Mechanism of cellular uptake and cytotoxicity of paclitaxel loaded lipid nanocapsules in breast cancer cells (original) (raw)
Related papers
Dexamethasone simulates the anticancer effect of nano-formulated paclitaxel in breast cancer cells
Bioorganic Chemistry, 2020
Although the chemosensitizing effect of Dexamethasone (DEX) and its ability to increase the sensitivity of breast cancer cells to chemotherapy were previously reported, this study aimed to explore how far cotreatment of breast cancer cells with paclitaxel (PTX) and DEX mimics the anticancer effect of nanoformulated PTX. To establish this goal, PTX was nanoformulated with poly (lactic-co-glycolic acid) (PLGA) and the nanoparticles (PTX-NPs) were physically authenticated. Breast cancer cells (MCF-7) were treated with PTX or PTX-NPs in presence or absence of low concentration (10 nM) of DEX. Cells viability (assessed by MTT assay), apoptosis (assessed by flow cytometry) and the expression of PTX resistance gene (TRX1) and PTX metabolizing genes (CYP2C8 and CYP3A4) were investigated. The results showed that nanoformulated PTX was validated by nanosize assessment, increased the anionic surface charge and prober conjugation with the biodegradable carrier (PLGA), as indicated by the FTIR spectroscopy. Initially, the IC 50 value of PTX was 19.3 μg/ml and cotreatment with DEX minimized it to 5.22 μg/ml, whereas PTX-NPs alone inhibited cell proliferation with IC 50 6.67 μg/ml. Also, in presence of DEX, PTX-NPs further decreased the IC 50 to 5 μg/ml. In parallel, DEX has increased the responsiveness of cells to PTX without potentiating its apoptotic effect. Moreover, the glucocorticoid (with PTX or PTX-NPs) downregulated TXR1 gene by 26% (P < 0.01) and 28.4% (P < 0.05) respectively. Similarly, the mRNA level of CYP3A4 significantly decreased in presence of DEX. The main PTX metabolizing gene CYP2C8, in contrast, was upregulated, especially in cells cotreated with PTX/DEX (P < 0.001). Conclusively, the study reports that cotreatment of breast cancer cells with submolar concentration of DEX acts as similar as the nanoformulated PTX, possibly through its modulatory effects on the expression of the main PTX metabolizing gene (CYP2C8) and downregulating Taxol resistance gene.
Efficacy and In Vitro Cytotoxicity of Nanostructured Lipid Carriers for Paclitaxel Delivery
Paclitaxel (PTX) is an anticancer drug having poor aqueous solubility and low bioavailability. Formulation of PTX into Nanostructure lipid carriers (NLC) could be a potential way to enhance PTX aqueous solubility and bioavailability hence increases efficacy and decreases side effects. Eight PTX-NLC formulae were prepared using homogenization-ultrasonication technique. Characterization of the nanoparticles was done by transmission electron microscopy and by measurement of particle size, poly dispersibility index and zeta potential. Encapsulation efficiency, drug loading, and In Vitro release were measured. Particle size ranged between 172.8 ± 0.8 to 378.2 ± 1.8 nm and zeta potential between-18.6 ± 0.4 to-28.1 ± 1.2 mV. High EE and DL were obtained due to incorporation of liquid lipid and the In Vitro release showed prolonged time dependent release compared to Taxol ®. NLC-3 had the best results among the eight prepared formulae. In Vitro cytotoxicity of NLC-3 was evaluated on MCF-7 cell line and compared to pure PTX powder and Taxol ®. These findings show that NLC is a potential carrier to improve efficacy and enhance PTX delivery.
International Journal of Nanomedicine, 2018
Background: Successfully overcoming obstacles due to anticancer drugs' toxicity and achieving effective treatment using unique nanotechnology is challenging. The complex nature of breast tumors is mainly due to chemoresistance. Successful docetaxel (DTX) delivery by nanoparticles (NPs) through inhibition of multidrug resistance (MDR) can be a bridge to enhance intracellular dose and achieve higher cytotoxicity for cancer cells. Purpose: This study tested primary patient breast cancer cells in vitro with traditional free DTX in comparison with polymeric nanocarriers based on poly lactic co-glycolic acid (PLGA) NPs. Materials and methods: Establishment of primary cell line from breast malignant tumor depends on enzymatic digestion. Designed DTX-loaded PLGA NPs were prepared with a solvent evaporation method; one design was supported by the use of folic acid (FA) conjugated to PLGA. The physical properties of NPs were characterized as size, charge potential, surface morphology, DTX loading, and encapsulation efficiency. In vitro cellular uptake of fluorescent NPs was examined visually with confocal fluorescence microscopy and quantitatively with flow cytometry. In vitro cytotoxicity of all DTX designed NPs against cancer cells was investigated with MTT assay. RT-PCR measurements were done to examine the expression of chemoresistant and apoptotic genes of the tested DTX NPs. Results: Cellular uptake of DTX was time dependent and reached the maximum after loading on PLGA NPs and with FA incorporation, which activated the endocytosis mechanism. MTT assay revealed significant higher cytotoxicity of DTX-loaded FA/PLGA NPs with higher reduction of IC50 (8.29 nM). In addition, PLGA NPs, especially FA incorporated, limited DTX efflux by reducing expression of ABCG2 (3.2-fold) and MDR1 (2.86-fold), which were highly activated by free DTX. DTX-loaded FA/PLGA NPs showed the highest apoptotic effect through the activation of Caspase-9, Caspase-3, and TP53 genes by 2.8-, 1.6-, and 1.86-fold, respectively. Conclusion: FA/PLGA NPs could be a hopeful drug delivery system for DTX in breast cancer treatment.
Biological response and cytotoxicity induced by lipid nanocapsules
Journal of Nanobiotechnology
Background Lipid nanocapsules (LNCs) are promising vehicles for drug delivery. However, since not much was known about cellular toxicity of these nanoparticles in themselves, we have here investigated the mechanisms involved in LNC-induced intoxication of the three breast cancer cell lines MCF-7, MDA-MD-231 and MDA-MB-468. The LNCs used were made of Labrafac™ Lipophile WL1349, Lipoid® S75 and Solutol® HS15. Results High resolution SIM microscopy showed that the DiD-labeled LNCs ended up in lysosomes close to the membrane. Empty LNCs, i.e. without encapsulated drug, induced not only increased lysosomal pH, but also acidification of the cytosol and a rapid inhibition of protein synthesis. The cytotoxicity of the LNCs were measured for up to 72 h of incubation using the MTT assay and ATP measurements in all three cell lines, and revealed that MDA-MB-468 was the most sensitive cell line and MCF-7 the least sensitive cell line to these LNCs. The LNCs induced generation of reactive free o...
Drug Development and Industrial Pharmacy, 2015
The aim of this study was to fabricate docetaxel loaded nanocapsules (DTX-NCs) with a high payload using Layer-by-Layer (LbL) technique by successive coating with alternate layers of oppositely charged polyelectrolytes. Developed nanocapsules (NCs) were characterized in terms of morphology, particle size distribution, zeta potential (z-potential), entrapment efficiency and in vitro release. The morphological characteristics of the NCs were assessed using transmission electron microscopy (TEM) that revealed coating of polyelectrolytes around the surface of particles. The developed NCs successfully attained a submicron particle size while the z-potential of optimized NCs alternated between (+) 34.64 ± 1.5 mV to (À) 33.25 ± 2.1 mV with each coating step. The non-hemolytic potential of the NCs indicated the suitability of the developed formulation for intravenous administration. A comparative study indicated that the cytotoxicity of positively charged NCs (F4) was significant higher (p50.05) rather than negative charged NCs (F3), plain drug (DTX) and marketed preparation (Taxotere Õ ) when evaluated in vitro on MCF-7 cells. Furthermore, cell uptake studies evidenced a higher uptake of positive NCs (!1.2 fold) in comparison to negative NCs. In conclusion, formulated NCs are an ideal vehicle for passive targeting of drugs to tumor cells that may result in improved efficacy and reduced toxicity of encapsulated drug moiety.
Studies in Medical Sciences
Background & Aims: Owing to its anti-cancer and anti-oxidant properties, Kaempferol (KAE) has become an ideal candidate to be more welcome into clinical practice. However, Due to its low water solubility and bioavailability, we aimed to design and address a new liposomal formulation with KAE and evaluate its anti-cancer activity against MDA-MB 468 breast cancer cells. Materials & Methods: To characterize the physicochemical features, pharmaceutical parameters such as nanoparticle size, morphology of particles under scanning electron microscopy (SEM), and zeta potential were measured. The optimum liposomal formulation along with paclitaxel was incubated to investigate their biological activity against breast cancer cells. Furthermore, molecular mechanisms related to program cell death (apoptosis) and their gene expression were measured by flowcytometric and real-time PCR, respectively. Results: SEM images showed narrow distributed and scattered particles with the size of 80.3 nm (KAE) formulated in liposomes. IC50 values for KAE and paclitaxel were determined to be as 44 ± 0.52 μM and 1.75 ± 0.36 nM, respectively. Cell proliferation averaged from 44 ± 3.9% to 56 ± 26.8% (p <0.05) after treatment with KAE-loaded liposomes. Co-administration of nanoparticles containing KAE and paclitaxel in cancer cells significantly increased the percentage of apoptosis (P <0.05). Conclusion: Taking our data into consideration, we suggest that insertion of KAE into liposomal carriers not only improved the bioavailability of this flavonoid but also surged the anti-cancer efficacy of paclitaxel.
The aim of this study was to investigate the cytotoxicity of paclitaxel solid lipid nanoparticles (SLN) modified with stearic acid octaarginine (SA-R 8 -PTX-SLN) as well as the cellular uptake of coumarin-6-loaded SLN modified with SA-R 8 (SA-R 8 -C 6 -SLN) in human lung cancer cells, A549. SLN were prepared using a film dispersion method; and then their particle size, zeta potential, morphology, bound efficiency of SA-R 8 , drug loading efficiency, and in vitro release were characterized. SA-R 8 -PTX-SLN and SA-R 8 -C 6 -SLN were incubated with A549 cells to measure their cytotoxicity and cellular uptake, respectively. The results indicated that the cytotoxicity of SA-R 8 -PTX-SLN was enhanced significantly with the increasing amount of SA-R 8 and the cellular uptakes of SLN increased with the incubated concentrations and the incubated time of SLN. In contrast, SA-R 8 -SLN could significantly enhance the cellular uptake of SLN and the cytotoxicity of PTX in A549 cells. These in vitro results suggest that SA-R 8 -SLN could be proposed as alternative drug delivery system.
Materials Science and Engineering: C, 2018
Adenocarcinoma is the most lethal gynecologic tumor and treatment usually consists in surgery followed by chemotherapy. However, the chemotherapy benefits are eventually limited due to drug toxicity to normal tissues and cells, which cause several and harsh side effects. Paclitaxel (PCX) is the drug of first choice for ovarian cancer treatment, but it has low aqueous solubility, which reduces its bioavailability. Thus, in the commercial drug, Taxol ® , PCX is solubilized in a mixture of toxic surfactants. The development of drug nanocarriers has been investigated to promote the reduction of toxic effects and increase the safety and therapeutic efficacy of PCX. The aim of this work was the development and characterization of PCX loaded nanoparticles (PNPCX) and evaluation of in vitro efficacy of developed system using adenocarcinoma cell line. The nanocarrier was successfully obtained using nanoprecipitation technique. The results showed that the PNPCX-A had a particle size distribution around 140 nm and polydispersity index smaller than 0.1, with high PCX encapsulation efficiency. The results obtained were suitable for the intravenous administration route and promotion of passive targeting in the tumor microenvironment. The in vitro cytotoxicity assays of SKOV-3 cell line demonstrated that PNPCX-A was able to release PCX and reduce cell viability. The flow cytometry assays first reported that a nanostructured system with such composition (PNPCX-A) presented a time dependent cellular uptake, showing the ability of nanocarrier to be internalized. PNPCX-A present a distinguish potential for ovarian cancer therapy optimization. In vivo studies are needed to confirm the in vitro results and provide additional data regarding safety and efficacy of ovarian cancer treatment.
Effectiveness of liposomal paclitaxel against MCF-7 breast cancer cells
Canadian journal of physiology and pharmacology, 2010
Paclitaxel is an effective chemotherapeutic agent that is widely used for the treatment of several cancers, including breast, ovarian, and non-small-cell lung cancer. Due to its high lipophilicity, paclitaxel is difficult to administer and requires solubilization with Cremophor EL (polyethoxylated castor oil) and ethanol, which often lead to adverse side effects, including life-threatening anaphylaxis. Incorporation of paclitaxel in dimyristoylphosphatidylcholine:dimyristoylphosphatidylglycerol (DPPC:DMPG) liposomes can facilitate its delivery to cancer cells and eliminate the adverse reactions associated with the Cremophor EL vehicle. Accordingly, the effectiveness of liposomal paclitaxel on MCF-7 breast cancer cells was examined. The results from this study showed that (i) the lipid components of the liposomal formulation were nontoxic, (ii) the cytotoxic effects of liposomal paclitaxel were improved when compared with those seen with conventional paclitaxel, and (iii) the intracellular paclitaxel levels were higher in MCF-7 cells treated with the liposomal paclitaxel formulation. The results of these studies showed that delivery of paclitaxel as a liposomal formulation could be a promising strategy for enhancing its chemotherapeutic effects.
Pharmaceutics, 2020
Background: Paclitaxel (PTX) is a widely used anti-cancer drug for treating various types of solid malignant tumors including breast, ovarian and lung cancers. However, PTX has a low therapeutic response and is linked with acquired resistance, as well as a high incidence of adverse events, such as allergic reactions, neurotoxicity and myelosuppression. The situation is compounded when its complex chemical structure contributes towards hydrophobicity, shortening its circulation time in blood, causing off-target effects and limiting its therapeutic activity against cancer cells. Formulating a smart nano-carrier may overcome the solubility and toxicity issues of the drug and enable its more selective delivery to the cancerous cells. Among the nano-carriers, natural polymers are of great importance due to their excellent biodegradability, non-toxicity and good accessibility. The aim of the present research is to develop self-assembled sodium caseinate nanomicelles (NaCNs) with PTX loaded into the hydrophobic core of NaCNs for effective uptake of the drug in cancer cells and its subsequent intracellular release. Methods: The PTX-loaded micelle was characterized with high-performance liquid chromatography (HPLC), Fourier Transform Infrared Spectra (FTIR), High Resolution-Transmission Electron Microscope (HR-TEM), Field Emission Scanning Electron Microscope (FESEM) and Energy Dispersive X-Ray (EDX). Following treatment with PTX-loaded NaCNs, cell viability, cellular uptake and morphological changes were analyzed using MCF-7 and MDA-MB 231 human breast cancer cell lines. Results: We found that PTX-loaded NaCNs efficiently released PTX in an acidic tumor environment, while showing an enhanced cytotoxicity, cellular uptake and in-vivo anti-tumor efficacy in a mouse model of breast cancer when compared to free drug and blank micelles. Additionally, the nanomicelles also presented improved colloidal stability for three months at 4 • C and −20 • C and when placed at a temperature of 37 • C. Conclusions: We conclude that the newly developed NaCNs is a promising carrier of PTX to enhance tumor accumulation of the drug while addressing its toxicity issues as well.