Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biomater Adv ; 159: 213822, 2024 May.
Article in English | MEDLINE | ID: mdl-38442461

ABSTRACT

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.


Subject(s)
Micelles , Triple Negative Breast Neoplasms , Humans , Lapatinib/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Glutamine/therapeutic use , Cell Line, Tumor , Apoptosis
2.
Biomater Adv ; 156: 213700, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38042001

ABSTRACT

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.


Subject(s)
Ferroptosis , Nanoparticles , Humans , HeLa Cells , Drug Delivery Systems , Simvastatin/pharmacology
3.
Int J Pharm ; 648: 123570, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37918494

ABSTRACT

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.


Subject(s)
Nanoparticles , Triple Negative Breast Neoplasms , Humans , Silybin , Triple Negative Breast Neoplasms/drug therapy , Cell Line, Tumor , Apoptosis , Albumins
4.
Int J Biol Macromol ; 253(Pt 8): 127254, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37813219

ABSTRACT

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.


Subject(s)
Ferroptosis , Nanoparticles , Sorafenib/pharmacology , Serum Albumin, Bovine , Simvastatin/pharmacology , Drug Carriers/pharmacokinetics , Particle Size
5.
ACS Biomater Sci Eng ; 9(6): 2879-2890, 2023 06 12.
Article in English | MEDLINE | ID: mdl-33587853

ABSTRACT

Amphotericin B (AmB) is gold standard therapy for leishmaniasis and fungal infections. Considering the global disease burden, nearly 90% of cases occur in economically vulnerable countries, making the cost of AmB therapy a critical healthcare challenge in controlling disease burden. All currently marketed AmB products are administered through an intravenous (i.v.) route and involve high treatment costs. Designing an orally effective AmB formulation can substantially reduce the cost of therapy and improve patient compliance. However, it is a challenging task because of the distinctive physicochemical properties of AmB. Previously, we developed a lipid-based prodrug of AmB, AmB-oleyl conjugate (AmB-OA), which showcased remarkable stability in the gastrointestinal (GI) environment and improved intestinal permeation. Hereby, we have developed self-nanoemulsifiying drug delivery system (SNEDDS) of AmB-OA to further enhance the oral bioavailability of AmB and potentiate its therapeutic benefits. SNEDDS was developed by screening a wide range of oils, surfactants, and cosurfactants, and formulation composition was optimized using extreme vertices design. AmB-OA SNEDDS possessed the ability of quick self-nanoemulsification on dilution (droplet size ∼56 nm) along with remarkable stability in the GI environment. Accelerated stability (40 °C/75% relative humidity) studies and freeze-thaw cycling studies proved that the formulation was stable at tropical conditions as well as temperature cycling stress. Drug transport analysis in Caco-2 cells revealed a remarkable increase in drug transport for AmB-OA SNEDDS compared to AmB along with minimal cellular toxicities. AmB-OA SNEDDS showcased ∼8.9-fold higher AUCTot than AmB in in vivo pharmacokinetic study, proving the effectiveness of formulation to enhance oral bioavailability. In vivo toxicity analysis highlighted the ameliorated toxicity risk associated with SNEDDS formulation. Therefore, the AmB-OA SNEDDS formulation may provide a cost-friendly and effective strategy to resolve the oral AmB drug delivery challenge.


Subject(s)
Prodrugs , Humans , Amphotericin B/pharmacology , Caco-2 Cells , Solubility , Drug Delivery Systems
6.
Drug Deliv Transl Res ; 13(4): 1074-1087, 2023 04.
Article in English | MEDLINE | ID: mdl-36528709

ABSTRACT

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.


Subject(s)
Nanoparticles , Triple Negative Breast Neoplasms , Humans , Paclitaxel/pharmacokinetics , Triple Negative Breast Neoplasms/drug therapy , Adenosine , Polylactic Acid-Polyglycolic Acid Copolymer , Cell Line, Tumor , Polyethylene Glycols , Drug Carriers/pharmacology
7.
Drug Deliv Transl Res ; 13(3): 839-851, 2023 03.
Article in English | MEDLINE | ID: mdl-36223029

ABSTRACT

Sildenafil (SLD) is employed for the management of erectile dysfunction and pulmonary arterial hypertension. It exhibits meagre water solubility and is available in the form of citrate salt hydrate to improve the solubility. However, it still exhibits moderate solubility, high first-pass metabolism, resulting in very less oral bioavailability. The present study demonstrates the preparation of self-nanoemulsifying drug delivery system for augmenting the oral bioavailability of SLD. Oleic acid and Capmul MCM C8 blend (oil phase), Cremophor® RH40 (surfactant), and Labrafil® M1944 CS (cosurfactant) were selected as main constituents for making liquid preconcentrate based on the solubility and emulsification study. The preconcentrate upon dilution and emulsification showed droplet size 52.03 ± 13.03 nm, PDI 0.143 ± 0.028, and % transmittance was 99.77 ± 1.86% with SLD load of 40 mg/g of formulation. The prepared formulation was further assessed for stability, in vitro release, Caco-2 cell uptake, and in vivo pharmacokinetic performance. SLD-SNEDDS formulation was found to be robust in terms of stability against several folds dilution in the gastrointestinal tract (GIT), freeze-thaw cycles, and had a storage stability of 3 months at 4 °C and 25 °C. SLD-SNEDDS showed ~4.7-fold and ~5-fold increase in time- and concentration-dependent cellular uptake as against SLD cultured with Caco-2 cells. In vivo pharmacokinetic study revealed ~5.8- and ~2.5-fold increase in AUC0-∞ values in case of SLD-SNEDDS as against SLD suspension and SLD citrate solution, respectively.


Subject(s)
Drug Delivery Systems , Nanoparticles , Rats , Male , Humans , Animals , Sildenafil Citrate , Rats, Wistar , Caco-2 Cells , Emulsions , Drug Delivery Systems/methods , Surface-Active Agents , Solubility , Biological Availability , Citrates , Administration, Oral , Particle Size
8.
ACS Biomater Sci Eng ; 8(8): 3473-3484, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35896042

ABSTRACT

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.


Subject(s)
Paclitaxel , Triple Negative Breast Neoplasms , Adenosine/pharmacology , Humans , Hydrogen-Ion Concentration , Liposomes , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Triple Negative Breast Neoplasms/drug therapy
9.
ACS Biomater Sci Eng ; 8(6): 2349-2362, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35522530

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Prodrugs , Triple Negative Breast Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cisplatin/pharmacology , Cisplatin/therapeutic use , Humans , Prodrugs/pharmacology , Triple Negative Breast Neoplasms/drug therapy
10.
Mol Pharm ; 19(5): 1325-1337, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35437994

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Biological Products , Prodrugs , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/chemistry , Cisplatin/pharmacology , Cisplatin/therapeutic use , Ligands , Platinum/chemistry , Prodrugs/chemistry , Tissue Distribution
11.
Colloids Surf B Biointerfaces ; 204: 111821, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33971612

ABSTRACT

Combination therapy, which combines anti-cancer drugs with different oligonucleotides, have shown potential in cancer treatment. However, delivering a hydrophobic anti-cancer drug and a hydrophilic oligonucleotide simultaneously is a herculean task. This study takes advantage of interactions between histidine-lauric acid-based green surfactant and poly(amidoamine) dendrimers to achieve this aim. The green surfactant was synthesized by carbodiimide chemistry and characterized by FTIR, 1H-NMR, and mass spectroscopy. Further, green surfactant-dendrimer aggregates encapsulating DTX and complexing SIRT 1 shRNA i.e., "aggreplexes" were developed and characterized. The term "aggreplexes" signifies complexes which are formed between green-surfactant-dendrimer aggregates and SIRT-1 shRNA via electrostatic interaction. The aggreplexes displayed particle size of 262.33 ± 3.87 nm, PDI of 0.25 and entrapment efficiency of 70.56 %. The TEM images revealed spherical shape of aggreplexes with irregular outer surface and corroborated particle size obtained from zetasizer. The in-vitro release study revealed biphasic release patterns of DTX from aggreplexes and were compatible for intravenous administration. Further, aggreplexes augmented cellular uptake in MDA-MB-231 cells by ∼1.87-fold compared to free DTX. Also, EGFP expression revealed significantly higher transfection of aggreplexes compared to naked shRNA and Superfect™ complexes. Further, aggreplexes showed higher cytotoxicity in MDA-MB-231 cells and ∼4.16-fold reduction in IC50 value compared to free DTX. Finally, apoptosis-index observed in case of aggreplexes was ∼3.57-fold higher than free DTX. These novel aggreplexes showed increased drug loading capacity and superior gene transfection potential. Thus, they open new avenues for co-delivery of hydrophobic anti-cancer drugs and hydrophilic therapeutic genes for improving current standards of cancer therapy.


Subject(s)
Antineoplastic Agents , Dendrimers , Nanoparticles , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Docetaxel , Drug Carriers , Neoplasms/drug therapy , Neoplasms/genetics , Particle Size , Surface-Active Agents
12.
Mol Pharm ; 17(7): 2473-2486, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32496783

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Docetaxel/administration & dosage , Drug Carriers/chemistry , Drug Compounding/methods , Nanoparticles/chemistry , Administration, Cutaneous , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/pathology , Cell Survival/drug effects , Disease Models, Animal , Docetaxel/blood , Docetaxel/pharmacokinetics , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Particle Size , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Skin/drug effects , Skin/metabolism , Swine , Tissue Distribution , Transplantation, Homologous , Treatment Outcome , Tumor Burden/drug effects
13.
Mol Pharm ; 16(11): 4519-4529, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31509418

ABSTRACT

Amphotericin B (AmB) is one of the most effective drugs used in the treatment of leishmaniasis and systemic fungal infections. Considering the global burden of leishmaniasis, ∼90% of disease cases occur in developing countries, suggestive of the need for an affordable AmB therapy. However, owing to the physicochemical properties of AmB, all the clinically available formulations must be administered by intravenous route, thereby creating a significant hurdle in patients' access to AmB due to pharmacoeconomic considerations. We have previously demonstrated that lipid conjugation (e.g., fatty acids) to AmB significantly decreases the toxicity of resulting prodrug by a favorable alteration in the aggregation pattern. The hypothesis of the present work was to investigate the potential of the previously established AmB-lipid conjugate [AmB-oleyl conjugate (AmB-OA)] in improving the physicochemical properties such as gastric instability and lower intestinal permeability that otherwise limits the oral delivery of AmB. The synthesized AmB-OA conjugate was remarkably stable at gastric pH in contrast to AmB and exhibited significantly higher permeation across the Caco-2 monolayer (indicative of intestinal permeability). Mechanistic studies revealed that AmB-OA retained an equivalent antifungal activity. Also, AmB-OA was found to interact preferentially with intracellular membranes of Saccharomyces cerevisiae, while AmB interacted with the plasma membrane. The results of Caco-2 monolayer permeation experiments were further confirmed by in vivo pharmacokinetics, which showed that AmB-OA exhibited a 3.13-fold increase in the Cmax and a 4.88-fold increase in AUCTot as compared to AmB. In conclusion, the lipid conjugation approach may provide an effective solution for current challenges in designing drug delivery systems intended for oral AmB therapy.


Subject(s)
Amphotericin B/chemistry , Amphotericin B/pharmacokinetics , Fatty Acids/chemistry , Gastrointestinal Tract/metabolism , Administration, Oral , Amphotericin B/pharmacology , Animals , Antifungal Agents/chemistry , Antifungal Agents/pharmacokinetics , Antifungal Agents/pharmacology , Biological Availability , Caco-2 Cells , Cell Line, Tumor , Cell Membrane/metabolism , Chemistry, Pharmaceutical/methods , Drug Delivery Systems/methods , Humans , Lipids/chemistry , Male , Rats, Sprague-Dawley , Saccharomyces cerevisiae/drug effects
14.
Nanomedicine (Lond) ; 14(16): 2227-2241, 2019 08.
Article in English | MEDLINE | ID: mdl-31397197

ABSTRACT

Aim: To design a nanocarrier platform for enhanced transdermal drug permeation. Materials & methods: Gel-based high permeation vesicles (HPVs) were developed and their performance in terms of transdermal flux improvement, in vitro release and skin irritancy was assessed. The mechanistic insights of permeation enhancement were explored using confocal laser scanning microscopy, ATR-FTIR, DSC and P31 NMR. Results: HPVs exhibited as vesicles with uniform size (∼150 nm), extended drug-release profile (∼48 h) and improved transdermal flux. HPVs were also nontoxic and nonirritant to skin. Enhanced vesicle deformability, improved vesicle membrane fluidity and synergistic permeation enhancement action of synergistic combination of permeation enhancer components was found to be responsible for HPV-mediated permeation enhancement. Conclusion: Overall, the study established that HPVs demonstrate a promising therapeutic advantage over conventional transdermal drug carriers.


Subject(s)
Drug Delivery Systems/methods , Microscopy, Confocal/methods , Skin/metabolism , Animals , Cell Line, Tumor , Humans , Magnetic Resonance Spectroscopy , Rats , Rats, Sprague-Dawley , Skin Absorption , Spectroscopy, Fourier Transform Infrared , Swine
15.
AAPS PharmSciTech ; 20(2): 41, 2019 Jan 04.
Article in English | MEDLINE | ID: mdl-30610658

ABSTRACT

Oral drug delivery route is one of the most convenient and extensively utilised routes for drug administration. But there exists class of drugs which exhibit poor bioavailability on oral drug administration. Designing of drug-lipid conjugates (DLCs) is one of the rationale strategy utilised in overcoming this challenge. This review extensively covers the various dimensions of drug modification using lipids to attain improved oral drug delivery. DLCs help in improving oral delivery by providing benefits like improved permeability, stability in gastric environment, higher drug loading in carriers, formation of self-assembled nanostructures, etc. The clinical effectiveness of DLCs is highlighted from available marketed drug products along with many DLCs in phase of clinical trials. Conclusively, this drug modification strategy can potentially help in augmenting oral drug delivery in future.


Subject(s)
Drug Delivery Systems , Lipids/chemistry , Administration, Oral , Animals , Drug Delivery Systems/methods , Humans , Nanostructures/chemistry , Permeability
16.
ACS Biomater Sci Eng ; 5(7): 3361-3372, 2019 Jul 08.
Article in English | MEDLINE | ID: mdl-33405578

ABSTRACT

PEGylation (i.e., attachment of polyethylene-glycol) of carbon nanotubes (CNTs) is one of the most widely used strategies to improve its biocompatibility and aqueous dispersion stability, which are critical for their successful clinical application. However, PEGylation of nanomaterials has recently been associated with production of anti-PEG antibody, low cellular uptake, and degradation. Herein, we explore surface functionalization of CNTs using the bovine-milk-derived protein succinylated ß-lactoglobuline (Sblg) as an alternative strategy to PEGylation. The aqueous dispersion stability, in vitro cell uptake and biocompatibility of Sblg-functionalized multiwalled CNTs (Sblg-f-MWCNTs) was compared to PEGylated MWCNTs (PEG-f-MWCNTs). The surface functionalization with Sblg was found to improve the IC50 values of CNTs by ∼5- to 6-fold in comparison with pristine CNTs in various cell lines. Both Sblg-f-MWCNTs and PEG-f-MWCNTs improved the aqueous colloidal stability of CNTs, which remained suspended for a period of one month. Our study concluded that the Sblg provides a cost-effective alternative to the PEG-based CNT functionalization with significant improvement in the biocompatibility and dispersion stability of CNTs.

17.
Mol Pharm ; 15(6): 2391-2402, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29747513

ABSTRACT

miR-34a is a master tumor suppressor playing a key role in the several signaling mechanisms involved in cancer. However, its delivery to the cancer cells is the bottleneck in its clinical translation. Herein we report cationic amphiphilic copolymers grafted with cholesterol (chol), N, N-dimethyldipropylenetriamine (cation chain) and 4-(2-aminoethyl)morpholine (morph) for miR-34a delivery. The copolymer interacts with miR-34a at low N/P ratios (∼2/1) to form nanoplexes of size ∼108 nm and a zeta potential ∼ +39 mV. In vitro studies in 4T1 and MCF-7 cells indicated efficient transfection efficiency. The intracellular colocalization suggested that the copolymer effectively transported the FAM labeled siRNA into the cytoplasm within 2 h and escaped from the endo-/lysosomal environment. The developed miR-34a nanoplexes inhibited the breast cancer cell growth as confirmed by MTT assay wherein 28% and 34% cancer cell viability was observed in 4T1 and MCF-7 cells, respectively. Further, miR-34a nanoplexes possess immense potential to induce apoptosis in both cell lines.


Subject(s)
Breast Neoplasms/therapy , Drug Carriers/chemistry , Genetic Therapy/methods , MicroRNAs/administration & dosage , Animals , Apoptosis/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Survival/genetics , Cholesterol/chemistry , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , MicroRNAs/genetics , Morpholines/chemistry , Polymers/chemistry , Transfection
18.
J Control Release ; 271: 60-73, 2018 02 10.
Article in English | MEDLINE | ID: mdl-29273320

ABSTRACT

Cancer remained a major cause of death providing diversified challenges in terms of treatment including non-specific toxicity, chemoresistance and relapse. Nanotechnology- based delivery systems grabbed tremendous attention for delivering cancer therapeutics as they provide benefits including controlled drug release, improved biological half-life, reduced toxicity and targeted delivery. Majority of the nanocarriers consists of either a polymer or a lipid component along with other excipients to stabilize the colloidal system. Lipid-based systems provide advantages like better entrapment efficiency, scalability and low- cost raw materials, however, suffer from limitations including instability, a burst release of the drug, and limited surface functionalization. On the other hand, polymeric systems provide an excellent diversity of chemical modifications, stability, controlled release, however limited drug loading capacities and scale up limit their use. Hybrid nanocarriers consisting of lipid and polymer were able to overcome some of these disadvantages while retaining the advantages of both the systems. Designing a stable lipid-polymer hybrid system requires a thorough understanding of the material properties and their behavior in in vitro and in vivo environments. This review highlights the current status and future prospects of lipid-polymer hybrid systems with a particular focus on cancer nanotherapeutics.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/administration & dosage , Lipids/administration & dosage , Nanoparticles/administration & dosage , Polymers/administration & dosage , Animals , Graphite/administration & dosage , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...