Repurposing an Erstwhile Cancer Drug: A Quantitative and Therapeutic Evaluation of Alternative Nanosystems for the Delivery of Colchicine to Solid Tumors

نویسندگان

  • Shifalika Tangutoori
  • Akio Ohta
  • Robert B. Campbell
چکیده

Background: Antimitotic drugs represent some of the most popular vascular disrupting agents used today in the fight against cancer. Colchicine is the first known antimitotic alkaloid. The drug binds to tubulin proteins and depolymerizes microtubules. Despite the impressive therapeutic activity as an antimitotic agent, colchicine is fatally toxic when administered to cancer patients intravenously due to its low therapeutic index. This study supports the early development of a relatively safe and target-specific nanoparticle for the IV administration of colchicine to lung tumors. Methods: For in vitro studies, lung cancer (LLC, Chago-k-1 and MCA-205), and endothelial cell lines (MS1VEGF, HMEC-1) were employed. The qualitative and quantitative analyses of the cytoskeleton and nuclear areas were performed using FITC-labeled β-tubulin antibody; the mean area of cytoskeleton and nucleus per cell was determined by fluorescence microscopy and BIOQUANT. In vivo, the biodistribution and therapeutic efficacy studies of the pegylated cationic liposomes (PCLs) were performed in C57BL/6 mice bearing pseudo-orthotopic lung tumors. The biodistribution of colchicine and PCL-colchicine was determined by dual labeling. 111In labeled PCLs and 3H-Colchicine were employed to simultaneously track the vehicle and drug, respectively. The therapeutic efficacy was determined by monitoring animal survival. Results: The disruption of microtubules was most evident when colchicine was loaded in DOTAP-PCLs. We report a ~2 fold (p=0.0214, 95% CI is 2.923 to 13.247) increase in the accumulation of PCL-colchicine in tumorbearing lung compared to the normal lung, resulting in significantly extended survival times in PCL treated group (p=0.0052, Log Rank (Mantel-Cox test)). Conclusions: The PCL-based colchicine-loaded nanosystem can renew the clinical potential of abandoned antimitotic agents such as colchicine. The PCL platform can enable the accumulation of clinically relevant doses of colchicine in solid tumors. This will ensure antimitotic activity while decreasing the uptake of the drug in healthy tissues. Citation: Tangutoori S, Ohta A, Gatley S, Campbell RB (2014) Repurposing an Erstwhile Cancer Drug: A Quantitative and Therapeutic Evaluation of Alternative Nanosystems for the Delivery of Colchicine to Solid Tumors. J Cancer Sci Ther 6: 236-246. doi:10.4172/1948-5956.1000277 Volume 6(7) 236-246 (2014) 237 J Cancer Sci Ther ISSN:1948-5956 JCST, an open access journal inhibitor of angiogenesis, but only when administered above the maximum tolerated dose [11]. The risk associated with intravenous use of colchicine is due to widely variable patient responses to therapy, and metronomic doses required for effective disease management [12,13]. True to its poisonous nature, many suicides and homicides resulted due to the consumption of doses higher than those prescribed [12,14-18]. Currently, the ISMP (Institute for Safe Medication Practices) has listed the intravenous use of colchicine as a high toxicity-alert medication [19,20]. In general, the α/β tubulin heterodimers, polymerize into dynamically unstable microtubules, which play an important role in various sub-cellular motility functions, including the events during spindle formation and chromosomal rearrangements during mitosis [19]. Mechanistically, colchicine binds to the interface of α/β tubulin heterodimers and thus inhibits all the subsequent functions in the cancer cell. Colchicine poisoning occurs due to its large volume of distribution, and its potent antimitotic activity towards every nonspecific mitotic cell it encounters. Clinical indications of colchicine poisoning are typically anaplastic anemia accompanied by high fever, loss of scalp hair, severe agranulocytic leucopenia, hemorrhage and multisystem failure within 24 to 48 hours of acute over-dosage [8,15]. Hence, efforts to improve targeted delivery of colchicine to lung tumors represent a promising clinical approach [21]. Nanoliposome is a well characterized platform used to deliver many toxic chemotherapeutic drugs [22]. Based on previous works, we postulated that the synthetic cationic lipids (i.e., DOTAP, DODAP, DDAB, DMTAP, DSTAP) are potentially useful in the development of colchicine nanoliposomal formulations for cancer therapy [23,24]. These second generation liposomes have previously been used to selectively target drugs to tumor endothelia [21,25]. We now report the early development and evaluation of Pegylated Cationic Liposomal-colchicine (PCL-colchicine) to achieve superior and selective pulmonary targeting. This investigation details the qualitative and quantitative analyses in vitro, and biodistribution and efficacy evaluation studies performed on a pulmonary metastatic tumor model of fibrosarcoma. Methods Preparation of nanoliposomes All lipids were purchased from Avanti Polar Lipids (Alabaster, AL). Liposomes were prepared as previously reported [26,27]. The PCLcolchicine was prepared at 3 and 10 mole% concentrations for in vitro and in vivo studies (Table S2). A lipid mixture consisting of DOPC: Cationic lipid: Cholesterol: DOPE-PEG5000 (35:50:10:5) was evaporated to dryness at 42°C with the aid of a rotary evaporator to obtain a thin lipid film. Any residual organic solvent was removed by freeze drying the lipid film for an additional 2 hrs using Labconco freeze dryer (Labconco Corporation, Kansas City, MO), followed by hydration and PCL size manipulation as previously reported [28]. Cell lines LLC, MS1-VEGF [29], Chago-k-1 [30], HMEC-1 [31,32] and maintenance media DMEM, RPMI 1640, EBM-2,VEGF and hFGF-b, and FBS were all purchased from ATCC (American Type Culture Collection, Manassas, VA). PBS 1X was purchased from Cambrex, NJ. MCA-205 [33] was a gift in kind from New England Inflammation & Tissue Protection Institute, Northeastern University (NU), Boston, MA. Analysis of cytoskeleton disruption using PCL-colchicine Slow Fade® Gold antifade reagent with DAPI was purchased from Molecular Probes (Invitrogen Life Technologies, Carlsbad, California). Monoclonal Anti β tubulin FITC conjugate and purified mouse immunoglobulin (F 2043) were purchased from Sigma Aldrich (St. Louis, MO). Ammonium acetate and BSA were purchased from Sigma Aldrich (St. Louis, MO). Fluorescence microscopic analyses were carried out using a BX61 W1 Olympus fluorescence microscope from Optical Analysis Corporation (Melville, NY). Cells were seeded at a density of 5x105 onto sterile coverslips in a 6 well plate (Corning, NY). After 24 hrs of incubation at 37°C free drug or liposomal drug was added to selected wells. The well plate was incubated for an additional 24 hrs after which the coverslips were prepared for mounting. Cell fixation and staining with antibody was performed according to specifications provided with the MSDS of FITC-conjugated β tubulin antibody. Briefly, the coverslips were washed twice with 1X PBS to remove cellular debris and then fixed with cold absolute methanol. Coverslips were then incubated at -20oC for 10 min. The methanol was washed off with cold acetone and the cells were rehydrated in 1X PBS for at least 30 min. The rehydrated cells were then incubated with FITC-conjugated β-tubulin antibody for 1 hr, suitably diluted with 1X PBS containing 1% BSA. A working dilution of 1:25 was used. Excess antibody was removed with 1X PBS, and samples were mounted onto a glass slide using Slowfade® Gold antifade reagent DAPI as the mounting media. The images were randomly selected at five different fields of view (FOV). DIC microscopy was used to observe cell shape and morphology prior to obtaining all fluorescence images. The FITC and DAPI images were merged to observe the area of cytoskeleton and nucleus using LLC and MS1-VEGF cell lines. All images were captured at 40X magnification using a BX61 W1 Olympus Fluorescence microscope from Optical Analysis Corporation (Melville, NY) BIOQUANT for quantitative analysis BIOQUANT (Image Analysis Corporation) was used to quantify the merged images of the cell nucleus and cytoskeleton. Images were selected randomly at five different FOV on the coverslip. Areas with a fixed threshold of fluorescence intensities for FITC (cytoskeleton) and DAPI (nucleus) were selected and measured (Figure 1). We determined the total number of cells in each field, and the area occupied per cell. Mean cytoskeleton and nuclear areas per cell was plotted as a scatter plot. Each dot in the plot represents the Mean ± 95% CI of single field in the cell sample. Statistics were performed using one way ANOVA (Graph Pad Prism 5) at a significance level of 0.05, and the Bonferroni post hoc test was used to compare pairs of column in the study. Animal studies: All of the studies involving animals were performed in accordance with NU-IACUC policies and procedures. C57BL/6 female mice, 8 wks old, weighing ~20 gm, were purchased from Charles River Laboratories Inc. (Wilmington, MA). The radioactive isotopes 111In, 3H-colchicine, SolvableTM, UltimaGoldTM XR used for the biodistribution studies were purchased from PerkinElmer Inc (Waltham, MA). Tissue distribution and therapeutic efficacy studies of formulated and free drug were carried out in C57BL/6 female mice. Generation of pulmonary metastatic tumor nodules: A well characterized pulmonary metastatic tumor model was developed in female C57BL6 mice as described previously using MCA-205 cells [34]. This pseudo-orthotopic model has been shown to reliably produce pulmonary metastases that can be visualized by microscopic Citation: Tangutoori S, Ohta A, Gatley S, Campbell RB (2014) Repurposing an Erstwhile Cancer Drug: A Quantitative and Therapeutic Evaluation of Alternative Nanosystems for the Delivery of Colchicine to Solid Tumors. J Cancer Sci Ther 6: 236-246. doi:10.4172/1948-5956.1000277 Volume 6(7) 236-246 (2014) 238 J Cancer Sci Ther ISSN:1948-5956 JCST, an open access journal evaluation. Briefly, 3x105 MCA-205 cells were injected via tail vein into mice and the lungs were harvested at various stages of tumor growth (Table 1 and 2). Mice were anesthetized with ketamine-xylazine (i.p. 9 mg/100 g-0.9-100 g), and excessive inhalation of 5% Isoflourane was used to euthanize the mice Biodistribution of dual radiolabeled 111InPCL (3H-Colchicine) Synthesis of dual radiolabeled [111In PCL (3H-Colchicine)]: To establish the biodistribution profile of colchicine and PCL-colchicine, two stable radioactive isotopes were employed. PCLs labeled with 111In have been used to detect the path followed by PCLs in vivo [35] . Back in 1930’s, 3H-colchicine and 14C-colchicine were used for tracking the drug in various tissues in mice [36,37]. Hence, we formulated the PCLcolchicine using two different radioisotopes. 111In and 3H were used for tracking PCLs and colchicine, respectively. Unlabeled colchicine was added to trace amounts (5 μCi/mouse) of 3H-colchicine to maintain the required drug to lipid ratio with comparable drug loaded. 111In labeling was carried out as described previously [35,38]. Briefly, the required lipids including DTPA-DOPE at 5 mole% were added and a thin film was generated by rotary evaporation. The film was hydrated in (1X) PBS overnight in warm water bath to produce PCLs. The 3H-colchicine-PCL was acidified using citric acid buffer (pH-4.5), and labeled with 111In (2 μCi/mouse). At pH 4.5 the 111In was covalently attached to the DTPA in the PCL bilayer. Excess and unbound 111In was separated from the formulation by dialysis overnight in 4L of HEPES buffer (pH 7.4). For free drug, 50 μCi of 3Hcolchicine (from stock of 60-87 Ci/mmol), was measured out, diluted in PBS and injected into mice via tail vein. Biodistribution studies-study design: To study the infiltration of drug in the tumor nodules at various stages of development, tumor cells were injected on different days and the formulations on a fixed (the same) day. Tumor cells were injected at 1, 5 and 8 days to generate the tumors that were 12, 8 and 5 days old (Groups 3-5), respectively, on the day of treatment (Table-1). One mouse from every group was euthanized, and the lungs were harvested to assess metastasis. Analysis of 111In labeled PCL accumulation Measurement of 111In labeled PCLs was carried as explained elsewhere [27]. Briefly, after the mice were sacrificed, various organs including blood, liver, spleen, tail, kidney, lung/tumor and heart were harvested in groups 2-5 and collected into pre-weighed test tubes. The half-life of 111In is 2.8 days, predominantly emitting high energy gamma radiations. The amount of radioactivity thus retained by individual organs was quantified as counts per minute (CPMs) using a Beckman 5500B gamma counter (Beckmann instruments, Fullerton, CA), and the amount of the accumulated radioactivity per gram of tissue was calculated (% D/M). The radioactivity retained by the tumor-bearing lung was compared with the healthy lung controls. Analysis of 3H-colchicine accumulation: The 3H measurements were acquired based in part on previous reports using radioactive colchicine to determine organ-specific uptake [36,37]. The protocol was modified to meet the goals of the present study. After the mice were sacrificed, various organs were harvested for all groups, and organs were collected into pre-weighed scintillation vials. The halflife of 3H is 12.3 years, emitting low energy β radiations. To rule out the interference of gamma radiations of the 111In during the 3H measurements, the tissue samples collected were stored at 4°C for at least 20 days (10 t0.5 of 111In) to allow for sufficient 111In decay. The tissue samples were subsequently digested with SolvableTM (Perkin Elmer, Controls Saturated Cationic Liposomes Unsaturated Cationic Liposomes Figure 1: Fluorescence images of endothelial cell, HMEC-1, 24 hrs following exposure to colchicine (free drug) or various PCLs (with colchicine). The images show qualitative effect of different cationic lipids of PCLs on cytoskeleton (green) and the nucleus (blue) cell areas. Images were captured using 40X magnification. Magnification bar = 20 μm. Citation: Tangutoori S, Ohta A, Gatley S, Campbell RB (2014) Repurposing an Erstwhile Cancer Drug: A Quantitative and Therapeutic Evaluation of Alternative Nanosystems for the Delivery of Colchicine to Solid Tumors. J Cancer Sci Ther 6: 236-246. doi:10.4172/1948-5956.1000277 Volume 6(7) 236-246 (2014) 239 J Cancer Sci Ther ISSN:1948-5956 JCST, an open access journal Boston, MA), an aqueous based solubulizer. The highly perfused tissues (i.e., liver, lung, heart, and blood) were treated with 10% H2O2 overnight to decolorize dark brown color owing to the blood retention in these tissues. Measures were taken to prevent any escape of tritiated water due to oxidation reactions related to hydrogen peroxide. After all the samples were sufficiently decolorized (light yellowish tinge), 10 mL of liquid scintillation fluid, Ultima GoldTM XR (Perkin Elmer, Boston, MA) was added to each vial, the samples were stored in dark overnight and the following day the CPM’s were measured in liquid scintillation counter (Beckman CoulterTM, LS-6500 Scintillation Systems, Fullerton, CA). The raw CPM’s were used to calculate the % injected dose per gram of the tissue sample collected (% D/G). The radioactivity retained by the tumor-bearing lung was compared with the healthy lung tissue control groups. The distribution profile of free colchicine and PCLcolchicine was also compared. Therapeutic response -survival studies Study design: The animal survival study was performed using the pulmonary pseudo-metastatic model. The MCA-205 cells (3x105) were injected via tail vein on day 1. The tumor-bearing mice were separated into 3 different experimental groups. Colchicine or PCL-colchicine was administered on 9, 12 and 15 days following the injection of tumor cells (Table-2). All mice were monitored daily for changes in body weight. To visualize the tumor nodules the lungs were harvested and fixed in Fakete’s fixing solution. The Kaplan-meier curves were generated using Graph Pad Prism 5 (Graph Pad Prism software, Inc, La Jolla, CA). Imaging of tumor nodules: Once the mice were sacrificed they were dissected at the tracheal region, and the diaphragm was removed to clearly expose the lungs. A small nick was made on the trachea and 15% black India ink (HigginsTM, Boston. MA) was injected retro-tracheally until the lungs were inflated to their full volume. The lungs insufflated with ink were now black providing a counterstain for the white tumor nodules on the surface. Lungs were then harvested, washed in distilled water to remove excess ink or blood and fixed overnight in Fakete’s fixing solution [34]. An in vivo imaging microscope (Fisher Scientific, 4.5X) was used to capture the images of the harvested lungs. Statistical analysis Statistical analyses were carried out using one way ANOVA with bonforroni post hoc test, log rank Mantel-Cox test (for Kaplan Meier curves) and two tailed t-tests at α=0.05 with 95% CI values as errors. GraphPad Prism 5 was used to assign the significant values as * p< 0.05, ** p<0.001, *** p<0.0001 and to report the 95% confidence intervals. Results DOTAP-PCL-colchicine qualitatively altered the cytoskeletal area of endothelial cells to the greatest extent PCL-colchicine was optimized to increase the potential for tumor vascular disruption. We screened for the synthetic cationic lipid most capable of inducing significant structural changes in endothelial cells when employed in the preparation of PCL-colchicine. HMEC1 (the endothelial cellular model) was exposed to the various PCLcolchicine formulations composed of both saturated and unsaturated cationic lipids. We analyzed qualitative changes in the cytoskeleton. Qualitatively, DOTAP-PCL-colchicine was the most effective among all the preparations evaluated. The effect of DOTAP-PCL-colchicine was similar also to the effect of free colchicine. This can be attributed to the spacio-temporal availability of higher concentration of the drug, possibly due to relatively rapid release of colchicine when delivered via unsaturated DOTAP-PCLs. In comparison to other PCL varieties, DOTAP was the most effective at altering the structural and morphological properties of the HMEC-1 cells (Figure 2). PCL-colchicine induced significant quantitative changes to the cytoskeletal area of endothelial cells Since colchicine and PCL-colchicine altered cell morphology, fluorescence microscopy was next used to quantify the extent of cytoskeletal disruption (Figures 1,3). The results from this study revealed that free colchicine reduced the cytoskeleton area of the endothelial cells without altering the nuclear area. We determined that HMEC-1 had the greatest cytoskeleton area per cell (Figure 4E) (1946 ± 306), which was significantly reduced following exposure to colchicine (1073 ± 390, 95% CI-512 to 1234). MS1-VEGF (1692 ± 425), followed the same trend, and exposure to free drug reduced the area to 590 ± 265 (95% CI-276 to 955). Both endothelial cells showed significant decrease in cytoskeleton area per cell when compared to the effect of free colchicine (232 ± 173, 95% CI-1033 to 1839) (Figure 3, 4A, 4C). In cancer cellular models evaluated, PCL-colchicine significantly altered both the cytoskeleton and nuclear areas per cell in comparison to untreated cells, except for MCA-205 (Figure 1). Although promising, the qualitative effect observed in cytoskeletal disruption of MCA205 was not translated quantitatively since there was no significant difference observed compared to the untreated samples (Figure 1E,1F). This can be attributed to several factors including the inherent resistance of the cell line to cytotoxic drug therapy. Effective delivery and biodistribution profile of PCL-loaded colchicine We next evaluated the therapeutic effect of PCL-colchicine in a pulmonary metastatic model of MCA-205, as this cell line was derived from metastatic lung cancer. Our rationale was that, the MCA-205 tumor model reliably induces metastatic nodules in the female C57BL6 mice, and is the most 6 k1 resistant cell line of the many evaluated in this study to the effects of colchicine. In order to determine the changes in the biodistribution profile due to altered pharmacokinetics of the PCLcolchicine, we simultaneously tracked PCL-colchicine or colchicine using 111In and 3H isotopes, three hours following a single i.v. injection. We determined that recovery of 111In PCL and 3H colchicine in the liver (~40-58%) was the highest among all of the tissues evaluated in the PCL-colchicine treated group. The recovery of both labels from Group Day of Tumor Injection Age of tumor (days old) Single i.v. injection received on day 12 (100 c.c) Radiolabels Analyzed 1 Control-No tumor N/A 3H-colchicine 3H 2 Control-No tumor N/A [111In PCL ( 3H colchicine)] 111In and 3H 3 Day 7 5 [111In PCL ( 3H colchicine)] 111In and 3H 4 Day 4 8 [111In PCL ( 3H colchicine)] 111In and 3H 5 Day 1 12 [111In PCL ( 3H colchicine)] 111In and 3H 6 Day 1 12 3H-colchicine 3H Table 1: Biodistribution study design. Group Days of treatment Dose of drug (mg/ kg) # of mice per group 1 Untreated controls 5 2 Colchicine treated 9,12,15 2, 2, 4 5 3 PCL-Colchicine treated 9,12,15 2, 2, 4 5 Table 2: Experimental Design for therapeutic response survival study. Citation: Tangutoori S, Ohta A, Gatley S, Campbell RB (2014) Repurposing an Erstwhile Cancer Drug: A Quantitative and Therapeutic Evaluation of Alternative Nanosystems for the Delivery of Colchicine to Solid Tumors. J Cancer Sci Ther 6: 236-246. doi:10.4172/1948-5956.1000277 Volume 6(7) 236-246 (2014) 240 J Cancer Sci Ther ISSN:1948-5956 JCST, an open access journal the same site suggests that the carrier and the drug co-exist (Figure 5A and 5B). The recovery of 3H label from blood and tumor in PCLcolchicine treated mice was ~10-18% and 10-20% of the injected dose, respectively. The accumulation of the label gradually increased with tumor age (Figure 5B). The lung to blood ratio in healthy tissue and in tumor-bearing mice treated with PCL-colchicine and colchicine showed ratios of 0.7 and 1.6, respectively. The >2 fold decrease of accumulation in healthy lung compared to the tumor group suggests minimal potential off-targeting effects of PCLs (Figure 6F). In contrast, there was minimal difference in free colchicine accumulation between healthy lungs and in the tumor (Figure 6E). The experimental findings support enhanced tumor targeting in the lung (as supported by a ~10 fold increase in tumor uptake over free drug treated group), and a favorable decrease in nonspecific accumulation in healthy lung tissue (Figure 6B). PCL-colchicine uptake positively correlated with tumor size and age We next evaluated the uptake of PCL-colchicine as a function of tumor burden size/age. The tumors were selected at various stages of growth, and PCL-colchicine was administered via tail vein injection (Figure 6A,6D). We observed a higher tumor to blood ratio in advanced tumors (8 and 12 day), when compared to no tumor group or early stages of tumor development (day 5). The group consisting of 12 day old tumors revealed a significantly higher accumulation of the drug (19% of injected dose) when compared to day 5 (~11% of injected dose), and no tumor (~10% of injected dose). The experimental finding suggested that the overall drug recovery by the tumor mass increased with tumor age (Figure 6C). PCL-colchicine significanlty enhanced survival of tumorbearing mice We next determined the therapeutic effect of PCL-colchicine compared to the colchicine treatment. The Kaplan Meier survival curves were used to demonstrate the therapeutic benefit of formulating colchicine in PCLs. The tumor nodules in the untreated control group were larger and greater in number compared to treated groups (Figure 7A). The specific observations are summarized in Table S3. The PCLcolchicine treated groups showed 80% survival on day 23, 40% survival on day 25 and 20% survival on day 29. Therefore, PCLs significantly prolonged effect of colchicine and hence the animal survival compared to other groups (Figure 7B). By day 21, the untreated group showed severe respiratory distress. The mice were extremely moribund, and experienced severe body weight loss (Figure 7C). Following two injections of 2 mg/kg (0.05mg/mouse/injection) in the free and PCLcolchicine treated groups, the mice initially displayed weight loss, but eventually showed signs of recovery (Figure 7D,7E). The third dose was administered to the two groups at 4 mg/kg (0.1 mg/mouse/injection) on day 15. Again, a drastic decrease in the body weight was observed initially followed by a period of recovery. Approximately 48 hours following the final injection, >20% body weight loss was observed in the entire free drug treatment group (Figure 7D), and the mice were subsequently removed from the study according to IACUC regulations. Although the PCL-colchicine showed an initial decrease in body weight (Figure 7E), only one mouse from this group reached the 20% weight loss limit, the remaining subjects demonstrated definite signs of recovery. Discussion Colchicine is a potent alkaloid with a highly impressive antimitotic C A Figure 2: The fluorescence images show the qualitative effect of DOTAP-PCLs, free colchicine and DOTAP-PCL-colchicine against various cancer and endothelial cell lines. The cytoskeleton (green) and nucleus (blue) of the cell were captured using 40X magnification. Magnification bar = 7 μm. Citation: Tangutoori S, Ohta A, Gatley S, Campbell RB (2014) Repurposing an Erstwhile Cancer Drug: A Quantitative and Therapeutic Evaluation of Alternative Nanosystems for the Delivery of Colchicine to Solid Tumors. J Cancer Sci Ther 6: 236-246. doi:10.4172/1948-5956.1000277 Volume 6(7) 236-246 (2014) 241 J Cancer Sci Ther ISSN:1948-5956 JCST, an open access journal profile. The drug is currently employed for the treatment of gouty arthritis [7,17], familial mediterranean fever [39] and other vasculardependent diseases [3,5,6]. The systemic administration of colchicine is not deemed clinically safe due to toxic effects exerted against various tissues. PCLs offer an attractive array of benefits to encapsulate and deliver the toxic drugs like colchicine, including preferential tumor vascular targeting [40]. The structural and chemical differences of various cationic lipids strongly influence physicochemical characteristics of PCLs such as particle size, zeta potential, loading efficiency of the drug and overall extent of liposome uptake [26,28]. Cationic lipids are uniquely identified by their acyl chain length, head group composition, degree of chain saturation, phase transition temperature (Tm) and the spatial arrangement of the head and tail groups in relation to the other lipids. These properties ultimately govern the stability and targeting efficiency of the eventual formulation [25,41]. The prerequisites for an ideal drug carrier for tumor vascular targeting are particle size (ranging from 100-200 nm), zeta potential (ranging from 20-25 mv) and the capacity for incorporating and delivering the drug payload to desired locations [41]. For any PCL another important consideration is to strike the right balance between the cationic surface charge, and partial shielding of the cationic charge potential afforded by PEG [42]. Therefore, to maximize vascular targeting efficiency we evaluated PCLs as a function of cationic lipid content and type, varied degree of saturation and unsaturation, considered the amount of PEG to employ, the loading efficiency and drug to lipid ratios [25,27,38,40,43,44]. We achieved a loading efficiency of ~80% of the initial drug concentration, the highest degree of loading reported for colchicine in nanoparticle platforms to date. The degree of incorporation is highly dependent on methodology and is lipid-composition dependent [45A (C ha go -k -1 )-C yt os ke le to n ar ea /c el l (L LC )-C yt os ke le to n ar ea /c el l (M C A -2 05 )-C yt os ke le to n ar ea /c el l (M C A -2 05 )-N uc le ar a re a/ ce ll (L LC )-N uc le ar a re a/ ce ll (C ha go -k -1 )-N uc le ar a re a/ ce ll 4000

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

منابع مشابه

Repurposing of drug candidates for treatment of skin cancer

Skin cancers are highly prevalent malignancies that affect millions of people worldwide. These include melanomas and nonmelanoma skin cancers. Melanomas are among the most dangerous cancers, while nonmelanoma skin cancers generally exhibit a more benign clinical pattern; however, they may sometimes be aggressive and metastatic. Melanomas typically appear in body regions exposed to the sun, alt...

متن کامل

Nanotechnology application in cancer treatment

Chemotherapy has been the main known treatment for cancer diseases. However, its achievement rate remains low, mainly because of the restricted accessibility of drugs to the tumor tissue, their painful toxicity, and development of multi-drug resistance. In recent years, either better understanding of tumor biology or development of the ever-growing field of nanotechnology has proposed new treat...

متن کامل

State of the art of stimuli-responsive liposomes for cancer therapy

Specific delivery of therapeutic agents to solid tumors and their bioavailability at the target site are the most clinically important and challenging goals in cancer therapy. Liposomes are promising nanocarriers and have been well investigated for cancer therapy. In spite of preferred accumulation in tumors via the enhanced permeability and retention (EPR) effect, inefficient drug release at t...

متن کامل

Nanomedicine – The role of newer drug delivery technologies in cancer

Nanotechnology has slowly but steadily revolutionized the diagnosis, imaging and treatment of cancer. Detecting cancer at earliest stages, locating the tumor at different areas in the body and specific delivery of the drugs to malignant cells including surgically inaccessible tumors are the core areas of medical and pharmaceutical research across the world. In this endeavour, Nanodevices have e...

متن کامل

State of the art of stimuli-responsive liposomes for cancer therapy

Specific delivery of therapeutic agents to solid tumors and their bioavailability at the target site are the most clinically important and challenging goals in cancer therapy. Liposomes are promising nanocarriers and have been well investigated for cancer therapy. In spite of preferred accumulation in tumors via the enhanced permeability and retention (EPR) effect, inefficient drug release at t...

متن کامل

ذخیره در منابع من


  با ذخیره ی این منبع در منابع من، دسترسی به آن را برای استفاده های بعدی آسان تر کنید

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

عنوان ژورنال:

دوره   شماره 

صفحات  -

تاریخ انتشار 2014