Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Mol Pharm ; 21(6): 2699-2712, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38747900

RESUMEN

This study aims to encapsulate gemcitabine (GEM) using a phospholipid complex (PLC) in lipid nanoparticles (NPs) to achieve several desirable outcomes, including high drug loading, uniform particle size, improved therapeutic efficacy, and reduced toxicities. The successful preparation of GEM-loaded lipid NPs (GEM-NPs) was accomplished using the emulsification-solidification method, following optimization through Box-Behnken design. The size of the GEM-NP was 138.5 ± 6.7 nm, with a low polydispersity index of 0.282 ± 0.078, as measured by a zetasizer and confirmed by transmission electron and atomic force microscopy. GEM-NPs demonstrated sustained release behavior, surpassing the performance of the free GEM and phospholipid complex. Moreover, GEM-NPs exhibited enhanced cytotoxicity, apoptosis, and cell uptake in Panc-2 and Mia PaCa cells compared to the free GEM. The in vivo pharmacokinetics revealed approximately 4-fold higher bioavailability of GEM-NPs in comparison with free GEM. Additionally, the pharmacodynamic evaluation conducted in a DMBA-induced pancreatic cancer model, involving histological examination, serum IL-6 level estimation, and expression of cleaved caspase-3, showed the potential of GEM-NPs in the management of pancreatic cancer. Consequently, the lipid NP-based approach developed in our investigation demonstrates high stability and uniformity and holds promise for enhancing the therapeutic outcomes of GEM.


Asunto(s)
Desoxicitidina , Gemcitabina , Nanopartículas , Neoplasias Pancreáticas , Fosfolípidos , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Desoxicitidina/farmacología , Desoxicitidina/farmacocinética , Desoxicitidina/administración & dosificación , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Nanopartículas/química , Animales , Humanos , Línea Celular Tumoral , Fosfolípidos/química , Ratones , Tamaño de la Partícula , Apoptosis/efectos de los fármacos , Portadores de Fármacos/química , Lípidos/química , Liberación de Fármacos , Masculino , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacología , Estabilidad de Medicamentos , Ratas , Liposomas
2.
Bioconjug Chem ; 34(9): 1528-1552, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37603704

RESUMEN

Most cancer patients rarely benefit from monodrug therapy because of both cancer complexity and tumor environment. One of the main reasons for this failure is insufficient accumulation of the optimal dose at the tumorous site. Our investigation implies a promising strategy to engineer prodrug nanoparticles (NPs) of bortezomib (BTZ) and selenium (Se) using sialic acid (SAL) as a ligand to improve breast cancer therapy. BTZ was conjugated with SAL and HPMA (N-2-hydroxypropyl methacrylamide) to prepare a prodrug conjugate; BTZ-SAL-HPMA (BSAL-HP) and then fabricated into prodrug NPs with Se (Se_BSAL-HP prodrug NPs). The self-assembly of prodrug NPs functionalized with Se showed size (204.13 ± 0.02 nm) and zeta potential (-31.0 ± 0.11 mV) in dynamic light scattering (DLS) experiments and spherical shape in TEM and SEM analysis. Good stability and low pH drug release profile were characterized by Se_BSAL-HP prodrug NPs. The tumor-selective boronate-ester-based prodrug NPs of BTZ in combination with Se endowed a synergistic effect against cancer cells. Compared to prodrug conjugate, Se_BSAL-HP prodrug NPs exhibited higher cell cytotoxicity and enhanced cellular internalization with significant changes in mitochondria membrane potential (MMP). Elevated apoptosis was observed in the (G2/M) phase of the cell cycle for Se_BSAL-HP prodrug NPs (2.7-fold) higher than BTZ. In vivo studies were performed on Sprague-Dawley rats and resulted in positive trends. The increased therapeutic activity of Se_BSAL-HP prodrug NPs inhibited primary tumor growth and showed 43.05 fold decrease in tumor volume than the control in 4T1 tumor bearing mice. The surprising and remarkable outcomes for Se_BSAL-HP prodrug NPs were probably due to the ROS triggering effect of boronate ester and selenium given together.


Asunto(s)
Neoplasias , Profármacos , Selenio , Ratas , Animales , Ratones , Ratas Sprague-Dawley , Profármacos/uso terapéutico , Ácido N-Acetilneuramínico , Bortezomib/farmacología , Bortezomib/uso terapéutico , Ésteres
3.
Mol Pharm ; 20(1): 524-544, 2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36306447

RESUMEN

Breast cancer leads to the highest mortality among women resulting in a major clinical burden. Multidrug therapy is more efficient in such patients compared to monodrug therapy. Simultaneous combinatorial or co-delivery garnered significant interest in the past years. Caffeic acid (CFA) (a natural polyphenol) has received growing attention because of its anticarcinogenic and antioxidant potential. Bortezomib (BTZ) is a proteasome inhibitor and may be explored for treating breast cancer. Despite its high anticancer activity, the low water solubility and chemical instability restrict its efficacy against solid tumors. In the present study, we designed and investigated a HP-PCL (N-2-hydroxypropylmethacrylamide-polycaprolactone) polymeric micellar (PMCs) system for the simultaneous delivery of BTZ and CFA in the treatment of breast cancer. The designed BTZ+CFA-HP-PCL PMCs were fabricated, optimized, and characterized for size, zeta potential, surface morphology, and in vitro drug release. Developed nanosized (174.6 ± 0.24 nm) PMCs showed enhanced cellular internalization and cell cytotoxicity in both MCF-7 and MDA-MB-231 cells. ROS (reactive oxygen species) levels were highest in BTZ-HP-PCL PMCs, while CFA-HP-PCL PMCs significantly (p < 0.001) scavenged the ROS generated in 2',7'-dichlorofluorescein diacetate (DCFH-DA) assay. The mitochondrial membrane potential (MMP) assay revealed intense and significant green fluorescence in both types of cancer cells when treated with BTZ-HP-PCL PMCs (p < 0.001) indicating apoptosis or cell death. The pharmacokinetic studies revealed that BTZ-HP-PCL PMCs and BTZ+CFA-HP-PCL PMCs exhibited the highest bioavailability, enhanced plasma half-life, decreased volume of distribution, and lower clearance rate than the pure combination of drugs. In the organ biodistribution studies, the combination of BTZ+CFA showed higher distribution in the spleen and the heart. Overall findings of in vitro studies surprisingly resulted in better therapeutic efficiency of BTZ-HP-PCL PMCs than BTZ+CFA-HP-PCL PMCs. However, the in vivo tumor growth inhibition study performed in tumor-induced mice concluded that the tumor growth was inhibited by both BTZ-HP-PCL PMCs and BTZ+CFA-HP-PCL PMCs (p < 0.0001) more efficiently than pure BTZ and the combination (BTZ+CFA), which may be due to the conversion of boronate ester into boronic acid. Henceforth, the combination of BTZ and CFA provides further indications to be explored in the future to support the hypothesis that BTZ may work with polyphenol (CFA) in the acidic environment of the tumor.


Asunto(s)
Antineoplásicos , Inhibidores de Proteasoma , Femenino , Ratones , Animales , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Micelas , Especies Reactivas de Oxígeno , Distribución Tisular , Quimioterapia Combinada , Leprostáticos/uso terapéutico , Bortezomib/farmacología , Bortezomib/química , Polímeros/química , Línea Celular Tumoral , Antineoplásicos/química
4.
AAPS PharmSciTech ; 24(7): 180, 2023 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-37697085

RESUMEN

Ferroptosis, a pathway dependent on oxygen and iron catalysts, holds promise as a therapeutic approach for cancer treatment due to its manageable regulation, direct control, and immunogenic properties. The sensitivity of cancer cells to ferroptosis induction varies based on their metabolic, genetic, and signalling pathways, prompting the use of combination therapy. In this study, we conducted a screening of drug combinations, including sorafenib (SOR) with simvastatin (SIM), phenethyl isothiocyanate, and trigonelline, in MDA-MB-231, A549, and HeLa cells to assess their cytotoxicity. The SOR-SIM combination exhibited a synergistic effect in MDA-MB-231, A549, and HeLa cells, with calculated CI values of ~ 0.66, 0.53, and 0.59, respectively. Furthermore, co-treatment with ferrostatin-1 resulted in a concentration-dependent increase in the IC50 values. Additionally, SOR + SIM demonstrated a significant reduction in GSH levels, an increase in MDA levels, and mitochondrial membrane depolarization across all three cell lines, indicating their ferroptosis inducing potential. In-vivo studies showed a significant reduction in tumor volume by 3.53-, 2.55-, and 1.47-fold compared to control, SIM, and SOR, respectively. Toxicity assessments revealed insignificant changes in biomarker levels and no observable deformations in isolated organs, except for erythrocyte shrinkage and membrane scrambling effects caused by the SOR + SIM combination. Overall, our findings highlight the potential of the SOR + SIM combination as an effective strategy for cancer treatment, emphasizing the importance of further research in targeted drug delivery systems to ensure its safety.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Detección Precoz del Cáncer , Células HeLa , Sorafenib/farmacología , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico
5.
Mol Pharm ; 19(5): 1325-1337, 2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35437994

RESUMEN

Cisplatin is a platinum (Pt)-based anticancer drug with broad-scale clinical utility. However, due to its hydrophilic nature and high kinetic reactivity, it offers numerous drug delivery challenges. Limitations such as severe systemic toxicities, chemoresistance, extensive cisplatin-plasma protein interaction, and limited cellular drug uptake reduce the therapeutic impact of cisplatin therapy. Cisplatin(IV) prodrug formation can effectively resolve these challenges. The selection of axial ligands could play a key role in determining the fate of cisplatin(IV) prodrugs by modulating the therapeutic and biopharmaceutical outcomes of therapy. Hereby, three cisplatin(IV) derivatives were developed utilizing valproate, tocopherol, and chlorambucil as axial ligands, and their biopharmaceutical performance was compared along with cisplatin. The impact of cisplatin(IV) derivative formation on their kinetic stability, drug-albumin interaction, cytotoxicity profile, cellular uptake pattern, self-assembling behavior, hemotoxicity, and tumor biodistribution pattern was analyzed to establish the correlation between the structural properties of cisplatin(IV) agents and their biopharmaceutical outcomes. The kinetic inertness of the designed cisplatin(IV) compounds helped in minimizing their plasma protein interactions and ensuring their stability in the blood environment. The lipophilicity enhancement due to Pt(IV) prodrug formation critically helped in enhancing the cellular drug uptake and reduced the dependence on transporters for drug uptake. The lipophilicity and activity of axial ligands were the key drivers governing the biopharmaceutical performance of the Pt(IV) derivatives. The properties of the axial ligand, such as its therapeutic activity, chemical backbone, and functional groups present in its structure, were the critical factors determining their plasma protein interaction, cellular uptake, anticancer activity, and self-assembly pattern. Cisplatin(IV) derivative formation further improved the amount of platinum accumulated in tumors after intravenous injection compared to free cisplatin therapy (2.7-5.4 folds increment) and reduced drug-erythrocyte interactions. Overall, the results highlighted the potential of cisplatin(IV) agents in resolving cisplatin drug delivery challenges and denoted the critical role of axial ligand selection in Pt(IV) prodrug designing.


Asunto(s)
Antineoplásicos , Productos Biológicos , Profármacos , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Cisplatino/química , Cisplatino/farmacología , Cisplatino/uso terapéutico , Ligandos , Platino (Metal)/química , Profármacos/química , Distribución Tisular
6.
AAPS PharmSciTech ; 23(7): 238, 2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-36002600

RESUMEN

The current study elucidates the improved drug loading of paclitaxel (PTX) in lipid- and D-α-tocopheryl polyethylene glycol succinate (TPGS)-based core-shell-type lipid nanocapsules (PTX-TPGS-LNC) for augmenting the therapeutic efficacy and curbing the toxicity. PTX-TPGS-LNCs were formulated by employing anti-solvent precipitation technique and displayed a particle size of 162.1 ± 4.70 nm and % practical drug loading of 15.04 ± 2.44%. Electron microscopy revealed that PTX-TPGS-LNCs have spherical morphology and the inner core was surrounded by a relatively lighter region, i.e., layer of lipids and TPGS. The nature of loaded PTX inside the PTX-TPGS-LNC was also confirmed using DSC and PXRD analysis. The in vitro release study showed biphasic and sustained release pattern of PTX from PTX-TPGS-LNC and it showed ~ threefold higher PTX uptake in MCF-7 cell line in comparison to free PTX. Moreover, it was apparent from the cytotoxicity assay that PTX-TPGS-LNC displayed higher cytotoxicity in MCF-7 cells and revealed ~ 2.92-fold decrease in IC50 value as against free PTX when incubated for 72 h. The apoptotic index in case of PTX-TPGS-LNC was ~ twofold higher than free PTX. The pharmacokinetic profile of PTX-TPGS-LNC revealed a ~ 3.18-fold increase in t1/2 and a ~ 2.62-fold higher AUC(0→∞) compared to Intaxel®. Finally, treatment with PTX-TPGS-LNC demonstrated significant lowering in the % tumor burden and serum toxicity markers compared to marketed formulation Intaxel®. Thus, the lipid- and TPGS-based core-shell-type LNC with high PTX loading can advance the existing standards of therapy for overshadowing cancer.


Asunto(s)
Nanocápsulas , Paclitaxel , Línea Celular Tumoral , Humanos , Lípidos , Polietilenglicoles , Vitamina E , alfa-Tocoferol
7.
Mol Pharm ; 17(7): 2473-2486, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32496783

RESUMEN

The currently available systemic chemotherapy for treating breast cancer often results in serious systemic side effects and compromises patient compliance. The distinct anatomical features of human breasts (e.g., embryological origin of breast skin, highly developed internal lymphatic and venous circulation, and the presence of mammary fat layers) help in preferential accumulation of drugs into breasts after topical application on breast region. This unique feature is termed as localized transdermal delivery which could be utilized for effectively delivering anticancer agents to treat breast cancer and reducing the systemic side effects by limiting their presence in blood. However, the clinical effectiveness of this drug delivery approach is highly limited by barrier properties of skin reducing the permeation of anticancer drugs. In the present work, we have developed high permeation vesicles (HPVs) using phospholipids and synergistic combination of permeation enhancers (SCOPE) to improve the skin permeation of drugs. Docetaxel (DTX) was used as a model drug for hypothesis testing. The optimized SCOPE mixture composed of sodium oleate/sodium lauryl ether sulfate/propylene glycol in 64:16:20% w/w ratio. DTX HPVs were prepared using phospholipid: SCOPE, 8:2% w/w ratio. DTX HPVs exhibited as a uniform deformable vesicles with size range 124.2 ± 7.6 nm, significantly improved skin permeation profile, and sustained drug release until 48 h. Superior vesicle deformability, better vesicle membrane fluidization, and SCOPE mediated enhancement in skin fluidization were the prime factors behind enhancement of DTX permeation. The improved cellular uptake, reduced IC50 values, and higher apoptotic index of DTX HPVs in MCF-7 and MDA-MB-231 cells ensured the therapeutic effectiveness of HPV based therapy. Also, HPVs were found to be predominantly internalized inside cells through clathrin and caveolae-dependent endocytic pathways. Bioimaging analysis in mice confirmed the tumor penetration potential and effective accumulation of HPVs inside tumors after topical application. In vivo studies were carried out in comparison with marketed intravenous DTX injection (Taxotere) to compare the effectiveness of topical chemotherapy. The topical application of DTX HPV gel in tumor bearing mice resulted in nearly 4-fold tumor volume reduction which was equivalent to intravenous Taxotere therapy. Toxicity analysis of DTX HPV gel in comparison with intravenous Taxotere dosing showcased remarkably lower levels of toxicity biomarkers (aspartate transaminase (AST), alanine transaminase (ALT), blood urea nitrogen (BUN), and creatinine), indicating improved safety of topical chemotherapy. Overall results warranted the effectiveness of topical DTX chemotherapy to reduce tumor burden with substantially reduced risk of systemic toxicities in breast cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Docetaxel/administración & dosificación , Portadores de Fármacos/química , Composición de Medicamentos/métodos , Nanopartículas/química , Administración Cutánea , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Docetaxel/sangre , Docetaxel/farmacocinética , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Permeabilidad/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Piel/efectos de los fármacos , Piel/metabolismo , Porcinos , Distribución Tisular , Trasplante Homólogo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
8.
AAPS PharmSciTech ; 20(2): 43, 2019 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-30610392

RESUMEN

Among many, the oral route of delivery is considered to be the most favorable route with the highest patient compliance. The main issue with oral delivery is the environmental vulnerability of gastro intestinal tract (G.I.T). The bioavailability could further decrease when drug has poor aqueous solubility and permeability through biological membrane. This drawback could be resolved by employing drug-phospholipid complex strategy, as they utilize mechanism which is similar to the absorption mechanism of nutritional constituents form G.I.T. The drug-phospholipid complexes are considered ideal for oral delivery as they are biodegradable and non-toxic, which enable them to be employed as solubilizer, emulsifier, and as a matrix forming excipient for dugs with poor solubility and/or permeability. The present review compiles the basic know how about the phospholipids and the mechanism through which it improves the bioavailability of drugs. Further, it also compiles the crucial formulation aspects and methods of preparations of drug-phospholipid complex along with its physical and in silico characterization techniques. The increase in number of recent reports involving the utilization of drug-phospholipid complex to improve oral bioavailability of drugs thus explains how vital the strategy is for a successful oral delivery.


Asunto(s)
Sistemas de Liberación de Medicamentos , Fosfolípidos/química , Administración Oral , Animales , Disponibilidad Biológica , Humanos , Permeabilidad , Solubilidad
9.
Biomater Adv ; 156: 213700, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38042001

RESUMEN

In this study, we investigated the potential of the sorafenib (SOR) and simvastatin (SIM) combination to induce ferroptosis-mediated cancer therapy. To enhance targeted drug delivery, we encapsulated the SOR + SIM combination within 4-carboxy phenylboronic acid (CPBA) modified PLGA nanoparticles (CPBA-PLGA(SOR + SIM)-NPs). The developed CPBA-PLGA(SOR + SIM)-NPs exhibited a spherical shape with a size of 213.1 ± 10.9 nm, a PDI of 0.22 ± 0.03, and a Z-potential of -22.9 ± 3.2 mV. Notably, these nanoparticles displayed faster drug release at acidic pH compared to physiological pH. In cellular experiments, CPBA-PLGA(SOR + SIM)-NPs demonstrated remarkable improvements, leading to a 2.51, 2.69, and 2.61-fold decrease in IC50 compared to SOR alone, and a 7.50, 16.71, and 5.11-fold decrease in IC50 compared to SIM alone in MDA-MB-231, A549, and HeLa cells, respectively. Furthermore, CPBA-PLGA(SOR + SIM)-NPs triggered a reduction in glutathione (GSH) levels, an increase in malondialdehyde (MDA) levels, and mitochondrial membrane depolarization in all three cell lines. Pharmacokinetic evaluation revealed a 2.50- and 2.63-fold increase in AUC0-∞, as well as a 1.53- and 2.46-fold increase in mean residence time (MRT) for SOR and SIM, respectively, compared to the free drug groups. Notably, the CPBA-PLGA(SOR + SIM)-NPs group exhibited significant reduction in tumor volume, approximately 9.17, 2.45, and 1.63-fold lower than the control, SOR + SIM, and PLGA(SOR + SIM)-NPs groups, respectively. Histological examination and biomarker analysis showed no significant differences compared to the control group, suggesting the biocompatibility of the developed particles for in-vivo applications. Altogether, our findings demonstrate that CPBA-PLGA(SOR + SIM)-NPs hold tremendous potential as an efficient drug delivery system for inducing ferroptosis, providing a promising therapeutic option for cancer treatment.


Asunto(s)
Ferroptosis , Nanopartículas , Humanos , Células HeLa , Sistemas de Liberación de Medicamentos , Simvastatina/farmacología
10.
Biomater Adv ; 159: 213822, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38442461

RESUMEN

Certain aggressive cancers, such as triple-negative breast cancer (TNBC), heavily bank on glutamine for their proliferation and survival. In this context, TNBC functions as a "glutamine trap," extracting circulating glutamine at a rate surpassing that of any other organ. Moreover, the overexpression of Alanine, Serine, Cysteine Transporter 2 (ASCT2), a key player in glutamine uptake, further underscores the significance of targeted therapy to enhance TNBC treatment. This led to the exploration of a novel approach involving hydrophobized Pluronic-based mixed micelles achieved through the use of docosahexaenoic acid and stapled with glutamine for displaying inherent ASCT2 targeting ability-a formulation termed LPT G-MM. LPT G-MM exhibited optimal characteristics, including a size of 163.66 ± 10.34 nm, a polydispersity index of 0.237 ± 0.083, and an enhanced drug loading capacity of approximately 15 %. Transmission electron microscopy validated the spherical shape of these micelles. In vitro release studies demonstrated drug release in a sustained manner without the risk of hemolysis. Importantly, LPT G-MM displayed heightened cellular uptake, increased cytotoxicity, a lower IC50 value, elevated reactive oxygen species, induced mitochondrial membrane depolarization, and a greater apoptosis index in TNBC cell lines compared to free LPT. The pharmacokinetic profile of LPT G-MM revealed a substantial rise in half-life (t1/2) by approximately 1.48-fold and an elevation in the area under the curve [AUC(0→∞)] by approximately 1.19-fold. Moreover, there was a significant reduction in the percentage of tumor volume by approximately 7.26-fold, along with decreased serum toxicity markers compared to free LPT. In summary, LPT G-MM demonstrated promising potential in boosting payload capacities and targeting specificity in the context of TNBC treatment.


Asunto(s)
Micelas , Neoplasias de la Mama Triple Negativas , Humanos , Lapatinib/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Glutamina/uso terapéutico , Línea Celular Tumoral , Apoptosis
11.
Int J Biol Macromol ; 252: 126565, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37640185

RESUMEN

This study investigates the impact of charge and chain length of bile salts in the bilosomes on the oral bioavailability of insulin (IN) by examining their uptake via the apical sodium-dependent bile acid transporter (ASBT). Deoxycholic acid bile salt was conjugated with different amino acids to create conjugates with varying charge and chain length, which were then embedded in liposomes. The resulting bilosomes had a particle size <400 nm, a PDI of 0.121 ± 0.03, and an entrapment efficiency of ∼70 %, while maintaining the chemical and conformational integrity of the loaded IN. Bilosomes also provided superior protection in biological fluids without compromising their biophysical attributes. Quantitative studies using the Caco-2 cell line demonstrated that anionic bilosomes were taken up more efficiently through ASBT than cationic bilosomes with 4- and 1.3-fold increase, respectively. Ex-vivo permeability studies corroborated these findings. In-vivo efficacy studies revealed a 1.6-fold increase in the AUC of IN with bilosomes compared to subcutaneous IN. The developed bilosomes were able to reduce blood glucose levels by ∼65 % at 6 h, with a cumulative hypoglycemic value of 35 % and a BAR of ∼30 %. These results suggest that ASBT can be a suitable target for improving the oral bioavailability of bilosomes containing IN.


Asunto(s)
Insulina , Liposomas , Humanos , Disponibilidad Biológica , Células CACO-2 , Liposomas/química , Ácidos y Sales Biliares
12.
Drug Deliv Transl Res ; 13(4): 1074-1087, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36528709

RESUMEN

In present investigation, we developed paclitaxel (PTX)-loaded adenosine (ADN)-conjugated PLGA nanoparticles for combating triple-negative breast cancer (TNBC), where ADN acts as a substrate for adenosine receptors (AR) that are overexpressed in TNBC. Using synthesized PLGA-PEG-ADN, PTX-loaded nanoparticles (PTX ADN-PEG-PLGA NPs) were prepared via emulsion diffusion evaporation process that rendered particles of size 135 ± 12 nm, PDI of 0.119 ± 0.03, and entrapment-efficiency of 79.26 ± 2.52%. The NPs showed higher %cumulative release at pH 5.5 over 7.4 with Higuchi release kinetics. The PTX ADN-PEG-PLGA NPs showed ~ 4.87- and 5.22-fold decrease in %hemolysis in comparison to free PTX and Intaxel®, indicating their hemocompatible nature. The ADN modification assisted cytoplasmic internalization of particles via AR-mediated endocytosis that resulted in ~ 3.77- and 3.51-fold reduction in IC50 and showed apoptosis index of 0.93 and 1.18 in MDA-MB-231 and 4T1 cells respectively. The pharmacokinetic profile of ADN-PEG-PLGA NPs revealed higher AUC and t1/2 than Intaxel® and Nanoxel® pharmacodynamic activity showed ~ 18.90-fold lower %tumor burden than control. The kidney and liver function biomarkers showed insignificant change in the levels, when treated with PTX ADN-PEG-PLGA NPs and exhibited no histological alterations in the liver, spleen, and kidney. Overall, the optimized particles were found to be biocompatible with improved anti-TNBC activity.


Asunto(s)
Nanopartículas , Neoplasias de la Mama Triple Negativas , Humanos , Paclitaxel/farmacocinética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adenosina , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Línea Celular Tumoral , Polietilenglicoles , Portadores de Fármacos/farmacología
13.
Adv Drug Deliv Rev ; 199: 114901, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37257756

RESUMEN

Long-acting injectable (LAIs) delivery systems sustain the drug therapeutic action in the body, resulting in reduced dosage regimen, toxicity, and improved patient compliance. Lipid-based depots are biocompatible, provide extended drug release, and improve drug stability, making them suitable for systemic and localized treatment of various chronic ailments, including psychosis, diabetes, hormonal disorders, arthritis, ocular diseases, and cancer. These depots include oil solutions, suspensions, oleogels, liquid crystalline systems, liposomes, solid lipid nanoparticles, nanostructured lipid carriers, phospholipid phase separation gel, vesicular phospholipid gel etc. This review summarizes recent advancements in lipid-based LAIs for delivering small and macromolecules, and their potential in managing chronic diseases. It also provides an overview of the lipid depots available in market or clinical phase, as well as patents for lipid-based LAIs. Furthermore, this review critically discusses the current scenario of using in vitro release methods to establish IVIVC and highlights the challenges involved in developing lipid-based LAIs.


Asunto(s)
Nanoestructuras , Fosfolípidos , Humanos , Preparaciones de Acción Retardada
14.
Int J Biol Macromol ; 253(Pt 8): 127254, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37813219

RESUMEN

Ferroptosis is a non-apoptotic cell death pathway characterized by the accumulation of lipid-peroxy radicals within the affected cells. Here, we investigate the synergistic capacity of sorafenib (SOR) and simvastatin (SIM) to trigger ferroptosis for cancer therapy. For precise in-vivo delivery, SOR + SIM was ratiometrically loaded in bovine serum albumin nanoparticles (BSA-NPs) modified with 4-carboxy phenylboronic acid (CPBA). The developed CPBA-BSA(SOR + SIM)-NPs revealed size of 175.2 ± 12.8 nm, with PDI of 0.22 ± 0.03 and Z-potential of -29.6 ± 4.8 mV. Significantly, CPBA-BSA(SOR + SIM)-NPs exhibited > 2 and > 5-fold reduction in IC50 values compared to individual SOR and SIM treatments respectively, in all tested cell lines. Moreover, CPBA-BSA(SOR + SIM)-NPs treated cells exhibited decrease in glutathione levels, increase in malonaldehyde levels and depolarization of mitochondrial membrane potential (JC-1 assay). Pharmacokinetic analysis revealed enhanced AUC0-∞ and MRT levels for SOR and SIM when administered as CPBA-BSA(SOR + SIM)-NPs compared to free drugs. Crucially, in in-vivo experiments, CPBA-BSA(SOR + SIM)-NPs led to a significant reduction in tumor volume compared to various control groups. Histological and biomarker analyses underscore their biocompatibility for clinical applications. In conclusion, this study highlights the potential of CPBA-BSA(SOR + SIM)-NPs as a promising strategy for inducing ferroptosis in cancer cells, concurrently improving drug delivery and therapeutic efficacy. This approach opens new avenues in cancer treatment.


Asunto(s)
Ferroptosis , Nanopartículas , Sorafenib/farmacología , Albúmina Sérica Bovina , Simvastatina/farmacología , Portadores de Fármacos/farmacocinética , Tamaño de la Partícula
15.
Int J Pharm ; 648: 123570, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37918494

RESUMEN

Triple negative breast cancer (TNBC) cells resist chemotherapy by hijacking apoptosis. Alternative cell death forms like ferroptosis offer new treatment options. A combined therapy using neratinib (NTB; ferroptosis inducer) and silibinin (SLB; apoptosis inducer) via albumin-based nanocarriers (N-S Alb NPs) was explored to target TNBC. N-S Alb NPs had optimal size (134.26 ± 10.23 nm), PDI (0.224 ± 0.01), and % entrapment efficiency (∼80 % for NTB and ∼87 % for SLB). Transmission electron microscopy confirmed their spherical shape. In vitro release studies showed sustained drug release without hemolysis risk. N-S Alb NPs had higher cellular uptake and cytotoxicity than individual drugs or their mixture. IC50 values for N-S Alb NPs were significantly reduced in MDA-MB-231 (∼2.23-fold) and 4T1 (∼1.85-fold) cell lines and apoptosis index were significantly higher in MDA-MB-231 (∼1.31-fold) and 4T1 cell line (∼1.35-fold) than the physical mixture of both drugs (NTB + SLB). N-S Alb NPs generated more reactive oxygen species (ROS) and caused mitochondrial membrane depolarization, indicating increased cell death. They also exhibited better ferroptosis induction by reducing glutathione (GSH), increasing Fe2+ activity and MDA levels in TNBC cells. Thus, N-S Alb NPs had the ability to promote "mixed" type cell death, showed promise in enhancing the payload capabilities and targeting in TNBC.


Asunto(s)
Nanopartículas , Neoplasias de la Mama Triple Negativas , Humanos , Silibina , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Línea Celular Tumoral , Apoptosis , Albúminas
16.
ACS Biomater Sci Eng ; 8(6): 2349-2362, 2022 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-35522530

RESUMEN

Triple-negative breast cancer (TNBC) cells show improved sensitivity for cisplatin therapy due to their defective DNA damage repair system. However, the clinical utilization of cisplatin is limited by dose-dependent systemic toxicities and chemoresistance. Cisplatin Pt(IV) derivatives having kinetically inert octahedral geometry provide an effective strategy to overcome these limitations. Upon cellular reduction, these derivatives release cisplatin and axial ligands, acting as dual-action prodrugs. Hereby, we have developed three cisplatin(IV) conjugates using distinct bioactive axial moieties (valproate, tocopherol, and chlorambucil), which can synergistically complement cisplatin activity and attack multiple cellular targets. The designed derivatives showcased enhanced antiproliferative activity and improved therapeutic synergism along with a noteworthy cisplatin dose reduction index in a panel of six cancer cells. These Pt(IV) derivatives remarkably improved cellular drug uptake and showed lower dependency on copper transporter 1 (Ctr1) for uptake than cisplatin. The results of enhanced in vitro activity were well corroborated by in vivo efficacy testing in the 4T1 cell-based TNBC model, showcasing ∼2-7-folds higher tumor volume reduction for Pt(IV) derivatives than cisplatin. In addition, the designed derivatives significantly reduced the nephrotoxicity risk involved in cisplatin therapy, indicated by systemic toxicity biomarkers and organ histopathology. The results indicated that cisplatin(IV) derivatives could open new avenues for safer synergistic chemotherapy in TNBC.


Asunto(s)
Antineoplásicos , Profármacos , Neoplasias de la Mama Triple Negativas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Cisplatino/farmacología , Cisplatino/uso terapéutico , Humanos , Profármacos/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
17.
ACS Biomater Sci Eng ; 8(8): 3473-3484, 2022 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-35896042

RESUMEN

Triple-negative breast cancer (TNBC) belongs to the category of the most destructive forms of breast cancer. Being a highly potent chemotherapeutic agent, paclitaxel (PTX) is extensively utilized in the management of various cancers. Commercially available PTX formulations contain non-targeted drug carriers that result in low antitumor activity because of non-specific tissue distribution. Thus, to resolve this issue, we designed PTX-loaded pH-sensitive liposomes (pH Lipos) in the present investigation and used adenosine (ADN) as a targeting ligand. Further, d-α-tocopheryl polyethylene glycol succinate (TPGS) was incorporated into the liposomes to impart a stealth effect to the system. For the development of these pH Lipos, different conjugates were synthesized (ADN-CHEMS and TPGS-ADN) and further utilized for the preparation of ADN-PEG-pH Lipo and ADN-pH Lipo by a thin-film hydration method. DOPE:HSPC:CHEMS:cholesterol at a molar ratio of 3:3:2:2 was selected for the preparation of pH-Lipo possessing 7.5% w/w drug loading. They showed a particle size below 140 nm, a PDI below 0.205, and a % EE greater than 60%. All of the pH Lipos displayed a biphasic pattern of PTX release at pH 7.4 and 5.5. However, the percent drug release at pH 5.5 was substantially greater because of the pH-sensitive nature of the liposomes. The MDA MB 231 and 4T1 cell lines depicted improvement in the qualitative as well as quantitative cellular uptake of PTX ADN-PEG-pH Lipo with a substantial decrease in the IC50 value. Moreover, a higher apoptotic index was observed with pH Lipo compared to free PTX. PTX ADN-PEG-pH Lipo revealed a 3.98- and 3.41-fold rise in the AUC and t1/2 values of PTX compared to Intaxel, respectively. Overall, characteristic decreases in tumor volume and serum toxicity marker levels were observed, which confirmed the development of an efficient and safe formulation.


Asunto(s)
Paclitaxel , Neoplasias de la Mama Triple Negativas , Adenosina/farmacología , Humanos , Concentración de Iones de Hidrógeno , Liposomas , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
18.
Carbohydr Polym ; 286: 119305, 2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35337491

RESUMEN

Chondroitin Sulfate (CS) is an anionic hetero polysaccharide possessing anti-inflammatory, antioxidant, antitumor, anticoagulant and antithrombogenic activities. It is biodegradable and biocompatible in nature. Further, it inherits the ability of active and subcellular targeting due to its affinity for CD 44 receptors and glycosylation enzymes, which are overexpressed on the surface of tumor cells and intracellular organelles respectively. CS is known to degrade in presence of physiological stimuli, the hyaluronidase (HAase) enzyme and reactive oxygen species (ROS), assisting in site specific drug release. Due to these properties, it serve as a promising biomaterial for drug delivery and tissue engineering. In this review, the fundamental theory of CS, CS-based nanocarriers for the delivery of biopharmaceuticals and stimuli sensitive delivery systems such as HAase and ROS responsive nanocarriers for tumor targeted delivery are discussed critically. In addition, the manuscript describes the application of CS-based tissue constructs in tissue engineering and wound healing.


Asunto(s)
Productos Biológicos , Sulfatos de Condroitina , Materiales Biocompatibles/farmacología , Sistemas de Liberación de Medicamentos , Ingeniería de Tejidos
19.
Drug Deliv Transl Res ; 12(3): 562-576, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33774776

RESUMEN

The present investigation demonstrates the development of crosslinked ß-cyclodextrin nanoparticles (ß-CD NPs) for enhancing the therapeutic efficacy of docetaxel (DTX) against breast cancer. Initially, a partial inclusion complex between ß-CD and polypropylene glycol (PPG) was formed to induce self-assembly. This was followed by crosslinking of ß-CDs using epichlorohydrin (EPI) and removal (by solubilization) of PPG to yield uniform ß-CD NPs. The formed particles were used for loading DTX to form DTX ß-CD NPs. The resultant DTX ß-CD NPs exhibited particle size of 223.36 ± 17.73 nm with polydispersity index (PDI) of 0.13 ± 0.09 and showed entrapment efficiency of 54.53 ± 2%. Increased cell uptake (~5-fold), cytotoxicity (~3.3-fold), and apoptosis were observed in MDA-MB-231 cells when treated with DTX ß-CD NPs in comparison to free DTX. Moreover, pharmacokinetic evaluation of DTX ß-CD NPs revealed ~2 and ~5-fold increase in AUC0-∞ and mean residence time (MRT) of DTX when compared to Docepar®. Further, the anti-tumor activity using DMBA-induced cancer model showed that DTX ß-CD NPs were capable of reducing the tumor volume to ~40%, whereas Docepar® was able to reduce tumor volume till ~80%. Finally, the toxicity evaluation of DTX ß-CD NPs revealed no short-term nephrotoxicity and was confirmed by estimating the levels of biomarkers and histopathology of the organs. Thus, the proposed formulation strategy can yield uniformly formed ß-CD NPs which can be effectively utilized for improving the therapeutic efficacy of DTX.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Nanopartículas , beta-Ciclodextrinas , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Docetaxel/farmacología , Portadores de Fármacos/farmacocinética , Femenino , Humanos
20.
J Control Release ; 330: 72-100, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33321156

RESUMEN

Triple negative breast cancer (TNBC) is one of the most aggressive form of breast cancer. It is characterized by the absence of estrogen, progesterone and human epidermal growth factor receptors. The main issue with TNBC is that it exhibits poor prognosis, high risk of relapse, short progression-free survival and low overall survival in patients. This is because the conventional therapy used for managing TNBC has issues pertaining to poor bioavailability, lower cellular uptake, increased off-target effects and development of resistance. To overcome such pitfalls, several other approaches are explored. In this context, the present manuscript showcases three of the most widely used approaches which are (i) nanotechnology-based approach; (ii) gene therapy approach and (iii) Phytochemical-based approach. The ultimate focus is to present and explain the insightful reports based on these approaches. Further, the review also expounds on the identified molecular targets and novel targeting ligands which are explored for managing TNBC effectively. Thus, in a nutshell, the review tries to highlight these existing treatment approaches which might inspire for future development of novel therapies with a potential of overshadowing TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA