using nonviral carriers is discussed including pharmacokinetic considerations.
BIODISTRIBUTION CHARACTERISTICS OF NAKED PDNA Although naked pDNA is a powerful tool for nonviral gene therapy, pDNA exhibits poor cellular uptake and it is biologically unstable to nucleases. Therefore, it is necessary for the successful clinical use of pDNA to develop optimized delivery systems, which will enhance the cellular uptake, offer protection from enzymatic degradation and provide cell-specific delivery. In this section, we will discuss the physicochemical properties, cellular uptake mechanism and biodistribution characteristics of naked pDNA.
Pharmacokinetic Characteristics of Naked pDNA It has been reported that intravenous administration of naked pDNA actually induces no gene expression.1 To develop a strategy for establishing nonviral gene carrier systems, however, it is necessary to understand the in vivo disposition characteristics of naked pDNA. pDNA is rapidly eliminated from the plasma involving extensive uptake by the liver after intravenous injection of [32P] labeled-pDNA.1 Pharmacokinetic analysis has demonstrated that the hepatic uptake clearance is almost identical to the plasma flow rate in the liver, suggesting highly effective elimination by this organ. In addition, pDNA is taken up preferentially by the liver nonparenchymal cells via a scavenger receptor (SRA)-mediated process, in a manner specific for polyanions.1–3 DOI 10.1002/jps
727
Uptake Mechanism of Naked pDNA Studies involving in vitro binding and uptake using cultured mouse peritoneal macrophages have shown that the binding of pDNA is significantly inhibited by polyinosinic acid (poly [I]) and dextran sulfate that are substrates of SRA, but not by polycytidylic acid (poly [C]), dextran and EDTA that are not substrates of this receptor. These data suggest that pDNA is taken up by macrophages via a mechanism mediated by a receptor like the macrophage SRA. Involvement of SRAs in the hepatic uptake of pDNA has also been supported by single-pass rat perfusion experiments using [32P] pDNA.2 The class A SRA, the best characterized SR, which recognizes a wide variety of anionic macromolecules based on their three dimensional structure, seems likely to be responsible for pDNA uptake. Takakura et al.3 reported an in vitro study of [32P] pDNA binding and uptake using cultured CHO cells expressing SRA (CHO (SRA) cells) and peritoneal macrophages from SRA-knockout mice. The [32P] pDNA binding and uptake by CHO (SRA) cells were minimal and almost identical to that by wild-type CHO cells. Macrophages from knockout mice showed pronounced pDNA binding and uptake, as did the control macrophages. In both types of macrophage [32P] pDNA binding was significantly inhibited by the pDNA, poly [I] and dextran sulfate but not by poly [C] or Ac-LDL. These results provide direct evidence that SRA is not responsible for any significant binding and subsequent uptake of pDNA by mouse peritoneal macrophages. Therefore, pDNA binding and uptake by mouse peritoneal macrophages are mediated by a specific mechanism involving some defined polyanions and not by the SRAs. These findings form an important basis for further studies to elucidate the mechanisms of pDNA uptake by macrophages.
IN VIVO DELIVERY APPROACHES FOR PDNA Owing to rapid degradation by nucleases in the serum and rapid clearance, the clinical use of naked pDNA is generally limited. Therefore, some efforts had been devoted to develop nonviral pDNA delivery systems, which allow high gene expression in target tissues or cells. These techniques are categorized into two groups: (i) naked DNA delivery by a physical method such as electroporation and the gene gun and JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
728
KAWAKAMI, HIGUCHI, AND HASHIDA
(ii) delivery mediated by a carrier, such as cationic polymers or liposomes. These systems improve transfection efficacy of pDNA in vitro. Recently, several types of in vivo gene delivery systems have been developed and investigated in clinical situations (Tab. 1). In this section, we will discuss the in vivo gene delivery systems using nonviral approaches.
Physical Approaches for Transfection Direct injection of naked pDNA into tissues or systemic injection is the simplest and safest form of administration. In order to enhance gene expression, several physical approaches has been developed, such as the hydrodynamics method, gene gun, electroporation, sonoporation, and laser irradiation. Dramatic improvements in pDNA gene transfer and expression have been achieved in vivo by these technologies. The hydrodynamic method consists of the very rapid injection through the mouse-tail vein of a large volume of pDNA solution: this results in a very efficient transfection of liver cells. Although this method is a useful laboratory tool even although the mouse sustains some damage, its clinical application is limited. However, other physical methods discussed following have a potential for clinical use as human gene therapy although further optimization will be needed.
The gene delivery system using gene gun is useful for the percutaneous administration. Electroporation The first pioneering demonstration that pDNA could be introduced into living cells by means of electric pulses was published in 1982.17 This technique, termed electroporation or electropermiabilization, exposed the cell membrane to highintensity electrical pulses that can causes transient and localized destabilization of the cell membrane. The instrumentation required for electroporation consists of a pulse generator and an ‘‘applicator’’ which includes the electrodes. The efficiency of gene transfer by this method is influenced by several physical factors (electrode shape, electrode number, pulse duration, and electric field strength), pDNA concentration, conformation, and cell size.18,19 Different electrodes have been developed for several types of in vivo application: surface electrodes, needle electrodes for deep tissues, and electroparation catheters for hollow organs, such as blood vessels.20 Skin,21,22 muscle,22,23 and solid tumors24,25 are ideal targets for electroporation. Effective transfer of intravenously injected pDNA to the liver using electroporation has also been reported.26,27 Sonoporation
The Gene Gun This biological and ballistic delivery uses heavy metal particles coated with pDNA, which are propelled at a high velocity into the target cell. This technique was used first in 1987 in vitro 4 and then extended to mammalian cells and living tissues in the early 1990s. The particles must be nontoxic, nonreactive, and smaller than the diameter of the target cell. Several particles (e.g., heavy metal and tungsten) have been developed, and gold beads are usually employed. The efficiency of this delivery depends on the kind of particle, its size and the timing of delivery.5 Now, the transfection area is limited by the shallow penetration of pDNA, therefore skin has been the major target site for vaccine and immune-therapy.6–9 Some reports have involved other target sites for in vivo application, including the liver10,11 and brain.12,13 As far as clinical trials are concerned, gene gun applied for the hepatitis,14 and DNA vaccine therapy to treat influenza15 and melanoma16 without sever toxicity. JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
Sonoporation enhances cell permeability via the application of ultrasound, which was developed in the mid-1990s.28–30 Ultrasound is in general clinical use for both therapeutic and diagnostic purposes. Low-level ultrasound is used for diagnostic imaging, and ultrasonic shock waves are used in the treatment of kidney stones and highintensity focused ultrasound is used for the thermal destruction of tumors. Gene transfection by sonoporation involves acoustic cavitation, which can cause mechanical perturbation and collapse of active bubbles, and the associated energy release can permiabilize adjacent cell membranes. It has been shown that ultrasound contrast agent, which consists of elastic and compressible gas-filled microbubbles, can enhance cavitation. Bubble implosion may form localized jets that would open pathways through the cell membrane.31 Several studies have examined a wide range of contrast agents, concluding that albumin-coated octa-fluoropropane gas microbubbles (Option) are preferable because of high gene DOI 10.1002/jps
DOI 10.1002/jps
Intramuscular (naked) Intradermal þ intramuscular (naked) Intratumoral Intratumoral
HER-2 Melanoma antigen gp100
Intramuscular (naked) Intramuscular (naked)
Intramuscular (naked) Intratumoral injection (1 naked) Intramuscular (naked) Intramyocardial (naked)
NV1FGF P53 Vascular endothelial growth factor (VEGF)
Interferon-alpha (IFN-a) polyvinylpyrrolidone Interferon-gamma (IFN-g), interleukin-2 (IL-2), polyvinylpyrrolidone Hepatocyte growth factor (HGF) Myelin basic protein (hMBP)
Intramuscular (naked)
Intraperitoneal injection (lipofection)
E1A
Fibroblast growth factor (FGF)
Intratumoral injection (lipofection)
Interleukin-2 (IL-2)
Intramascular injection (lipofection)
Intramascular (naked) Intramyocardial (naked) Intratumoral injection (lipofection)
Hepatocyte growth factor (HGF) Vascular endothelial growth factor (VEGF) HER-2
Poloxamer 188/Del-1
Intratumoral injection (lipofection)
Administration Route
HLA-B7/beta 2-microglobulin
Target Molecular of Action
Angioendothelioma (II) Squamous cell carcinoma of the head and neck (II) Peripheral artery disease (II) Multiple sclerosis (MS) (II)
Melanoma, colorectal carcinoma, renal cell carcinoma, breast cancer, lymphoma, breast adenocarcinoma, renal cell carcinoma, colorectal adenocarcinoma, nonHodgkin’s lymphoma, renal cell cancaer, head and neck cancer, squamous cell carcinoma of the head and neck (III/II) Peripheral arterial disease (III) Refractory angina pectoris (II/III) Squamous cell carcinoma of the head and neck (II) Renal cell cancer, prostate cancer, squamous cell carcinoma of the head and neck, renal cell carcinoma (II) Ovarian cancer, squamous cell carcinoma of the head and neck (II) Intermittent claudication secondary to peripheral arterial disease, peripheral artery disease (II) Severe peripheral artery occlusive disease (PAOD), peripheral arterial occlusive disease (II) Intermittent claudication (II) Posthepatitis liver cancer (II) Peripheral vascular disease (II) Coronary artery disease, ischemic myocardium (II) Breast cancer (II) Melanoma (II)
Clinical Development (Phase 2 and Beyond)
Table 1. Nonviral Approach Based pDNA-Delivery Therapeutics Currently in Advanced Stages of Clinical Development (Phase 2 and Beyond)
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
729
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
730
KAWAKAMI, HIGUCHI, AND HASHIDA
transfection efficacy. Gene transfection efficiency using sonoporation depends on several factors, including transducer frequency, acoustic pressure, pulse duration, and exposure duration. In addition, the ultrasound contrast agent concentration and its formulation are also important factors. Recently, in vivo pDNA transfection mediated by ultrasound has been reported, first using a lithotripter,32 and then using focused sinusoidal sonoporation.33 To date, sonoporation has been shown to improve gene expression in muscle,34,35 carotid artery,36,37 and solid tumors.38
formed by complexes (lipo- or polyplexes) with pDNA. Since cationic charge on the surface of the complexes affect on their biodistribution, we discussed about it at the section 3.3.2. Focusing on clinical development, lipid- based pDNA delivery is also being examined for nonviral gene therapy. On the other hand there have been no clinical applications of polymers, until now. The therapeutic nonviral pDNA delivery trials currently at advanced stages of clinical development are listed in Table 1.
Cationic Polymers Laser Irradiation The laser beam is commonly focused onto the target cell via a lens. It has been reported that laser irradiation creates transient pores in the membrane, which can be quite large in size (approximately 2 mm in diameter) but apparently self repair within a fairly short period of time (of the order of seconds).39,40 However, the mechanism of gene expression enhancement in vivo by laser is not clear. It has been predicted that the thermal effect at the cite of laser beam impact causes a difference in osmotic pressure between the cytoplasm and the medium surrounding the cell which changes the permeability of the cell membrane.41 To date, gene delivery via laser irradiation has not been widely used. This may be because of the high cost, the physical size of laser sources, and the difficulties in controlling the appropriate condition until now. Recently, it has been reported that gene transfer into muscle could be enhanced using a femtosecond infrared laser.42 Further studies will be needed before application to human gene therapy.
Carrier Approaches for Transfection Cationic compounds are the most promising carriers for pDNA because the positive charge can interact in an electrostatic manner with the negative charge of pDNA to form complexes. Cationic polymers and liposomes are the two major types of nonviral gene delivery vectors for pDNA currently being investigated. Since the excess positive charge of complexes enable the complexes to interact with negative charge on the cell surface through electrostatic interaction, successful transfection of pDNA has been achieved using such cationic polymers and liposomes JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
Cationic polymers used for pDNA delivery are of two types, natural polymers including chitosan43,44 and atelocollagen45,46 or synthetic polymers including poly(L-lysin) (PLL),47,48 49–51 polyethylenimine (PEI), and dendrimer.52,53 Natural polymers generally have the advantage of being nontoxic, even in large doses, with good mucoadhesive, biocompatibility and are biodegradable.54 On the other hand, synthetic polymers can provide flexibility in formulation design and can be tailored to fit the size and topology of the pDNA. Typical cationic polymers are shown in Figure 1A.
Cationic Liposomes Cationic liposomes are generally composed of positively charged lipids, such as N-[1-(2,3dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), dioleoyl trimethylammonium propane (DOTAP), or dimethylaminoethane carbamoyl cholesterol (DC-Chol), and so called ‘‘helper lipid’’, such as dioleoyl phosphatidylethanolamine (DOPE) and cholesterol, which confer additional fusogenicity and/or stability on the lipoplexes. Cationic liposomes are also useful for enhancing the cellular uptake of pDNA or ODN in vitro. However, intravenous administration of cationic liposome/pDNA complexes leads to systemic gene expression, particularly in the lung in mice.55–57 Typical cationic lipids are shown in Figure 1B.
HVJ Liposomes Hemagglutinating virus of Japan (HVJ)-liposomes58 are another type of liposome containing two fusigenic proteins such as HVJ and artificial DOI 10.1002/jps
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
731
complexes, because they have been studied in great detail. The Size of the Complexes
Figure 1. The structures of typical cationic polymer and lipid composing cationic liposome.
viral envelope, which could transfer encapsulated pDNA into cells. Since HVJ-liposome transfer pDNA by fusion with cell membrane, pDNA could be directory transferred into cytoplasm escaping from the digestion in endosome or lysosome. As far as in vivo application is concerned, HVJ liposomes are taken up in a nonspecific manner after intravenous injection. Therefore, HVJ liposomes are directly injected into target organs, such as liver59 and heart.60
The Factors Affecting Transfection Efficacy by Carrier Methods To date, several reports have discussed the factors, which govern the gene expression in vivo and in vitro. To achieve a therapeutic effect using pDNA or ODN, it is important to discuss these factors, especially in the in vivo case. We will focus on the factors governing the in vivo gene expression of pDNA and nonviral vector DOI 10.1002/jps
The size is an important factor for controlling targeted gene delivery systems because the structure of the blood capillary wall varies greatly in different organs and tissues. Because of its large molecular weight, pDNA does not effectively penetrate endothelial and epithelial barriers and there is very little extravasation from the vascular to the interstitial space. The complex formation by cationic carriers not only condenses the pDNA but also enhances its uptake by the cells via electrostatic interaction. pDNA/carrier complexes are often prepared in a nonionic solution due to their well-known tendency to aggregate out of solution as the salt concentration is increased.61 Aggregation during lipoplex formation in ionic solution might be due to neutralization of the surface positive charge of the lipoplex intermediate by the associated counter ion. The size of the pDNA complexed with cationic liposomes at a charge ratio (:þ) of 1.0:2.3 is very condensed (100–200 nm) when it is prepared in a nonionic solvent, such as dextrose and sucrose, compared with an ionic solvent.62 The endothelium is a monolayer of cells that are metabolically very active and mediate the bidirectional exchange of fluid between plasma and interstitial fluid. Thus, the endothelium has a profound influence on the extravasation of macromolecules. Discontinuous capillaries, also known as sinusoidal capillaries, are common in the liver, spleen, and bone marrow. Tumor tissues are characterized by increased interstitial pressure, which may retard the extravasation of macromolecules. Capillary vessels in a human tumor inoculated into SCID mice are permeable even by liposomes of up to 400 nm in a diameter.63 Hence, targeted gene delivery systems can be applied to liver, spleen, and tumors by ligand modifications because of the size factor. In vivo receptor-mediated gene delivery is listed in Table 1. The Charge Ratio of the Complexes Neutralization of the cationic charge of the pDNA/ cationic gene carrier complexes by negatively charged serum proteins seems to reduce the in vitro and/or in vivo transfection efficacy. Yang and Huang64 reported that neutralization of the excess positive charge in pDNA/cationic liposome JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
732
KAWAKAMI, HIGUCHI, AND HASHIDA
complexes by negatively charged serum proteins is likely to reduce the transfection efficiency in vitro. They also demonstrated that this can be overcome by increasing the cationic charge ratio of pDNA/cationic liposome complexes, and the optimal charge ratio (:þ) was 1.0:4.0 for efficient transfection, even in the presence of 20% serum. After intravenous administration, the higher cationic charge of pDNA/cationic liposome complexes enhances gene expression in the lung via electrostatic interaction. However, the too much cationic charge of pDNA/cationic liposome complexes makes it difficult to deliver pDNA to the targeted organs except for lung, because of high aggregation caused by nonspecific interaction with blood cells. We have evaluated the effect of the cationic charge on asialoglycoprotein receptor-mediated gene transfection systems for hepatocytes using pDNA/galactosylated cationic liposome complexes after intraportal administration.62,65 As far as the pDNA/galactosylated cationic liposome complexes were concerned, they were prepared at a charge ratio (:þ) of 1.0:2.3 and/or 1.0:3.1 for selective gene expression in the liver. At a charge ratio of 1.0:7.0, on the other hand, the gene expression in the lung exceeded that in the liver, suggesting a highly nonspecific interaction. Therefore, a cationic charge ratio (:þ) of 1.0:2.3 and/or 1.0:3.1 for pDNA to galactosylated cationic liposomes seems to be optimum for receptor-mediated in vivo gene transfection. We have also evaluated the effect of the cationic charge on mannose receptor-mediated gene transfection systems using pDNA/mannosylated cationic liposome complexes after intravenous administration.66 The transfection efficiency after intravenous administration of complex at a charge ratio (:þ) of 1.0:2.3 and/or 1.0:3.1 in liver and spleen that expressed a mannose receptor on the cell surface was higher than that in lung. On the other hand, when complexes were formed at a charge ratio (:þ) of 1.0:4.7, the transfection efficiency in the lung was the highest. These results suggest that a complex with a charge ratio (:þ) of 1.0:2.3 and/or 1.0:3.1 is optimum for receptor-mediated gene delivery systems by ligand-modified gene carrier systems. To prepare more stable complexes of pDNA and cationic liposomes, we developed a new method of preparing the complexes in ionic buffer containing 10 mM sodium chloride (NaCl), which results in a surface charge regulated (SCR) lipoplex.67 After intravenous injection, pDNA/galactosylated SCR JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
liposome complexes significantly delayed aggregation and showed10–20-fold higher gene expression in liver than pDNA/conventional liposome complexes. Since SCR liposomes could approach nearer each other than conventional liposomes due to partial neutralization, SCR liposome would make more stable complexes with pDNA. Interactions With Blood Components The first obstacle to the use of pDNA and carrier complexes in vivo is their interaction with the biological milieu at the site of injection (i.e., blood in the case of intravenous injection). Even in the case of transfection in vitro, the efficacy of early transfection kits using cationic lipids was inhibited by the presence of the biological milieu, such as serum and plasma. Therefore, such carriers could not produce in vivo gene expression, and this might be due to physical changes occurring in lipoplexes in the biological milieu. We and other groups have demonstrated that interactions between erythrocytes and lipoplexes or polyplexes lead to lung accumulation and gene expression.68,69 In addition, interaction with opsonins in the blood lead rapid uptake by the reticuloendothelial system (RES), resulting in rapid bloodstream clearance. Recently, various approaches have been introduced in order to overcome the interaction with blood to develop new cationic lipids that are more serum-resistant,70,71 including changing helper lipids,72,73 stabilization of the complexes by DNA-condensing agents such as polyamines and protamine sulfate, and steric stabilization of lipoplexes by poly (ethylene glycol) (PEG) phospholipid conjugate,74–76 then achieve the longer retention time in blood due to escape from the uptake of RES.
Targeted Delivery of pDNA by Carrier Methods Several studies have involved the development of carriers for improving the cellular membrane permeability of pDNA and they have been successful in achieving effective pDNA transfer to cells. However, because such carriers and pDNA complexes are taken up by cells in a nonspecific manner, their in vivo use is limited to local administration. Therefore, there is a need to develop targeted carriers, which can control tissue disposition after intravenous injection. Moreover, the delivery to the target cell could help in reducing the pDNA dose. To cell-specific delivery of pDNA, ligand-mediated uptake is a promising DOI 10.1002/jps
DOI 10.1002/jps
91,92,93
90
Tumor cell Tumor cell
66,84,85,86,87
62,65
81
Liver (hepatocyte) Liver (hepatocyte) Liver (nonparenchymal cell)
733
Transferiin-PEI Folate-liposome
Mannose receptor-mediated endocytosis
Transferrin receptor-mediated endocytosis Folate receptor-mediated endocytosis
Intraportal injection Intraportal injection Intravenous or intraperitleal injection Intraperitleal injection Intravenous injection
Administration Route Liposome Mechanism
Table 2. Cell-Targeted Carriers for In Vivo pDNA Delivery
Liver parenchymal cell exclusively express large numbers of high affinity cell-surface receptors that can bind asialoglycoproteins and subsequently internalize them in the cell interior. In order to achieve liver parenchymal cell-specific gene transfection, the galactose moiety is introduced into either cationic polymers or cationic liposomes because of high expression of asialoglycoprotein receptors on liver parenchymal cells. In the late 1980s, Wu et al.77,78 demonstrated successful in vivo gene transfer to liver using PLL linked with asialoorosomucoid. Successful in vivo gene expression after intravenous injection has been also reported for glycosylated PLL79 and galactosylated poly-L-ornithine conjugated with a fusogenic peptide.80 In general, the transfection efficacy of cationic liposomes is higher than that of cationic polymers; therefore, this mechanism would be an effective way of achieving hepatocyte targeting using galactosylated cationic liposomes. The galactosylation of liposomes can be achieved by coating with either glycoproteins or galactose conjugated synthetic lipids. As far as targeted gene delivery by liposomes is concerned, asialofetuin-labeled liposomes were developed and achieved high gene expression in cultured hepatocytes and liver after intraportal injection with a preload of EDTA.81 However, the introduction of asialoglycoproteins to liposomes is complicated and there are a number of problems associated with the carriers, such as reproducibility and immunogenicity. Therefore, using low-molecular-weight glycolipids, galactose-presenting lipopolyamine vectors were developed and produced high gene transfer into hepatoma cells under in vitro conditions.82 Inclusion of galactose residues in the electrically neutral complex increased transgene expression to that approaching the value obtained with a large excess of cationic liposomes alone. The authors stressed that the galactose-presenting DNA particles may avoid interaction with serum proteins because of their electric neutrality. However, in order to achieve successful in vivo gene delivery systems, the pharmacokinetics, physicochemical properties of the complex, and
Target Tissue (Cell)
Asialoglycoprotein Receptor-Mediated Gene Transfection
Asialofetuin-liposome Galactose-liposome Mannose-liposome
approach and several types of ligand-modified carriers have been developed. In this section, we will focus on the delivery of pDNA in vivo. Table 2 summarizes the cell-targeted carriers that have been used in vivo.
Asialoglycoprotein receptor madiated endocytosis
Reference
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
734
KAWAKAMI, HIGUCHI, AND HASHIDA
theoretical design of galactosylated lipids need to be examined. Taking these into consideration, we synthesized cholesten-5-yloxy-N-(4-((1-imino2-D-thiogalactosylethyl)amino)alkyl)formamide (Gal-C4-Chol), which possess the cationic charge necessary for pDNA binding, galactose residues as a targetable ligand for liver parenchymal cells and cholesterol residues as an anchor for stable insertion into the lipid layer, and examined the in vivo gene transfer by optimizing the pharmacokinetics and physicochemical properties.62 The hepatic gene expression of pDNA complexed with Gal-C4-Chol liposomes was more than a 10-folds greater than that of pDNA complexed with conventional cationic liposomes after intraportal injection. Moreover, high gene expression was observed in hepatocytes but not in liver nonparenchymal cells.
be efficiently transfected into dendritic cells, which express a large number of mannose receptors. Recently, Hattori et al. demonstrated that the targeted delivery of DNA vaccine by ManC4-Chol liposomes is a potent vaccination method for DNA vaccine therapy.85 High gene expression in antigen presenting cells (APCs) in liver, spleen, greater omentum, and peritoneal exudate cells was observed after intraperitonearl injection of pDNA complexed with Man-C4-Chol liposomes.86 Furthermore, applying this system to OVA coding pDNA as a vaccine, a stronger OVA-specific CTL response could be obtained compared with pDNA complexed with conventional liposomes and naked pDNA. Antitumor effects in terms of the survival rate of tumor bearing mice were also achieved after intraperitonearl injection of OVA coding pDNA complexed with Man-C4-Chol liposomes.87
Mannose Receptor-Mediated Gene Transfection Macrophages are important targets for the gene therapy of diseases such as Gaucher’s disease and human immunodeficiency virus (HIV) infection, but the process of gene transfection in such cases is not easy. The use of nonviral vectors is attractive for in vivo gene delivery because it is simpler than using viral systems and is free from some of the risks inherent in the latter. One of the most promising nonviral gene delivery systems developed to date involves cationic liposomes as far as in vivo transfection efficiency is concerned. Hence, cationic liposomebased targeted gene delivery systems are ideal for in vivo conditions. Recently, we synthesized a novel mannosylated cholesterol derivative, ManC4-Chol, for mannose receptor-mediated gene transfection to macrophages,83,84 which are known to express large numbers of mannose receptors on their surface. In primary cultured mouse peritoneal macrophages, pDNA complexed with Man-C4-Chol liposomes showed higher transfection activity than that complexed with conventional cationic liposomes. The presence of 20 mM mannose significantly inhibited the transfection efficiency of pDNA complexed with Man-C4-Chol liposomes, suggesting that the complexes of pDNA and mannosylated cationic liposomes are recognized and taken up by the mannose receptors on macrophages. As far as the application to gene therapy using targeted gene delivery by mannosylated liposomes is concerned, DNA vaccine therapy is suitable because the antigen encoded pDNA must JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
Transferrin Receptor-Mediated Gene Transfection Transferrin, an iron-binding glycoprotein, is a ligand that has been studied in detail for tumortargeting. Iron-loaded transferrin is recognized and binds to transferrin receptors on the cell surface. The expression of transferrin receptors is increased in rapidly dividing cells due to a need for iron; therefore, transferrin receptors are often unregulated on the surface of malignant cells and this has been used as a tumor-targeting ligand for several drug delivery systems. Recently, Wagner and coworkers88,89 developed transferrin-linked PEI for tumor-selective gene transfection. To block undesired, nonspecific interactions with blood components or nontarget cells, the surface charge of the complexes was masked by either covalently attached hydrophilic PEG or a higher density of attached transferrin. After intravenous injection, the gene expression in the tumor was approximately 100-times higher than that in other tissues. More recently, Kursa et al.90 have demonstrated that intravenous injection of PEG-PEI-transferrin containing pDNA encoding for tumor necrosis factor (TNF-a) inhibited tumor growth in murine tumor models. Folate Receptor-Mediated Gene Transfection The folate receptor is known to be overexpressed in a large fraction of human tumors, but it is only minimally distributed in normal tissues. Therefore, the folate receptor also has been used as a tumor-targeting ligand for several drug delivery DOI 10.1002/jps
DOI 10.1002/jps
Colon cancer rectal cancer pancreatic cancer (II) Ovarian epithelial cancer recurrent breast cancer (II) Atopic dermatitis (II)
Macular degeneration (IV)
Intravenous infusion Intravenous infusion Intravenous infusion
Intravenous infusion Intravenous infusion
Intradarmal injection
Intravitreal injection
Bcl-2 antisense ODN (G3139) ICAM-1 antisense ODN (ISIS 2302) PKC-k antisense ODN (ISIS 3521, LY900003) H-ras antisense ODN (ISIS 2503) c-raf kinase antisense ODN (ISIS 5132)
Subcutaneous injection Hepatitis C (II) Intratumoral injection Glioblastoma (II) Intravitreal injection Choroid neovascularization age-related macular degeneration (II)
Cytomegalovirus retinitis in AIDS (FDA approved) Nonsmall cell lung cancer (II/III) Crohn’s disease (III) Carcinoma, nonsmall-cell lung (III) Intravitreal injection IE2 antisense ODN
Clinical Development (Phase 2 and Beyond)
735
Decoy ODN (double stranded short NFkB decoy ODN DNA) Aptamar (single or double short AntiVEGF pegylated aptamer RNA or DNA) CpG ODN (single stranded short DNA) (CPG10101) (CpG-ODN) siRNA (double stranded short RNA) VEGF (AGN211745)
Rapid elimination is observed after intravenous injection of oligonucleotides. Not only hepatic uptake and degradation by nucleases but also urinary excretion is a major elimination pathway for oligonucleotides due to their smaller molecular weight.94 After intravenous injection, 30 (% of dose/g tissue) kidney accumulation was observed95 while accumulation in other tissues was extremely low.95 To clarify the pharmacokinetic characteristics of oligonucleotides at the organ level of liver96 and kidney,97,98 perfusion experiments were carried out. In contrast to pDNA, both liver parencymal and
Antisense ODN (single stranded short DNA)
Biodistribution Characteristics of ODN
Administration Route
During the past few decades, there has been great interest in exploiting pDNA or single or doublestranded ODNs, relatively small fragments of synthetic DNA or RNA, for therapeutic purposes. They can be classified in terms of the mechanism by which they elicit their action (Tab. 3). Unlike pDNA, there has been no clinical development using carrier (Tab. 3). The use of chemically synthesized ODN might be able to improve the pharmacokinetic properties and therapeutic effect. Compared with pDNA, ODNs are easy to synthesize chemically since each strand is of short length (about 20–30 bp). In this section, we will discuss the chemical modification and biodistribution characteristics of naked ODN.
Target Molecular of Action
BIODISTRIBUTION CHARACTERISTICS OF NAKED ODN
Type of DNA
systems. Hofland et al.91 synthesized folate-PEGlipid derivatives for preparing folate modified cationic liposomes. After intravenous injection of folate liposome complexes, the gene expression in the tumors was unchanged while the gene expression in the lung was reduced compared with conventional complexes. However, after intraperitoneal injection in a murine disseminated peritoneal tumor model, folate liposome complex formulations produced approximately a 10-folds increase in tumor-associated gene expression compared with conventional complex.92 Recently, Ward et al. synthesized folic acidPEG-NH2 and make conjugation with PLL/pDNA polyplexes. This polyplexes covered with folic acid-PEG-NH2 more accumulated to the liver than polyplexes alone.93
Table 3. ODN-Based Therapeutics Currently in Advanced Stages of Clinical Development (Phase 2 and Beyond)
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
736
KAWAKAMI, HIGUCHI, AND HASHIDA
nonparenchymal cells were involved in the hepatic uptake of oligonucleotides and this was a nonspecific process. The rat kidney perfusion experiments showed that the renal uptake of oligonucleotides was due to tubular reabsorption and direct uptake from the capillary side. Moreover, the uptake from the blood side was a saturated process and coadoministration of dextran sulfate and polyinosinic acid inhibited the renal uptake while polycytidic acid did not. This suggests that oligonucleotides are taken up via a process like the SRA-mediated one as far as polyanion specificity is concerned. Biodistribution Characteristics of Chemically Modified ODN Natural ODNs, which have a phosphodiester type backbone, have disadvantages for clinical use, including their instability to nucleases and the low cellular uptake. One strategy to overcome these problems is the chemical modification of ODNs because of the nuclease attack to the phosphoric ester bonds. Another strategy is the structural modification of natural ODNs. However, chemical modification of the backbone and structural modification may lead to a loss of the base pairing ability. Many types of chemically modified ODNs have been produced and investigated to date, such as modification of the ODN backbone, the sugar moiety and the nucleic bases.99,100 These modifications have been classified as three generations. The modified ODN of the first generation was designed to form internucleotide linkages, to the backbone of which the nucleotide bases were covalently attached, making them more resistant to the nonbridging oxygen atoms in the phosphate group with a sulfur, a methyl group, etc. (Fig. 2A). Phosphorothioate ODN is the most widely used to date in a number of disease models both in vivo and in vitro, although it produces undesirable side effects in vivo, such as immune stimulation, complement activation and cellular toxicity.101–103 The seemingly negative property of phosphorothioate ODN to interact with serum proteins proved to be advantageous as far as the pharmacokinetic profile was concerned. Their binding to plasma proteins protects them from filtration in the kidney and is responsible for an increased serum half-life.104 The second generation involves ODNs with alkyl modification at the 20 position of the ribose (Fig. 2B). These type of ODNs made of these building blocks at the 20 position are less toxic JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
Figure 2. The structures of backbone modification of oligonucleotide (A, B) and nuclear acid analogs (C).
than phosphorothioate ODNs and have a slightly enhanced affinity towards their complementary RNAs. The third generation ODNs contains DNA and RNA analogs with modified linkages or riboses as well as nucleotides with a completely different chemical moiety replacing the furanose ring. Peptide nucleic acids (PNA),105,106 morpholino phosphoroamidates (MF),107,108 and locked nucleic acids (LNA)109,110 are the most important analogs in this group (Fig. 2C). Typical chemical modifications are shown in Figure 2. Terminal modifications of ODN significantly enhance resistance to degradation by exonucleases in serum and tissue homogenates. Recently, Soutschek et al.111 prepared chemically modified siRNA with partial phosphorothioate linkages, 20 -o-methyl sugars and a cholesterol moiety at the 30 end of the sense strand. After intravenous injection of cholesterol-modified siRNA, the elimination half-life (two compartment) and plasma clearance increased compared with naked siRNA. DOI 10.1002/jps
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
As far as solid tumor-targeting is concerned, a lack of functional lymphatic drainage will result in the accumulation of macromolecules by a passive mechanism, called an enhanced permeability and retention effect (EPR effect).112,113 Therefore, passive targeting is one of the strategies for targeted delivery of ODN. Jeong et al.114 synthesized antisense ODN–PEG conjugate and made complex with PEI and ODN–PEG conjugate. After intravenous injection, ODN–PEG/PEI complex showed 2.5 times higher accumulation to tumor than naked ODN in tumor bearing mouse.
737
to tumor119 and kidney.120In vivo pDNA transfer using ultrasound was first reported in 1998, and recently sonoporation has been reported. However, the therapeutic effect in animal models via ultrasound-mediated transfer has been reported using decoy ODN to the kidney for renal transplantation121 and arteries for balloon injury,122,123 or using antisense ODN to tumor for inhibition of tumor growing124 and heart for ischemia/reperfusion injury.125 At present, ODN delivery via laser irradiation is not widely used. Most recently, in vivo ODN transfer using laser irradiation has been reported to improve the uptake of ODN to the skin126 and eye.127
IN VIVO DELIVERY APPROACHES WITH ODN Similar to pDNA, ODNs also do not readily cross the cell membrane on their own because of their large molecular mass and their high negative charge. In addition, instability to nucleases and rapid clearance of naked ODN has also limited the in vivo use of ODN.115,116 There are two types of delivery strategies to overcome these obstacle, physical transfer and carrier-mediated delivery.
Physical Methods To obtain an ODN therapeutic effect, appropriate drug concentrations must be achieved and maintained at the site of action of ODN. The therapeutic target cite of ODN is the cytoplasm and nucleolus. However, naked ODN tends to remain trapped inside endocytic vesicles and ends up being degraded in the lysosomes. For this reason, physical transfer, which could make transient pores on the cell membrane, resulted in nonendosomal uptake of ODN and avoided degradation in the lysosomes. Several approaches including electroporation, sonoporation, and laser irradiation, the detailed mechanisms of which have been described above, have been reported and improved ODN transfer has been obtained in vivo and in vitro. Although there are still some limitations with regard to use, such as the lack of targeting ability, the limitation of applicable tissue, and the limited distribution of transfection area, physical transfer has the potential to be used as a new ODN therapy. In this section, we focus on ODN physical transfer in vivo. Electroporation is widely used for physical transfer and its therapeutic application has already been examined in preclinical trials involving antisense ODN to liver117 muscle,118 siRNA DOI 10.1002/jps
Carrier-Mediated Methods Cationic polymers and liposomes are two major types of nonviral gene delivery vectors for ODN that are currently being investigated. Similar to pDNA, cationic polymers and liposomes form complexes with the negative charges of ODN in elecctrostatic interactions. Moreover, the excess positive charge of the complex of ODN and carriers could enhance the cellular uptake because of the electrostatic interaction with the negative charge on the cell membrane in vivo. Cationic polymers used for ODN delivery are classified into two types, natural polymers including chitosan128,129 and atelocollagen130,131 or synthetic polymers including PLL,132,133 PEI,134,135 and dendrimer.136,137 Since polymers could protect ODN from enzymatic degradation, ODN was stable in the serum or tissue. In addition, chitosan and atelocollagen slowly released ODN; consequently serum concentration of ODN was kept longer. Therefore, to achieve targeted delivery, ligand modification would be necessary. ODNs complexed with cationic liposomes are mainly and initially taken up by lung and then gradually move to the liver.138 Therefore, as far as in vivo use is concerned, cationic liposomes are useful for lung- or liver-targeted delivery by intravenous injection138 or direct administration to the target tissue.139 HVJ-liposomes140 are another type of liposome containing two fusigenic proteins, such as HVJ and artificial viral envelope, which can transfer encapsulated ODNs into cells. After intravenous injection, HVJ liposomes are taken up in a nonspecific manner. Therefore, HVJ liposomes are directly injected into target organs such as liver141 and heart.142 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
148
HIV1 envelop or ErbB2 expressing cells (cancer) Intratumoral or intravenous injection Protamine-antibody (HIV1 envelop or ErbB2) fusion protein
147
Liver parenchymal cell Intravenous injection Galactose-liposome
Kupffer cells (hepatitis) Intravenous injection Fucose-liposome
146
95
Liver nonparenchymal cell (hepatitis) Intravenous injection Mannose-liposome
siRNA
Among the several transcriptional factors for targeted decoy ODNs, NFkB decoy is the most widely used in reports about therapeutic effects. NFkB decoy, double stranded ODN containing NFkB binding sequence, can bind to activated NFkB and inhibit the transcription of NFkB which regulates the production of immune response related proteins and inflammation cytokines, such as TNFa, IL-1b, IFNg, and IL-12 (Fig. 3).143–145 Therefore, immune response and inflammation related molecule producing cells, including macrophage dendritic cells and Kupffer cells, are the target cells for NFkB decoy therapy. Recently, we have demonstrated targeted delivery by systemic injection and the therapeutic effect of NFkB decoy using Man-C4-Chol liposomes.95 After intravenous injection, NFkB decoy complexed with Man-C4-Chol liposomes rapidly and highly accumulated in the liver and spleen, which contains a large number of macrophages and dendritic cells. On the other hand, NFkB decoy complexed with bare cationic liposomes highly accumulated to lung and gradually moved to liver, although their liver accumulation was lower than that of NFkB decoy complexed with Man-C4-Chol liposomes. A therapeutic effect was observed in LPS-induced hepatic injury in mice. More recently, we have reported that fucosylated cationic liposomes prepared by Fuc-C4-Chol can deliver NFkB decoy targeting Kupffer cells,
Table 4. Cell-Targeted Carriers for In Vivo ODN Delivery
Targeted Delivery of Decoy ODN
Liposome
Administration Route
Target Tissue or Cell (Application)
Physical methods and cationic carriers could result in improved cellular uptake and an enhanced therapeutic effect on tissues, such as skin, muscle, and lung. In addition, intravenously injected ODN is widely and rapidly dispersed into tissues and, as a result, it is difficult to achieve effective levels of ODN at the target tissue or cells. Therefore, a suitable strategy to control the pharmacokinetic properties needs to be developed for therapy involving other organs. Furthermore, targeted delivery generally leads to a reduction in the dose and avoids side effects. For in vivo active targeted delivery systems, ligand recognition including receptor and antibody offers the potential to deliver ODN to the target cells, similar to pDNA delivery. To date, there are, as yet, few reports about the targeted delivery of ODN. In this section, we will discuss the in vivo targeted delivery system for each type of ODN. In vivo targeted delivery systems are shown in Table 4.
Mannose receptor-mediated endocytosis Fucose receptor-mediated endocytosis Asialoglycoprotein receptor-mediated endocytosis Antibody mediated endocytosis
Targeted Delivery of ODN by Carrier Methods
NFkB decoy
Reference
KAWAKAMI, HIGUCHI, AND HASHIDA
Mechanism
738
DOI 10.1002/jps
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
739
ope-negative B16 cells. Using this system, intratumorally or intravenously injected siRNA targeting c-myc, MDM2, and VEGF mRNA inhibited envelope-expressing subcutaneous B16 cells.
CONCLUSION
Figure 3. The decoy oligonucleotide approach to block the function of transcription factor NFkB.
which involves macrophages in the liver.146 After intravenous injection of NFkB decoy complexed with Fuc-C4-Chol liposomes, high and rapid accumulation in liver nonparenchymal cells was observed. Moreover, GdCl3 pretreatment significantly inhibited the liver accumulation, suggesting the contribution of Kupffer cells to liver accumulation. NFkB decoy complexed with FucC4-Chol liposomes also effectively inhibited cytokine production and liver injury induced by LPS. Targeted Delivery of siRNA Asialoglycoprotein receptor-mediated uptake is an attractive tool for liver hepatocyte-targeted delivery since asialoglycoprotein receptors are highly expressed on hepatocytes as mentioned above. Recently, Sato et al.147 reported that intravenously injected siRNA complexed with galactosylated cationic liposomes were effectively taken up by hepatocytes and significantly reduced target mRNA. To improve tumor-targeting and intracellular delivery of siRNA, Song et al.148 designed a protamine-antibody fusion protein to deliver siRNA to HIV-infected or envelope-transfected cells. The fusion protein was designed with the protemine coding sequence linked to the C terminus of the heavy chain Fab fragment of an HIV-1 envelope antibody. Intratumorally or intravenously injected protamine-antibody fusion protein complexed with FITC labeled siRNA in mouse targeted HIV envelope-expressing B16 meranoma cells, but not normal tissue or envelDOI 10.1002/jps
Successful gene therapy depends on the development of efficient delivery systems. Although pDNA and ODN are novel candidates for nonviral gene therapy, owing to their rapid degradation by nucleases in the serum and rapid clearance, their clinical use is generally limited. Therefore, targeting of pDNA and ODN is a promising strategy for gene therapy. Here, we have described physical and carrier-mediated approaches for targeting pDNA and ODN. These approaches could achieve efficient gene therapy for a number of refractory diseases in the future.
REFERENCE 1. Kawabata K, Takakura Y, Hashida M. 1995. The fate of plasmid DNA after intravenous injection in mice: Involvement of scavenger receptors in its hepatic uptake. Pharm Res 12:825–830. 2. Yoshida M, Mahato RI, Kawabata K, Takakura Y, Hashida M. 1996. Disposition characteristics of plasmid DNA in the single-pass rat liver perfusion system. Pharm Res 13:599–603. 3. Takakura Y, Takagi T, Hashiguchi M, Nishikawa M, Yamashita F, Doi T, Imanishi T, Suzuki H, Kodama T, Hashida M. 1999. Characterization of plasmid DNA binding and uptake by peritoneal macrophages from class A scavenger receptor knockout mice. Pharm Res 16:503–508. 4. Klein TM, Wolf ED, Wu R, Sanford JC. 1987. Highvelocity microprojectiles for delivering nucleic acids into living cells. Nature 327:70–73. 5. Matthews KE, Mills GB, Horsfall W, Hack N, Skorecki K, Keating A. 1993. Bead transfection: Rapid and efficient gene transfer into marrow stromal and other adherent mammalian cells. Exp Hematol 21:697–702. 6. Eisenbraun MD, Fuller DH, Haynes JR. 1993. Examination of parameters affecting the elicitation of humoral immune responses by particle bombardment-mediated genetic immunization. DNA Cell Biol 12:791–797. 7. Lin MT, Pulkkinen L, Uitto J, Yoon K. 2000. The gene gun: Current applications in cutaneous gene therapy. Int J Dermatol 39:161–170. JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
740
KAWAKAMI, HIGUCHI, AND HASHIDA
8. Wang S, Joshi S, Lu S. 2004. Delivery of DNA to skin by particle bombardment. Methods Mol Biol 245:185–196. 9. Alvarez D, Harder G, Fattouh R, Sun J, Goncharova S, Sta¨ mpfli MR, Coyle AJ, Bramson JL, Jordana M. 2005. Cutaneous antigen priming via gene gun leads to skin-selective Th2 immuneinflammatory responses. J Immunol 174:1664– 1674. 10. Muangmoonchai R, Wong SC, Smirlis D, Phillips IR, Shephardl EA. 2002. Transfection of liver in vivo by biolistic particle delivery: Its use in the investigation of cytochrome P450 gene regulation. Mol Biotechnol 20:145–151. 11. Kuriyama S, Mitoro A, Tsujinoue H, Nakatani T, Yoshiji H, Tsujimoto T, Yamazaki M, Fukui H. 2000. Particle-mediated gene transfer into murine livers using a newly developed gene gun. Gene Ther 7:1132–1136. 12. Sato H, Hattori S, Kawamoto S, Kudoh I, Hayashi A, Yamamoto I, Yoshinari M, Minami M, Kanno H. 2000. In vivo gene gun-mediated DNA delivery into rodent brain tissue. Biochem Biophys Res Commun 270:163–170. 13. Zhang G, Selzer ME. 2001. In vivo transfection of lamprey brain neurons by gene gun delivery of DNA. Exp Neurol 167:304–311. 14. Roberts LK, Barr LJ, Fuller DH, McMahon CW, Leese PT, Jones S. 2005. Clinical safety and efficacy of a powdered Hepatitis B nucleic acid vaccine delivered to the epidermis by a commercial prototype device. Vaccine 23:4867– 4878. 15. Drape RJ, Macklin MD, Barr LJ, Jones S, Haynes JR, Dean HJ. 2006. Epidermal DNA vaccine for influenza is immunogenic in humans. Vaccine 24:4475–4481. 16. Cassaday RD, Sondel PM, King DM, Macklin MD, Gan J, Warner TF, Zuleger CL, Bridges AJ, Schalch HG, Kim KM, Hank JA, Mahvi DM, Albertini MR. 2007. A phase I study of immunization using particle-mediated epidermal delivery of genes for gp100 and GM-CSF into uninvolved skin of melanoma patients. Clin Cancer Res 13:540– 549. 17. Neumann E, Schaefer-Ridder M, Wang Y, Hofschneider PH. 1982. Gene transfer into mouse lyoma cells by electroporation in high electric fields. EMBO J 1:841–845. 18. Andreason GL, Evans GA. 1989. Optimization of electroporation for transfection of mammalian cell lines. Anal Biochem 180:269–275. 19. Mir LM, Moller PH, Andre´ F, Gehl J. 2005. Electric pulse-mediated gene delivery to various animal tissues. Adv Genet 54:83–114. 20. Wells DJ. 2004. Gene therapy progress and prospects: Electroporation and other physical methods. Gene Ther 11:1363–1369.
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
21. Denet AR, Vanbever R, Pre´ at V. 2004. Skin electroporation for transdermal and topical delivery. Adv Drug Deliv Rev 56:659–674. 22. Vanbever R, Pre´ at V. 1999. In vivo efficacy and safety of skin electroporation. Adv Drug Deliv Rev 35:77–88. 23. Hoover F, Magne Kalhovde J. 2000. A doubleinjection DNA electroporation protocol to enhance in vivo gene delivery in skeletal muscle. Anal Biochem 285:175–178. 24. Bettan M, Ivanov MA, Mir LM, Boissiere F, Delaere P, Scherman D. 2000. Efficient DNA electrotransfer into tumors. Bioelectrochemistry 52:83–90. 25. Jaroszeski MJ, Heller LC, Gilbert R, Heller R. 2004. Electrically mediated plasmid DNA delivery to solid tumors in vivo. Methods Mol Biol 245:237– 244. 26. Liu F, Huang L. 2002. Electric gene transfer to the liver following systemic administration of plasmid DNA. Gene Ther 9:1116–1119. 27. Sakai M, Nishikawa M, Thanaketpaisarn O, Yamashita F, Hashida M. 2005. Hepatocyte-targeted gene transfer by combination of vascularly delivered plasmid DNA and in vivo electroporation. Gene Ther 12:607–616. 28. Kim HJ, Greenleaf JF, Kinnick RR, Bronk JT, Bolander ME. 1996. Ultrasound-mediated transfection of mammalian cells. Hum Gene Ther 7: 1339–1346. 29. Tata DB, Dunn F, Tindall DJ. 1997. Selective clinical ultrasound signals mediate differential gene transfer and expression in two human prostate cancer cell lines: LnCap and PC-3. Biochem Biophys Res Commun 234:64–67. 30. Bao S, Thrall BD, Miller DL. 1997. Transfection of a reporter plasmid into cultured cells by sonoporation in vitro. Ultrasound Med Biol 23:953–959. 31. Miller MW. 2000. Gene transfection and drug delivery. Ultrasound Med Biol 26:S59–62. 32. Bao S, Thrall BD, Gies RA, Miller DL. 1998. In vivo transfection of melanoma cells by lithotripter shock waves. Cancer Res 58:219–221. 33. Huber PE, Pfisterer P. 2000. In vitro and in vivo transfection of plasmid DNA in the Dunning prostate tumor R3327-AT1 is enhanced by focused ultrasound. Gene Ther 7:1516–1525. 34. Taniyama Y, Tachibana K, Hiraoka K, Aoki M, Yamamoto S, Matsumoto K, Nakamura T, Ogihara T, Kaneda Y, Morishita R. 2002. Development of safe and efficient novel nonviral gene transfer using ultrasound: Enhancement of transfection efficiency of naked plasmid DNA in skeletal muscle. Gene Ther 9:372–380. 35. Lu QL, Liang HD, Partridge T, Blomley MJ. 2003. Microbubble ultrasound improves the efficiency of gene transduction in skeletal muscle in vivo with reduced tissue damage. Gene Ther 10:396–405.
DOI 10.1002/jps
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
36. Chen S, Shohet RV, Bekeredjian R, Frenkel P, Grayburn PA. 2003. Optimization of ultrasound parameters for cardiac gene delivery of adenoviral or plasmid deoxyribonucleic acid by ultrasoundtargeted microbubble destruction. J Am Coll Cardiol 42:301–308. 37. Taniyama Y, Tachibana K, Hiraoka K, Namba T, Yamasaki K, Hashiya N, Aoki M, Ogihara T, Yasufumi K, Morishita R. 2002. Local delivery of plasmid DNA into rat carotid artery using ultrasound. Circulation 105:1233–1239. 38. Miller DL, Song J. 2003. Tumor growth reduction and DNA transfer by cavitation-enhanced highintensity focused ultrasound in vivo. Ultrasound Med Biol 29:887–893. 39. Kurata S, Tsukakoshi M, Kasuya T, Ikawa Y. 1986. The laser method for efficient introduction of foreign DNA into cultured cells. Exp Cell Res 162:372–378. 40. Shirahata Y, Ohkohchi N, Itagak H, Satomi S. 2001. New technique for gene transfection using laser irradiation. J Investig Med 49: 184–190. 41. Palumbo G, Caruso M, Crescenzi E, Tecce MF, Roberti G, Colasanti A. 1996. Targeted gene transfer in eucaryotic cells by dye-assisted laser optoporation. J Photochem Photobiol B 36: 41–46. 42. Zeira E, Manevitch A, Khatchatouriants A, Pappo O, Hyam E, Darash-Yahana M, Tavor E, Honigman A, Lewis A, Galun E. 2003. Femtosecond infrared laser-an efficient and safe in vivo gene delivery system for prolonged expression. Mol Ther 8:342–350. 43. Borchard G. 2001. Chitosans for gene delivery. Adv Drug Deliv Rev 52:145–150. 44. Mansouri S, Lavigne P, Corsi K, Benderdour M, Beaumont E, Fernandes JC. 2004. Chitosan-DNA nanoparticles as non-viral vectors in gene therapy: Strategies to improve transfection efficacy. Eur J Pharm Biopharm 57:1–8. 45. Sano A, Maeda M, Nagahara S, Ochiya T, Honma K, Itoh H, Miyata T, Fujioka K. 2003. Atelocollagen for protein and gene delivery. Adv Drug Deliv Rev 55:1651–1677. 46. Ochiya T, Nagahara S, Sano A, Itoh H, Terada M. 2001. Biomaterials for gene delivery: Atelocollagen-mediated controlled release of molecular medicines. Curr Gene Ther 1:31–52. 47. Hashida M, Takemura S, Nishikawa M, Takakura Y. 1998. Targeted delivery of plasmid DNA complexed with galactosylated poly(L-lysine). J Control Release 53:301–310. 48. Oupicky´ D, Howard KA, Kona´ k C, Dash PR, Ulbrich K, Seymour LW. 2000. Steric stabilization of poly-L-Lysine/DNA complexes by the covalent attachment of semitelechelic poly[N-(2-hydroxypropyl)methacrylamide]. Bioconjug Chem 11: 492–501.
DOI 10.1002/jps
741
49. Boussif O, Lezoualc’h F, Zanta MA, Mergny MD, Scherman D, Demeneix B, Behr JP. 1995. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: Polyethylenimine. Proc Natl Acad Sci USA 92:7297–7301. 50. Morimoto K, Nishikawa M, Kawakami S, Nakano T, Hattori Y, Fumoto S, Yamashita F, Hashida M. 2003. Molecular weight-dependent gene transfection activity of unmodified and galactosylated polyethyleneimine on hepatoma cells and mouse liver. Mol Ther 7:254–261. 51. Kunath K, von Harpe A, Petersen H, Fischer D, Voigt K, Kissel T, Bickel U. 2002. The structure of PEG-modified poly(ethylene imines) influences biodistribution and pharmacokinetics of their complexes with NFkB decoy in mice. Pharm Res 19: 810–817. 52. Kukowska-Latallo JF, Bielinska AU, Johnson J, Spindler R, Tomalia DA, Baker JR. 1996. Efficient transfer of genetic material into mammalian cells using Starburst polyamidoamine dendrimers. Proc Natl Acad Sci USA 93:4897–4902. 53. Bielinska AU, Yen A, Wu HL, Zahos KM, Sun R, Weiner ND, Baker JR, Jr, Roessler BJ. 2000. Application of membrane-based dendrimer/DNA complexes for solid phase transfection in vitro and in vivo. Biomaterials 21:877–887. 54. Ratner BD, Bryant SJ. 2004. Biomaterials: Where we have been and where we are going. Annu Rev Biomed Eng 6:41–75. 55. Zhu N, Liggitt D, Liu Y, Debs R. 1993. Systemic gene expression after intravenous DNA delivery into adult mice. Science 261:209–211. 56. Liu F, Qi H, Huang L, Liu D. . Factors controllimg the efficiency of cationic lipid-mediated transfection in vivo via intravenous administration. Gene Ther 4:517–523. 57. Song YK, Liu F, Liu D. 1998. Enhanced gene expression in mouse lung by prolonging the retention time of intravenously injected plasmid DNA. Gene Ther 5:1531–1537. 58. Dzau VJ, Mann MJ, Morishita R, Kaneda Y. 1996. Fusigenic viral liposome for gene therapy in cardiovascular diseases. Proc Natl Acad Sci USA 93: 11421–11425. 59. Hirano T, Kaneko S, Kaneda Y, Saito I, Tamaoki T, Furuyama J, Tamaoki T, Kobayashi K, Ueki T, Fujimoto J. 2001. HVJ-liposome-mediated transfection of HSVtk gene driven by AFP promoter inhibits hepatic tumor growth of hepatocellular carcinoma in SCID mice. Gene Ther 8:80–83. 60. Ellison KE, Bishopric NH, Webster KA, Morishita R, Gibbons GH, Kaneda Y, Sato B, Dzau VJ. 1996. Fusigenic liposome-mediated DNA transfer into cardiac myocytes. J Mol Cell Cardiol 28:1385– 1399. 61. Ogris M, Steinlein P, Kursa M, Mechtler K, Kircheis R, Wagner E. 1998. The size of DNA/
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
742
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
KAWAKAMI, HIGUCHI, AND HASHIDA
transferrin-PEI complexes is an important factor for gene expression in cultured cells. Gene Ther 5:1425–1433. Kawakami S, Fumoto S, Nishikawa M, Yamashita F, Hashida M. 2000. In vivo gene delivery to the liver using novel galactosylated cationic liposomes. Pharm Res 17:306–313. Yuan F, Dellian M, Fukumura D, Leunig M, Berk DA, Torchilin VP, Jain RK. 1995. Vascular permeability in a human tumor xenograft: Molecular size dependence and cutoff size. Cancer Res 55:3752– 3756. Yang JP, Huang L. 1997. Overcoming the inhibitory effect of serum on lipofection by increasing the charge ratio of cationic liposome to DNA. Gene Ther 4:950–960. Fumoto S, Nakadori F, Kawakami S, Nishikawa M, Yamashita F, Hashida M. 2003 Analysis of hepatic disposition of galactosylated cationic liposome/plasmid DNA complexes in perfused rat liver. Pharm Res 20:1452–1459. Kawakami S, Hattori Y, Lu Y, Higuchi Y, Yamashita F, Hashida M. 2004. Effect of cationic charge on receptor-mediated transfection using mannosylated cationic liposome/plasmid DNA complexes following the intravenous administration in mice. Pharmazie 59:405–408. Fumoto S, Kawakami S, Ito Y, Shigeta K, Yamashita F, Hashida M. 2004. Enhanced hepatocyteselective in vivo gene expression by stabilized galactosylated liposome/plasmid DNA complex using sodium chloride for complex formation. Mol Ther 10:719–729. Sakurai F, Nishioka T, Saito H, Baba T, Okuda A, Matsumoto O, Taga T, Yamashita F, Takakura Y, Hashida M. 2001. Interaction between DNA-cationic liposome complexes and erythrocytes is an important factor in systemic gene transfer via the intravenous route in mice: The role of the neutral helper lipid. Gene Ther 8:677–686. Eliyahu H, Servel N, Domb AJ, Barenholz Y. 2002. Lipoplex-induced hemagglutination: Potential involvement in intravenous gene delivery. Gene Ther 9:850–858. Lewis JG, Lin KY, Kothavale A, Flanagan WM, Matteucci MD, DePrince RB, Mook RA, Jr, Hendren RW, Wagner RW. 1996. A serum-resistant cytofectin for cellular delivery of antisense oligodeoxynucleotides and plasmid DNA. Proc Natl Acad Sci USA 93:3176–3181. Escriou V, Ciolina C, Lacroix F, Byk G, Scherman D, Wils P. 1998. Cationic lipid-mediated gene transfer: Effect of serum on cellular uptake and intracellular fate of lipopolyamine/DNA complexes. Biochim Biophys Acta 1368:276–288. Hong K, Zheng W, Baker A, Papahadjopoulos D. 1997. Stabilization of cationic liposome-plasmid DNA complexes by polyamines and poly(ethylene
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
glycol)-phospholipid conjugates for efficient in vivo gene delivery. FEBS Lett 400:233–237. Liu Y, Mounkes LC, Liggitt HD, Brown CS, Solodin I, Heath TD, Debs RJ. 1997. Factors influencing the efficiency of cationic liposome-mediated intravenous gene delivery. Nat Biotechnol 15:167– 173. Sternberg B, Hong K, Zheng W, Papahadjopoulos D. 1998. Ultrastructural characterization of cationic liposome-DNA complexes showing enhanced stability in serum and high transfection activity in vivo. Biochim Biophys Acta 1375:23–35. Sorgi FL, Bhattacharya S, Huang L. 1997. Protamine sulfate enhances lipid-mediated gene transfer. Gene Ther 4:961–968. Li S, Huang L. 1997. In vivo gene transfer via intravenous administration of cationic lipid-protamine-DNA (LPD) complexes. Gene Ther 4:891–900. Wu GY, Wu CH. 1988. Receptor-mediated gene delivery and expression in vivo. J Biol Chem 263:14621–14624. Chowdhury NR, Wu CH, Wu GY, Yerneni PC, Bommineni VR, Chowdhury JR. 1993. Fate of DNA targeted to the liver by asialoglycoprotein receptor-mediated endocytosis in vivo. Prolonged persistence in cytoplasmic vesicles after partial hepatectomy. J Biol Chem 268:11265–11271. Perales JC, Ferkol T, Beegen H, Ratnoff OD, Hanson RW. 1994. Gene transfer in vivo: Sustained expression and regulation of genes introduced into the liver by receptor-targeted uptake. Proc Natl Acad Sci USA 91:4086–4090. Nishikawa M, Yamauchi M, Morimoto K, Ishida E, Takakura Y, Hashida M. 2000. Hepatocytetargeted in vivo gene expression by intravenous injection of plasmid DNA complexed with synthetic multi-functional gene delivery system. Gene Ther 7:548–555. Hara T, Aramaki Y, Takada S, Koike K, Tsuchiya S. 1995. Receptor-mediated transfer of pSV2CAT DNA to mouse liver cells using asialofetuinlabeled liposomes. Gene Ther 2:784–788. Remy JS, Kichler A, Mordvinov V, Schuber F, Behr JP. 1995. Targeted gene transfer into hepatoma cells with lipopolyamine-condensed DNA particles presenting galactose ligands: A stage toward artificial viruses. Proc Natl Acad Sci USA 92:1744–1748. Kawakami S, Sato A, Nishikawa M, Yamashita F, Hashida M. 2000. Mannose receptor-mediated gene transfer into macrophages using novel mannosylated cationic liposomes. Gene Ther 7:292– 299. Kawakami S, Sato A, Yamada M, Yamashita F, Hashida M. 2001. The effect of lipid composition on receptor-mediated in vivo gene transfection using mannosylated cationic liposomes in mice. STP Pharma Sci 11:117–120.
DOI 10.1002/jps
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
85. Hattori Y, Kawakami S, Suzuki S, Yamashita F, Hashida M. 2004. Enhancement of immune responses by DNA vaccination through targeted gene delivery using mannosylated cationic liposome formulations following intravenous administration in mice. Biochem Biophys Res Commun 317:992–999. 86. Hattori Y, Kawakami S, Nakamura K, Yamashita F, Hashida M. 2006. Efficient gene transfer into macrophages and dendritic cells by in vivo gene delivery with mannosylated lipoplex via the intraperitoneal route. J Pharmacol Exp Ther 318:828– 834. 87. Hattori Y, Kawakami S, Lu Y, Nakamura K, Yamashita F, Hashida M. 2006. Enhanced DNA vaccine potency by mannosylated lipoplex after intraperitoneal administration. J Gene Med 8: 824–834. 88. Ogris M, Brunner S, Schuller S, Kircheis R, Wagner E. 1999. PEGylated DNA/transferrinPEI complexes: Reduced interaction with blood components, extended circulation in blood and potential for systemic gene delivery. Gene Ther 6:595–605. 89. Kircheis R, Blessing T, Brunner S, Wightman L, Wagner E. 2001. Tumor targeting with surfaceshielded ligand—Polycation DNA complexes. J Control Release 72:165–170. 90. Kursa M, Walker GF, Roessler V, Ogris M, Roedl W, Kircheis R, Wagner E. 2003. Novel shielded transferrin-polyethylene glycol-polyethylenimine/DNA complexes for systemic tumor-targeted gene transfer. Bioconjug Chem 14:222–231. 91. Hofland HE, Masson C, Iginla S, Osetinsky I, Reddy JA, Leamon CP, Scherman D, Bessodes M, Wils P. 2002. Folate-targeted gene transfer in vivo. Mol Ther 5:739–744. 92. Reddy JA, Abburi C, Hofland H, Howard SJ, Vlahov I, Wils P, Leamon CP. 2002. Folatetargeted, cationic liposome-mediated gene transfer into disseminated peritoneal tumors. Gene Ther 9:1542–1550. 93. Ward CM, Pechar M, Oupicky D, Ulbrich K, Seymour LW. 2002 Modification of pLL/DNA complexes with a multivalent hydrophilic polymer permits folate-mediated targeting in vitro and prolonged plasma circulation in vivo. J Gene Med 4:536–547. 94. Miyao T, Takakura Y, Akiyama T, Yoneda F, Sezaki H, Hashida M. 1995. Stability and pharmacokinetic characteristics of oligonucleotides modified at terminal linkages in mice. Antisense Res Dev 5:115–121. 95. Higuchi Y, Kawakami S, Oka M, Yabe Y, Yamashita F, Hashida M. 2006. Intravenous administration of mannosylated cationic liposome/NFkB decoy complexes effectively prevent LPS-induced
DOI 10.1002/jps
96.
97.
98.
99.
100.
101.
102.
103.
104.
105.
106. 107.
108. 109.
110.
111.
743
cytokine production in a murine liver failure model. FEBS Lett 580:3706–3714. Takakura Y, Mahato RI, Yoshida M, Kanamaru T, Hashida M. 1996. Uptake characteristics of oligonucleotides in the isolated rat liver perfusion system. Antisense Nucleic Acid Drug Dev 6:177–183. Sawai K, Miyao T, Takakura Y, Hashida M. 1995. Renal disposition characteristics of oligonucleotides modified at terminal linkages in the perfused rat kidney. Antisense Res Dev 5:279–287. Sawai K, Mahato RI, Oka Y, Takakura Y, Hashida M. 1996. Disposition of oligonucleotides in isolated perfused rat kidney: Involvement of scavenger receptors in their renal uptake. J Pharmacol Exp Ther 279:284–290. Kurreck J. 2003. Antisense technologies. Improvement through novel chemical modifications. Eur J Biochem 270:1628–1644. Chen X, Dudgeon N, Shen L, Wang JH. 2005. Chemical modification of gene silencing oligonucleotides for drug discovery and development. Drug Discov Today 10:587–593. Campbell JM, Bacon TA, Wickstrom E. 1990. Oligodeoxynucleoside phosphorothioate stability in subcellular extracts, culture media, sera, and cerebrospinal fluid. J Biochem Biophys Methods 20:259–267. Eckstein F. 2000. Phosphorothioate oligodeoxynucleotides: What is their origin and what is unique about them? Antisense Nucleic Acid Drug Dev 10: 117–121. Braasch DA, Jensen S, Liu Y, Kaur K, Arar K, White MA, Corey DR. 2003. RNA interference in mammalian cells by chemically-modified RNA. Biochemistry 42:7967–7975. Levin AA. 1999. A review of the issues in the pharmacokinetics and toxicology of phosphorothioate antisense oligonucleotides. Biochim Biophys Acta 1489:69–84. Nielsen PE, Egholm M, Berg RH, Buchardt O. 1991. Sequence-selective recognition of DNA by strand displacement with a thymine-substituted polyamide. Science 254:1497–1500. Nielsen PE. 2004. PNA Technology. Mol Biotechnol 26:233–248. Summerton J. 1999. Morpholino antisense oligomers: The case for an RNase H-independent structural type. Biochim Biophys Acta 1489:141–158. Heasman J. 2002. Morpholino oligos: Making sense of antisense? Dev Biol 243:209–214. Braasch DA, Corey DR. 2001. Locked nucleic acid (LNA): Fine-tuning the recognition of DNA and RNA. Chem Biol 8:1–7. Vester B, Wengel J. 2004. LNA (locked nucleic acid): High-affinity targeting of complementary RNA and DNA. Biochemistry 43:13233–13241. Soutschek J, Akinc A, Bramlage B, Charisse K, Constien R, Donoghue M, Elbashir S, Geick A,
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
744
112.
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
123.
KAWAKAMI, HIGUCHI, AND HASHIDA
Hadwiger P, Harborth J, John M, Kesavan V, Lavine G, Pandey RK, Racie T, Rajeev KG, Ro¨ hl I, Toudjarska I, Wang G, Wuschko S, Bumcrot D, Koteliansky V, Limmer S, Manoharan M, Vornlocher HP. 2004. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature 432:173–178. Matsumura Y, Maeda H. 1986. A new concept for macromolecular therapeutics in cancer chemotherapy: Mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 46:6387–6392. Maeda H, Matsumura Y. 1989. Tumoritropic and lymphotropic principles of macromolecular drugs. Crit Rev Ther Drug Carrier Syst 6:193–210. Jeong JH, Kim SH, Kim SW, Park TG. 2005. Polyelectrolyte complex micelles composed of craf antisense oligodeoxynucleotide-poly(ethylene glycol) conjugate and poly(ethylenimine): Effect of systemic administration on tumor growth. Bioconjug Chem 16:1034–1037. Akhtar S, Agrawal S. 1997. In vivo studies with antisense oligonucleotides. Trends Pharmacol Sci 18:12–18. Mahato RI, Takakura Y, Hashida M. 1997. Development of targeted delivery systems for nucleic acid drugs. J Drug Target 4:337–357. Baba M, Iishi H, Tatsuta M. 2000. In vivo electroporetic transfer of bcl-2 antisense oligonucleotide inhibits the development of hepatocellular carcinoma in rats. Int J Cancer 85:260–266. Wells KE, Fletcher S, Mann CJ, Wilton SD, Wells DJ. 2003. Enhanced in vivo delivery of antisense oligonucleotides to restore dystrophin expression in adult mdx mouse muscle. FEBS Lett 552:145– 149. Nakai N, Kishida T, Shin-Ya M, Imanishi J, Ueda Y, Kishimoto S, Mazda O. 2007. Therapeutic RNA interference of malignant melanoma by electrotransfer of small interfering RNA targeting Mitf. Gene Ther 14:357–365. Takabatake Y, Isaka Y, Mizui M, Kawachi H, Shimizu F, Ito T, Hori M, Imai E. 2005. Exploring RNA interference as a therapeutic strategy for renal disease. Gene Ther 12:965–973. Azuma H, Tomita N, Kaneda Y, Koike H, Ogihara T, Katsuoka Y, Morishita R. 2003. Transfection of NFkB-decoy oligodeoxynucleotides using efficient ultrasound-mediated gene transfer into donor kidneys prolonged survival of rat renal allografts. Gene Ther 10:415–425. Inagaki H, Suzuki J, Ogawa M, Taniyama Y, Morishita R, Isobe M. 2006. Ultrasound-microbubble-mediated NF-kB decoy transfection attenuates neointimal formation after arterial injury in mice. J Vasc Res 43:12–18. Hashiya N, Aoki M, Tachibana K, Taniyama Y, Yamasaki K, Hiraoka K, Makino H, Yasufumi K,
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008
124.
125.
126.
127.
128.
129.
130.
131.
132.
133.
Ogihara T, Morishita R. 2004. Local delivery of E2F decoy oligodeoxynucleotides using ultrasound with microbubble agent (Optison) inhibits intimal hyperplasia after balloon injury in rat carotid artery model. Biochem Biophys Res Commun 317:508–514. Haag P, Frauscher F, Gradl J, Seitz A, Scha¨ fer G, Lindner JR, Klibanov AL, Bartsch G, Klocker H, Eder IE. 2006. Microbubble-enhanced ultrasound to deliver an antisense oligodeoxynucleotide targeting the human androgen receptor into prostate tumours. J Steroid Biochem Mol Biol 102:103– 113. Erikson JM, Freeman GL, Chandrasekar B. 2003. Ultrasound-targeted antisense oligonucleotide attenuates ischemia/reperfusion-induced myocardial tumor necrosis factor-alpha. J Mol Cell Cardiol 35:119–130. Lee WR, Shen SC, Liu CR, Fang CL, Hu CH, Fang JY. 2006. Erbium:YAG laser-mediated oligonucleotide and DNA delivery via the skin: An animal study. J Control Release 115:344–353. Normand N, Valamanesh F, Savoldelli M, Mascarelli F, BenEzra D, Courtois Y, Behar-Cohen F. 2005. VP22 light controlled delivery of oligonucleotides to ocular cells in vitro and in vivo. Mol Vis 11:184–191. Springate CM, Jackson JK, Gleave ME, Burt HM. 2005. Efficacy of an intratumoral controlled release formulation of clusterin antisense oligonucleotide complexed with chitosan containing paclitaxel or docetaxel in prostate cancer xenograft models. Cancer Chemother Pharmacol 56:239– 247. Howard KA, Rahbek UL, Liu X, Damgaard CK, Glud SZ, Andersen MØ, Hovgaard MB, Schmitz A, Nyengaard JR, Besenbacher F, Kjems J. 2006. RNA interference in vitro and in vivo using a novel chitosan/siRNA nanoparticle system. Mol Ther 14:476–484. Takeshita F, Minakuchi Y, Nagahara S, Honma K, Sasaki H, Hirai K, Teratani T, Namatame N, Yamamoto Y, Hanai K, Kato T, Sano A, Ochiya T. 2005. Efficient delivery of small interfering RNA to bone-metastatic tumors by using atelocollagen in vivo. Proc Natl Acad Sci USA 102:12177– 12182. Hanai K, Kurokawa T, Minakuchi Y, Maeda M, Nagahara S, Miyata T, Ochiya T, Sano A. 2004. Potential of atelocollagen-mediated systemic antisense therapeutics for inflammatory disease. Hum Gene Ther 15:263–272. Clarenc JP, Degols G, Leonetti JP, Milhaud P, Lebleu B. 1993. Delivery of antisense oligonucleotides by poly(L-lysine) conjugation and liposome encapsulation. Anticancer Drug Des 8:81–94. Sugano M, Makino N, Sawada S, Otsuka S, Watanabe M, Okamoto H, Kamada M, Mizushima A.
DOI 10.1002/jps
NONVIRAL DELIVERY FOR PLASMID DNA AND OLIGONUCLEOTIDE
134.
135.
136.
137.
138.
139.
140.
141.
1998. Effect of antisense oligonucleotides against cholesteryl ester transfer protein on the development of atherosclerosis in cholesterol-fed rabbits. J Biol Chem 273:5033–5036. Boussif O, Lezoualc’h F, Zanta MA, Mergny MD, Scherman D, Demeneix B, Behr JP. 1995. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: Polyethylenimine. Proc Natl Acad Sci USA 92:7297–7301. Williams JH, Sirsi SR, Latta DR, Lutz GJ. 2006. Induction of dystrophin expression by exon skipping in mdx mice following intramuscular injection of antisense oligonucleotides complexed with PEG-PEI copolymers. Mol Ther 14:88–96. Yoo H, Juliano RL. 2000. Enhanced delivery of antisense oligonucleotides with fluorophore-conjugated PAMAM dendrimers. Nucleic Acids Res 28:4225–4231. Marano RJ, Toth I, Wimmer N, Brankov M, Rakoczy PE. 2005. Dendrimer delivery of an anti-VEGF oligonucleotide into the eye: A longterm study into inhibition of laser-induced CNV, distribution, uptake, and toxicity. Gene Ther 12: 1544–1550. Higuchi Y, Kawakami S, Oka M, Yamashita F, Hashida M. 2006. Suppression of TNFa production in LPS induced liver failure in mice after intravenous injection of cationic liposomes/NFkB decoy complex. Pharmazie 61:144–147. Griesenbach U, Scheid P, Hillery E, de Martin R, Huang L, Geddes DM, Alton EW. 2000. Antiinflammatory gene therapy directed at the airway epithelium. Gene Ther 7:306–313. Nakamura H, Morishita R, Kaneda Y. 2002. Molecular therapy via transcriptional regulation with double-stranded oligodeoxynucleotides as decoys. In Vivo 16:45–48. Kupatt C, Habazettl H, Goedecke A, Wolf DA, Zahler S, Boekstegers P, Kelly RA, Becker BF.
DOI 10.1002/jps
142.
143.
144.
145.
146.
147.
148.
745
1999. Tumor necrosis factor-alpha contributes to ischemia- and reperfusion-induced endothelial activation in isolated hearts. Circ Res 84:392–400. Ogushi I, Iimuro Y, Seki E, Son G, Hirano T, Hada T, Tsutsui H, Nakanishi K, Morishita R, Kaneda Y, Fujimoto J. 2003. Nuclear factor kappa B decoy oligodeoxynucleotides prevent endotoxin-induced fatal liver failure in a murine model. Hepatology 38:335–344. Bielinska A, Shivdasani RA, Zhang LQ, Nabel GJ. 1990. Regulation of gene expression with double-stranded phosphorothioate oligonucleotides. Science 250:997–1000. Morishita R, Higaki J, Tomita N, Ogihara T. 1998. Application of transcription factor ‘‘decoy’’ strategy as means of gene therapy and study of gene expression in cardiovascular disease. Circ Res 82: 1023–1028. Morishita R, Sugimoto T, Aoki M, Kida I, Tomita N, Moriguchi A, Maeda K. 1997. In vivo transduction of cis element decoy against nuclear factor-kB binding siteprevents myocardial infraction. Nat Med 3:894–899. Higuchi Y, Kawakami S, Yamashita F, Hashida M. 2007. The potential role of fucosylated cationic liposome/NFkB decoy complexes in the treatment of cytokine-related liver disease. Biomaterials 28:532–539. Sato A, Takagi M, Shimamoto S, Kawakami S, Hashida M. 2007. Small interfering RNA delivery to the liver by intravenous administration of galactosylated cationic liposomes in mice. Biomaterials 28:1434–1442. Song E, Zhu P, Lee SK, Chowdhury D, Kussman S, Dykxhoorn DM, Feng Y, Palliser D, Weiner DB, Shankar P, Marasco WA, Lieberman J. 2005. Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors. Nat Biotechnol 23:709–717.
JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 2, FEBRUARY 2008