Open Access

Effect of integrin receptor‑targeted liposomal paclitaxel for hepatocellular carcinoma targeting and therapy

Retraction in: /10.3892/ol.2021.12611

  • Authors:
    • Liyu Chen
    • Yanbin Liu
    • Weiya Wang
    • Kai Liu
  • View Affiliations

  • Published online on: May 20, 2015     https://doi.org/10.3892/ol.2015.3242
  • Pages: 77-84
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The major aim of the present study was to develop an integrin receptor‑targeted liposomal paclitaxel (PTX) to enhance the targeting specificity and therapeutic effect of PTX on hepatocellular carcinoma (HCC) cells. The specific Arg‑Gly‑Asp (RGD) ligand was conjugated to 1,2‑distearoylphosphatidylethanolamine‑polyethylene glycol 2000 to prepare the RGD‑modified liposomes (RGD‑LP). Furthermore, physicochemical characteristics of RGD‑LP, including particle size, ζ potential, encapsulation efficiency and in vitro PTX release, were evaluated. RGD‑modified liposomes were selected as the carrier for the present study, as they exhibit good biocompatibility and are easy to modify using RGD. The cellular uptake efficacy of RGD‑LP by HepG2 cells was 3.3‑fold higher than that of liposomes without RGD, indicating that RGD‑LP may specifically target HepG2 cells by overexpressing integrin αvβ3 receptors. The RGD modification appeared to enhance the anti‑proliferative activity of LP‑PTX against HepG2 cells, with the extent of anti‑proliferative activity dependent on the concentration of PTX and the incubation time. Additionally, evaluation of the homing specificity and anticancer efficacy of RGD‑LP on the tumor spheroids indicated that solid tumor penetration was enhanced by the modification of RGD. In agreement with these in vitro findings, in vivo investigations demonstrated that RGD‑LP‑PTX exhibited a greater inhibitory effect on tumor growth in HepG2‑bearing mice than LP‑PTX or free PTX. Thus, RGD‑LPs may represent an efficient targeted PTX delivery system for the treatment of patients with HCC.

Introduction

Hepatocellular carcinoma (HCC) is the most malignant type of cancer and the leading cause of cancer-associated mortality, worldwide (1). According to 2013 statistics, >80% of HCC cases occur in developing countries, 55% of which occur in China (2). No effective therapeutic treatment is currently available, particularly when HCC is diagnosed at an advanced stage (3). Surgical excision is always the primary choice in the clinical treatment of HCC; however, total resection of the tumor is typically not possible, therefore, surgical treatment is supplemented with radiotherapy and chemotherapy to kill the remaining tumor cells (4). The single-agent activity of paclitaxel (PTX), a widely used anticancer agent, has gained increasing attention in recent years (57). As well as promoting the assembly and stabilization of microtubules, PTX appears to interfere with essential cellular functions, for example mitosis, cell transport and cell motility (810). Due to its poor water solubility, single-agent PTX is currently administered as Taxol [PTX dissolved in Cremophor EL® (polyethoxylated castor oil) and ethanol (1:1, v/v)]. However, various reports have indicated that Cremophor EL may induce a number of serious side effects, including acute neurotoxicity, hypersensitivity, cardio toxicity and nephrotoxicity (11). Recently, an albumin-bound PTX nanoparticle (Abraxane™) became the first Cremophor EL-free PTX agent approved for the treatment of various types of cancer, including lung, breast and pancreatic cancer, by the Food and Drug Administration (Silver Spring, MA, USA) (5,9). The application of this agent in cancer treatment may reduce the toxicities associated with PTX-based therapy and increase the loading efficiency of PTX to >85% [1:9 (w/w) PTX and albumin mixture].

Over the last two decades, liposomal drug delivery systems have exhibited significant potential for the delivery of therapeutic agents to tumors, and various strategies have been used to improve their targeting specificity and cellular uptake. For example, polyethylene glycol (PEG)ylation has been extensively employed to increase the accumulation of liposomes in tumor tissues via enhanced permeability and retention (EPR) effects (i.e., passive targeting) (11). To increase the specificity of the interactions between liposomes and tumor cells, various studies have focused on the development of active tumor-targeted liposomes modified with specific ligands, including transferrin (12), folic acid (13), peptides (1418) or antibodies (1921). These modified liposomes are able to selectively recognize and bind to specific receptors overexpressed on tumor cells. Clinically, these modifications are able to increase targeting efficiency and reduce toxicity. The cyclic arginine-glycine-aspartic acid-phenylalanine-lysine (cRGDFK/RGD) peptide has been widely used with various anticancer agents and nanocarriers to facilitate specific tumor targeting (22,23). The RGD motif, comprised of extracellular matrix proteins, functions as a cell adhesion site for various types of integrin, particularly αvβ3 and αvβ5. These two integrins are overexpressed on the angiogenic endothelium of diseased tissues and various types of malignant tumor.

In the present study, an RGD-conjugated PEG-modified liposome (RGD-LP) was constructed as a nanoplatform to deliver tumor-targeted PTX contrast agent, yielding PTX-loaded RGD-LP (RGD-LP-PTX; Fig. 1). The current study subsequently aimed to evaluate the in vitro and in vivo targeting and anti-tumor efficiency of RGD-LP-PTX in the treatment of HCC.

Materials and methods

Materials

Soybean lecithin consisting of 90–95% phosphatidylcholine (SPC), and 1,2-distearoylphosphatidylethanolamine methylated polyethylene glycol 2000 (DSPE-mPEG2000), and DSPE-PEG2000-Maleimide (Mal) were purchased from Avanti Polar Lipids, Inc. (Alabaster, AL, USA). Additionally, cholesterol was purchased from Chengdu Kelong Chemical Co. Ltd. (Chengdu, China) and carboxyfluorescein phosphatidylethanolamine (CFPE) was purchased from Avanti Polar Lipids, Inc. The RGD peptide with a terminal cysteine was synthesized according to the standard solid phase peptide synthesis protocol described by the manufacturer (Shanghai Jier Bio-Pharmaceutical Co., Ltd., Shanghai, China). Furthermore, DAPI was obtained from Beyotime Institute of Biotechnology (Haimen, China), 1,10-dioctadecyl-3,3, 30,30-tetramethylindotricarbocyanineiodide was purchased from Biotium, Inc. (Hayward, CA, USA) and cell culture plates were purchased from Wuxi Nest Biotechnology Co., Ltd. (Wuxi, China). All other chemicals and reagents were of analytical grade and obtained commercially.

BALB/c male athymic nude mice (weight, ~20 g; n=40) were purchased from the Experimental Animal Center of Sichuan University (Chengdu, China). They were housed at 25°C and 50% humidity under sterilized conditions, with a 12 h light/dark cycle and free access to food and water. All animal procedures were approved by the Experimental Animal Administrative Committee of Sichuan University.

DSPE-PEG2000-RGD synthesis

DSPE-PEG2000-RGD was synthesized by conjugating the cysteine residue of the three RGD peptides to DSPE, PEG2000 and Mal, respectively. Briefly, DSPE-PEG2000-Mal and Cys-RGD (molar ratio, 1:1.5) were mixed in 8 ml chloroform/4 ml methanol (2:1, v/v), and 0.3 ml triethylamine was added as a catalyst. The mixture was gently stirred for 24 h in the dark at room temperature. Once thin layer chromatography (DCM/MeOH/H2O; 3:0.5:0.001) demonstrated that no DSPE-PEG2000-Mal remained, indicating the synthesis of DSPE-PEG2000-RGD was completed, the mixture was evaporated by a Yamato RE200B rotary evaporator (Yarong Biolab Co., Ltd., Shanghai, China) in a vacuum. The residue was redissolved in chloroform and the solution was filtered to purify the product. The filtrate was then evaporated again by rotary evaporation to obtain the final product (DSPE-PEG2000-RGD), which was stored at <-20°C.

Preparation and characterization of the liposome

RGD-LP-PTX was prepared using thin film hydration, as previously described (24,25). Briefly, SPC, cholesterol, PTX (10% of the weight of SPC plus cholesterol; Haizheng Pharmaceutical Co., Ltd., Zhejiang, China), DSPE-PEG2000 and DSPE-PEG2000-RGD were dissolved in chloroform (DSPE-PEG2000:DSPE-PEG2000-RGD molar ratio, 9.5:0.5; final phospholipid:cholesterol molar ratio, 3:2; Chengdu KeLong Chemical Co., Ltd., Chengdu, China). Chloroform was then evaporated by rotary evaporation and the residual organic solvent was removed in a vacuum overnight. The thin film was subsequently hydrated in phosphate-buffered saline (PBS; pH 7.4; Chengdu KeLong Chemical Co., Ltd.) for 1 h at 37°C, followed by intermittent probe sonication on a JY92-II sonicator (Xinhi Biolab Co., Ltd., Ningbo, China) for 50 sec at 100 W.

CFPE-labeled LP-PTX and RGD-LP-PTX were prepared by adding 100 µg CFPE to the organic lipid solution. The entrapment efficiency of PTX was determined by high performance liquid chromatography (Agilent 1200; Agilent Technologies, Inc., Santa Clara, CA, USA). Furthermore, the mean size, polydispersity index (PDI) and ζ potential of LP-PTX and RGD-LP-PTX were detected using a nanoparticle analyzer at a fixed angle of 90° and a temperature of 20°C (ZetaSizer Nano ZS90; Malvern Instruments Ltd., Malvern, UK).

To demonstrate the serum stability of the liposomes, turbidity variations were monitored in the presence of fetal bovine serum (FBS; Chengdu KeLong Chemical Co., Ltd.) (26,27). Briefly, liposomes were mixed with an equal volume of FBS at a temperature of 37°C with gentle shaking (500 × g). As described in a previous study (28), at predetermined time-points (1, 2, 4, 8, 24 and 48 h), a 200 µl sample was pipetted onto a 96-well plate and the transmittance was measured at a wavelength of 750 nm using a microplate reader (Varioskan™ Flash Multimode Reader; Thermo Fisher Scientific, Waltham, MA, USA).

In vitro PTX release analysis was performed using the dialysis method (25), with PBS (pH 7.4) containing 0.1% (v/v) Tween 80 (Chengdu KeLong Chemical Co., Ltd.), which was used as the release media. LP-PTX or free PTX (5 ml) were loaded into dialysis tubes (pore size, 8,000 Da MWCO; KeLong Chemical Co., Ltd.) and tightly sealed. Release media (50 ml) was added to the dialysis tubes and incubated at 37°C with gentle oscillation for 72 h. At the predetermined time-points, 0.1 ml release media was sampled and replaced with an equal volume of fresh release media. The samples were then analyzed by high-performance liquid chromatography (HPLC) using a 1200 HPLC System (Agilent Technologies, Inc., Santa Clara, CA, USA) to determine the concentration of PTX remaining in the dialysis medium.

Cell culture

HepG2 and HeLa human cervical carcinoma cells obtained from the American Type Culture Collection (Manassas, VA, USA) were cultured in RPMI-1640 medium containing 10% FBS, 100 U/ml penicillin and 100 mg/ml streptomycin (all from Chengdu KeLong Chemical Co., Ltd.). The cells were incubated at 37°C in a 5% CO2 humidified atmosphere (Thermo Fisher Scientific).

Cellular uptake

HepG2 and HeLa cells were plated in six-well plates at a density of 5×105 cells/well and cultured for 24 h at 37°C in the RPMI-1640 culture medium as described above. CFPE-labeled LP-PTX and RGD-LP-PTX were prepared as described above, then added to the plates to a final CFPE concentration of 2.0 mg/ml. Following incubation for 4 h at 37°C, the cells were washed three times with cold PBS, trypsinized and resuspended in 0.5 ml PBS. The fluorescence intensity of cells treated with the various liposomes was measured using a flow cytometer (Cytomics FC 500; Beckman Coulter, Inc., Brea, CA, USA); the fluorescence was excited at 465 nm and recorded at 502 nm.

For qualitative analysis, HepG2 and HeLa cells were plated at a density of 1×105 cells/well on gelatin-coated cover slips in six-well plates and cultured for 24 h. CFPE-labeled liposomes were added to the cover slips as described in the quantitative cellular uptake experiments. Following 4 h of incubation at 37°C, the cells were washed three times with cold PBS and fixed with 4% paraformaldehyde for 30 min at room temperature for cytoplasm staining. Subsequently, DAPI was added for 5 min for nuclei staining. Finally, the cells were imaged using a confocal microscope (FV1000; Olympus Corp., Tokyo, Japan).

Uptake mechanism

To determine the uptake mechanism of RGD-LP-PTX, HepG2 cells were preincubated with various endocytosis inhibitors, including penicilloyl polylysine (400 mg/ml), sodium azide (6.0 mg/ml), chlorpromazine (10 mg/ml) and filipin (10 mg/ml). In addition, the inhibitory effects of free RGD peptide (100 mg/ml) and temperature (4°C) were studied. Following a 30 min pre-incubation with the aforementioned inhibitory agents, CFPE-labeled RGD-LP-PTX were added and incubated for an additional 4 h at 37°C. The cells were washed three times with cold PBS, trypsinized and resuspended in 0.5 ml PBS. The fluorescence intensity of the cells treated with various inhibitors was measured using a Cytomics FC 500 flow cytometer.

Tumor spheroid penetration

HepG2 tumor spheroids were established by seeding HepG2 cells in 96-well plates at a density of 2×103 cells/200 µl per well. The plates were coated with 80 µl 2% low-melting temperature agarose (Chengdu KeLong Chemical Co., Ltd.). Following 7 days of culture at 37°C, the tumor spheroids were treated with 10 µg/ml CFPE-labeled LP-PTX or RGD-LP-PTX and incubated for an additional 4 h at 37°C. Following this incubation period, the spheroids were washed three times with ice-cold PBS and fixed in 4% paraformaldehyde for 30 min. The spheroids were subsequently transferred to glass slides and covered with glycerophosphate (Chengdu KeLong Chemical Co., Ltd.). Fluorescence intensity was evaluated using a TCS SP5 laser scanning confocal microscope with Acousto Optical Bream Splitter (Leica Microsystems GmbH, Wetzlar, Germany).

In vitro cytotoxicity and anti-proliferation assay

The cytotoxicity of PTX-loaded liposomes was measured by performing an MTT assay. HepG2 cells were plated in 96-well plates at a density of 2×103 cells/well and cultured for 24 h at 37°C. LP-PTX and free-PTX were diluted to predetermined concentrations with PBS, and added to each well for a 24-h incubation. The final concentrations of PTX ranged between 0.3 and 30 mg/ml. Subsequently, 20 ml MTT (concentration, 5 mg/ml in PBS) was added to each well and incubated for 4 h at a temperature of 37°C. Finally, the medium was removed and replaced with 150 ml dimethyl sulfoxide. Absorbance was measured using a Varioskan Flash multimode microplate reader (Thermo Fisher Scientific, Waltham, MA, USA) at a wavelength of 570 nm. The cells treated with PBS were evaluated as controls. Cell viability was calculated using the following formula: Cell viability (%) = Atreated / Acontrol × 100, where Atreated and Acontrol represent the absorbance of treated and control cells at 570 nm, respectively.

In vivo tumor growth inhibition

Nude mouse HCC xenograft models were established by subcutaneously injecting HepG2 cells (1×107 cells per mouse) into the subcutaneous tissue on the back of 4–6 week-old BALB/c male athymic nude mice (n=40). The nude mouse HCC xenograft models were divided into four groups. Tumor volume (mm3) was measured with vernier calipers, and when the tumors reached a volume of 100–200 mm3, the mice were administered with PBS, free PTX, LP-PTX or RGD-LP-PTX once every other day for 10 days (total, 10 mg/kg), via tail vein injection. Tumor volumes were also measured every other day.

Statistical analysis

Analysis of variance was performed to determine the variance of the whole values in each group. Statistical comparisons of the experimental groups were performed using a Student's t-test. SPSS software, version 21.0 (IBM SPSS, Armonk, NY, USA) was used for statistical analysis, and P<0.05 was considered to indicate a statistically significant difference.

Results and Discussion

Table I indicates the particle size, PDI and ζ potential of the liposomal samples, as determined by a nanoparticle analyzer. RGD-LP-PTX had a mean particle size of 123.6 nm. This value was not significantly different from the LP-PTX mean particle size (117.4 nm). A narrow PDI range was identified for all the liposomal samples, indicating homogeneity of dispersion. The drug encapsulation efficiency (EE) of nanoparticles is crucial for determining their clinical applications; an EE of >80% is considered sufficient for the preparation of efficient liposomes (29). Furthermore, HPLC determined that the mean EE of PTX was 87.79 and 88.48% for LP-PTX and RGD-LP-PTX, respectively. The similar EEs indicate that peptide modification did not affect the loading of PTX into the liposome.

Table I.

Characteristics of PTX-loaded LP and RGD-LP (n=3).

Table I.

Characteristics of PTX-loaded LP and RGD-LP (n=3).

GroupParticle size, nmPolydispersity indexζ potential, mVEncapsulation efficiency,%
LP-PTX123.6±11.80.140±0.08−2.56±1.4987.79±2.45
RGD-LP-PTX117.4±12.10.190±0.132.85±1.7788.48±3.61

[i] PTX, paclitaxel; LP, liposome; RGD, Arg-Gly-Asp. Values are presented as the mean ± standard deviation.

Particle stability under physiological conditions is a prerequisite for the in vivo application of particles; therefore, 50% FBS was used to mimic in vivo conditions. To evaluate the serum stability of liposomes, the current study monitored transmittance variations. As indicated in Fig. 2, LP-PTX and RGD-LP-PTX exhibited transmittance of >90%, with minimal change over 48 h. These observations indicated that no liposome aggregation occurred in the presence of serum.

The release profile of PTX from non-targeted (LP-PTX) and targeted (RGD-LP-PTX) liposomal samples was determined following treatment with PBS (pH 7.4) by HPLC. The difference between LP-PTX and RGD-LP-PTX was the modification of RGD, while the release of the PTX from LP-PTX and RGD-LP-PTX were similar. Thus, the rate and extent of PTX release from the prepared liposomes did not appear to be affected by peptide modification or density (Fig. 3). For free PTX, >95% of the PTX was released from the dialysis tubes following 5 h of incubation, while <20% of the PTX content was released into the dialysis medium from LP-PTX and RGD-LP-PTX.

The selectivity and internalization of various liposomes was also investigated. HeLa cells were selected for use in the present study due to their low integrin receptor expression levels compared with those of HepG2 cells, which overexpress integrin receptors (30). Fig. 4 indicates the uptake of LP-PTX and RGD-LP-PTX by HeLa and HepG2 cells. No significant difference in the cellular uptake of LP-PTX and RGD-LP-PTX was identified in HeLa cells, however, RGD-LP-PTX uptake was significantly higher than LP-PTX uptake in HepG2 cells (~3.3-fold higher; P<0.001). This difference was potentially due to the targeting capacity of the integrin receptors that are highly expressed in HepG2 cells. As the cellular uptake results were consistent with the integrin expression levels on the cell surface, it was suggested that the RGD motif may be able to recognize and target cell-surface integrin receptors. As indicated in Fig. 5, the fluorescence intensity of LP-PTX was lower than that of RGD-LP-PTX in the two cell types analyzed. In HeLa cells, the fluorescence intensity of RGD-LP-PTX was did not significantly differ from that of LP-PTX. However, in HepG2 cells, the fluorescence intensity of RGD-LP-PTX was markedly greater than that of LP-PTX. Thus, there was agreement between the quantitative (Fig. 4) and qualitative fluorescence imaging (Fig. 5) results. It was, therefore, proposed that integrin receptor-mediated endocytosis (RME) facilitates the cellular uptake of RGD-LP-PTX into HepG2 cells, resulting in a higher uptake efficiency compared with that of LP-PTX. In HeLa cells, the uptake efficiency of RGD-LP-PTX is similar to that of LP-PTX, predominantly due to the small number of integrin receptors expressed by HeLa cells.

To identify the uptake mechanism of RGD-LP-PTX, HepG2 cells were pre-incubated with a series of endocytosis inhibitors, and the rate of inhibition was calculated. As indicated in Fig. 6, the presence of free RGD peptide significantly decreased the cellular uptake of RGD-LP-PTX (P<0.01). This supported the theory that the RGD facilitates an increase in cellular internalization by specifically binding to integrin receptors expressed on HepG2 cells. When targeting sites were competitively bound by free RGD, the cellular uptake of RGD-LP-PTX was decreased. In addition, penicilloyl polylysine was used as positive charge inhibitor; 4°C and sodium azide were selected to analyze the effect of energy on uptake; and chlorpromazine and filipin were used to block clathrin- and caveolin-mediated endocytosis, respectively. As demonstrated in Fig. 6, each inhibitor induced varying levels of inhibition. Polylysine did not significantly alter uptake, indicating that the cellular uptake of RGD-LP-PTX was not dependent on positive charge. Similarly, chlorpromazine and filipin did not significantly change the cellular uptake of RGD-LP-PTX, indicating that uptake was independent of clathrin- and caveolin-mediated endocytosis. By contrast, a temperature of 4°C and treatment with sodium azide had a significant impact on cellular uptake [reduced from 100 to 14.5 (P<0.001) and 33.7% (P<0.01) uptake, respectively], indicating the energy-dependent properties of integrin RME.

Hypoxic and avascular regions are observed in numerous types of solid tumor. The poor permeation of delivery systems associated with such tumors results in a low concentration of PTX reaching the inside of solid tumors (31,32). Tumor spheroids were prepared, as the lack of blood vessels makes them effective models for the evaluation of the in vivo status of solid tumors (33) and the solid tumor penetration effect of liposomes. Fig. 7 exhibits confocal laser scanning microscopic images of three-dimensional tumor spheroids 4 h after the application of CFPE-labeled LP-PTX and RGD-LP-PTX. LP-PTX almost entirely lacked efficient penetration of the HepG2 spheroids, with no fluorescence observed in the centre of the spheroid. This observation indicated that unmodified liposomes are weakly penetrating. By contrast, enhanced fluorescence was observed in the centre of the spheroids treated with RGD-LP-PTX, indicating that solid tumor penetration was enhanced by RGD modification.

Fig. 8A indicates that the viabilities (survival rates) of HepG2 HCC cells incubated with 0.3, 3.0, 10.0 and 30.0 mg/ml free PTX for 24 h were 82.12, 79.48, 76.36 and 72.54%, respectively. Following 48 h of treatment with identical concentrations of PTX, cell viabilities were decreased to 67.49, 63.70, 57.74 and 53.62%, respectively (Fig. 8B). Thus, an increasing PTX concentration and extending the incubation time resulted in a reduction in cell viability (or, equivalently, an increase in cell death). To determine the cytotoxicity of LP-PTX and RGD-LP-PTX, 0.3, 3.0, 10.0 and 30.0 mg/ml PTX encapsulated in the liposomes was applied to HepG2 cells. Following treatment for 24 h, cell viability was identified to be 68.72, 65.44, 59.26 and 55.06% for LP-PTX, and 56.53, 48.86, 41.12 and 31.28% for RGD-LP-PTX, respectively. However, following 48 h of treatment, cell viability was found to be decreased to 54.27, 51.60, 45.47 and 39.26% for LP-PTX, and 25.94, 25.94, 19.81 and 17.91% for RGD-LP-PTX, respectively. These data indicated that the anti-proliferative effect of the PTX-loaded liposomes was markedly enhanced by modification with RGD. Furthermore, the increased cytoxicity of RGD-LP-PTX compared with that of free PTX may be associated with the rapid internationalization of this structure and the successive release of PTX from RGD-LP-PTX to reach a therapeutic concentration within the cells.

To determine the efficacy of the functionalized nanoparticles in vivo, HCC nude mouse xenograft models were established. As indicated in Fig. 9, LP-PTX and RGD-LP-PTX significantly inhibited the growth of the tumor compared with physiological PBS and free PTX (P<0.001). The superior in vivo efficacy of these liposomes compared with that of PTX alone may be attributed to their sustained release profiles, prolonged blood circulation time and EPR effect. In addition, RGD-LP-PTX exhibited a greater effect on tumor inhibition compared with LP-PTX, potentially due to the integrin receptor targeting efficiency of the RGD peptide. Thus, the in vivo and in vitro results of the present study demonstrated that RGD-LP-PTX may be effectively used to treat HCC and represents a potential targeted drug delivery system for patients with HCC.

In conclusion, the present study successfully developed tumor-targeting liposomes modified with the integrin receptor-specific RGD ligand (RGD-LP-PTX). This liposomal delivery system exhibited increased cellular uptake efficiency and targeting specificity in HepG2 cells with high integrin receptor expression levels, compared with PTX alone or LP-PTX, which are two commonly used paclitaxel dosage forms within the clinic. In addition, efficient targeted delivery of the therapeutic agent was achieved in HepG2 tumor cells and HepG2 tumor-bearing nude mice, ultimately achieving high therapeutic efficacy in the tumor-bearing mice. Based on the investigations conducted in the current study, it was hypothesized that RGD-modified liposomes may be used as a potential anti-tumor PTX delivery system for the effective treatment of patients with HCC.

References

1 

Farazi PA and DePinho RA: Hepatocellular carcinoma pathogenesis: From genes to environment. Nat Rev Cancer. 6:674–687. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Liu X, Wang W and Wang Z: Recent progress in understanding the effects of autophagy in hepatocellular carcinoma. Chin J Hepatobil Surg. 20:69–73. 2014.

3 

Pan Y, Ye S, Yuan D, et al: Hydrogen sulfide (H2S)/cystathionine γ-lyase (CSE) pathway contributes to the proliferation of hepatoma cells. Mutat Res. 763–764:10–18. 2014. View Article : Google Scholar

4 

Zhang C, He H, Zhang H, et al: The blockage of Ras/ERK pathway augments the sensitivity of SphK1 inhibitor SKI II in human hepatoma HepG2 cells. Biochem Biophys Res Commun. 434:35–41. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Baek JS, So JW, Shin SC and Cho CW: Solid lipid nanoparticles of paclitaxel strengthened by hydroxypropyl-β-cyclodextrin as an oral delivery system. Int J Mol Med. 30:953–959. 2012.PubMed/NCBI

6 

Sun J, Deng L, Duan Y, et al: Inhibitory effect of endostatin combined with paclitaxel-cisplatin on breast cancer in xenograft-bearing mice. Exp Ther Med. 3:159–164. 2012.PubMed/NCBI

7 

Kimura K, Tanaka S, Iwamoto M, et al: Safety of nanoparticle albumin-bound paclitaxel administered to breast cancer patients with clinical contraindications to paclitaxel or docetaxel: Four case reports. Oncol Lett. 6:881–884. 2013.PubMed/NCBI

8 

Tanaka T, Toujima S and Tanaka J: Differential sensitivity to paclitaxel-induced apoptosis and growth suppression in paclitaxel-resistant cell lines established from HEC-1 human endometrial adenocarcinoma cells. Int J Oncol. 41:1837–1844. 2012.PubMed/NCBI

9 

Tsukada T, Fushida S, Harada S, et al: Low-dose paclitaxel modulates tumour fibrosis in gastric cancer. Int J Oncol. 42:1167–1174. 2013.PubMed/NCBI

10 

Horwitz SB: Taxol (paclitaxel): Mechanisms of action. Ann Oncol. 5 (Suppl 6):S3–S6. 1994.PubMed/NCBI

11 

Liebmann J, Cook JA and Mitchell JB: Cremophor EL, solvent for paclitaxel and toxicity. Lancet. 342:14281993. View Article : Google Scholar : PubMed/NCBI

12 

Gou Y, Wang L, Lv P and Zhang P: Transferrin-conjugated doxorubicin-loaded lipid-coated nanoparticles for the targeting and therapy of lung cancer. Oncol Lett. 9:1065–1072. 2015.PubMed/NCBI

13 

Despierre E, Lambrechts D, Neven P, et al: The molecular genetic basis of ovarian cancer and its roadmap towards a better treatment. Gynecol Oncol. 117:358–365. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Curtin JP, Blessing JA, Webster KD, Rose PG, Mayer AR, Fowler WC Jr, Malfetano JH and Alvarez RD: Paclitaxel, an active agent in nonsquamous carcinomas of the uterine cervix: A Gynecologic Oncology Group study. J Clin Oncol. 19:1275–1278. 2001.PubMed/NCBI

15 

McGuire WP, Blessing JA, Moore D, Lentz SS and Photopulos G: Paclitaxel has moderate activity in squamous cervix cancer. A Gynecologic Oncology Group study. J Clin Oncol. 14:792–795. 1996.PubMed/NCBI

16 

Rose PG, Blessing JA, Gershenson DM and McGehee R: Paclitaxel and cisplatin as first-line therapy in recurrent or advanced squamous cell carcinoma of the cervix: A gynecologic oncology group study. J Clin Oncol. 17:2676–2680. 1999.PubMed/NCBI

17 

Chua DT, Sham JS and Au GK: A phase II study of docetaxel and cisplatin as first line chemotherapy in patients with metastatic nasopharyngeal carcinoma. Oral Oncol. 41:589–595. 2005. View Article : Google Scholar : PubMed/NCBI

18 

McCarthy JS, Tannock IF, Degendorfer P, Panzarella T, Furlan M and Siu LL: A Phase II trial of docetaxel and cisplatin in patients with recurrent or metastatic nasopharyngeal carcinoma. Oral Oncol. 38:686–690. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Yin Y, Wu X, Yang Z, et al: The potential efficacy of R8-modified paclitaxel-loaded liposomes on pulmonary arterial hypertension. Pharm Res. 30:2050–2062. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Wender PA, Galliher WC, Goun EA, Jones LR and Pillow TH: The design of guanidinium-rich transporters and their internalization mechanisms. Adv Drug Deliv Rev. 60:452–472. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Lee JH, Engler JA, Collawn JF and Moore BA: Receptor mediated uptake of peptides that bind the human transferrin receptor. Eur J Biochem. 268:2004–2012. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Oba M, Fukushima S, Kanayama N, et al: Cyclic RGD peptide-conjugated polyplex micelles as a targetable gene delivery system directed to cells possessing alphavbeta3 and alphavbeta5 integrins. Bioconjug Chem. 18:1415–1423. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Li S, Wei J, Yuan L, et al: RGD-modified endostatin peptide 530 derived from endostatin suppresses invasion and migration of HepG2 cells through the αvβ3 pathway. Cancer Biother Radiopharm. 26:529–538. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Kibria G, Hatakeyama H, Ohga N, et al: Dual-ligand modification of PEGylated liposomes shows better cell selectivity and efficient gene delivery. J Control Release. 153:141–148. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Sharma G, Modgil A, Sun C and Singh J: Grafting of cell-penetrating peptide to receptor-targeted liposomes improves their transfection efficiency and transport across blood-brain barrier model. J Pharm Sci. 101:2468–2478. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Jiang T, Zhang Z, Zhang Y, et al: Dual-functional liposomes based on pH-responsive cell-penetrating peptide and hyaluronic acid for tumor-targeted anticancer drug delivery. Biomaterials. 33:9246–9258. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Maeda N, Takeuchi Y, Takada M, et al: Anti-neovascular therapy by use of tumor neovasculature-targeted long-circulating liposome. J Control Release. 100:41–52. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Zhang Q, Tang J, Fu L, et al: A pH-responsive α-helical cell penetrating peptide-mediated liposomal delivery system. Biomaterials. 34:7980–7993. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Qin Y, Chen H, Yuan W, et al: Liposome formulated with TAT-modified cholesterol for enhancing the brain delivery. Int J Pharm. 419:85–95. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Borgne-Sanchez A, Dupont S, Langonné A, et al: Targeted Vpr-derived peptides reach mitochondria to induce apoptosis of alphaVbeta3-expressing endothelial cells. Cell Death Differ. 14:422–435. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Lewis CE and Pollard JW: Distinct role of macrophages in different tumor microenvironments. Cancer Res. 66:605–612. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Fukumura D, Xu L, Chen Y, et al: Hypoxia and acidosis independently up-regulate vascular endothelial growth factor transcription in brain tumors in vivo. Cancer Res. 61:6020–6024. 2001.PubMed/NCBI

33 

Jain RK: Delivery of molecular and cellular medicine to solid tumors. Adv Drug Deliv Rev. 46:149–168. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 10 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen L, Liu Y, Wang W and Liu K: Effect of integrin receptor‑targeted liposomal paclitaxel for hepatocellular carcinoma targeting and therapy Retraction in /10.3892/ol.2021.12611. Oncol Lett 10: 77-84, 2015
APA
Chen, L., Liu, Y., Wang, W., & Liu, K. (2015). Effect of integrin receptor‑targeted liposomal paclitaxel for hepatocellular carcinoma targeting and therapy Retraction in /10.3892/ol.2021.12611. Oncology Letters, 10, 77-84. https://doi.org/10.3892/ol.2015.3242
MLA
Chen, L., Liu, Y., Wang, W., Liu, K."Effect of integrin receptor‑targeted liposomal paclitaxel for hepatocellular carcinoma targeting and therapy Retraction in /10.3892/ol.2021.12611". Oncology Letters 10.1 (2015): 77-84.
Chicago
Chen, L., Liu, Y., Wang, W., Liu, K."Effect of integrin receptor‑targeted liposomal paclitaxel for hepatocellular carcinoma targeting and therapy Retraction in /10.3892/ol.2021.12611". Oncology Letters 10, no. 1 (2015): 77-84. https://doi.org/10.3892/ol.2015.3242