Open access

Targeted Gene Delivery: Importance of Administration Routes

Written By

Shintaro Fumoto, Shigeru Kawakami, Mitsuru Hashida and Koyo Nishida

Submitted: 01 May 2012 Published: 13 February 2013

DOI: 10.5772/54741

From the Edited Volume

Novel Gene Therapy Approaches

Edited by Ming Wei and David Good

Chapter metrics overview

2,931 Chapter Downloads

View Full Metrics

1. Introduction

Gene therapy is a promising approach to treat intractable and refractory diseases at the genetic level. Basically, in gene therapy, target gene expression is induced by delivering foreign genes. Downregulation of target gene expression or gene silencing can also be performed using miRNA, siRNA or shRNA expression vectors [1]. Gene therapy is useful for both genetic and acquired diseases. For genetic diseases, the first clinical trial was performed for adenosine deaminase deficiency in 1990 [2]. Subsequently, numerous clinical trials were carried out for other congenital genetic defects such as familial hypercholesterolemia and cystic fibrosis [3]. Gene therapy clinical trials were also performed for acquired diseases such as cancers, cardiovascular diseases and infectious diseases [3].

There are two strategies to perform gene therapy, that is, ex vivo methods and in vivo methods. In ex vivo gene transfer, once cells are taken from a patient, in vitro gene transfer is performed, and then transfected cells are introduced into the patient. Since ex vivo gene transfer requires a cell culture facility, the procedure is cumbersome. On the other hand, in vivo gene transfer is performed by directly administering genetic medicine into the patient. When foreign genes are administered into systemic circulation as a naked form, they are rapidly taken up by the reticuloendothelial system and degraded by nuclease in the blood [4]; thus, foreign genes themselves are generally inactive in gene transfer. As such, to achieve in vivo gene transfer, both viral and non-viral vectors have been utilized. In both cases, the selectivity of transgene expression in target organs/sites/cells would determine the therapeutic outcome. Uncontrolled transgene expression in non-target organs/sites/cells is problematic due to high biological activities of transgene products. Furthermore, undesirable biodistribution of vectors leads to their loss and vector-dependent side effects. Thus, gene delivery systems that are targeted to specific organs/sites/cells are important for not only efficacy but also safety.

Advertisement

2. Overview of targeted gene delivery

There are several strategies to achieve targeted gene delivery. Among them, modification with a ligand for specific receptors on target cells is a rational approach. Viral vectors natively utilize specific receptors. For example, adenoviral vector serotype 5 utilizes coxsackievirus and adenovirus receptor (CAR) and integrin, which are abundant on mouse hepatocytes [5, 6]. On the other hand, the receptor for adenoviral vector serotype 35 is CD34, which is expressed on human hematopoietic stem cells [7]. As another good example, sugar modification of vectors is useful. Galactosylation of vectors is useful for targeting to hepatocytes via asialoglycoprotein receptors [8], whereas mannosylation is useful for targeting to macrophages [9]. Furthermore, antibodies against cell surface proteins are also a useful tool for targeting. Antibody against transferrin receptors is utilized for targeting to the brain [10, 11].

Activation of vectors by target cell-specific enzymes is also a rational strategy. In most tumor cells, protein kinase Cα (PKCα) is hyper-activated. A cationic polymer having a peptide substrate of PKCα is specifically phosphorylated in tumor cells; subsequently, the polymer is detached from DNA and transgene expression is turned on [12]. As a similar strategy, a polymer having HIV proteinase-cleavable cationic residues has been developed [13].

To regulate transgene expression in target cells, a tissue-selective promoter can be utilized. For example, albumin promoter and human α1-antitrypsin promoter selectively work in liver hepatocytes [14]. Tumor-selective promoters such as AFP promoter [15] and CAE promoter [16] are useful to improve tumor-selective transgene expression.

Selection of administration routes is a simple and useful way to control the in vivo fate of both viral and non-viral vectors. Selection of administration routes can be combined with other strategies. Depending on the administration routes, accessibilities of vectors to target organs/sites/cells vary significantly. Thus, selection of administration routes is important.

Figure 1.

Scheme of administration routes for targeted gene delivery.

Advertisement

3. Administration routes

Figure 1 shows a schematic representation of administration routes for targeted gene delivery. When target cells are distributed throughout the body, various administration routes can be chosen. Antigen-presenting cells such as macrophages and dendritic cells are good examples. Factors affecting transgene expression, such as interaction with blood components and retention time, are different in each administration route. In addition, transfected cell types are dependent on administration routes. When target cells have polarity, secretion polarity of transgene products is subject to the route of transfection, that is, apical or basal route. Thus, we should cautiously select administration routes in accordance with the purpose. We explain the characteristics of each administration route below.

3.1. Oral route

The oral route is one of the most attractive and challenging routes. Non-invasive administration could be theoretically achieved by the oral route. The potential for daily intake of genetic medicine is also one of the merits of oral administration. Cells in the gastrointestinal tract are transfected via oral routes. Using foreign genes encoding secretion proteins, the transgene products can be secreted into systemic circulation. However, the epithelial barrier, acidic pH in the stomach and digestive fluids are major obstacles for gene transfer via the oral route.

The in vivo stability of a recombinant adeno-associated virus (rAAV) type 2 vector could be improved by gastric acid neutralization with sodium bicarbonate and protease inhibition with aprotinin [17]. Despite these changes, the transduction efficiency after oral administration of this vector remained low. We also failed to detect transgene expression after intragastric injection of plasmid DNA in mice [18]. To overcome these obstacles, microparticles and nanoparticles are a promising approach. Chitosan-DNA microparticles could protect the encapsulated plasmid DNA from nuclease degradation [19]. In in vivo animal studies, a blue color was observed upon X-gal staining of histological stomach and small intestine sections after oral administration of chitosan-DNA microparticles. Furthermore, chitosan nanoparticles using quaternized chitosan (60% trimethylated chitosan) that were given via a gastric feeding tube exhibited green fluorescent protein expression in the mucosa of the stomach, duodenum, jejunum, ileum and large intestine [20]. Bhavsar and Amiji developed a hybrid system dubbed the nanoparticles-in-microsphere oral system (NiMOS), which consists of gelatin nanoparticles containing plasmid DNA and a poly(epsilon-caprolactone) outer shell [21]. NiMOS resided in the stomach and small intestine for longer than gelatin nanoparticles alone.

In the case of DNA vaccines, transfection into only a subset of antigen-presenting cells may be sufficient for the vaccination to exhibit its required effect. The feasibility of DNA vaccination via the oral route may be high since one or a few administrations is theoretically enough to maintain immunity. In fact, oral DNA vaccines against Mycobacterium tuberculosis using liposome [22] and attenuated Salmonella vector [23] were developed and elicited immune responses.

3.2. Intravenous route

Various targeted gene delivery systems via the intravenous route have been developed worldwide. By intravenous administration, various organs and cells can be targeted. However, undesirable and broad biodistribution of vectors can easily lead to side effects.

Adenoviral vectors have liver tropism after intravenous injection [24]. If the target is not the liver, it is necessary to reduce hepatic transgene expression. Fiber-shaft exchange from adenovirus serotype 5 to serotype 35 in combination with both CAR- and αv integrin-binding ablation by mutation reduced liver tropism [25]. Such mutation may be suitable for retargeting from the liver to other organs/tissues. Capsid engineering of adenoviral fibers from serotype 19p based on phage display technology is useful for targeting to the kidney [26]. On the other hand, when cationic liposome/plasmid DNA complex (lipoplex) was injected intravenously, transgene expression mainly occurred in the lung [27]. Galactosylation of the lipoplex reduced transgene expression in the lung after intravenous injection, while it maintained transgene expression in the liver; however, it remained unselective to the liver [28]. In contrast, we successfully delivered foreign genes to the liver Kupffer cells via the intravenous route by mannosylation of the lipoplex [9].

Innate and adaptive immune responses caused by vector administration are problematic. Recombinant adenoviral vectors induce the production of neutralizing antibodies by single administration [29]. Moreover, neutralizing antibodies to human adenovirus serotype 5 have a prevalence of 60% in Europe [30, 31], 35–70% in North America [32, 33] and 75–100% in Asia [34]; thus, many patients already have neutralizing antibodies before administration of recombinant adenoviral vectors. Neutralizing antibodies also induce complement activation upon administration of recombinant adenoviruses [35]. In addition, an alternative pathway is also activated by recombinant adenoviruses [36]. Neutrophils recognize opsonized adenoviral vectors [37]. These immune responses can cause adverse side effects. In fact, administration of recombinant adenoviral vectors causes liver damage and elevates c-reactive protein in cynomolgus monkey [38]. Moreover, human mortality upon the administration of recombinant adenoviral vectors was reported [39]. On the other hand, non-viral vectors also induce immune responses. Plasmid DNA generally contains an immunostimulatory CpG motif, which is recognized by Toll-like receptor 9 [40, 41]. Lipoplex containing plasmid DNA causes the production of inflammatory cytokines and subsequent liver damage [42, 43]. Immunostimulatory CpG motifs in plasmid DNA also inhibit transgene expression by lipoplex [44]. In addition, dexamethasone treatment was found to improve transgene expression by lipoplex [44]. Here, immunostimulatory CpG motifs can be depleted from plasmid DNA. As expected, depletion of immunostimulatory CpG motifs from plasmid DNA improves the safety and transgene expression over a long period [45].

When using the intravenous route, it should be considered that interaction with blood components can affect transfection using viral and non-viral vectors. A low level of neutralizing antibodies against adenovirus inhibits CAR-dependent transfection, whereas neutralized adenoviral vector can transfect Fcγ receptor-positive cells [46]. However, this Fcγ receptor-mediated delivery of adenoviral vectors can induce liver inflammation [37, 47]. Binding of coagulation factor X to adenoviral vector serotype 5 determines liver and spleen tropism via heparan sulfate proteoglycan [48-50]. On the other hand, the lipoplex interacts with various blood components due to its cationic nature. Interaction of the lipoplex with serum inhibits in vitro transfection, but the inhibitory effect of serum can be overcome by increasing the charge ratio, which is the molar ratio of cationic residues of lipids to anionic residues of DNA [51]. The inhibitory effect of serum on transfection can also be overcome by increasing the lipoplex particle size [52-54]. The lipoplex interacts with complement proteins after intravenous administration in mice; however, the lipofection efficiency and biodistribution of the lipoplex did not change when complement proteins were depleted from mice [55]. Interaction of the lipoplex with plasma lipoproteins decreased transfection efficiency [56, 57]. In contrast, interaction of the lipoplex with erythrocytes greatly inhibited in vivo transfection, whereas interaction with serum did not [58, 59]. The lipoplex also induced hemagglutination upon an increase in the charge ratio [60]. Thus, it is necessary to control interaction with blood components for successful and safe in vivo transfection using lipoplex. To prevent hemagglutination, coating of cationic carriers with anionic polymers such as γ-polyglutamic acid [61, 62] and chondroitin sulfate [63, 64] is a useful strategy.

Physicochemical properties such as surface charge and particle size of vectors affect in vivo transfection, as mentioned above. The size of lipoplex is dependent on the charge ratio and can determine pulmonary transfection efficiency after intravenous injection [65]. In addition, neutral lipids, so-called ‘helper lipids’, are also important for in vivo transfection using lipoplex. While incorporation of DOPE to liposomes is effective in cell culture, incorporation of cholesterol to liposomes enhances pulmonary transfection efficiency [66]. The combination of mannosylated cationic cholesterol derivative with DOPE exhibited superior in vivo disposition and transgene expression in the liver than that with DOPC [67]. Incorporation of N-lauroylsarcosine into cationic liposomes in addition to cholesterol inhibited hemagglutination observed in the case of incorporation of DOPE, and increased the pulmonary transfection efficiency [68].

3.3. Local administration

For transfection into a specific organ/tissue/site, local administration is a useful strategy. Local administration can be categorized into the following two routes: vasculature route and non-vasculature route.

Administration routes Target organs/tissues Vectors References
ia Liver Naked plasmid DNA [69]
ia Pancreas Adenoviral vector [70]
ia Hind limb Naked plasmid DNA [71]
ia Cecum AAV [72]
ia Brain tumor Adenoviral vector and lipoplex [73]
ip Liver Lipoplex [28]
riv Kidney Naked plasmid DNA [74]

Table 1.

Administration routes for targeted gene delivery to specific organs/tissues

Abbreviations: ia, intra-arterial; ip, intraportal; riv, retrograde intravenous


3.3.1. Vasculature route

Intra-arterial, intraportal and retrograde intravenous routes have been investigated for transfection into a specific target organ. Table 1 summarizes the administration routes and tested target organs.

We developed galactosylated cationic lipoplex targeted to the liver parenchymal cells [8, 28]. Liver-selective transgene expression was observed after intraportal injection of the galactosylated lipoplex, whereas transgene expression was ineffective and non-selective to the liver after intravenous injection [9]. We also developed galactosylated polyethylenimine (PEI)/plasmid DNA complex (polyplex) and analyzed the molecular weight dependence of PEI [75]. For targeted delivery to the liver parenchymal cells, penetration through fenestrated endothelium is one of the major obstacles. We analyzed the intrahepatic disposition characteristics of galactosylated lipoplex [76] and galactosylated PEI polyplex [77]. While galactosylation of carriers was useful to deliver plasmid DNA to the liver, it was proposed that reduction of the particle size of lipoplex would further improve parenchymal cell selectivity by enhancing the penetration through fenestrated endothelium. Here, larger lipoplex exhibited superior transfection efficiency; however, liver parenchymal cell selectivity was low in large lipoplex [78]. In terms of the particle size of lipoplex and polyplex, the composition of the solution is important. Particle sizes of lipoplex and polyplex in non-ionic solution are smaller than those in ionic solution [79, 80]. In the case of siRNA, the particle size of lipoplex is relatively small; using such lipoplexes, several reported studies succeeded in delivering siRNA to hepatocytes in vivo [81, 82].

In terms of interaction of the lipoplex with serum, we reported that transgene expression in the liver after intraportal injection of galactosylated lipoplex was increased by pre-incubation of the lipoplex with serum [83]. This enhancement of transgene expression in the liver was also observed in conventional lipoplex [84]. Multiple components in serum including calcium ion, aggregation-inhibiting components, fibronectin and complement component C3 were responsible for increased transgene expression in the liver [84].

Target organs/tissues Vectors References
Skeletal muscle Naked plasmid DNA [85]
Heart Naked plasmid DNA [86]
Heart AAV [87]
Liver Naked plasmid DNA [88]
Kidney Lentiviral vector [89]
Spleen Naked plasmid DNA [90]
Stomach Naked plasmid DNA [91]
Thymus Adenoviral vector and others [92]
Tumor Naked plasmid DNA [93]
Tumor Naked plasmid DNA and lipoplex [94, 95]

Table 2.

Direct injection for targeted gene delivery to specific organs/tissues

3.3.2. Non-vasculature route

Direct injection to the target organ such as the liver or spleen has been investigated (Table 2). By direct injection to the target organ, the use of naked plasmid DNA without carrier systems is sufficient to detect transgene expression. However, in general, transgene expression is limited to the injection site. To overcome a limited transfection area, electroporation after intramuscular injection of plasmid DNA increased the number of transfected myofibers [96].

For other routes of gene transfer, retrograde intrabiliary injection of naked plasmid DNA, polyethylenimine-plasmid DNA complex and chitosan-plasmid DNA complex resulted in transgene expression in the liver [97]. Intranasal administrations of adenoviral vector [98], lipoplex and polyplex [99] were also tested. In addition, inhalation of chitosan/plasmid DNA nanoparticles resulted in pulmonary transgene expression [100]. Intracerebroventricular administration of lentiviral vector was utilized to deliver foreign genes to the brain [101]. Gene gun bombardment of plasmid DNA with gold particles resulted in efficient gene transfer to the skin [102]. After intraperitoneal injection of adenoviral vector, not only mesothelium but also parenchymal cells of the liver were transduced [103]. This non-specific biodistribution was overcome by ablation of native CAR and integrin receptor binding [103].

Figure 2.

Scheme of organ surface instillation. Panel (A) represents the proposed drug distribution after systemic administration and organ surface instillation of drugs. Panel (B) represents attachment of a glass-made cylindrical diffusion cell onto the organ surface.

3.4. Organ surface route

We developed a novel route for targeted gene delivery to intra-abdominal and intra-thoracic organs, namely, the organ surface route (Fig. 2A). When diseases are limited to a certain region, the organ surface route enables us to target the diseased region, while drugs are distributed to the whole organ via the normal route. Naked plasmid DNA was utilized to transfect target organs/sites. As a first report of this approach, the liver was targeted and successfully transfected in mice [104]. Selectivity of transgene expression in the applied liver lobe was high. Laparotomy was performed in the first reported study, but it is not essential since catheter-based administration through the abdominal wall is available [105]. This catheter-based administration is essential to the safety of liver surface instillation of plasmid DNA [106].

We developed an experimental system using a glass-made cylindrical diffusion cell attached to the organ surface (Fig. 2B) [107]. Using this experimental system, we can precisely limit the area of drug application. Specific transgene expression in the applied area of the liver was achieved [108]. The effect of solution composition on naked plasmid DNA transfer was also examined [109]. Use of hypotonic solution enhanced the transfection efficiency in the applied site of the liver. As for the mechanism of transfection, we analyzed endocytic routes for naked plasmid DNA transfer in vivo. While the lipoplex and polyplex are taken up via clathrin- and caveolae-mediated endocytosis [110-113], macropinocytosis is essential for naked plasmid DNA uptake in mesothelial cells in mice [114].

As for other organs, unilateral kidney [115], unilateral lung [116], spleen [117] and stomach surface [118, 119] were transfected with naked plasmid DNA in mice. To improve organ selectivity, microinstillation of naked plasmid DNA onto the stomach was performed [18]. Since specific transgene expression in the stomach was observed in rats [120], organ size would be an important factor for target selectivity of gene transfer. Moreover, specific transgene expression in the applied liver lobe was also achieved in mice by controlling instillation speed using an infusion pump [121].

3.5. Comparison of administration routes

We summarize the advantages and disadvantages of each administration route for targeted gene delivery in Table 3.

Administration routes Advantages Disadvantages
Oral Ease of administration,
Frequent dosing (daily intake)
Barriers (epithelium, digestive fluids), Low selectivity
Intravenous Frequent dosing,
Vast distribution
Non-specificity
Intra-arterial,
Intraportal,
Retrograde intravenous
Selective delivery
Necessity of cannulation
Direct injection Effective gene transfer,
High selectivity
Physical force against the organ,
Limited region,
Limited frequency of dosing
Intraperitoneal Effective gene transfer Low selectivity
Organ surface Effective gene transfer,
High selectivity
Necessity of laparoscopy

Table 3.

Advantages and disadvantages of vector transfer routes.

Direct injection of rAAV vector to the liver exhibited faster and stronger transgene expression than intravenous and intraportal injections of rAAV vector [122]. Similar results were obtained for direct injection of the lipoplex into localized intrahepatic tumors [123]. Moreover, direct intrahepatic injection of adenoviral vector reduced inflammation and increased transgene expression in comparison with intravenous injection [124]. On the other hand, retrograde infusion of lentiviral vector into the ureter, injection into the renal vein or artery, and direct injection into the renal parenchyma were compared [89]. Parenchymal or ureteral administration appeared to be more efficient than other routes of administration.

Figure 3.

Scheme of administration routes for targeted delivery of foreign genes to the stomach

Figure 3 depicts the administration routes for targeted gene delivery to the stomach. Via the oral route, there are many barriers such as digestive fluids and acidic pH that hamper effective gene transfer. Although effective gene transfer can be achieved by direct injection, it is necessary to consider tissue damage. In contrast, safe and effective gene transfer is possible by serosal surface instillation of naked plasmid DNA. Although transgene expression is limited to the surface layer in the case of serosal surface instillation, limited vertical distribution of transgene products can be overcome by the use of the secretory form of proteins [121].

Advertisement

4. Improving methods for targeted gene delivery

Various strategies have been tested to improve targeted gene delivery. Methods for improved targeted gene delivery can be categorized as physical approaches and chemical approaches.

Physical forces such as electroporation, sonoporation and mechanical massage have been employed to improve targeted gene delivery. Naked plasmid DNA can be delivered to the liver by intravenous injection with electroporation [125, 126]. Intravenous injection of naked plasmid DNA with tissue electroporation resulted in significant transgene expression in the liver, spleen and kidney, but not in the skin or muscle [127].

Utilization of microbubbles with ultrasound exposure can deliver naked plasmid DNA to the muscle [128, 129], liver [130] and lung [131]. Use of PEGylated liposomal bubbles containing perfluoropropane with ultrasound exposure was also effective to deliver naked plasmid DNA via the femoral artery [132]. Mannosylated lipoplex and liposomal bubbles with ultrasound exposure can transfect the liver and spleen [133]. In addition, mannosylated PEGylated bubble lipoplexes selectively transfected antigen-presenting cells in vivo [134]. DNA vaccination by this type of lipoplex with ultrasound exposure resulted in suppression of melanoma growth and metastasis [135]. The timing of ultrasound exposure was important [136]. As a mechanism of high transgene expression, a transcriptional process activated by ultrasound exposure was involved [137].

Hydrodynamics-based transfection, with rapid large volume injection of naked plasmid DNA via the intravascular route, is an efficient method to transfect the liver [138, 139]. It was also reported that pig liver can be transfected by retrograde hydrodynamic injection of plasmid DNA via an isolated segment of the inferior vena cava [140]. In terms of the mechanism of high efficiency of gene transfer in hydrodynamics-based transfection, both the generation of transient pores [141, 142] and a transcriptional process activated by hydrodynamic injection [143, 144] are important.

Naked plasmid DNA was also intravenously delivered to the liver by mechanical massage of the liver [145]. Pressure-mediated deliveries of naked plasmid DNA to the kidney [146], liver and spleen [147] were also achieved. As the mechanism of high transgene expression, a transcriptional process activated by pressure to the tissue was involved [148].

Chemical modification of gene carriers has also been investigated. PEGylation of carriers improves blood circulation of the carrier and tumor accumulation by the enhanced permeability and retention effects [149]. However, transfection efficiencies of PEGylated vectors are generally low. Although PEGylation of lipoplex reduced retention in the lung and heart, PEGylated lipoplex failed to deliver foreign gene into tumors [150]. PEGylation of adenoviral vectors generally prevents CAR recognition. Hexon-specific PEGylation of adenoviral vector improved in vitro transfection efficiency in the presence of neutralizing antibodies, in vivo blood retention and tumor accumulation after intravenous administration; however, transfection efficiency in tumor remained low [151]. To overcome this dilemma of PEGylation, that is, high retention and low uptake, cleavable PEG-lipids have been developed. PEG-lipids, which were designed to exhibit cleavage of the PEG moiety by tumor-specific matrix metalloproteinase, were incorporated into a multifunctional envelope-type nano-device [152]. As a result, transgene expression in the tumor was stimulated after intravenous injection of this carrier in comparison with that with normal PEGylated gene carrier.

It was reported that incorporation of human serum albumin to lipoplex enhanced the transfection efficiency in vitro and in vivo [153]. Moreover, utilization of serum components such as asialofetuin [154], transferrin [155] and fibronectin [156] was tested for the development of vectors.

Intravenous sequential injection of cationic liposome and plasmid DNA resulted in significant pulmonary transgene expression with reduced inflammatory cytokine production compared with those with the lipoplex [157]. Sequential injection resulted in lower DNA uptake by the liver and higher DNA levels in the lung than with the lipoplex administration [158]. Interaction with several serum proteins including albumin reduced inflammatory cytokine production by sequential complex (liposome mixed with serum proteins before mixing with plasmid DNA), whereas interaction of the lipoplex with serum proteins did not reduce inflammatory cytokine production by lipoplex [159].

Figure 4.

Schematic representation of surface charge-regulated lipoplex.

We successfully developed surface charge-regulated (SCR) lipoplex, which improved targeted gene delivery by stabilizing the lipoplex. Figure 4 shows a scheme of the salt-dependent formation of lipoplex. For in vivo preparation of the lipoplex, the concentrations of plasmid DNA and liposomes are high; consequently, a physiological concentration of salts induces aggregation of the lipoplex. This problem can be overcome using a non-ionic solution such as 5% glucose solution. Here, we hypothesized that repulsion between cationic liposomes was too strong to induce sufficient fusion of lipid membranes for stable lipoplex formation. Moderate concentration of salts in the solution of the lipoplex would reduce repulsion among cationic liposomes and enhance fusion of lipid membranes, while maintaining sufficient repulsion among lipoplex particles. This hypothesis was proved by a series of physicochemical experiments including fluorescent resonance energy transfer assessments and measurements of particle size changes in the presence of physiological concentration of salts [160]. This stable galactosylated SCR lipoplex exhibited superior hepatocyte-selective gene transfer than conventional lipoplex after intraportal injection [160]. Furthermore, the stabilization effect of SCR lipoplex was also evident in pulmonary gene transfer after intravenous injection [161].

As for the organ surface instillation method, we succeeded in enhancing the transfection efficiency of naked plasmid DNA by several strategies. Pretreatment with epidermal growth factor (EGF) enhanced transgene expression and increased transgene-positive cells on the stomach after instillation of naked plasmid DNA onto it [162]. Rubbing the gastric serosal surface with a medical spoon after instillation of naked plasmid DNA onto the stomach was more effective than EGF pretreatment [163]. However, rubbing the organ surface with a medical spoon may be impractical for future clinical application. Thus, we searched for various materials to reproduce the effect of rubbing an organ’s surface. Among them, concomitant use of calcium carbonate suspension with naked plasmid DNA was similarly effective as rubbing the gastric serosal surface [164]. Unfortunately, sedimentation of calcium carbonate suspension occurs rapidly and is problematic. To obtain slowly settling particles of calcium carbonate, we tested various conditions for calcium carbonate synthesis. We succeeded in synthesizing a novel form of calcium carbonate with a flower-like shape, named calcium carbonate microflowers [164]. Sedimentation of calcium carbonate microflowers was sufficiently slow to perform in vivo experiments. Fortunately, the suspension of calcium carbonate microflowers containing naked plasmid DNA was a more effective transfection reagent than commercially available calcium carbonate, especially at a low concentration of calcium carbonate. Intraperitoneal injection of the suspension of calcium carbonate microflowers containing naked plasmid DNA resulted in effective and peritoneal cavity-selective gene transfer. However, the mechanism of effective in vivo transfection remains to be elucidated.

Advertisement

5. Disease-dependent strategies in targeted gene delivery

Among the above-mentioned methods, intramuscular injection of naked plasmid is one of the simplest methods since it can be applied without surgery and carriers. Not only muscular diseases, such as dystrophy, but also systemic diseases may be cured using the secretory form of proteins. Intramuscular injection of plasmid DNA encoding hepatocyte growth factor rescued critical limb ischemia with high safety in a phase I/IIa clinical trial [165]. Muscular delivery of naked plasmid DNA encoding erythropoietin resulted in an increase of hematocrits [166]. In general, however, targeted gene delivery to specific organs/sites/cells is required since high biological activities of proteins may lead to side effects. For example, in suicide gene therapy to treat tumors, thymidine kinase gene expression should be restricted to tumor cells [167]. Since hepatocyte growth factor is mitogenic, liver-directed gene transfer is a rational approach to treat liver cirrhosis [168]. To treat inherited gene deficiency diseases such as familial hypercholesterolemia (LDL receptor deficiency in hepatocytes) [169] and Crigler-Najjar syndrome (uridine diphospho-glucuronosyl transferase 1A1 deficiency in hepatocytes) [170], targeted gene delivery is also reasonable due to its efficacy.

As for DNA vaccination, Kasinrerk et al. compared intramuscular, intraperitoneal, intravenous and intrasplenic immunizations with a single dose of naked plasmid DNA and observed that only the intrasplenic route induced specific antibody production [171]. In contrast, to develop DNA vaccine to induce cellular immunity, intradermal injection of naked plasmid DNA with electroporation was better than intrasplenic injection, even though there was high transfection efficiency in the spleen [172]. Gene gun bombardments of naked plasmid DNA to the skin were not effective to induce cellular immunity in comparison with intracutaneous injections of antigen-transduced dendritic cells [102]. Gene gun bombardments of naked plasmid DNA to the skin induced Th2 response and anaphylactic shock upon antigen recall [173]. On the other hand, transgene expression of fusion proteins of the immunodominant domain of human type XVII collagen and dendritic cell-specific antibody targeted to dendritic cells in the skin induced tolerance to human type XVII collagen in a skin transplantation model [174]. Intraperitoneal injection of mannosylated lipoplex resulted in efficient transgene expression in antigen-presenting cells and induced cellular immunity [175, 176]. As for the intravenous route, mannosylated lipoplex initiated a Th1 response [177]. As mentioned above, mannosylated PEGylated bubble lipoplexes with ultrasound exposure more effectively and selectively transfected antigen-presenting cells than an approach without ultrasound exposure after intravenous injection, and induced strong cellular immunity [134, 135]. Thus, the success or failure of DNA vaccination is dependent on transfection methods including transfection routes.

Advertisement

6. Conclusions

Selection of administration routes is important in targeted gene delivery for not only efficacy but also safety of the vector. Administration routes can be categorized as systemic routes and local routes. Via the systemic routes, administration is simple and does not require a complicated operation. However, very wide distribution of the vectors after their systemic administration may lead to systemic side effects. This problem can be overcome by changing the administration route from a systemic route to a local route. In addition, target selectivity can be improved by modification of the vectors with a ligand, combination with targeted application of physical forces and utilization of tissue-specific promoters. Importantly, selection of administration routes can be combined with these strategies to improve targeted gene delivery. The importance of selection of administration routes is dependent on the kind of target disease. Taking safety including germline conservativeness into consideration, further improvement of targeted gene delivery systems should be pursued.

Advertisement

Acknowledgments

This work was supported in part by a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan.

References

  1. 1. Boudreau RL, Davidson BL. Generation of hairpin-based RNAi vectors for biological and therapeutic application. Methods in Enzymology 2012;507 275-296.
  2. 2. Blaese RM, Culver KW, Miller AD, Carter CS, Fleisher T, Clerici M, Shearer G, Chang L, Chiang Y, Tolstoshev P, Greenblatt JJ, Rosenberg SA, Klein H, Berger M, Mullen CA, Ramsey WJ, Muul L, Morgan RA, Anderson WF. T lymphocyte-directed gene therapy for ADA- SCID: initial trial results after 4 years. Science 1995;270(5235) 475-480.
  3. 3. Edelstein ML, Abedi MR, Wixon J. Gene therapy clinical trials worldwide to 2007--an update. Journal of Gene Medicine 2007;9(10) 833-842.
  4. 4. Kawabata K, Takakura Y, Hashida M. The fate of plasmid DNA after intravenous injection in mice: involvement of scavenger receptors in its hepatic uptake. Pharmaceutical Research 1995;12(6) 825-830.
  5. 5. Wickham TJ, Mathias P, Cheresh DA, Nemerow GR. Integrins αv β3 and αvβ5 promote adenovirus internalization but not virus attachment. Cell 1993;73(2) 309-319.
  6. 6. Tomko RP, Xu R, Philipson L. HCAR and MCAR: the human and mouse cellular receptors for subgroup C adenoviruses and group B coxsackieviruses. Proceedings of the National Academy of Science of the United States of America 1997;94(7) 3352-3356.
  7. 7. Sakurai F, Mizuguchi H, Hayakawa T. Efficient gene transfer into human CD34+ cells by an adenovirus type 35 vector. Gene Therapy 2003;10(12) 1041-1048.
  8. 8. Kawakami S, Yamashita F, Nishikawa M, Takakura Y, Hashida M. Asialoglycoprotein receptor-mediated gene transfer using novel galactosylated cationic liposomes. Biochemical and Biophysical Research Communications 1998;252(1) 78-83.
  9. 9. Kawakami S, Sato A, Nishikawa M, Yamashita F, Hashida M. Mannose receptor-mediated gene transfer into macrophages using novel mannosylated cationic liposomes. Gene Therapy 2000;7(4) 292-299.
  10. 10. Shi N, Pardridge WM. Noninvasive gene targeting to the brain. Proceedings of the National Academy of Science of the United States of America 2000;97(13) 7567-7572.
  11. 11. Shi N, Zhang Y, Zhu C, Boado RJ, Pardridge WM. Brain-specific expression of an exogenous gene after i.v. administration. Proceedings of the National Academy of Science of the United States of America 2001;98(22) 12754-12759.
  12. 12. Toita R, Kang JH, Kim JH, Tomiyama T, Mori T, Niidome T, Jun B, Katayama Y. Protein kinase Cα-specific peptide substrate graft-type copolymer for cancer cell-specific gene regulation systems. Journal of Controlled Release 2009;139(2) 133-139.
  13. 13. Asai D, Kuramoto M, Shoji Y, Kang JH, Kodama KB, Kawamura K, Mori T, Miyoshi H, Niidome T, Nakashima H, Katayama Y. Specific transgene expression in HIV-infected cells using protease-cleavable transcription regulator. Journal of Controlled Release 2010;141(1) 52-61.
  14. 14. Jacobs F, Snoeys J, Feng Y, Van Craeyveld E, Lievens J, Armentano D, Cheng SH, De Geest B. Direct comparison of hepatocyte-specific expression cassettes following adenoviral and nonviral hydrodynamic gene transfer. Gene Therapy 2008;15(8) 594-603.
  15. 15. Dai M, Liu J, Chen DE, Rao Y, Tang ZJ, Ho WZ, Dong CY. Tumor-targeted gene therapy using Adv-AFP-HRPC/IAA prodrug system suppresses growth of hepatoma xenografted in mice. Cancer Gene Therapy 2012;19(2) 77-83.
  16. 16. Liu T, Zhang G, Chen YH, Chen Y, Liu X, Peng J, Xu MH, Yuan JW. Tissue specific expression of suicide genes delivered by nanoparticles inhibits gastric carcinoma growth. Cancer Biology and Therapy 2006;5(12) 1683-1690.
  17. 17. Shao G, Greathouse K, Huang Q, Wang CM, Sferra TJ. Gene transfer to the gastrointestinal tract after peroral administration of recombinant adeno-associated virus type 2 vectors. Journal of Pediatric Gastroenterology and Nutrition 2006;43(2) 168-179.
  18. 18. Nishi J, Fumoto S, Ishii H, Kodama Y, Nakashima M, Sasaki H, Nakamura J, Nishida K. Improved stomach selectivity of gene expression following microinstillation of plasmid DNA onto the gastric serosal surface in mice. European Journal of Pharmaceutics and Biopharmaceutics 2008;69(2) 633-639.
  19. 19. Guliyeva Ü, Öner F, Özsoy Ş, Haziroğlu R. Chitosan microparticles containing plasmid DNA as potential oral gene delivery system. European Journal of Pharmaceutics and Biopharmaceutics 2006;62(1) 17-25.
  20. 20. Zheng F, Shi XW, Yang GF, Gong LL, Yuan HY, Cui YJ, Wang Y, Du YM, Li Y. Chitosan nanoparticle as gene therapy vector via gastrointestinal mucosa administration: results of an in vitro and in vivo study. Life Sciences 2007;80(4) 388-396.
  21. 21. Bhavsar MD, Amiji MM. Gastrointestinal distribution and in vivo gene transfection studies with nanoparticles-in-microsphere oral system (NiMOS). Journal of Controlled Release 2007;119(3) 339-348.
  22. 22. Wang D, Xu J, Feng Y, Liu Y, Mchenga SS, Shan F, Sasaki J, Lu C. Liposomal oral DNA vaccine (mycobacterium DNA) elicits immune response. Vaccine 2010;28(18) 3134-3142.
  23. 23. Wang QL, Pan Q, Ma Y, Wang K, Sun P, Liu S, Zhang XL. An attenuated Salmonella-vectored vaccine elicits protective immunity against Mycobacterium tuberculosis. Vaccine 2009;27(48) 6712-6722.
  24. 24. Honigman A, Zeira E, Ohana P, Abramovitz R, Tavor E, Bar I, Zilberman Y, Rabinovsky R, Gazit D, Joseph A, Panet A, Shai E, Palmon A, Laster M, Galun E. Imaging transgene expression in live animals. Molecular Therapy 2001;4(3) 239-249.
  25. 25. Koizumi N, Mizuguchi H, Sakurai F, Yamaguchi T, Watanabe Y, Hayakawa T. Reduction of natural adenovirus tropism to mouse liver by fiber-shaft exchange in combination with both CAR- and αv integrin-binding ablation. Journal of Virology 2003;77(24) 13062-13072.
  26. 26. Denby L, Work LM, Seggern DJ, Wu E, McVey JH, Nicklin SA, Baker AH. Development of renal-targeted vectors through combined in vivo phage display and capsid engineering of adenoviral fibers from serotype 19p. Molecular Therapy 2007;15(9) 1647-1654.
  27. 27. Liu F, Qi H, Huang L, Liu D. Factors controlling the efficiency of cationic lipid-mediated transfection in vivo via intravenous administration. Gene Therapy 1997;4(6) 517-523.
  28. 28. Kawakami S, Fumoto S, Nishikawa M, Yamashita F, Hashida M. In vivo gene delivery to the liver using novel galactosylated cationic liposomes. Pharmaceutical Research 2000;17(3) 306-313.
  29. 29. Gahéry-Ségard H, Juillard V, Gaston J, Lengagne R, Pavirani A, Boulanger P, Guillet JG. Humoral immune response to the capsid components of recombinant adenoviruses: routes of immunization modulate virus-induced Ig subclass shifts. European Journal of Immunology 1997;27(3) 653-659.
  30. 30. Kostense S, Koudstaal W, Sprangers M, Weverling GJ, Penders G, Helmus N, Vogels R, Bakker M, Berkhout B, Havenga M, Goudsmit J. Adenovirus types 5 and 35 seroprevalence in AIDS risk groups supports type 35 as a vaccine vector. AIDS 2004;18(8) 1213–1216.
  31. 31. Mast TC, Kierstead L, Gupta SB, Nikas AA, Kallas EG, Novitsky V, Mbewe B, Pitisuttithum P, Schechter M, Vardas E, Wolfe ND, Aste-Amezaga M, Casimiro DR, Coplan P, Straus WL, Shiver JW. International epidemiology of human pre-existing adenovirus (Ad) type-5, type-6, type-26 and type-36 neutralizing antibodies: correlates of high Ad5 titers and implications for potential HIV vaccine trials. Vaccine 2010;28(4). pp. 950–957.
  32. 32. Nwanegbo E, Vardas E, Gao W, Whittle H, Sun H, Rowe D, Robbins PD, Gambotto A. Prevalence of neutralizing antibodies to adenoviral serotypes 5 and 35 in the adult populations of The Gambia, South Africa, and the United States. Clinical and Diagnostic Laboratory Immunology 2004;11(2) 351–357.
  33. 33. Sumida SM, Truitt DM, Lemckert AA, Vogels R, Custers JH, Addo MM, Lockman S, Peter T, Peyerl FW, Kishko MG, Jackson SS, Gorgone DA, Lifton MA, Essex M, Walker BD, Goudsmit J, Havenga MJ, Barouch DH. Neutralizing antibodies to adenovirus serotype 5 vaccine vectors are directed primarily against the adenovirus hexon protein. Journal of Immunology 2005;174(11) 7179–7185.
  34. 34. Pilankatta R, Chawla T, Khanna N, Swaminathan S. The prevalence of antibodies to adenovirus serotype 5 in an adult Indian population and implications for adenovirus vector vaccines. Journal of Medical Virology 2010;82(3) 407–414.
  35. 35. Cichon G, Boeckh-Herwig S, Schmidt HH, Wehnes E, Müller T, Pring-Akerblom P, Burger R. Complement activation by recombinant adenoviruses. Gene Therapy 2001;8(23) 1794-800.
  36. 36. Jiang H, Wang Z, Serra D, Frank MM, Amalfitano A. Recombinant adenovirus vectors activate the alternative complement pathway, leading to the binding of human complement protein C3 independent of anti-ad antibodies. Molecular Therapy 2004;10(6) 1140-1142.
  37. 37. Cotter MJ, Zaiss AK, Muruve DA. Neutrophils interact with adenovirus vectors via Fc receptors and complement receptor 1. Journal of Virology 2005;79(23) 14622-14631.
  38. 38. Sakurai F, Nakamura S, Akitomo K, Shibata H, Terao K, Kawabata K, Hayakawa T, Mizuguchi H. Transduction properties of adenovirus serotype 35 vectors after intravenous administration into nonhuman primates. Molecular Therapy 2008;16(4) 726-733.
  39. 39. Marshall E. Gene therapy death prompts review of adenovirus vector. Science 1999;286(5448) 2244-2245.
  40. 40. Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K, Akira S. A Toll-like receptor recognizes bacterial DNA. Nature 2000;408(6813) 740-745.
  41. 41. Cornélie S, Hoebeke J, Schacht AM, Bertin B, Vicogne J, Capron M, Riveau G. Direct evidence that toll-like receptor 9 (TLR9) functionally binds plasmid DNA by specific cytosine-phosphate-guanine motif recognition. Journal of Biological Chemistry 2004;279(15) 15124-15129.
  42. 42. Loisel S, Le Gall C, Doucet L, Ferec C, Floch V. Contribution of plasmid DNA to hepatotoxicity after systemic administration of lipoplexes. Human Gene Therapy 2001;12(6) 685-696.
  43. 43. Ito Y., Kawakami S, Charoensit P, Higuchi Y, Hashida M. Evaluation of proinflammatory cytokine production and liver injury induced by plasmid DNA/cationic liposome complexes with various mixing ratios in mice. European Journal of Pharmaceutics and Biopharmaceutics 2009;71(2) 303-309.
  44. 44. Tan Y, Li S, Pitt BR, Huang L. The inhibitory role of CpG immunostimulatory motifs in cationic lipid vector-mediated transgene expression in vivo. Human Gene Therapy 1999;10(13) 2153-2161.
  45. 45. Yew NS, Zhao H, Przybylska M, Wu IH, Tousignant JD, Scheule RK, Cheng SH. CpG-depleted plasmid DNA vectors with enhanced safety and long-term gene expression in vivo. Molecular Therapy 2002;5(6) 731-738.
  46. 46. Leopold PL, Wendland RL, Vincent T, Crystal RG. Neutralized adenovirus-immune complexes can mediate effective gene transfer via an Fc receptor-dependent infection pathway. Journal of Virology 2006;80(20) 10237-10247.
  47. 47. Muruve DA, Barnes MJ, Stillman IE, Libermann TA. Adenoviral gene therapy leads to rapid induction of multiple chemokines and acute neutrophil-dependent hepatic injury in vivo. Human Gene Therapy 1999;10(6) 965-976.
  48. 48. Bradshaw AC, Parker AL, Duffy MR, Coughlan L, van Rooijen N, Kähäri VM, Nicklin SA, Baker AH. Requirements for receptor engagement during infection by adenovirus complexed with blood coagulation factor X. PLoS Pathogens 2010;6(10), e1001142.
  49. 49. Zaiss AK, Lawrence R, Elashoff D, Esko JD, Herschman HR. Differential effects of murine and human factor X on adenovirus transduction via cell-surface heparan sulfate. Journal of Biological Chemistry 2011;286(28) 24535-24543.
  50. 50. Corjon S, Gonzalez G, Henning P, Grichine A, Lindholm L, Boulanger P, Fender P, Hong SS. Cell entry and trafficking of human adenovirus bound to blood factor X is determined by the fiber serotype and not hexon:heparan sulfate interaction. PLoS One 2011;6(5), e18205.
  51. 51. Yang JP, Huang L. Overcoming the inhibitory effect of serum on lipofection by increasing the charge ratio of cationic liposome to DNA. Gene Therapy 1997;4(9) 950-960.
  52. 52. Ross PC, Hui SW. Lipoplex size is a major determinant of in vitro lipofection efficiency. Gene Therapy 1999;6(4) 651-659.
  53. 53. Turek J, Dubertret C, Jaslin G, Antonakis K, Scherman D, Pitard B. Formulations which increase the size of lipoplexes prevent serum-associated inhibition of transfection. Journal of Gene Medicine 2000;2(1) 32-40.
  54. 54. Almofti MR, Harashima H, Shinohara Y, Almofti A, Li W, Kiwada H. Lipoplex size determines lipofection efficiency with or without serum. Molecular Membrane Biology 2003;20(1) 35-43.
  55. 55. Barron LG, Meyer KB, Szoka FC Jr. Effects of complement depletion on the pharmacokinetics and gene delivery mediated by cationic lipid-DNA complexes. Human Gene Therapy 1998;9(3) 315-323.
  56. 56. Tandia BM, Vandenbranden M, Wattiez R, Lakhdar Z, Ruysschaert JM, Elouahabi A. Identification of human plasma proteins that bind to cationic lipid/DNA complex and analysis of their effects on transfection efficiency: implications for intravenous gene transfer. Molecular Therapy 2003;8(2) 264-273.
  57. 57. Tandia BM, Lonez C, Vandenbranden M, Ruysschaert JM, Elouahabi A. Lipid mixing between lipoplexes and plasma lipoproteins is a major barrier for intravenous transfection mediated by cationic lipids. Journal of Biological Chemistry 2005;280(13) 12255-12261.
  58. 58. Sakurai F, Nishioka T, Saito H, Baba T, Okuda A, Matsumoto O, Taga T, Yamashita F, Takakura Y, Hashida M. 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 Therapy 2001;8(9) 677-686.
  59. 59. Sakurai F, Nishioka T, Yamashita F, Takakura Y, Hashida M. Effects of erythrocytes and serum proteins on lung accumulation of lipoplexes containing cholesterol or DOPE as a helper lipid in the single-pass rat lung perfusion system. European Journal of Pharmaceutics and Biopharmaceutics 2001;52(2) 165-172.
  60. 60. Eliyahu H, Servel N, Domb AJ, Barenholz Y. Lipoplex-induced hemagglutination: potential involvement in intravenous gene delivery. Gene Therapy 2002;9(13) 850-858.
  61. 61. Kurosaki T, Kitahara T, Fumoto S, Nishida K, Nakamura J, Niidome T, Kodama Y, Nakagawa H, To H, Sasaki H. Ternary complexes of pDNA, polyethylenimine, and γ-polyglutamic acid for gene delivery systems. Biomaterials 2009;30(14) 2846-2853.
  62. 62. Kurosaki T, Kitahara T, Kawakami S, Higuchi Y, Yamaguchi A, Nakagawa H, Kodama Y, Hamamoto T, Hashida M, Sasaki H. γ-polyglutamic acid-coated vectors for effective and safe gene therapy. Journal of Controlled Release 2010;142(3) 404-410
  63. 63. Kurosaki T, Kitahara T, Kawakami S, Nishida K, Nakamura J, Teshima M, Nakagawa H, Kodama Y, To H, Sasaki H. The development of a gene vector electrostatically assembled with a polysaccharide capsule. Biomaterials 2009;30(26) 4427-4434.
  64. 64. Kurosaki T, Kitahara T, Fumoto S, Nishida K, Yamamoto K, Nakagawa H, Kodama Y, Higuchi N, Nakamura T, Sasaki H. Chondroitin sulfate capsule system for efficient and secure gene delivery. Journal of Pharmacy & Pharmaceutical Sciences 2010;13(3) 351-361.
  65. 65. Templeton NS, Lasic DD, Frederik PM, Strey HH, Roberts DD, Pavlakis GN. Improved DNA: liposome complexes for increased systemic delivery and gene expression. Nature Biotechnology 1997;15(7) 647-652.
  66. 66. Liu Y, Mounkes LC, Liggitt HD, Brown CS, Solodin I, Heath TD, Debs RJ. Factors influencing the efficiency of cationic liposome-mediated intravenous gene delivery. Nature Biotechnology 1997;15(2) 167-173.
  67. 67. Hattori Y, Suzuki S, Kawakami S, Yamashita F, Hashida M. The role of dioleoylphosphatidylethanolamine (DOPE) in targeted gene delivery with mannosylated cationic liposomes via intravenous route. Journal of Controlled Release 2005;108(2-3) 484-495.
  68. 68. Kurosaki T, Kitahara T, Teshima M, Nishida K, Nakamura J, Nakashima M, To H, Hukuchi H, Hamamoto T, Sasaki H. Exploitation of De Novo helper-lipids for effective gene delivery. Journal of Pharmacy & Pharmaceutical Sciences 2008;11(4) 56-67.
  69. 69. Kanemura H, Iimuro Y, Takeuchi M, Ueki T, Hirano T, Horiguchi K, Asano Y, Fujimoto J. Hepatocyte growth factor gene transfer with naked plasmid DNA ameliorates dimethylnitrosamine-induced liver fibrosis in rats. Hepatology Research 2008;38(9) 930-939.
  70. 70. Tang MM, Zhu QE, Fan WZ, Zhang SL, Li DZ, Liu LZ, Chen M, Zhang M, Zhou J, Wei CJ. Intra-arterial targeted islet-specific expression of Sirt1 protects β cells from streptozotocin-induced apoptosis in mice. Molecular Therapy 2011;19(1) 60-66.
  71. 71. Zhang G, Ludtke JJ, Thioudellet C, Kleinpeter P, Antoniou M, Herweijer H, Braun S, Wolff JA. Intraarterial delivery of naked plasmid DNA expressing full-length mouse dystrophin in the mdx mouse model of duchenne muscular dystrophy. Human Gene Therapy 2004;15(8) 770-782.
  72. 72. Porvasnik SL, Mah C, Polyak S. Targeting murine small bowel and colon through selective superior mesenteric artery injection. Microsurgery 2010;30(6) 487-493
  73. 73. Rainov NG, Ikeda K, Qureshi NH, Grover S, Herrlinger U, Pechan P, Chiocca EA, Breakefield XO, Barnett FH. Intraarterial delivery of adenovirus vectors and liposome-DNA complexes to experimental brain neoplasms. Human Gene Therapy 1999;10(2) 311-318.
  74. 74. Maruyama H, Higuchi N, Nishikawa Y, Hirahara H, Iino N, Kameda S, Kawachi H, Yaoita E, Gejyo F, Miyazaki J. Kidney-targeted naked DNA transfer by retrograde renal vein injection in rats. Human Gene Therapy 2002;13(3) 455-468.
  75. 75. Morimoto K, Nishikawa M, Kawakami S, Nakano T, Hattori Y, Fumoto S, Yamashita F, Hashida M. Molecular weight-dependent gene transfection activity of unmodified and galactosylated polyethyleneimine on hepatoma cells and mouse liver. Molecular Therapy 2003;7(2) 254-261.
  76. 76. Fumoto S, Nakadori F, Kawakami S, Nishikawa M, Yamashita F, Hashida M. Analysis of hepatic disposition of galactosylated cationic liposome/plasmid DNA complexes in perfused rat liver. Pharmaceutical Research 2003;20(9) 1452-1459.
  77. 77. Fumoto S, Kawakami S, Ishizuka M, Nishikawa M, Yamashita F, Hashida M. Analysis of hepatic disposition of native and galactosylated polyethylenimine complexed with plasmid DNA in perfused rat liver. Drug Metabolism and Pharmacokinetics 2003;18(4) 230-237.
  78. 78. Higuchi Y, Kawakami S, Fumoto S, Yamashita F, Hashida M. Effect of the particle size of galactosylated lipoplex on hepatocyte-selective gene transfection after intraportal administration. Biological & Pharmaceutical Bulletin 2006;29(7) 1521-1523.
  79. 79. Li S, Rizzo MA, Bhattacharya S, Huang L. Characterization of cationic lipid-protamine-DNA (LPD) complexes for intravenous gene delivery. Gene Therapy 1998;5(7) 930-937.
  80. 80. Goula D, Remy JS, Erbacher P, Wasowicz M, Levi G, Abdallah B, Demeneix BA. Size, diffusibility and transfection performance of linear PEI/DNA complexes in the mouse central nervous system. Gene Therapy 1998;5(5) 712-717.
  81. 81. Sato A, Takagi M, Shimamoto A, Kawakami S, Hashida M. Small interfering RNA delivery to the liver by intravenous administration of galactosylated cationic liposomes in mice. Biomaterials 2007;28(7) 1434-1442.
  82. 82. Semple SC, Akinc A, Chen J, Sandhu AP, Mui BL, Cho CK, Sah DW, Stebbing D, Crosley EJ, Yaworski E, Hafez IM, Dorkin JR, Qin J, Lam K, Rajeev KG, Wong KF, Jeffs LB, Nechev L, Eisenhardt ML, Jayaraman M, Kazem M, Maier MA, Srinivasulu M, Weinstein MJ, Chen Q, Alvarez R, Barros SA, De S, Klimuk SK, Borland T, Kosovrasti V, Cantley WL, Tam YK, Manoharan M, Ciufolini MA, Tracy MA, de Fougerolles A, MacLachlan I, Cullis PR, Madden TD, Hope MJ. Rational design of cationic lipids for siRNA delivery. Nature Biotechnology 2010;28(2) 172-176.
  83. 83. Fumoto S, Kawakami S, Shigeta K, Higuchi Y, Yamashita F, Hashida M. Interaction with blood components plays a crucial role in asialoglycoprotein receptor-mediated in vivo gene transfer by galactosylated lipoplex. Journal of Pharmacology and Experimental Therapeutics 2005;315(2) 484-493.
  84. 84. Yoshikawa N, Sakamoto K, Mizuno S, Sakaguchi J, Miyamoto H, Mine T, Sasaki H, Fumoto S, Nishida K. Multiple components in serum contribute to hepatic transgene expression by lipoplex in mice. Journal of Gene Medicine 2011;13(11) 632-643.
  85. 85. Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, Felgner PL. Direct gene transfer into mouse muscle in vivo. Science 1990;247(4949 Pt. 1) 1465-1468.
  86. 86. Li K, Welikson RE, Vikstrom KL, Leinwand LA. Direct gene transfer into the mouse heart. Journal of Molecular and Cellular Cardiology 1997;29(5) 1499-1504.
  87. 87. Su H, Lu R, Kan YW. Adeno-associated viral vector-mediated vascular endothelial growth factor gene transfer induces neovascular formation in ischemic heart. Proceedings of the National Academy of Science of the United States of America 2000;97(25) 13801-13806.
  88. 88. Hickman MA, Malone RW, Lehmann-Bruinsma K, Sih TR, Knoell D, Szoka FC, Walzem R, Carlson DM, Powell JS. Gene expression following direct injection of DNA into liver. Human Gene Therapy 1994;5(12) 1477-1483.
  89. 89. Gusella GL, Fedorova E, Hanss B, Marras D, Klotman ME, Klotman PE. Lentiviral gene transduction of kidney. Human Gene Therapy 2002;13(3) 407-414.
  90. 90. White SA, LoBuglio AF, Arani RB, Pike MJ, Moore SE, Barlow DL, Conry RM. Induction of anti-tumor immunity by intrasplenic administration of a carcinoembryonic antigen DNA vaccine. Journal of Gene Medicine 2000;2(2) 135-140.
  91. 91. Takehara T, Hayashi N, Yamamoto M, Miyamoto Y, Fusamoto H, Kamada T. In vivo gene transfer and expression in rat stomach by submucosal injection of plasmid DNA. Human Gene Therapy 1996;7(5) 589-593.
  92. 92. DeMatteo RP, Raper SE, Ahn M, Fisher KJ, Burke C, Radu A, Widera G, Claytor BR, Barker CF, Markmann JF. Gene transfer to the thymus. A means of abrogating the immune response to recombinant adenovirus. Annals of Surgery 1995;222(3) 229-239.
  93. 93. Yang JP, Huang L. Direct gene transfer to mouse melanoma by intratumor injection of free DNA. Gene Therapy 1996;3(6) 542-548.
  94. 94. Nomura T, Nakajima S, Kawabata K, Yamashita F, Takakura Y, Hashida M. Intratumoral pharmacokinetics and in vivo gene expression of naked plasmid DNA and its cationic liposome complexes after direct gene transfer. Cancer Research 1997;57(13) 2681-2686.
  95. 95. Nomura T, Yasuda K, Yamada T, Okamoto S, Mahato RI, Watanabe Y, Takakura Y, Hashida M. Gene expression and antitumor effects following direct interferon (IFN)-gamma gene transfer with naked plasmid DNA and DC-chol liposome complexes in mice. Gene Therapy 1999;6(1) 121-129.
  96. 96. Mathiesen I. Electropermeabilization of skeletal muscle enhances gene transfer in vivo. Gene Therapy 1999;6(4) 508-514.
  97. 97. Dai H, Jiang X, Leong KW, Mao HQ. Transient depletion of kupffer cells leads to enhanced transgene expression in rat liver following retrograde intrabiliary infusion of plasmid DNA and DNA nanoparticles. Human Gene Therapy 2011;22(7) 873-878.
  98. 98. Gau P, Rodriguez S, De Leonardis C, Chen P, Lin DM. Air-assisted intranasal instillation enhances adenoviral delivery to the olfactory epithelium and respiratory tract. Gene Therapy 2011;18(5) 432-436.
  99. 99. Eliyahu H, Joseph A, Schillemans JP, Azzam T, Domb AJ, Barenholz Y. Characterization and in vivo performance of dextran-spermine polyplexes and DOTAP/cholesterol lipoplexes administered locally and systemically. Biomaterials 2007;28(14) 2339-2349.
  100. 100. Mohri K, Okuda T, Mori A, Danjo K, Okamoto H. Optimized pulmonary gene transfection in mice by spray-freeze dried powder inhalation. Journal of Controlled Release 2010;144(2) 221-226.
  101. 101. Deroose CM, Reumers V, Gijsbers R, Bormans G, Debyser Z, Mortelmans L, Baekelandt V. Noninvasive monitoring of long-term lentiviral vector-mediated gene expression in rodent brain with bioluminescence imaging. Molecular Therapy 2006;14(3) 423-431.
  102. 102. Gaffal E, Schweichel D, Tormo D, Steitz J, Lenz J, Basner-Tschakarjan E, Limmer A, Tüting T. Comparative evaluation of CD8+CTL responses following gene gun immunization targeting the skin with intracutaneous injection of antigen-transduced dendritic cells. European Journal of Cell Biology 2007;86(11-12) 817-26.
  103. 103. Akiyama M, Thorne S, Kirn D, Roelvink PW, Einfeld DA, King CR, Wickham TJ. Ablating CAR and integrin binding in adenovirus vectors reduces nontarget organ transduction and permits sustained bloodstream persistence following intraperitoneal administration. Molecular Therapy 2004;9(2) 218-230.
  104. 104. Kawakami S, Hirayama R, Shoji K, Kawanami R, Nishida K, Nakashima M, Sasaki H, Sakaeda T, Nakamura J. Liver- and lobe-selective gene transfection following the instillation of plasmid DNA to the liver surface in mice. Biochemical and Biophysical Research Communications 2002;294(1) 46-50.
  105. 105. Hirayama R, Kawakami S, Nishida K, Nakashima M, Sasaki H, Sakeda T, Nakamura J. Development of the liver- and lobe-selective nonviral gene transfer following the instillation of naked plasmid DNA using catheter on the liver surface in mice. Pharmaceutical Research 2003;20(2) 328-332.
  106. 106. Fumoto S, Furukawa H, Nakamura J, Nishida K. Safety of liver surface instillation of plasmid DNA in normal and carbon tetrachloride-induced hepatitis mice. Journal of Pharmacy & Pharmaceutical Sciences 2011;14(2) 274-282.
  107. 107. Nishida K, Sato N, Sasaki H, Nakamura J. Absorption of organic anions as model drugs following application to rat liver surface in-vivo. Journal of Pharmacy and Pharmacology 1994;46(11) 867-870.
  108. 108. Hirayama R, Nishida K, Fumoto S, Nakashima M, Sasaki H, Nakamura J. Liver site-specific gene transfer following the administration of naked plasmid DNA to the liver surface in mice. Biological & Pharmaceutical Bulletin 2004;27(10) 1697-1699.
  109. 109. Hirayama R, Fumoto S, Nishida K, Nakashima M, Sasaki H, Nakamura J. Effect of solution composition of plasmid DNA on gene transfection following liver surface administration in mice. Biological & Pharmaceutical Bulletin 2005;28(11) 2166-2169.
  110. 110. Zuhorn IS, Kalicharan R, Hoekstra D. Lipoplex-mediated transfection of mammalian cells occurs through the cholesterol-dependent clathrin-mediated pathway of endocytosis. Journal of Biological Chemistry 2002;277(20) 18021-18028.
  111. 111. Rejman J, Bragonzi A, Conese M. Role of clathrin- and caveolae-mediated endocytosis in gene transfer mediated by lipo- and polyplexes. Molecular Therapy 2005;12(3) 468-474.
  112. 112. Rejman J, Conese M, Hoekstra D. Gene transfer by means of lipo- and polyplexes: role of clathrin and caveolae-mediated endocytosis. Journal of Liposome Research 2006;16(3) 237-247.
  113. 113. von Gersdorff K, Sanders NN, Vandenbroucke R, De Smedt SC, Wagner E, Ogris M. The internalization route resulting in successful gene expression depends on both cell line and polyethylenimine polyplex type. Molecular Therapy 2006;14(5) 745-753.
  114. 114. Fumoto S, Nishi J, Ishii H, Wang X, Miyamoto H, Yoshikawa N, Nakashima M, Nakamura J, Nishida K. Rac-mediated macropinocytosis is a critical route for naked plasmid DNA transfer in mice. Molecular Pharmaceutics 2009;6(4) 1170-1179.
  115. 115. Hirayama R, Nishida K, Fumoto S, Nakashima M, Sasaki H, Nakamura J. Unilateral kidney-selective gene transfer following the administration of naked plasmid DNA to the kidney surface in mice. Biological & Pharmaceutical Bulletin 2005;28(1) 181-184.
  116. 116. Nakamura J, Fumoto S, Ariyoshi K, Kodama Y, Nishi J, Nakashima M, Sasaki H, Nishida K. Unilateral lung-selective gene transfer following the administration of naked plasmid DNA onto the pulmonary pleural surface in mice. Biological & Pharmaceutical Bulletin 2007;30(4) 729-732.
  117. 117. Nakamura J, Fumoto S, Kawanami R, Kodama Y, Nishi J, Nakashima M, Sasaki H, Nishida K. Spleen-selective gene transfer following the administration of naked plasmid DNA onto the spleen surface in mice. Biological & Pharmaceutical Bulletin 2007;30(5) 941-945.
  118. 118. Nakamur, J, Fumoto S, Shoji K, Kodama Y, Nishi J, Nakashima M, Sasaki H, Nishida K. Stomach-selective gene transfer following the administration of naked plasmid DNA onto the gastric serosal surface in mice. Biological & Pharmaceutical Bulletin 2006;29(10) 2082-2086.
  119. 119. Fumoto S, Nishi J, Nakamura J, Nishida K. Gene therapy for gastric diseases. Current Gene Therapy 2008;8(3) 187-200.
  120. 120. Nishi J, Fumoto S, Ishii H, Kodama Y, Nakashima M, Sasaki H, Nakamura J, Nishida K. Highly stomach-selective gene transfer following gastric serosal surface instillation of naked plasmid DNA in rats. Journal of Gastroenterology 2008;43(12) 912-919.
  121. 121. Fumoto S, Tsuchimochi M, Nishi J, Ishii H, Kodama Y, Nakashima M, Sasaki H, Nakamura J, Nishida K. Liver- and lobe-specific gene transfer following the continuous microinstillation of Plasmid DNA onto the liver surface in mice: effect of instillation speed. Biological & Pharmaceutical Bulletin 2009;32(7) 1298-302.
  122. 122. Berraondo P, Crettaz J, Ochoa L, Pañeda A, Prieto J, Trocóniz IF, González-Aseguinolaza G. Intrahepatic injection of recombinant adeno-associated virus serotype 2 overcomes gender-related differences in liver transduction. Human Gene Therapy 2006;17(6) 601-610.
  123. 123. Mohr L, Yoon SK, Eastman SJ, Chu Q, Scheule RK, Scaglioni PP, Geissler M, Heintges T, Blum HE, Wands JR. Cationic liposome-mediated gene delivery to the liver and to hepatocellular carcinomas in mice. Human Gene Therapy 2001;12(7) 799-809.
  124. 124. Crettaz J, Berraondo P, Mauleón I, Ochoa-Callejero L, Shankar V, Barajas M, van Rooijen N, Kochanek S, Qian C, Prieto J, Hernández-Alcoceba R, González-Aseguinolaza G. Intrahepatic injection of adenovirus reduces inflammation and increases gene transfer and therapeutic effect in mice. Hepatology 2006;44(3) 623-632.
  125. 125. Liu F, Huang L. Electric gene transfer to the liver following systemic administration of plasmid DNA. Gene Therapy 2002;9(16) 1116-1169.
  126. 126. Sakai M, Nishikawa M, Thanaketpaisarn O, Yamashita F, Hashida M.. Hepatocyte-targeted gene transfer by combination of vascularly delivered plasmid DNA and in vivo electroporation. Gene Therapy 2005;12(7) 607-616.
  127. 127. Thanaketpaisarn O, Nishikawa M, Yamashita F, Hashida M. Tissue-specific characteristics of in vivo electric gene: transfer by tissue and intravenous injection of plasmid DNA. Pharmaceutical Research 2005;22(6) 883-891.
  128. 128. Taniyama Y, Tachibana K, Hiraoka K, Aoki M, Yamamoto S, Matsumoto K, Nakamura T, Ogihara T, Kaneda Y, Morishita R. Development of safe and efficient novel nonviral gene transfer using ultrasound: enhancement of transfection efficiency of naked plasmid DNA in skeletal muscle. Gene Therapy 2002;9(6) 372-380.
  129. 129. Lu QL, Liang HD, Partridge T, Blomley MJ. Microbubble ultrasound improves the efficiency of gene transduction in skeletal muscle in vivo with reduced tissue damage. Gene Therapy 2003;10(5) 396-405.
  130. 130. Shen ZP, Brayman AA, Chen L, Miao CH. Ultrasound with microbubbles enhances gene expression of plasmid DNA in the liver via intraportal delivery. Gene Therapy 2008;15(16) 1147-1155.
  131. 131. Xenariou S, Griesenbach U, Liang HD, Zhu J, Farley R, Somerton L, Singh C, Jeffery PK, Ferrari S, Scheule RK, Cheng SH, Geddes DM, Blomley M, Alton EW. Use of ultrasound to enhance nonviral lung gene transfer in vivo. Gene Therapy 2007;14(9) 768-774.
  132. 132. Suzuki R, Takizawa T, Negishi Y, Hagisawa K, Tanaka K, Sawamura K, Utoguchi N, Nishioka T, Maruyama K. Gene delivery by combination of novel liposomal bubbles with perfluoropropane and ultrasound. Journal of Controlled Release 2007;117(1) 130-136.
  133. 133. Un K, Kawakami S, Suzuki R, Maruyama K, Yamashita F, Hashida M. Enhanced transfection efficiency into macrophages and dendritic cells by a combination method using mannosylated lipoplexes and bubble liposomes with ultrasound exposure. Human Gene Therapy 2010;21(1) 65-74.
  134. 134. Un K, Kawakami S, Suzuki R, Maruyama K, Yamashita F, Hashida M. Development of an ultrasound-responsive and mannose-modified gene carrier for DNA vaccine therapy. Biomaterials 2010;31(30) 7813-7826.
  135. 135. Un K, Kawakami S, Suzuki R, Maruyama K, Yamashita F, Hashida M. Suppression of melanoma growth and metastasis by DNA vaccination using an ultrasound-responsive and mannose-modified gene carrier. Molecular Pharmaceutics 2011;8(2) 543-554.
  136. 136. Un K, Kawakami S, Yoshida M, Higuchi Y, Suzuki R, Maruyama K, Yamashita F, Hashida M. The elucidation of gene transferring mechanism by ultrasound-responsive unmodified and mannose-modified lipoplexes. Biomaterials 2011;32(20) 4659-4669.
  137. 137. Un K, Kawakami S, Higuchi Y, Suzuki R, Maruyama K, Yamashita F, Hashida M. Involvement of activated transcriptional process in efficient gene transfection using unmodified and mannose-modified bubble lipoplexes with ultrasound exposure. Journal of Controlled Release 2011;156(3) 355-363.
  138. 138. Liu F, Song Y, Liu D. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA. Gene Therapy 1999;6(7) 1258-1266.
  139. 139. Zhang G, Budker V, Wolff JA. High levels of foreign gene expression in hepatocytes after tail vein injections of naked plasmid DNA. Human Gene Therapy 1999;10(10) 1735-1737.
  140. 140. Fabre JW, Grehan A, Whitehorne M, Sawyer GJ, Dong X, Salehi S, Eckley L, Zhang X, Seddon M, Shah AM, Davenport M, Rela M. Hydrodynamic gene delivery to the pig liver via an isolated segment of the inferior vena cava. Gene Therapy 2008;15(6) 452-462.
  141. 141. Zhang G, Gao X, Song YK, Vollmer R, Stolz DB, Gasiorowski JZ, Dean DA, Liu D. Hydroporation as the mechanism of hydrodynamic delivery. Gene Therapy 2004;11(8) 675-682.
  142. 142. Kobayashi N, Nishikawa M, Takakura Y. The hydrodynamics-based procedure for controlling the pharmacokinetics of gene medicines at whole body, organ and cellular levels. Advanced Drug Delivery Reviews 2005;57(5) 713-731.
  143. 143. Nishikawa M, Nakayama A, Takahashi Y, Fukuhara Y, Takakura Y. Reactivation of silenced transgene expression in mouse liver by rapid, large-volume injection of isotonic solution. Human Gene Therapy 2008;19(10) 1009-1020.
  144. 144. Takiguchi N, Takahashi Y, Nishikawa M, Matsui Y, Fukuhara Y, Oushiki D, Kiyose K, Hanaoka K, Nagano T, Takakura Y. Positive correlation between the generation of reactive oxygen species and activation/reactivation of transgene expression after hydrodynamic injections into mice. Pharmaceutical Research 2011;28(4) 702-711.
  145. 145. Liu F, Huang L. Noninvasive gene delivery to the liver by mechanical massage. Hepatology 2002;35(6) 1314-1319.
  146. 146. Mukai H, Kawakami S, Hashida M. Renal press-mediated transfection method for plasmid DNA and siRNA to the kidney. Biochemical and Biophysical Research Communications 2008;372(3) 383-387.
  147. 147. Mukai H, Kawakami S, Kamiya Y, Ma F, Takahashi H, Satake K, Terao K, Kotera H, Yamashita F, Hashida M. Pressure-mediated transfection of murine spleen and liver. Human Gene Therapy 2009;20(10) 1157-1167.
  148. 148. Mukai H, Kawakami S, Takahashi H, Satake K, Yamashita F, Hashida M. Key physiological phenomena governing transgene expression based on tissue pressure-mediated transfection in mice. Biological & Pharmaceutical Bulletin 2010;33(9) 1627-1632.
  149. 149. Fang J, Nakamura H, Maeda H. The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Advanced Drug Delivery Reviews 2011;63(3) 136-151.
  150. 150. Gjetting T, Arildsen NS, Christensen CL, Poulsen TT, Roth JA, Handlos VN, Poulsen HS. In vitro and in vivo effects of polyethylene glycol (PEG)-modified lipid in DOTAP/cholesterol-mediated gene transfection. International Journal of Nanomedicine 2010;5 371-383.
  151. 151. Suzuki-Kouyama E, Katayama K, Sakurai F, Yamaguchi T, Kurachi S, Kawabata K, Nakagawa S, Mizuguchi H. Hexon-specific PEGylated adenovirus vectors utilizing avidin-biotin interaction. Biomaterials 2011;32(6) 1724-1730.
  152. 152. Hatakeyama H, Akita H, Kogure K, Oishi M, Nagasaki Y, Kihira Y, Ueno M, Kobayashi H, Kikuchi H, Harashima H. Development of a novel systemic gene delivery system for cancer therapy with a tumor-specific cleavable PEG-lipid. Gene Therapy 2007;14(1), 68-77.
  153. 153. Simões S, Slepushkin V, Pires P, Gaspar R, Pedroso de Lima MC, Düzgüneş N. Human serum albumin enhances DNA transfection by lipoplexes and confers resistance to inhibition by serum. Biochimica et Biophysica Acta -Biomembranes 2000;1463(2) 459-469.
  154. 154. Arangoa MA, Düzgüneş N, Tros de Ilarduya C. Increased receptor-mediated gene delivery to the liver by protamine-enhanced-asialofetuin-lipoplexes. Gene Therapy 2003;10(1) 5-14.
  155. 155. Tros de Ilarduya C, Arangoa MA, Moreno-Aliaga MJ, Düzgüneş N. Enhanced gene delivery in vitro and in vivo by improved transferrin-lipoplexes. Biochimica et Biophysica Acta -Biomembranes 2002;1561(2) 209-221.
  156. 156. Hattori Y, Maitani Y. DNA/Lipid complex incorporated with fibronectin to cell adhesion enhances transfection efficiency in prostate cancer cells and xenografts. Biological, Pharmaceutical Bulletin 2007;30(3) 603-607.
  157. 157. Tan Y, Liu F, Li Z, Li S, Huang L. Sequential injection of cationic liposome and plasmid DNA effectively transfects the lung with minimal inflammatory toxicity. Molecular Therapy 2001;3(5, Pt. 1) 673-682.
  158. 158. Zhang JS, Liu F, Conwell CC, Tan Y, Huang L. Mechanistic studies of sequential injection of cationic liposome and plasmid DNA. Molecular Therapy 2006;13(2) 429-437.
  159. 159. Conwell CC, Liu F, Huang L. Several serum proteins significantly decrease inflammatory response to lipid-based non-viral vectors. Molecular Therapy 2008;16(2) 370-377.
  160. 160. Fumoto S, Kawakami S, Ito Y, Shigeta K, Yamashita F, Hashida M. Enhanced hepatocyte-selective in vivo gene expression by stabilized galactosylated liposome/plasmid DNA complex using sodium chloride for complex formation. Molecular Therapy 2004;10(4) 719-729.
  161. 161. Kawakami S, Ito Y, Fumoto S, Yamashita F, Hashida M. Enhanced gene expression in lung by a stabilized lipoplex using sodium chloride for complex formation. Journal of Gene Medicine 2005;7(12) 1526-1533.
  162. 162. Miyamoto H, Baba S, Nakajima S, Mine T, Yoshikawa N, Fumoto S, Nishida K. Pretreatment with Epidermal Growth Factor Enhances Naked Plasmid DNA Transfer onto Gastric Serosal Surface in Mice. Biological and Pharmaceutical Bulletin 2012;35(6) 903-908.
  163. 163. Mine T, Ishii H, Nakajima S, Yoshikawa N, Miyamoto H, Nakashima M, Nakamura J, Fumoto S, Nishida K. Rubbing gastric serosal surface enhances naked plasmid DNA transfer in rats and mice. Biological & Pharmaceutical Bulletin 2011;34(9) 1514-1517.
  164. 164. Fumoto S, Nakajima S, Mine T, Yoshikawa N, Kitahara T, Sasaki H, Miyamoto H, Nishida K. Efficient in Vivo Gene Transfer by Intraperitoneal Injection of Plasmid DNA and Calcium Carbonate Microflowers in Mice. Molecular Pharmaceutics 2012;9(7) 1962-1970.
  165. 165. Morishita R, Makino H, Aoki M, Hashiya N, Yamasaki K, Azuma J, Taniyama Y, Sawa Y, KanedaY, Ogihara T. Phase I/IIa clinical trial of therapeutic angiogenesis using hepatocyte growth factor gene transfer to treat critical limb ischemia. Arteriosclerosis, Thrombosis, and Vascular Biology 2011;31(3) 713-720.
  166. 166. Tripathy SK, Svensson EC, Black HB, Goldwasser E, Margalith M, Hobart PM, Leiden JM. Long-term expression of erythropoietin in the systemic circulation of mice after intramuscular injection of a plasmid DNA vector. Proceedings of the National Academy of Science of the United States of America 1996;93(20) 10876-10880.
  167. 167. Bonini C, Bondanza A, Perna SK, Kaneko S, Traversari C, Ciceri F, Bordignon C. The suicide gene therapy challenge: how to improve a successful gene therapy approach. Molecular Therapy 2007;15(7) 1248-1252.
  168. 168. Matsuno Y, Iwata H, Umeda Y, Takagi H, Mori Y, Kosugi A, Matsumoto K, Nakamura, T., Hirose, H. Hepatocyte growth factor gene transfer into the liver via the portal vein using electroporation attenuates rat liver cirrhosis. Gene Therapy 2003;10(18) 1559-1566.
  169. 169. Wilson JM, Grossman M, Wu CH, Chowdhury NR, Wu GY, Chowdhury JR. Hepatocyte-directed gene transfer in vivo leads to transient improvement of hypercholesterolemia in low density lipoprotein receptor-deficient rabbits. Journal of Biological Chemistry 1992;267(2) 963-967.
  170. 170. Miranda PS, Bosma PJ. Towards liver-directed gene therapy for Crigler-Najjar syndrome. Current Gene Therapy 2009;9(2) 72-82.
  171. 171. Kasinrerk W, Moonsom S, Chawansuntati K. Production of antibodies by single DNA immunization: comparison of various immunization routes. Hybridoma and Hybridomics 2002;21(4) 287-293.
  172. 172. Guan X, Nishikawa M, Takemoto S, Ohno Y, Yata T, Takakura Y. Injection site-dependent induction of immune response by DNA vaccine: comparison of skin and spleen as a target for vaccination. Journal of Gene Medicine 2010;12(3) 301-309.
  173. 173. Alvarez D, Harder G, Fattouh R, Sun J, Goncharova S, Stämpfli MR, Coyle AJ, Bramson JL, Jordana M. Cutaneous antigen priming via gene gun leads to skin-selective Th2 immune-inflammatory responses. Journal of Immunology 2005;174(3) 1664-1674.
  174. 174. Ettinger M, Gratz IK, Gruber C, Hauser-Kronberger C, Johnson TS, Mahnke K, Thalhamer J, Hintner H, Peckl-Schmid D, Bauer JW. Targeting of the hNC16A collagen domain to dendritic cells induces tolerance to human type XVII collagen. Experimental Dermatology 2012;21(5) 395-398.
  175. 175. Hattori Y, Kawakami S, Lu Y, Nakamura K, Yamashita F, Hashida M. Enhanced DNA vaccine potency by mannosylated lipoplex after intraperitoneal administration. Journal of Gene Medicine 2006;8(7) 824-834.
  176. 176. Hattori Y, Kawakami S, Nakamura K, Yamashita F, Hashida M. Efficient gene transfer into macrophages and dendritic cells by in vivo gene delivery with mannosylated lipoplex via the intraperitoneal route. Journal of Pharmacology and Experimental Therapeutics 2006;318(2) 828-834.
  177. 177. Hattori Y, Kawakami S, Suzuki S, Yamashita F, Hashida M. Enhancement of immune responses by DNA vaccination through targeted gene delivery using mannosylated cationic liposome formulations following intravenous administration in mice. Biochemical and Biophysical Research Communications 2004;317(4) 992-999.

Written By

Shintaro Fumoto, Shigeru Kawakami, Mitsuru Hashida and Koyo Nishida

Submitted: 01 May 2012 Published: 13 February 2013