Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
AAPS PharmSciTech ; 21(8): 313, 2020 Nov 09.
Article in English | MEDLINE | ID: mdl-33165766

ABSTRACT

A solid self-emulsifying drug delivery system (SEDDS) of paclitaxel (PTX) was developed that could enhance its oral bioavailability and neutralize other niggles associated with conventional delivery systems of PTX. TPGS-centered SEDDS containing PTX was optimized by Box-Behnken experimental design and then formulated as fumed colloidal silica-based solid SEDDS microparticles (Si-PTX-S-SEDDS). AFM analysis exhibited round-shaped microparticles of approximately 2-3 µM diameter, whereas after reconstitution, particle size measurement showed nanoemulsion droplets of 30.00 ± 2.00 nm with a zeta potential of 17.38 ± 2.88 mV. Si-PTX-S-SEDDS displayed improved efficacy proven by reduced IC50 of 0.19 ± 0.03 µM against MDA-MB-231 cells and a 45.83-fold higher cellular uptake in comparison to free PTX. Molecular mechanistic studies showed mitochondria-mediated intrinsic pathway of apoptosis following Akt/mTOR pathway, which is accompanied by survivin downregulation. Rhodamine 123 assay and chylomicron flow blocking studies revealed P-gp inhibition potential and lymphatic uptake of Si-PTX-S-SEDDS, responsible for over 4-fold increment in oral bioavailability compared to PTX administered as Taxol. In vivo anti-tumor studies in syngeneic mammary tumor model in SD rats revealed higher efficacy of Si-PTX-S-SEDDS as evident from significant reduction in tumor burden. In total, the developed Si-PTX-S-SEDDS formulation was found as an appropriate option for oral delivery of PTX.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Colloids/chemistry , Mammary Neoplasms, Animal/drug therapy , Paclitaxel/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Silicon Dioxide/chemistry , TOR Serine-Threonine Kinases/metabolism , Vitamin E/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Biological Availability , Cell Line, Tumor , Drug Delivery Systems , Emulsions/pharmacology , Humans , Paclitaxel/chemistry , Rats , Rats, Sprague-Dawley , Research Design
2.
Mol Pharm ; 16(9): 3744-3759, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31441308

ABSTRACT

We have devised a nanocarrier using "tocopheryl polyethylene glycol succinate (TPGS) conjugated to triphenylphosphonium cation" (TPP-TPGS) for improving the efficacy of doxorubicin hydrochloride (DOX). Triphenylphosphonium cation (TPP) has affinity for an elevated transmembrane potential gradient (mitochondrial), which is usually high in cancer cells. Consequently, when tested in molecular docking and cytotoxicity assays, TPP-TPGS, owing to its structural similarity to mitochondrially directed anticancer compounds of the "tocopheryl succinate" family, interferes specifically in mitochondrial CII enzyme activity, increases intracellular oxidative stress, and induces apoptosis in breast cancer cells. DOX loaded nanocarrier (DTPP-TPGS) constructed using TPP-TPGS was positively charged, spherical in shape, sized below 100 nm, and had its drug content distributed evenly. DTPP-TPGS offers greater intracellular drug delivery due to its rapid endocytosis and subsequent endosomal escape. DTPP-TPGS also efficiently inhibits efflux transporter P glycoprotein (PgP), which, along with greater cell uptake and inherent cytotoxic activity of the construction material (TPP-TPGS), cumulatively results in 3-fold increment in anticancer activity of DOX in resistant breast cancer cells as well as greater induction of necroapoptosis and arrest in all phases of the cell cycle. DTPP-TPGS after intravenous administration in Balb/C mice with breast cancer accumulates preferentially in tumor tissue, which produces significantly greater antitumor activity when compared to DOX solution. Toxicity evaluation was also performed to confirm the safety of this formulation. Overall TPP-TPGS is a promising candidate for delivery of DOX.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/drug effects , Mitochondria/metabolism , Vitamin E/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Disease Models, Animal , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/pharmacokinetics , Female , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Reactive Oxygen Species/metabolism , Tissue Distribution , Vitamin E/pharmacokinetics
3.
Int J Biol Macromol ; 122: 1100-1114, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30219515

ABSTRACT

Immunotherapeutic nanoparticles (NPs) could be a viable option for delivering cytotoxic agents in a manner which suppresses their toxic manifestations. Doxorubicin (DOX) loaded NPs were prepared using fucoidan (FCD), an immunomodulatory polysaccharide and evaluated against cancer. FCD was electrostatically assembled with cationic polyethylenimine (PEI) through intermolecular electrostatic interactions to develop an immunomodulatory platform to deliver DOX. FCD NPs offered improved cytotoxicity (2.64 folds), cell cycle arrest in G1-S phase (34.65%) and apoptosis (66.12%) in tumor cells compared to free DOX. The enhanced apoptosis was due to raised mitochondrial depolarization (88.00%). In vivo anticancer activity in 4T1 induced tumor bearing BALB/c mice demonstrated a 2.95 folds enhanced efficacy of NPs. Importantly, NPs treatment generated an immunotherapeutic response indicated by gradual increment of the plasma IL-12 levels and reversed polarization of tumor associated macrophages (TAMs) towards M1 subtype. Furthermore, pharmacokinetic study suggested that NPs administration in tumor infested mice caused serum DOX levels to vary in a biphasic pattern, with twin peaks occurring at 1 h and 6 h which help in maintaining preferential drug localization in tumor. Developed NPs would be an excellent approach for improved immune-chemotherapy (in terms of efficacy, safety and immunocompetency) against cancer.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin/chemistry , Doxorubicin/pharmacology , Immunologic Factors/pharmacology , Nanoparticles/chemistry , Polysaccharides/pharmacology , Static Electricity , Animals , Apoptosis/drug effects , Biological Transport , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Caspase 1/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/metabolism , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Synergism , G1 Phase/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , S Phase/drug effects , Tissue Distribution/drug effects
4.
Artif Cells Nanomed Biotechnol ; 46(sup3): S344-S358, 2018.
Article in English | MEDLINE | ID: mdl-30282466

ABSTRACT

Paclitaxel (PTX) in its commercial products exhibits adverse effects owing to excipients and also has poor oral bioavailability. Present work is directed towards development of tocopheryl polyethylene glycol succinate-assisted self-nanoemulsifying system (SEDDS) for oral delivery of PTX. Box-Behnken design of experiment was employed to optimize PTX-SEDDS and was characterized for droplet size (29.76 ± 2.64 nm), zeta potential (-21.46 ± 2.52 mV), PDI (0.177 ± 0.012), drug content (4.97 ± 0.98 mg), entrapment efficiency (98.33 ± 0.54%) and in vitro drug release (51.03 ± 2.23% PTX at 72 h). PTX-SEDDS exhibited IC50; 1.58 ± 0.12 µM and a 52.46-folds higher cell uptake in MDA-MB-231 cells along with cellular and nuclear morphology changes. Significantly higher G2M cell cycle arrest, apoptosis, mitochondrial membrane potential disruption and ROS production was exhibited by PTX-SEDDS in comparison to Taxol. Up-regulation of Bax, p21, cleaved-caspase 3, -caspase 9 and down-regulation of Bcl2 and survivin suggested apoptosis via intrinsic pathways. Pharmacokinetic study showed approximately 4-folds higher oral bioavailability of PTX-SEDDS than Taxol. Significant reduction in tumour volume and weight was observed in syngeneic mammary tumour in SD rats. Tumour histopathology and TUNEL assay showed apoptosis in tumour tissue. PTX-SEDDS caused low lung metastasis, and was safe and stable. Conclusively, PTX-SEDDS could be suitable option for oral delivery of PTX.


Subject(s)
Apoptosis/drug effects , Drug Delivery Systems , Mammary Neoplasms, Experimental , Micelles , Paclitaxel , Vitamin E , Animals , Emulsions , Female , Humans , Isografts , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Neoplasm Transplantation , Paclitaxel/chemistry , Paclitaxel/pharmacology , Rats , Rats, Sprague-Dawley , Vitamin E/chemistry , Vitamin E/pharmacology
5.
Pharm Res ; 35(3): 60, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29427248

ABSTRACT

PURPOSE: To fabricate, characterize and evaluate 3-O-sn-Phosphatidyl-L-serine (PhoS) anchored PLGA nanoparticles for macrophage targeted therapeutic intervention of VL. MATERIALS AND METHODS: PLGA-AmpB NPs were prepared by well-established nanoprecipitation method and decorated with Phos by thin film hydration method. Physico-chemical characterization of the formulation was done by Zetasizer nano ZS and atomic force microscopy. RESULTS: The optimized formulation (particle size, 157.3 ± 4.64 nm; zeta potential, - 42.51 ± 2.11 mV; encapsulation efficiency, ∼98%) showed initial rapid release up to 8 h followed by sustained release until 72 h. PhoS generated 'eat-me' signal driven augmented macrophage uptake, significant increase in in-vitro (with ∼82% parasite inhibition) and in-vivo antileishmanial activity with preferential accumulation in macrophage rich organs liver and spleen were found. Excellent hemo-compatibility justified safety profile of developed formulation in comparison to commercial formulations. CONCLUSION: The developed PhoS-PLGA-AmpB NPs have improved efficacy, and necessary stability which promisingly put itself as a better alternative to available commercial formulations for optimized treatment of VL.


Subject(s)
Amphotericin B/administration & dosage , Antiprotozoal Agents/administration & dosage , Drug Carriers/chemistry , Leishmaniasis, Visceral/drug therapy , Macrophages/drug effects , Animals , Cell Line , Delayed-Action Preparations/administration & dosage , Disease Models, Animal , Drug Compounding/methods , Drug Evaluation, Preclinical , Drug Stability , Humans , Leishmania donovani/drug effects , Macrophages/parasitology , Male , Mice , Nanoparticles/chemistry , Phosphatidylserines/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rats , Rats, Wistar , Treatment Outcome
6.
Int J Biol Macromol ; 105(Pt 1): 1220-1231, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28780414

ABSTRACT

The goal of study was to develop micellar formulation of Amphotericin B (AmB) to improve its antileishmanial efficacy. AmB loaded pluronic F127 (PF 127) micelles were developed and coated with chitosan (Cs-PF-AmB-M) to accord immunoadjuvant and macrophage targeting properties. Hemolysis and cytotoxicity studies demonstrated that Cs-PF-AmB-M was 7.93 fold (at 20µg/ml AmB concentration) and 9.35 fold less hemolytic and cytotoxic, respectively in comparison to AmB suspension. Flow cytometry studies indicated that Cs-PF-FITC-M was 21.97 fold higher internalized byJ774A.1 macrophage in comparison to PF-FITC-M.Cs-PF-AmB-M showed excellent in-vitro (1.82 fold in compared to AmB suspension) and in-vivo (75.84±7.91% parasitic inhibition) antileishmanial activity against macrophage resident intracellular promastigotes and Leishmania donovani infected Syrian hamsters, respectively. Chitosan coating stimulated a Th1 immune response mediating auxiliary immunotherapeutic action as judged by in-vitro and in-vivo cytokine and mRNA expression. Toxicity studies demonstrated normal blood urea nitrogen (BUN) and plasma creatinine (PC) level and no sign of abnormal histopathology upon intravenous administration of micellar formulations. Pharmacokinetic profiling and tissue distribution studies indicated that AmB was preferentially localized in macrophage harboring tissue instead of kidney, thereby circumventing the characteristic nephrotoxicity. Conclusively, Cs-PF-AmB-M could be a viable alternative for the current immuno and chemotherapy of visceral leishmaniasis (VL).


Subject(s)
Amphotericin B/chemistry , Amphotericin B/pharmacology , Chitosan/chemistry , Drug Carriers/chemistry , Leishmaniasis, Visceral/drug therapy , Micelles , Poloxamer/chemistry , Amphotericin B/pharmacokinetics , Amphotericin B/therapeutic use , Animals , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacokinetics , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Cell Line , Cricetinae , Cytokines/metabolism , Drug Carriers/toxicity , Drug Compounding , Female , Humans , Leishmania donovani/drug effects , Leishmania donovani/physiology , Macrophages/drug effects , Mice , Tissue Distribution
7.
Pharm Res ; 34(9): 1857-1871, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28608139

ABSTRACT

OBJECTIVE: To utilize nanoparticles produced by condensation of zymosan (an immunotherapeutic polysaccharide) with pegylated polyethylenimine (PEG-PEI) for dual intervention in breast cancer by modulating tumor microenvironment and direct chemotherapy. METHOD: Positively charged PEG-PEI and negatively charged sulphated zymosan were utilized for electrostatic complexation of chemoimmunotherapeutic nanoparticles (ChiNPs). ChiNPs were loaded with doxorubicin hydrochloride (DOX) for improved delivery at tumor site and were tested for in-vivo tolerability. Biodistribution studies were conducted to showcase their effective accumulation in tumor hypoxic regions where tumor associated macrophages (TAMs) are preferentially recruited. RESULTS: ChiNPs modulated TAMs differentiation resulting in decrement of CD206 positive population. This immunotherapeutic action was furnished by enhanced expression of Th1 specific cytokines. ChiNPs also facilitated an anti-angiogenetic effect which further reduces the possibility of tumor progression and metastasis.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Immunologic Factors/therapeutic use , Nanoparticles/chemistry , Zymosan/therapeutic use , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Breast/drug effects , Breast/immunology , Breast/pathology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cytokines/immunology , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Delivery Systems , Female , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacokinetics , Macrophages/drug effects , Macrophages/immunology , Mice, Inbred BALB C , Polyethyleneimine/chemistry , Static Electricity , Tissue Distribution , Zymosan/administration & dosage , Zymosan/pharmacokinetics
8.
Eur J Pharm Biopharm ; 113: 198-210, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28088632

ABSTRACT

OBJECTIVE: We explore a plausible method of targeting bendamustine hydrochloride (BM) to circulatory monocytes by exploiting their intrinsic endocytic/phagocytic capability. METHODS: We do so by complexation of sodium alginate and chitosan inside dioctyl sulfo succinate sodium (AOT) reverse micelles to form bendamustine hydrochloride loaded nanoparticles (CANPs). Dynamic light scattering, electrophoretic mobility and UV spectroscopy were used to detail intra-micellar complexation dynamics and to prove that drug was co-captured during interaction of carbohydrate polymers. A fluorescent conjugate of drug (RBM) was used to trace its intracellular fate after its loading into nanoparticles. RESULTS: CANPs were sized below 150nm, had 75% drug entrapment and negative zeta potential (-30mV). Confocal microscopy demonstrated that developed chitosan alginate nanoparticles had the unique capability to carry BM specifically to its site of action. Quantitative and mechanism based cell uptake studies revealed that monocytes had voracious capacity to internalize CANPs via simultaneous scavenger receptor based endocytic and phagocytic mechanism. Comparative in vitro pharmacokinetic studies revealed obtainment of significantly greater intracellular drug levels when cells were treated with CANPs. This caused reduction in IC50 (22.5±2.1µg/mL), enhancement in G2M cell cycle arrest, greater intracellular reactive oxygen species generation, and increased apopotic potential of bendamustine hydrochloride in THP-1 cells. CONCLUSION: Selective monocytic targeting of bendamustine hydrochloride using carbohydrate constructs can prove advantageous in case of leukemic disorders displaying overabundance of such cells.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Bendamustine Hydrochloride/administration & dosage , Drug Carriers , Micelles , Nanoparticles , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Bendamustine Hydrochloride/pharmacology , Cell Cycle/drug effects , Cell Line, Transformed , Endocytosis/drug effects , Fluorescent Dyes , Humans , Microscopy, Electron, Transmission , Phagocytosis/drug effects , Reactive Oxygen Species/metabolism
9.
J Chromatogr Sci ; 55(1): 30-39, 2017 01.
Article in English | MEDLINE | ID: mdl-27993861

ABSTRACT

Ion pair chromatography was used for quantifying bendamustine hydrochloride (BH) in its marketed vial. The permissive objective was to investigate time duration for which highly susceptible drug content of the marketed vial remained stable after reconstitution. However, the method could also be used to measure extremely low levels of drug in rat plasma and a pharmacokinetic study was accordingly conducted to further showcase method's applicability. Optimized separation was achieved on C-18 Purospher®STAR (250 mm × 4.6 mm, 5 µm particle size) column. Mobile phase flowing at 1.5 mL/min consisted of 5 mM sodium salt of octane sulfonic acid dissolved in methanol, water and glacial acetic acid (55:45:0.075) maintained at pH 6. Detection was carried out at 233 nm with BH eluting after 7.8 min. Validation parameters were determined as per ICH guidelines. Limit of detection and limit of quantification were found to be 0.1 µg/mL and 0.33 µg/mL, respectively. The recoveries were 98-102% in bulk and 85-91% in plasma. The developed method was specific for BH, and utilized for assessing its short-term stability in physiologic solvents and forced degradation products in acid, base, oxidative, light and temperature induced stress environments.


Subject(s)
Bendamustine Hydrochloride/analysis , Bendamustine Hydrochloride/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Chromatography, Reverse-Phase/methods , Animals , Bendamustine Hydrochloride/chemistry , Drug Stability , Female , Limit of Detection , Linear Models , Rats , Rats, Wistar , Reproducibility of Results
10.
J Nanosci Nanotechnol ; 16(4): 4180-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27451784

ABSTRACT

Although curcumin (Cur), has been poised to be an anticancer boon for quite some, its progress from bench to bed has been strained due to various pharmaceutical hurdles. Consequently curcumin has been entrapped in methoxy poly ethylene glycol and linoleic acid conjugated polymeric micelles (PMs) to not only tackle the routine issues but to also provide a synergetic effect against MCF-7 breast cancer cells. Optimized PMs of Cur had size 186.53 ± 12.10 nm with polydispersity index 0.143 ± 0.031 and zeta potential -30.1 ± 3.2 mV. Developed formulation (Mpeg-Cla-Cur PMs) was hemocompatible and had high cytotoxicity (IC50 55.80 ± 4.63 µ/mL) against MCF-7 cells in comparison to pure Cur suspension (IC50 75.05 ± 5.75 µg/mL). As postulated cell cycle arrest and apoptosis studies revealed synergetic effect of Mpeg-Cla-Cur PMs with higher cell population in G1 phase in addition to high apoptosis of MCF-7 cells as compared to pure Cur suspension and con- trol group. Pharmacokinetic studies also show PMs enhanced MRT and T1/2 of Cur indicating its longer retention time in body. Mpeg-Cla-Cur PMs might become as an excellent chemotherapeutic alternative candidate for treatment of breast cancer with higher commercial value.


Subject(s)
Apoptosis/drug effects , Cell Survival/drug effects , Curcumin/administration & dosage , Linoleic Acid/chemistry , Nanocapsules/chemistry , Polyethylene Glycols/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Curcumin/chemistry , Diffusion , Drug Synergism , Humans , MCF-7 Cells , Nanocapsules/administration & dosage
11.
Pharm Res ; 33(11): 2617-29, 2016 11.
Article in English | MEDLINE | ID: mdl-27401407

ABSTRACT

PURPOSE: To develop a biocompatible and bioresorbable calcium phosphate (CaP) nanoparticles (NPs) bearing Amphotericin B (AmB) with an aim to provide macrophage specific targeting in visceral leishmaniasis (VL). MATERIALS & METHODS: CaP-AmB-NPs were architectured through emulsion precipitation method. The developed formulation was extensively characterized for various parameters including in-vitro and in-vivo antileishmanial activity. Moreover, plasma pharmacokinetics, tissue biodistribution and toxicity profile were also assessed. RESULTS: Optimized CaP-AmB-NPs exhibited higher entrapment (71.1 ± 6.68%) of AmB. No trend related to higher hemolysis was apparent in the developed formulation as evidenced in commercially available colloidal and liposomal formulations. Cellular uptake of the developed CaP-AmB-NPs was quantified through flow cytometry in J774A.1 cell line, and a 23.90 fold rise in uptake was observed. Fluorescent microscopy also confirmed the time dependent rise in uptake. In-vivo multiple dose toxicity study demonstrated no toxicity upto 5 mg/kg dose of AmB. Plasma kinetics and tissue distribution studies established significantly higher concentration of AmB in group treated with CaP-AmB-NPs in liver and spleen as compared to CAmB, LAmB and AmB suspension group. In-vivo animal experimental results revealed that the CaP-AmB-NPs showed higher splenic parasite inhibition compared to CAmB and LAmB in leishmania parasite infected hamsters. CONCLUSIONS: The investigated CaP-AmB-NPs are effective in provoking macrophage mediated uptake and collectively features lower toxicity and offers a suitable replacement for available AmB-formulations for the obliteration of intra-macrophage VL parasite.


Subject(s)
Amphotericin B/administration & dosage , Antiprotozoal Agents/administration & dosage , Calcium Phosphates/chemistry , Drug Carriers/chemistry , Leishmaniasis, Visceral/drug therapy , Macrophages/drug effects , Nanoparticles/chemistry , Amphotericin B/pharmacokinetics , Animals , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacokinetics , Cell Line , Cricetinae , Drug Liberation , Emulsions , Erythrocytes/drug effects , Hemolysis , Leishmania donovani/drug effects , Leishmaniasis, Visceral/metabolism , Liver/drug effects , Liver/metabolism , Macrophages/metabolism , Male , Rats, Wistar , Spleen/drug effects , Spleen/metabolism , Tissue Distribution
12.
Drug Deliv ; 23(2): 479-88, 2016.
Article in English | MEDLINE | ID: mdl-24901205

ABSTRACT

CONTEXT: Nanoemulsions (NE) are one of the robust delivery tools for drugs due to their higher stability and efficacy. OBJECTIVES: The purpose of present investigation is to develop stable, effective and safe NE of docetaxel (DTX). METHODS: Soybean oil, lecithin, Pluronic F68, PEG 4000 and ethanol were employed as excipients and NEs were prepared by hot homogenization followed by ultra-sonication. NEs were optimized and investigated for different in vitro and in vivo parameters viz. droplet size, poly dispersity index, charge; zeta potential, drug content and in vitro drug release, in vitro cytotoxicity, in vitro cell uptake and acute toxicity. Transmission electron microscopy was performed to study morphology and structure of NEs. Stability studies of the optimized formulation were performed. RESULTS: Droplet size, poly dispersity index, zeta potential, drug content and in vitro drug release were found to be 233.23 ± 4.3 nm, 0.24 ± 0.010, -43.66 ± 1.9 mV, 96.76 ± 1.5%, 96.25 ± 2.1%, respectively. NE F11 exhibited higher cell uptake (2.83 times than control) and strong cytotoxic activity against MCF-7 cancer cells (IC50; 13.55 ± 0.21 µg/mL at 72 h) whereas no toxicity or necrosis was observed with liver and kidney tissues of mice at a dose of 20 mg/kg. Transmission electron microscopy ensured formation of poly-dispersed and spherical droplets in nanometer range. NE F11 (values indicated above) was selected as the optimized formulation based on the aforesaid parameters. CONCLUSION: Conclusively, stable, effective and safe NE was developed which might be used as an alternative DTX therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Drug Carriers , Nanoparticles , Taxoids/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival , Docetaxel , Dose-Response Relationship, Drug , Drug Compounding , Drug Stability , Emulsions , Ethanol/chemistry , Excipients/chemistry , Female , Hot Temperature , Humans , Inhibitory Concentration 50 , Lecithins/chemistry , MCF-7 Cells , Mice , Microscopy, Electron, Transmission , Nanotechnology , Particle Size , Poloxamer/chemistry , Polyethylene Glycols/chemistry , Solubility , Soybean Oil/chemistry , Surface Properties , Surface-Active Agents/chemistry , Taxoids/chemistry , Taxoids/metabolism , Taxoids/toxicity , Technology, Pharmaceutical/methods , Ultrasonics
13.
J Control Release ; 220(Pt A): 368-387, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26528900

ABSTRACT

The scope of RNAi based therapeutics is unquestionable. However, if we dissect the current trend of clinical trials for afore mentioned drug class, some stark trends appear: 1) naked siRNA only exerts influence in topical mode whilst systemic delivery requires a carrier and 2) even after two decades of extensive efforts, not even a single siRNA containing product is commercially available. It was therefore felt that a perspective simplifying the unique intricacies of working with a merger of siRNA and liposomes from a pharmaceutical viewpoint could draw the attention of a wider array of interested researchers. We begin from the beginning and attempt to conduit the gap between theoretical logic and experimental/actual constraints. This, in turn could stimulate the next generation of investigators, gearing them to tackle the conundrum, which is siRNA delivery.


Subject(s)
Gene Transfer Techniques , Lipids/chemistry , Nanoparticles , RNA Interference , RNA, Small Interfering/genetics , RNAi Therapeutics/methods , Animals , Endosomes/metabolism , Gene Expression Regulation , Humans , Lipids/immunology , Liposomes , RNA Stability , RNA, Small Interfering/chemistry , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism
14.
J Biomed Nanotechnol ; 11(10): 1747-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26502638

ABSTRACT

Docetaxel (DTX) is favoured option for breast cancer treatment; however its marketed formulation (Taxotere) generates therapeutic response at the cost of undue toxicity. In order to circumvent such limitations, DTX nanocrystals (DTX-NCs) were prepared through high pressure homogenization (HPH) technique using pluronic F-127 (PF-127) as a stabilizer. DTX-NCs presented higher efficacy against MCF-7 breast cancer cells with exposition of 1.75 and 2.13 fold lower inhibitory concentration (IC50) compared to free drug and Taxotere, respectively. DTX-NCs enhanced the DTX induce G2-M arrest by 1.24 and 1.79 fold compared to Taxotere and free DTX whereas highest apoptotic population (54.79%) of MCF-7 cells was also observed when cells were incubated with DTX-NCs for 24 h in comparison to free DTX (9.69%) and Taxotere (12.55%). The claims of improvement were substantiated by investigating the modulation in apoptotic mechanism induced by the subtle physical state variation of DTX in DTX-NCs. Results revealed that DTX-NCs induced apoptosis was linked to altered mitochondrial membrane potential. DTX-NCs caused highest (39.53%) depolarization of mitochondria compared to free DTX (9.34%) and Taxotere (18.72%). Further, safety of DTX-NCs was ascertained via haemolytic testing and in-vivo toxicity studies in mice. Developed formulation exhibited acceptable haemolytic potential which suggested its suitability towards parenteral administration. Moreover, in-vivo acute toxicity studies demonstrated that the developed NCs were safer than marketed Taxotere. These results elicit that DTX-NCs would be a viable alternative to commercial formulation for treatment of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Nanoparticles/administration & dosage , Poloxamer/chemistry , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Diffusion , Docetaxel , Drug Compounding/methods , Drug Stability , Drug Synergism , Excipients/chemistry , Female , Humans , MCF-7 Cells , Materials Testing , Membrane Potential, Mitochondrial/drug effects , Mice , Nanoparticles/chemistry , Nanoparticles/toxicity , Particle Size , Poloxamer/toxicity , Rats, Wistar , Taxoids/chemical synthesis , Treatment Outcome
15.
Int J Biol Macromol ; 79: 27-36, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25931395

ABSTRACT

Current leishmaniasis treatment is strangled due to concealed residence of parasite and reduced host cell mediated immune response. To circumvent above challenges, novel macrophage targeted oily core polymeric shell based doxorubicin (DOX) loaded nanocapsules (NCAPs) were fabricated employing chondroitin sulphate (CHD) for complimentary immunotherapy coupled chemotherapy against leishmaniasis. Excellent encapsulation efficiency along with pH dependent drug release was demonstrated by NCAPs. Improved cell cycle arrest at G1-S phase (1.56 folds) and apoptosis against promastigotes (6.26 folds), support the remarkable in-vitro antileishmanial activity of NCAPs (IC50: 0.254±0.038 µg/ml) compared to free DOX (IC50: 0.543±0.012 µg/ml). In-vivo antileishmanial activity in hamsters represented a significantly enhanced parasitic inhibition by NCAPs (1.42 folds). Improved activity was mediated via immunotherapeutic activity of NCAPs which up-regulated Th1 immune response (IL-12, INF-γ, and TNF-α) and down-regulated Th2 immune response (IL-4, IL-10, and TGF-ß). In conclusion, current novel nano-formulation could be a viable option against leishmaniasis.


Subject(s)
Antiprotozoal Agents/pharmacology , Chondroitin/chemistry , Doxorubicin/pharmacology , Immunity, Cellular/drug effects , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Animals , Apoptosis/drug effects , Cell Line , Cricetulus , Drug Compounding , Drug Liberation , G1 Phase Cell Cycle Checkpoints/drug effects , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Leishmania donovani/growth & development , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/parasitology , Nanocapsules , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/parasitology , Th1-Th2 Balance/drug effects , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/parasitology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
16.
Drug Dev Ind Pharm ; 41(11): 1759-68, 2015.
Article in English | MEDLINE | ID: mdl-25686725

ABSTRACT

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 (ζ-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 ζ-potential of optimized NCs alternated between (+) 34.64 ± 1.5 mV to (-) 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 (p < 0.05) rather than negative charged NCs (F3), plain drug (DTX) and marketed preparation (Taxotere®) 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.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Nanocapsules , Taxoids/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Chemistry, Pharmaceutical/methods , Docetaxel , Drug Compounding/methods , Drug Delivery Systems , Drug Liberation , Female , Humans , MCF-7 Cells , Microscopy, Electron, Transmission , Particle Size , Taxoids/pharmacokinetics , Taxoids/pharmacology
17.
Int J Pharm ; 478(2): 811-21, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25529434

ABSTRACT

Capecitabine (CAP) is an oral drug of choice for treatment of colorectal cancer. But its short plasma half-life limits clinical utility and the usually prescribed dosing regimen results in significant periods of therapeutically irrelevant concentration. To overcome this pharmacokinetic void a trichotomous gastroretentive (TRGDDS) system made up of CAP housed in xanthan gum microparticles (CXGMP) has been developed for extending CAP's gastric residence time thereby prolonging the subsequent elimination. TRGDDS was evaluated for particle size (243±25µm), surface morphology (porous) entrapment efficiency (87.72±7.31%), buoyancy (86.32±2.3%), mucoadhesiveness (88±4.3%), swelling index (80.37±4.65). X-ray diffraction (XRD) and differential scanning calorimetry (DSC) of CXGMP suggested CAP had been rendered amorphous, a property which unconventionally slows its dissolution. Significant control was offered by CXGMP compared to crystalline CAP in terms of drug release. Pharmacokinetic studies in Wistar rat further revealed that CXGMP increased the MRT (three times), elimination half-life (roughly 4 fold) and AUC (1.44 folds) of CAP at a dose of 5mg/kg in comparison to CAP solution of same strength. Conclusively the employment of TRGDDS had extended the duration for which CAP stayed in the rodent model, providing evidence for potentially obtaining a more efficacious dosing regimen in actual disease models.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Gastric Mucosa/metabolism , Adhesiveness , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/chemistry , Area Under Curve , Capecitabine , Deoxycytidine/blood , Deoxycytidine/chemistry , Deoxycytidine/pharmacokinetics , Excipients/chemistry , Fluorouracil/blood , Fluorouracil/chemistry , Fluorouracil/pharmacokinetics , Gastric Mucosa/chemistry , Goats , Half-Life , Polysaccharides, Bacterial/chemistry , Rats, Wistar , Solubility
18.
J Control Release ; 196: 295-306, 2014 Dec 28.
Article in English | MEDLINE | ID: mdl-25459427

ABSTRACT

Paclitaxel (PTX) is used as first line treatment for metastatic breast cancer but the relief comes at a heavy cost in terms of accompanying adverse effects. The pharmaceutical credentials of PTX are further dampened by the intrinsically low aqueous solubility. In order to sideline such insidious tendencies, PTX was incorporated in a vitamin E nanoemulsion using high pressure homogenization. The encapsulation efficiency of PTX in nanoemulsion was 97.81±2.7% and a sustained drug release profile was obtained. PTX loaded nanoemulsion exhibited higher cytotoxicity in breast cancer cell line (MCF-7) when compared to free PTX and marketed formulation (Taxol). Cell cycle arrest study depicted that MCF-7 cells treated with PTX loaded nanoemulsion showed high arrest in G2-M phase. Moreover blank nanoemulsion induced additional apoptosis in breast cancer cells through G1-S arrest by disrupting mitochondrial membrane potential. Cytokine estimation study in macrophages showed that both PTX loaded nanoemulsion and blank nanoemulsion enhanced secretion of IL-12 and downregulated secretion of IL-4 and IL-10. Results suggest that inclusion of vitamin E in nanoemulsion opened multiple complementary molecular effects which not only magnified the principle antiproliferative activity of PTX but also independently showcased potential in restoring the proactive nature of the breast cancer slackened chronic immune response. In-vivo anticancer activity showed significantly improved efficacy of PTX loaded nanoemlsion compare to Taxol and free PTX. The list of plausible advantages of PTX nanoemulsification was further substantiated by acceptable haemolytic potential, reduced in-vivo toxicity and conveniently modified pharmacokinetic profile in which the AUC and MRT were extended considerably. Overall, there were strong evidences that developed formulation can serve as a viable alternative to currently available PTX options.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Breast Neoplasms/drug therapy , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Th1 Cells/immunology , Th2 Cells/immunology , Vitamin E/administration & dosage , Vitamin E/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Compounding , Emulsions , Female , Hemolysis/drug effects , Humans , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Nanoparticles , Rats , Rats, Wistar , Th1 Cells/drug effects , Th2 Cells/drug effects
19.
J Control Release ; 196: 168-83, 2014 Dec 28.
Article in English | MEDLINE | ID: mdl-25305562

ABSTRACT

Delivery of proteins/peptides to the gastrointestinal (GI) tract via peroral/oral route involves tremendous challenges due to unfavorable environmental conditions like harsh pH, presence of proteolytic enzymes and absorption barriers. Detailed research is being conducted at the academic and industrial levels to diminish these troubles and various products are under clinical trials. Several approaches have been established to optimize oral delivery of proteins and peptides and can be broadly categorized into chemical and physical strategies. Chemical strategies include site specific mutagenesis, proteinylation, glycosylation, PEGylation and prodrug approaches, whereas physical strategies comprise formulation based approaches including application of absorption enhancers and metabolism modifiers along with delivering them via colloidal carrier systems such as nanoparticles, liposomes, microparticles, and micro- and nano-emulsions. This review stands to accomplish the diverse aspects of oral delivery of proteins/peptides and summarizes the key concepts involved in targeting the biodrugs to specific sites of the GI tract such as the intestine and colon. Furthermore some light has also been shed on the current industrial practices followed in developing oral formulations of such bioactives.


Subject(s)
Gastrointestinal Diseases/drug therapy , Gastrointestinal Tract/metabolism , Peptides/administration & dosage , Peptides/therapeutic use , Proteins/administration & dosage , Proteins/therapeutic use , Animals , Biological Transport, Active , Chemistry, Pharmaceutical , Drug Delivery Systems , Humans
20.
Appl Biochem Biotechnol ; 174(4): 1309-1330, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25106894

ABSTRACT

Solid lipid nanoparticles (SLNs) have emerged as an excellent substitute over polymeric nanoparticles and, when incorporated with chitosan which activates the macrophage to impart an immune response, produce excellent results to fight against deleterious diseases like leishmaniasis where its parasite diminishes the immunity of the host to induce resistance. Based upon this hypothesis, chitosan-coated SLNs were developed and loaded with amphotericin B (AmB) for immunoadjuvant chemotherapy of Leishmania infection. Both uncoated and chitosan-coated AmB-loaded SLNs (AmB-SLNs) were fabricated using solvent emulsification and evaporation method. The various processes and formulation parameters involved in AmB-SLN preparation were optimized with respect to particle size and stability of the particles. In vitro hemolytic test credited the formulations to be safe when injected in the veins. The cellular uptake analysis demonstrated that the chitosan-coated AmB-SLN was more efficiently internalized into the J774A.1 cells. The in vitro antileishmanial activity revealed their high potency against Leishmania-infected cells in which chitosan-coated AmB-SLNs were distinguishedly efficacious over commercial formulations (AmBisome and Fungizone). An in vitro cytokine estimation study revealed that chitosan-coated AmB-SLNs activated the macrophages to impart a specific immune response through enhanced production of TNF-α and IL-12 with respect to normal control. Furthermore, cytotoxic studies in macrophages and acute toxicity studies in mice evidenced the better safety profile of developed formulation in comparison to marketed formulations. This study indicates that the AmB-SLNs are a safe and efficacious drug delivery system which promises strong competence in antileishmanial chemotherapy and immunotherapy.


Subject(s)
Amphotericin B/pharmacology , Antiprotozoal Agents/pharmacology , Chitosan/pharmacology , Leishmania donovani , Leishmaniasis, Visceral/drug therapy , Nanoparticles , Amphotericin B/chemistry , Animals , Antiprotozoal Agents/chemistry , Cell Line , Emulsions/chemistry , Emulsions/pharmacology , Immunotherapy , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/pathology , Lipids/chemistry , Lipids/pharmacology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...