Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
EJNMMI Radiopharm Chem ; 9(1): 61, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39162901

RESUMO

BACKGROUND: This study aimed to develop a novel positron emission tomography (PET) tracer, [68Ga]Ga-TD-01, for CXCR4 imaging. To achieve this goal, the molecular scaffold of TIQ15 was tuned by conjugation with the DOTA chelator to make it suitable for 68Ga radiolabeling. METHODS: A bifunctional chelator was prepared by conjugating the amine group of TIQ15 with p-NCS-Bz-DOTA, yielding TD-01, with a high yield (68.92%). TD-01 was then radiolabeled with 68Ga using 0.1 M ammonium acetate at 60 °C for 10 min. A 1-h dynamic small animal PET/MRI study of the labeled compound in GL261-luc2 tumor-bearing mice was performed, and brain tumor uptake was assessed. Blocking studies involved pre-administration of TIQ15 (10 mg/kg) 10 min before the PET procedure started. RESULTS: [68Ga]Ga-TD-01 exhibited a radiochemical yield (RCY) of 36.33 ± 1.50% (EOS), with a radiochemical purity > 99% and a molar activity of 55.79 ± 1.96 GBq/µmol (EOS). The radiotracer showed in vitro stability in PBS and human plasma for over 4 h. Biodistribution studies in healthy animals revealed favorable kinetics for subsequent PET pharmacokinetic modeling with low uptake in the brain and moderate uptake in lungs, intestines and spleen. Elimination could be assigned to a renal-hepatic pathway as showed by high uptake in kidneys, liver, and urinary bladder. Importantly, [68Ga]Ga-TD-01 uptake in glioblastoma (GBM)-bearing mice significantly decreased upon competition with TIQ15, with a baseline tumor-to-background ratios > 2.5 (20 min p.i.), indicating high specificity. CONCLUSION: The newly developed CXCR4 PET tracer, [68Ga]Ga-TD-01, exhibited a high binding inhibition for CXCR4, excellent in vitro stability, and favorable pharmacokinetics, suggesting that the compound is a promising candidate for full in vivo characterization of CXCR4 expression in GBM, with potential for further development as a tool in cancer diagnosis.

2.
J Drug Target ; 32(6): 587-605, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38634290

RESUMO

Nanoparticle (NP) functionalization with specific ligands enhances targeted cancer therapy and imaging by promoting receptor recognition and improving cellular uptake. This review focuses on recent research exploring the interaction between cancer cell-expressed chemokine receptor 4 (CXCR4) and ligand-conjugated NPs, utilising small molecules, peptides, and antibodies. Active NP targeting has shown improved tumour targeting and reduced toxicity, enabling precision therapy and diagnosis. However, challenges persist in the clinical translation of targeted NPs due to issues with biological response, tumour accumulation, and maintaining NP quality at an industrial scale. Biological and intratumoral barriers further hinder efficient NP accumulation in tumours, hampering translatability. To address these challenges, the academic community is refocusing efforts on understanding NP biological fate and establishing robust preclinical models. Future studies should investigate NP-body interactions, develop computational models, and identify optimal preclinical models. Establishing central NP research databases and fostering collaboration across disciplines is crucial to expediting clinical translation. Overcoming these hurdles will unlock the transformative potential of CXCR4-ligand-NP conjugates in revolutionising cancer treatment.


Assuntos
Nanopartículas , Neoplasias , Receptores CXCR4 , Receptores CXCR4/metabolismo , Receptores CXCR4/antagonistas & inibidores , Humanos , Neoplasias/tratamento farmacológico , Animais , Medicina de Precisão/métodos , Ligantes , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos/métodos
3.
ACS Pharmacol Transl Sci ; 7(1): 28-41, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38230275

RESUMO

Alpha7 nicotinic acetylcholine receptor (α7 nAChR) is an ion-gated calcium channel that plays a significant role in various aspects of cancer pathogenesis, particularly in lung cancer. Preclinical studies have elucidated the molecular mechanism underlying α7 nAChR-associated lung cancer proliferation, chemotherapy resistance, and metastasis. Understanding and targeting this mechanism are crucial for developing therapeutic interventions aimed at disrupting α7 nAChR-mediated cancer progression and improving treatment outcomes. Drug research and discovery have determined natural compounds and synthesized chemical antagonists that specifically target α7 nAChR. However, approved α7 nAChR antagonists for clinical use are lacking, primarily due to challenges related to achieving the desired selectivity, efficacy, and safety profiles required for effective therapeutic intervention. This comprehensive review provided insights into the molecular mechanisms associated with α7 nAChR and its role in cancer progression, particularly in lung cancer. Furthermore, it presents an update on recent evidence about α7 nAChR antagonists and addresses the challenges encountered in drug research and discovery in this field.

4.
ACS Appl Bio Mater ; 6(12): 5426-5441, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-37956113

RESUMO

In this study, we synthesized hollow porous iron oxide nanoparticles (HPIONPs) with surface modifications using polymers, specifically chitosan (Chi), polyethylene glycol (PEG), and alginate (Alg), to improve colloidal stability and biocompatibility. For colloidal stability, Alg-coated HPIONPs maintained size stability up to 24 h, with only an 18% increase, while Chi, PEG, and uncoated HPIONPs showed larger size increases ranging from 64 to 140%. The biocompatibility of polymer-coated HPIONPs was evaluated by assessing their cell viability, genotoxicity, and hemocompatibility. Across tested concentrations from 6.25 to 100 µg/mL, both uncoated and polymer-coated HPIONPs showed minimal cytotoxicity against three normal cell lines: RAW264.7, 3T3-L1, and MCF10A, with cell viability exceeding 80% at the highest concentration. Notably, polymer-coated HPIONPs exhibited nongenotoxicity based on the micronucleus assay and showed hemocompatibility, with only 2-3% hemolysis in mouse blood, in contrast to uncoated HPIONPs which exhibited 4-5%. Furthermore, we evaluated the cytotoxicity of HPIONPs on MDA-MB-231 breast cancer cells after a 2 h exposure to a stationary magnetic field, and the results showed the highest cell death of 38 and 29% when treated with uncoated and polymer-coated HPIONPs at 100 µg/mL, respectively. This phenomenon is attributed to iron catalyzing the Fenton and Haber-Weiss reactions, leading to reactive oxygen species (ROS)-dependent cell death (r ≥ 0.98). In conclusion, the hydrothermal synthesis and subsequent surface modification of HPIONPs with polymers showed improved colloidal stability, nongenotoxicity, and hemocompatibility compared to uncoated HPIONPs while maintaining closely similar levels of cytotoxicity against both normal and cancer cells. This research has paved the way for further exploration of polymer coatings to enhance the overall performance and safety profile of magnetic nanoparticles in delivering anticancer drugs.


Assuntos
Antineoplásicos , Quitosana , Camundongos , Animais , Polímeros/química , Porosidade , Polietilenoglicóis/química , Quitosana/química , Nanopartículas Magnéticas de Óxido de Ferro
5.
MethodsX ; 11: 102318, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37608960

RESUMO

The current literature mostly contains relatively vague descriptions of techniques for implementing in vitro magnetic targeting delivery of iron oxide nanoparticles (IONPs), leading to irreproducible processes and incomparable findings. This discrepancy often arises from the varying exposure of IONPs to the non-uniform magnetic field and differences in the concentration of the polymer-coated IONPs. Hence, we meticulously designed and built a system comprising a platform constructed from polyoxymethylene sheets, which securely holds the permanent magnets, and the cell culture plate. We also tailored the preparation process of the IONPs and the in vitro toxicity studies. The inherent characteristics of IONPs are further enhanced by their coating with natural polymers, alginate (Alg) and chitosan (CS).•The design and construction of the platform were carried out using a laser engraving/cutting machine along with graphic design software. The precise locations of the permanent magnets relative to the cell culture plate were determined via a Gaussmeter.•The quantities of the components in the formulation and the method for fabricating the CS/Alg-coated IONPs (CS/Alg-IONPs) were optimized to ensure that the desired physicochemical properties were obtained.•The cultivation and cytotoxicity evaluation of the fabricated CS/Alg-IONPs against MCF-7 breast cancer cells were described.

6.
Int J Biol Macromol ; 242(Pt 1): 124673, 2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37137353

RESUMO

Magnetic drug targeting can be a strategy for effectively delivering phytochemicals in cancer treatment. Here, we demonstrate the benefit of magnetic targeting with superparamagnetic iron oxide nanoparticles for cytotoxicity enhancement of lutein (LUT) against breast cancer cells. Fabrication of LUT-loaded chitosan/alginate iron oxide nanoparticles (LUT-CS/Alg-Fe3O4-NPs) was optimized by a statistical approach using response surface methodology based on the Box-Behnken design. The optimized LUT-CS/Alg-Fe3O4-NPs with a balance among LUT concentration, copolymer coating, and iron ion concentration exhibited controlled size, narrow size distribution, better crystallinity, excellent saturation magnetization, and sustained-release profile. The negligible magnetic coercivity and remanent magnetization confirmed the superparamagnetism of the prepared NPs. The optimized LUT-CS/Alg-Fe3O4-NPs were biocompatible while exhibiting a significantly enhanced cytotoxicity towards breast cancer MCF-7 cells upon exposure to a permanent magnet compared to free LUT with a 4-fold increase, suggesting the potential of LUT-CS/Alg-Fe3O4-NPs as magnetically targeted delivery for breast cancer.


Assuntos
Neoplasias da Mama , Quitosana , Nanopartículas , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Alginatos , Luteína
7.
Polymers (Basel) ; 14(24)2022 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-36559930

RESUMO

This study shows the effectiveness of magnetic-guide targeting in the delivery of curcumin diethyl γ-aminobutyrate (CUR-2GE), a prodrug of curcumin (CUR) previously synthesized to overcome unfavorable physicochemical properties of CUR. In this study, chitosan (Ch)-coated iron oxide nanoparticles (Ch-IONPs) were fabricated and optimized using Box-Behnken design-based response surface methodology for delivery of CUR-2GE. Ch was used as a coating material on the nanoparticle surface to avoid aggregation. The optimized condition for preparing Ch-IONPs consisted of using 4 mg Ch fabricated at pH 11 under a reaction temperature of 85 °C. The optimized Ch-IONPs were successfully loaded with CUR-2GE with sufficient loading capacity (1.72 ± 0.01%) and encapsulation efficiency (94.9 ± 0.8%). The obtained CUR-2GE-loaded Ch-IONPs (CUR-2GE-Ch-IONPs) exhibited desirable characteristics including a particle size of less than 50 nm based on TEM images, superparamagnetic property, highly crystalline IONP core, sufficient stability, and sustained-release profile. In the presence of permanent magnets, CUR-2GE-Ch-IONPs significantly increased cellular uptake and cytotoxicity toward MDA-MB-231 with a 12-fold increase in potency compared to free CUR-2GE, indicating the potential of magnetic-field assisted delivery of CUR-2GE-Ch-IONPs for the treatment of triple-negative breast cancer.

8.
ACS Pharmacol Transl Sci ; 5(9): 774-790, 2022 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-36110378

RESUMO

Curcumin is a naturally occurring polyphenol compound with potential analgesic effects. It has been shown to improve pain-like behaviors in numerous models of pain. Despite its potential, curcumin exhibits poor physicochemical and pharmacokinetic properties, which hinder its oral therapeutic efficacy. Curcumin diethyl γ-aminobutyrate (CUR-2GE), a carbamate prodrug of curcumin, was designed to overcome these limitations and demonstrated greater anti-neuroinflammatory effects compared to curcumin in vitro. Thus, this study evaluated the effect of CUR-2GE and its parent compound on pain-like behaviors in carrageenan- and LPS-induced mouse models. The possible side effects of CUR-2GE were also assessed by exploring its effects on motor coordination and spontaneous locomotor activity after acute and chronic treatments. The results showed that CUR-2GE improved mechanical and thermal hyperalgesia and locomotor activity to a greater extent than curcumin in carrageenan-induced mice. These results are in line with the ability of CUR-2GE to suppress peripheral inflammation in the paw tissue of carrageenan-induced mice, indicated by a significant decrease in TNF-α and IL-6 expression levels. Similarly, in LPS-induced mice, CUR-2GE improved sickness and pain-like behaviors (exploratory behaviors and long-term locomotor activity) to a greater extent than curcumin. Furthermore, CUR-2GE significantly reduced the level of proinflammatory cytokines in both the plasma and spinal cord tissue of LPS-induced mice, exhibiting significantly higher inhibition than curcumin. Moreover, the motor coordination, and locomotive behaviors of mice were not affected by both acute and chronic administration of CUR-2GE, indicating no potential CNS side effects. Thus, CUR-2GE demonstrated enhanced therapeutic efficacy in mouse models of inflammatory pain without any possible CNS side effects, suggesting its potential to be developed as an analgesic agent against inflammatory pain.

9.
Sci Rep ; 12(1): 9713, 2022 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-35690654

RESUMO

Metformin is a well-tolerated antidiabetic drug and has recently been repurposed for numerous diseases, including pain. However, a higher dose of metformin is required for effective analgesia, which can potentiate its dose-dependent gastrointestinal side effects. Curcumin is a natural polyphenol and has beneficial therapeutic effects on pain. Curcumin has been used as an analgesic adjuvant with several analgesic drugs, allowing synergistic antinociceptive effects. Nevertheless, whether curcumin can exert synergistic analgesia with metformin is still unknown. In the present study, the nature of curcumin-metformin anti-inflammatory interaction was evaluated in in vitro using lipopolysaccharide-induced RAW 264.7 macrophage and BV-2 microglia cells. In both macrophage and microglia, curcumin effectively potentiates the anti-inflammatory effects of metformin, indicating potential synergistic effects in both peripheral and central pathways of pain. The nature of the interaction between curcumin and metformin was further recapitulated using a mouse model of formalin-induced pain. Coadministration of curcumin and metformin at a 1:1 fixed ratio of their ED50 doses significantly reduced the dose required to produce a 50% effect compared to the theoretically required dose in phase II of the formalin test with a combination index value of 0.24. Besides, the synergistic interaction does not appear to involve severe CNS side effects indicated by no motor alterations, no alterations in short-term and long-term locomotive behaviors, and the general well-being of mice. Our findings suggest that curcumin exerts synergistic anti-inflammation with metformin with no potential CNS adverse effects.


Assuntos
Curcumina , Metformina , Analgésicos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Curcumina/farmacologia , Curcumina/uso terapêutico , Humanos , Metformina/uso terapêutico , Dor/tratamento farmacológico
10.
Life Sci ; 303: 120661, 2022 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-35643380

RESUMO

AIM: The C-X-C chemokine-receptor type 4 (CXCR4) is an emerging target for cancer drug discovery due to its high expression in cancer cells. The present study aimed to produce CXCR4 overexpressing HEK293T cells for a non-radioactive binding assay as a platform to identify drug candidates targeting CXCR4. MAIN METHODS: HEK293T cells stably expressing human CXCR4 were constructed by transfection of CXCR4 plasmids from the human CXCR4 gene. The CXCR4 overexpressing HEK293T cells were obtained by fluorescence-activated sorting and verified by conducting the competition binding assay of a known CXCR4 inhibitor, AMD3100 (plerixafor), to determine the IC50 value against monoclonal anti-human CD184 (hCD184) antibody tagged with fluorescence probe, phycoerythrin (PE). The non-radioactive binding assay using CXCR4 overexpressing HEK293T cells and PE-anti hCD184 was applied as a platform for identifying the target of natural compounds that exhibited cytotoxicity against cancer cell lines. KEY FINDINGS: The CXCR4 overexpressing HEK293T cells were produced with high expression (99.8%). The IC50 value of plerixafor determined by fluorescence tagged antibody-based competition assay using our developed cells agree with previously reported values using a radioligand binding assay. We observed no significant displacement of bound PE-anti-hCD184 by the test natural compounds which could be due to non-specific binding to other functional targets or organelles, low potency of the natural compounds, or binding to CXCR4 at deeper pockets. SIGNIFICANCE: The verified non-radioactive binding assay can serve as an alternative screening tool for anticancer lead compounds targeting CXCR4 and an essential tool for proof of mechanism study in the drug discovery.


Assuntos
Ciclamos , Compostos Heterocíclicos , Quimiocina CXCL12/metabolismo , Células HEK293 , Mobilização de Células-Tronco Hematopoéticas , Compostos Heterocíclicos/farmacologia , Humanos , Rim/metabolismo , Receptores CXCR4/metabolismo
11.
Molecules ; 27(12)2022 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-35745046

RESUMO

Oxyresveratrol (OXY) has been reported for its anti-inflammatory activity; however, the pharmaceutical applications of this compound are limited by its physicochemical properties and poor pharmacokinetic profiles. The use of an ester prodrug is a promising strategy to overcome these obstacles. In previous researches, several carboxylate esters of OXY were synthesized and oxyresveratrol tetraacetate (OXY-TAc) was reported to possess anti-melanogenic and anti-skin-aging properties. In this study, in addition to OXY-TAc, two novel ester prodrugs of OXY, oxyresveratrol tetrapropionate (OXY-TPr), and oxyresveratrol tetrabutyrate (OXY-TBu), were synthesized. Results from the Caco-2-permeation assay suggested that synthesized ester prodrugs can improve the membrane-permeation ability of OXY. The OXY-TAc exhibited the most significant profile, then this prodrug was chosen to observe anti-inflammatory activities with lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Our results showed that OXY-Tac significantly alleviated secretion of several pro-inflammatory mediators (nitric oxide (NO), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α)), mitigated expression of enzyme-regulated inflammation (inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2)), and suppressed the MAPK cascades. Interestingly, the observed anti-inflammatory activities of OXY-TAc were more remarkable than those of its parent compound OXY. Taken together, we demonstrated that OXY-TAc improved physicochemical and pharmacokinetic profiles and enhanced the pharmacological effects of OXY. Hence, the results in the present study would strongly support the clinical utilities of OXY-TAc for the treatment of inflammation-related disorders.


Assuntos
Lipopolissacarídeos , Pró-Fármacos , Animais , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/farmacologia , Células CACO-2 , Ciclo-Oxigenase 2/metabolismo , Ésteres/metabolismo , Ésteres/farmacologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Extratos Vegetais , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Células RAW 264.7 , Estilbenos
12.
Polymers (Basel) ; 14(9)2022 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-35567007

RESUMO

Turmeric oil (TO) exhibits various biological activities with limited therapeutic applications due to its instability, volatility, and poor water solubility. Here, we encapsulated TO in chitosan/alginate nanocapsules (CS/Alg-NCs) using o/w emulsification to enhance its physicochemical characteristics, using poloxamer 407 as a non-ionic surfactant. TO-loaded CS/Alg-NCs (TO-CS/Alg-NCs) were prepared with satisfactory features, encapsulation efficiency, release characteristics, and cytotoxicity against breast cancer cells. The average size of the fabricated TO-CS/Alg-NCs was around 200 nm; their distribution was homogenous, and their shapes were spherical, with smooth surfaces. The TO-CS/Alg-NCs showed a high encapsulation efficiency, of 70%, with a sustained release of TO at approximately 50% after 12 h at pH 7.4 and 5.5. The TO-CS/Alg-NCs demonstrated enhanced cytotoxicity against two breast cancer cells, MDA-MB-231 and MCF-7, compared to the unencapsulated TO, suggesting that CS/Alg-NCs are potential nanocarriers for TO and can serve as prospective candidates for in vivo anticancer activity evaluation.

13.
Carbohydr Polym ; 288: 119401, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35450653

RESUMO

Chitosan (Ch)-coated nanostructured lipid carriers (NLCs) have great potential for transdermal delivery with high localization of chemotherapeutics in breast cancer. This study used tetrahydrocurcumin (THC), a primary metabolite of curcumin with enhanced antioxidant and anticancer properties, as a model compound to prepare NLCs. Response surface methodology was employed to optimize THC-loaded Ch-coated NLCs (THC-Ch-NLCs) fabricated by high-shear homogenization. The optimized THC-Ch-NLCs had particle size of 244 ± 18 nm, zeta potential of -17.5 ± 0.5 mV, entrapment efficiency of 76.6 ± 0.2% and drug loading of 0.28 ± 0.01%. In vitro release study of THC-Ch-NLCs showed sustained release following the Korsmeyer-Peppas model with Fickian and non-Fickian diffusion at pH 7.4 and 5.5, respectively. THC-Ch-NLCs demonstrated significantly enhanced in vitro skin permeation, cell uptake, and remarkable cytotoxicity toward MD-MBA-231 breast cancer cells compared to the unencapsulated THC, suggesting Ch-NLCs as potential transdermal nanocarriers of THC for triple-negative breast cancer treatment.


Assuntos
Neoplasias da Mama , Quitosana , Curcumina , Nanoestruturas , Feminino , Humanos , Neoplasias da Mama/tratamento farmacológico , Quitosana/química , Curcumina/análogos & derivados , Curcumina/farmacologia , Portadores de Fármacos/química , Lipídeos/química , Nanoestruturas/química , Tamanho da Partícula
14.
PLoS One ; 17(3): e0265689, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35303012

RESUMO

Curcumin is a polyphenol compound that alleviates several neuroinflammation-related diseases including Alzheimer's disease, Parkinson's disease, multiple sclerosis, epilepsy and cerebral injury. However, the therapeutic efficacy of curcumin is limited by its poor physicochemical properties. The present study aimed to develop a new carrier-linked curcumin prodrug, curcumin diethyl γ-aminobutyrate (CUR-2GE), with improved physicochemical and anti-neuroinflammatory properties. CUR-2GE was designed and synthesized by conjugating curcumin with gamma-aminobutyric acid ethyl ester (GE) via a carbamate linkage. The carbamate linkage was selected to increase stability at acidic pH while GE served as a promoiety for lipophilic enhancement. The synthesized CUR-2GE was investigated for solubility, partition coefficient, stability, and bioconversion. The solubility of CUR-2GE was less than 0.05 µg/mL similar to that of curcumin, while the lipophilicity with log P of 3.57 was significantly increased. CUR-2GE was resistant to chemical hydrolysis at acidic pH (pH 1.2 and 4.5) as anticipated but rapidly hydrolyzed at pH 6.8 and 7.4. The incomplete hydrolysis of CUR-2GE was observed in simulated gastrointestinal fluids which liberated the intermediate curcumin monoethyl γ-aminobutyric acid (CUR-1GE) and the parent curcumin. In plasma, CUR-2GE was sequentially converted to CUR-1GE and curcumin within 1 h. In lipopolysaccharide (LPS)-stimulated BV-2 microglial cells, CUR-2GE effectively attenuated the pro-inflammatory mediators by decreasing the secretion of nitric oxide and cytokines (TNF-α and IL-6) to a greater extent than curcumin due to an increase in cellular uptake. Altogether, the newly developed acid-stable CUR-2GE prodrug is a potential pre-clinical and clinical candidate for further evaluation on neuroprotective and anti-neuroinflammatory effects.


Assuntos
Curcumina , Pró-Fármacos , Aminobutiratos , Carbamatos , Curcumina/química , Curcumina/farmacologia , Ésteres/farmacologia , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Ácido gama-Aminobutírico
15.
Pharmaceutics ; 15(1)2022 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-36678739

RESUMO

Folate receptors (FRs) highly expressed in breast cancers can be used as a recognized marker for preventing off-target delivery of chemotherapeutics. In this study, folic acid (FA)-grafted chitosan-alginate nanocapsules (CS-Alg-NCs) loaded with turmeric oil (TO) were developed for breast cancer targeting. CS was successfully conjugated with FA via an amide bond with a degree of substitution at 12.86%. The TO-loaded FA-grafted CS-Alg-NCs (TO-FA-CS-Alg-NCs) optimized by Box-Behnken design using response surface methodology had satisfactory characteristics with homogenous particle size (189 nm) and sufficient encapsulation efficiency and loading capacity (35.9% and 1.82%, respectively). In vitro release study of the optimized TO-FA-CS-Alg-NCs showed a sustained TO release following the Korsmeyer-Peppas model with a Fickian diffusion mechanism at pH 5.5 and 7.4. The TO-FA-CS-Alg-NCs showed lower IC50 than ungrafted TO-CS-Alg-NCs and unencapsulated TO against MDA-MB-231 and MCF-7 breast cancer cells, suggesting that FA-CS-Alg-NCs can improve anticancer activity of TO through its active targeting to the high FRs expressing breast cancers.

16.
Pharmaceutics ; 13(7)2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34201974

RESUMO

Curcumin (CUR) has been used as adjuvant therapy for therapeutic application in the treatment of psoriasis through several mechanisms of action. Due to the poor oral bioavailability of CUR, several approaches have been developed to overcome the limitations of CUR, including the prodrug strategy. In this study, CUR was esterified with mycophenolic acid (MPA) as a novel conjugate prodrug. The MPA-CUR conjugate was structurally elucidated using FT-IR, 1H-NMR, 13C-NMR, and MS techniques. Bioavailable fractions (BFs) across Caco-2 cells of CUR, MPA, and MPA-CUR were collected for further biological activity evaluation representing an in vitro cellular transport model for oral administration. The antipsoriatic effect of the BFs was determined using antiproliferation and anti-inflammation assays against hyperproliferation of tumor necrosis factor-alpha (TNF-α)-induced human keratinocytes (HaCaT). The BF of MPA-CUR provided better antiproliferation than that of CUR (p < 0.001). The enhanced hyperproliferation suppression of the BF of MPA-CUR resulted from the reduction of several inflammatory cytokines, including IL-6, IL-8, and IL-1ß. The molecular mechanisms of anti-inflammatory activity were mediated by an attenuated signaling cascade of MAPKs protein, i.e., p38, ERK, and JNK. Our results present evidence for the MPA-CUR conjugate as a promising therapeutic agent for treating psoriasis by antiproliferative and anti-inflammatory actions.

17.
Eur J Pharmacol ; 899: 174008, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33705800

RESUMO

Neuropathic pain is a debilitating chronic pain condition, and its treatment remains a clinical challenge. Curcumin, a naturally occurring phenolic compound, possesses diverse biological and pharmacological effects but has not yet been approved as a drug due to its low bioavailability. In order to overcome this limitation, we synthesized a potential ester prodrug of curcumin, curcumin diethyl diglutarate (CurDDG). In this study, we evaluated the pharmacological advantages of CurDDG over curcumin in a mouse model of chronic constriction injury (CCI), and the anti-inflammatory effect of CurDDG in LPS-induced RAW 264.7 macrophage cells was accessed to clarify the underline mechanism. Mice were treated with various oral doses of curcumin (25, 50, 100 and 200 mg/kg/day, daily for 14 days) or equimolar doses of CurDDG. CurDDG at all doses tested significantly attenuated CCI-induced thermal hyperalgesia and mechanical allodynia compared with the CCI-control group. CurDDG at 25, 50 and 100 mg/kg demonstrated significantly greater efficacy on both mechanical and thermal hypersensitivities compared to that of curcumin. The effect of CurDDG correlated well with the inhibition of TNF-α and IL-6 levels in both the sciatic nerve and the spinal cord, as compared to its respective control groups. Similarly, in the in vitro study, CurDDG significantly reduced the LPS-induced expression of TNF-α and IL-6. Moreover, CurDDG significantly decreased COX-2 and iNOS levels and attenuated p38, JNK, and ERK1/2 phosphorylation as compared to the curcumin-treated cells. Altogether, this study demonstrated the improved pharmacological effects of curcumin by its diglutarate conjugate, CurDDG.


Assuntos
Analgésicos/farmacologia , Anti-Inflamatórios/farmacologia , Curcumina/análogos & derivados , Curcumina/farmacologia , Glutaratos/farmacologia , Hiperalgesia/prevenção & controle , Limiar da Dor/efeitos dos fármacos , Pró-Fármacos/farmacologia , Nervo Isquiático/efeitos dos fármacos , Ciática/prevenção & controle , Medula Espinal/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , Células RAW 264.7 , Nervo Isquiático/metabolismo , Nervo Isquiático/fisiopatologia , Ciática/metabolismo , Ciática/fisiopatologia , Transdução de Sinais , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Succinatos , Fator de Necrose Tumoral alfa/metabolismo
18.
Pharmaceuticals (Basel) ; 13(9)2020 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-32867013

RESUMO

The drug treatment for neuropathic pain remains a challenge due to poor efficacy and patient satisfaction. Curcumin has been reported to alleviate neuropathic pain, but its clinical application is hindered by its low solubility and poor oral bioavailability. Curcumin diglutaric acid (CurDG) is a curcumin prodrug with improved water solubility and in vivo antinociceptive effects. In this study, we investigated the anti-inflammatory mechanisms underlying the analgesic effect of CurDG in the chronic constriction injury (CCI)-induced neuropathy mouse model. Repeated oral administration of CurDG at a low dose equivalent to 25 mg/kg/day produced a significant analgesic effect in this model, both anti-allodynic activity and anti-hyperalgesic activity appearing at day 3 and persisting until day 14 post-CCI surgery (p < 0.001) while having no significant effect on the motor performance. Moreover, the repeated administration of CurDG diminished the increased levels of the pro-inflammatory cytokines: TNF-α and IL-6 in the sciatic nerve and the spinal cord at the lowest tested dose (equimolar to 25 mg/kg curcumin). This study provided pre-clinical evidence to substantiate the potential of pursuing the development of CurDG as an analgesic agent for the treatment of neuropathic pain.

19.
Biochem Biophys Res Commun ; 521(4): 977-983, 2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31727368

RESUMO

Lung cancer, one of the most commonly found carcinoma type, has the highest mortality rate in cancer patients worldwide. Therapeutic interventions targeting to lung cancer become remaining the world significant challenge. Recently, the α7-nicotinic acetylcholine receptor (α7-nAChR) was reported to play an important role in the mechanism underlying lung cancer progression, being intriguing drug target for lung cancer therapy. Hence, the top four α7-nAChR antagonists (QND7, PPRD10, PPRD11 and PPRD12) among our previously developed ligands were proceeded to the in vitro anti-cancer evaluations in non-small cell lung cancer (NSCLC) cell lines (H460 and A549). In this study, we found that QND7 exhibited the highest cytotoxic effect and induced cell apoptosis in both cell lines at a level comparable to cisplatin, whereas the PPRD compounds showed much lower cytotoxicity. Low doses of QND7 and PPRD11 were able to suppress H460 and A549 cell proliferation, whereas PPRD10 and PPRD12 were considered ineffective. In an in vitro wound healing assay, QND7-treatment showed the greatest suppression of H460 and A549 cell migration. The variations in the anti-cancer activities of PPRD compounds might be, at least in part of, their non-selective antagonisms to serotonin receptor (5-HT3) and α4ß2-nAChR. Further investigation revealed that QND7 was able to minimize protein kinase B/mammalian target of rapamycin (Akt/mTOR) activity, in correlating to its anti-cancer effects. These findings warrant QND7 for further preclinical evaluation and demonstrate the potential of α7-nAChR as cancer drug target.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Movimento Celular/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinuclidinas/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Triazóis/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Quinuclidinas/química , Transdução de Sinais , Triazóis/química , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
20.
Molecules ; 21(9)2016 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-27657036

RESUMO

Vascular endothelial growth factor receptor 2 (VEGFR2) is a vital target for therapeutic intervention in cancer. We have recently described a computer-based drug design for a small molecule VEGFR2 inhibitor named VH02 (1-((1-(1H-indazol-6-yl)-1H-1,2,3-triazol-4-yl)methyl)-3-(3-chloromethylphenyl)urea). This study aimed to further explore the anti-angiogenic activity of VH02 both in vitro and in vivo. The in vitro assays include cell viability, capillary-like tube formation, MMP activity, and western blot analyses of signaling through VEGFR2 while the in vivo anti-angiogenic response were performed to evaluate the effect on vascularization in Matrigel plug applied in C57BL/6L mice. VH02 reduced angiogenesis behavior of EA.hy926 including cell viability, migration, adhesion, capillary-like tube formation, and MMP-2 activity induced by VEGF. Furthermore, VH02 regulated angiogenesis by directly inhibiting VEGFR2 on Tyr1175 signaling pathway leading to the inhibition of Akt-mediated cell survival and migration. Disruption of phosphorylation at VEGFR2-Tyr1175 by VH02 abolished FAK-Tyr397 signaling but not phosphorylation of p38 MAPK. This suggests that blockade of FAK by VH02 apparently associated with reduction of endothelial cell motility. Actin cytoskeleton rearrangement was diminished by VH02 in human endothelial cells. The anti-angiogenic effect of VH02 was confirmed in the in vivo model, revealing the reduction of vascular density in Matrigel plug after VH02 treatment. Additionally, the pericyte-like cells surrounding blood vessels in the plugs were significantly reduced as well as vascular density and p-Akt intensity. Our findings indicate that VH02 successfully inhibits VEGF-induced angiogenesis both in vitro and in vivo models. The compound could be further developed as an antiangiogenesis agent for cancer therapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA