Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Pharm ; 21(7): 3240-3255, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38785196

RESUMO

Inhibitors of a DNA repair enzyme known as polynucleotide kinase 3'-phosphatase (PNKP) are expected to show synergistic cytotoxicity in combination with topoisomerase I (TOP1) inhibitors in cancer. In this study, the synergistic cytotoxicity of a novel inhibitor of PNKP, i.e., A83B4C63, with a potent TOP1 inhibitor, i.e., SN-38, against colorectal cancer cells was investigated. Polymeric micelles (PMs) for preferred tumor delivery of A83B4C63, developed through physical encapsulation of this compound in methoxy poly(ethylene oxide)-poly(α-benzyl carboxylate-ε-caprolactone) (mPEO-b-PBCL) micelles, were combined with SN-38 in free or PM form. The PM form of SN-38 was prepared through chemical conjugation of SN-38 to the functional end group of mPEO-b-PBCL and further assembly of mPEO-b-PBCL-SN-38 in water. Moreover, mixed micelles composed of mPEO-b-PBCL and mPEO-b-PBCL-SN-38 were used to co-load A83B4C63 and SN-38 in the same nanoformulation. The loading content (% w/w) of the SN-38 and A83B4C63 to mPEO-b-PBCL in the co-loaded formulation was 7.91 ± 0.66 and 16.13 ± 0.11% (w/w), respectively, compared to 15.67 ± 0.34 (% w/w) and 23.06 ± 0.63 (% w/w) for mPEO-b-PBCL micelles loading individual drugs. Notably, the average diameter of PMs co-encapsulating both SN-38 and A83B4C63 was larger than that of PMs encapsulating either of these compounds alone but still lower than 60 nm. The release of A83B4C63 from PMs co-encapsulating both drugs was 76.36 ± 1.41% within 24 h, which was significantly higher than that of A83B4C63-encapsulated micelles (42.70 ± 0.72%). In contrast, the release of SN-38 from PMs co-encapsulating both drugs was 44.15 ± 2.61% at 24 h, which was significantly lower than that of SN-38-conjugated PMs (74.16 ± 3.65%). Cytotoxicity evaluations by the MTS assay as analyzed by the Combenefit software suggested a clear synergy between PM/A83B4C63 (at a concentration range of 10-40 µM) and free SN-38 (at a concentration range of 0.001-1 µM). The synergistic cytotoxic concentration range for SN-38 was narrowed down to 0.1-1 or 0.01-1 µM when combined with PM/A83B4C63 at 10 or 20-40 µM, respectively. In general, PMs co-encapsulating A83B4C63 and SN-38 at drug concentrations within the synergistic range (10 µM for A83B4C63 and 0.05-1 µM for SN-38) showed slightly less enhancement of SN-38 anticancer activity than a combination of individual micelles, i.e., A83B4C63 PMs + SN-38 PMs at the same molar concentrations. This was attributed to the slower release of SN-38 from the SN-38 and A83B4C63 co-encapsulated PMs compared to PMs only encapsulating SN-38. Cotreatment of cells with TOP1 inhibitors and A83B4C63 formulation enhanced the expression level of γ-HA2X, cleaved PARP, caspase-3, and caspase-7 in most cases. This trend was more consistent and notable for PMs co-encapsulating both A83B4C63 and SN-38. The overall result from the study shows a synergy between PMs of SN-38 and A83B4C63 as a mixture of two PMs for individual drugs or PMs co-encapsulating both drugs.


Assuntos
Neoplasias Colorretais , Irinotecano , Micelas , Inibidores da Topoisomerase I , Humanos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Irinotecano/farmacologia , Irinotecano/administração & dosagem , Inibidores da Topoisomerase I/farmacologia , Inibidores da Topoisomerase I/administração & dosagem , Inibidores da Topoisomerase I/química , Linhagem Celular Tumoral , Animais , Camundongos , Nanomedicina/métodos , Sinergismo Farmacológico , DNA Topoisomerases Tipo I/metabolismo , Nanopartículas/química , Ensaios Antitumorais Modelo de Xenoenxerto , Poliésteres/química , Fosfotransferases (Aceptor do Grupo Álcool) , Enzimas Reparadoras do DNA
2.
Eur Biophys J ; 53(4): 171-181, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38597963

RESUMO

Polymeric micelles are nanocarriers for drug, protein and gene delivery due to their unique core/shell structure, which encapsulates and protects therapeutic cargos with diverse physicochemical properties. However, information regarding the micellar nanoenvironment's fluidity can provide unique insight into their makeup. In this study, we used electron paramagnetic resonance (EPR) spectroscopy to study free radical spin probe (5-doxylstearate methyl ester, 5-MDS, and 16-doxylstearic acid, 16-DS) behaviour in methoxy-poly(ethylene oxide)-poly(α-benzyl carboxylate-ε-caprolactone) (PEO-PBCL) and methoxy-poly(ethylene oxide)-poly(ε-caprolactone) (PEO-PCL) polymeric micelles. Spin probes provided information about the spectroscopic rotational correlation time (τ, s) and the spectroscopic partition parameter F. We hypothesized that spin probes would partition into the polymeric micelles, and these parameters would be calculated. The results showed that both 5-MDS and 16-DS spectra were modulated in the presence of polymeric micelles. Based on τ values, 5-MDS revealed that PEO-PCL (τ = 3.92 ± 0.26 × 10-8 s) was more fluid than PEO-PBCL (τ = 7.15 ± 0.63 × 10-8 s). The F parameter, however, could not be calculated due to the rotational hindrance of the probe within the micelles. With 16-DS, more probe rotation was observed, and although the F parameter could be calculated, it was not helpful to distinguish the micelles' fluidity. Also, doxorubicin-loading interfered with the spin probes, particularly for 16-DS. However, using simulations, we could distinguish the hydrophilic and hydrophobic components of the 16-DS probe. The findings suggest that EPR spectroscopy is a valuable method for determining core fluidity in polymeric micelles.


Assuntos
Micelas , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Poliésteres/química , Polietilenoglicóis/química , Marcadores de Spin , Polímeros/química
3.
Mol Pharm ; 20(11): 5359-5382, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37769017

RESUMO

Local drug delivery to the eye through conventional means has faced many challenges due to three essential barriers: (a) the complex structure of the cornea limiting drug absorption, (b) the capacity of ocular absorptive cells in drug metabolism, and (c) the washing effect of eye tears. Polymeric micelles (PMs) have been the focus of much interest for ocular drug delivery due to several advantages they provide for this application, including the capacity for the solubilization of hydrophobic drugs, nonirritability, nanoscopic diameter, and the clarity of their aqueous solution not interfering with vision. The potential to increase the release and residence time of incorporated medication at the site of absorption is also a bonus advantage for these delivery systems. This Review covers research conducted on single or mixed micelles prepared from small amphiphilic molecules, copolymers (diblock, triblock, and graft), and gel systems containing micelles. The purpose of this review is to provide an update on the status of micellar ocular delivery systems for different indications, with a focus on preclinical and clinical drug development. In this context, we are discussing the anatomy of the eye, various ocular barriers, different micellar formulations, and their benefits in ocular drug delivery, as well as the role of PMs in the management of ocular diseases both in preclinical models and in clinic. The encouraging preclinical effectiveness findings from experiments conducted in both laboratory settings and live animals have paved the way for the advancement of micellar systems in clinical trials for ocular administration and the first nanomicallar formulation approved for clinical use by the United States Food and Drug Administration (marketed as Cequa by Sun Pharmaceuticals).


Assuntos
Portadores de Fármacos , Micelas , Animais , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Polímeros/química , Córnea
4.
Mol Pharm ; 20(7): 3570-3577, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37307328

RESUMO

Selective delivery of chemotherapy to the tumor site while sparing healthy cells and tissues is an attractive approach for cancer treatment. Carriers such as peptides can facilitate selective tumor targeting and payload delivery. Peptides with specific affinity for the overexpressed cell-surface receptors in cancer cells are conjugated to chemotherapy to afford peptide-drug conjugates (PDCs) that show selective uptake by cancer cells. Using a 10-mer linear peptide (WxEAAYQrFL) called 18-4 that targets and binds breast cancer cells, we designed a peptide 18-4-doxorubicin (Dox) conjugate with high specific toxicity toward triple-negative breast cancer (TNBC) MDA-MB-231 cells and 30-fold lower toxicity to normal breast MCF10A epithelial cells. Here, we elucidate the in vivo activity of this potent and tumor-selective peptide 18-4-Dox conjugate in mice bearing orthotopic MDA-MB-231 tumors. Mice treated with four weekly injections of the conjugate showed significantly lower tumor volumes compared to mice treated with free Dox at an equivalent Dox dose. Immunohistochemical (IHC) analysis of mice tissues revealed that treatment with a low dose of PDC (2.5 mg/kg of Dox equiv) reduced the expression of proliferation markers (PCNA and Ki-67) and increased apoptosis (evidenced by increased caspase-3 expression). At the same dose of free Dox (2.5 mg/kg), the expression of these markers was similar to that of saline treatment. Accordingly, significantly more Dox accumulated in tumors of conjugate-treated mice (7-fold) compared to the Dox-treated mice, while lower levels of Dox were observed in the liver, heart, and lungs of peptide-Dox conjugate-treated mice (up to 3-fold less) than Dox-treated mice. The IHC analysis of keratin 1 (K1), the receptor for peptide 18-4, revealed K1 upregulation in tumors and low levels in normal mammary fat pad and liver tissues from mice, suggesting preferential uptake of PDCs by TNBC to be K1 receptor-mediated. Taken together, our data support the use of a PDC approach to deliver chemotherapy selectively to the TNBC to inhibit tumor growth.


Assuntos
Neoplasias da Mama , Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Feminino , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Queratina-1 , Sistemas de Liberação de Medicamentos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Peptídeos/uso terapêutico , Linhagem Celular Tumoral , Neoplasias da Mama/tratamento farmacológico
5.
Mol Pharm ; 19(6): 1825-1838, 2022 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-35271294

RESUMO

The disruption of polynucleotide kinase/phosphatase (PNKP) in colorectal cancer (CRC) cells deficient in phosphatase and tensin homolog (PTEN) is expected to lead to the loss of cell viability by a process known as synthetic lethality. In previous studies, we have reported on the encapsulation of a novel inhibitor of PNKP, namely, A83B4C63, in polymeric micelles and its activity in slowing the growth of PTEN-deficient CRC cells as well as subcutaneous xenografts. In this study, to enhance drug delivery and specificity to CRC tumors, the surface of polymeric micelles carrying A83B4C63 was modified with GE11, a peptide targeting epidermal growth factor receptor (EGFR) overexpressed in about 70% of CRC tumors. Using molecular dynamics (MD) simulations, we assessed the binding site and affinity of GE11 for EGFR. The GE11-modified micelles, tagged with a near-infrared fluorophore, showed enhanced internalization by EGFR-overexpressing CRC cells in vitro and a trend toward increased primary tumor homing in an orthotopic CRC xenograft in vivo. In line with these observations, the GE11 modification of polymeric micelles was shown to positively contribute to the improved therapeutic activity of encapsulated A83B4C63 against HCT116-PTEN-/- cells in vitro and that of orthotopic CRC xenograft in vivo. In conclusion, our results provided proof of principle evidence for the potential benefit of EGFR targeted polymeric micellar formulations of A83B4C63 as monotherapeutics for aggressive and metastatic CRC tumors but at the same time highlighted the need for the development of EGFR ligands with improved physiological stability and EGFR binding.


Assuntos
Neoplasias Colorretais , Micelas , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , Receptores ErbB/metabolismo , Xenoenxertos , Humanos , Fosfotransferases (Aceptor do Grupo Álcool) , Polímeros/química , Distribuição Tecidual
6.
Curr Issues Mol Biol ; 43(1): 313-323, 2021 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-34201211

RESUMO

Primary myeloma (PM) cells are short-lived in conventional culture, which limited their usefulness as a study model. Here, we evaluated if three-dimensional (3D) culture can significantly prolong the longevity of PM cells in-vitro. We employed a previously established 3D model for culture of bone marrow mononuclear cells isolated from 15 patients. We assessed the proportion of PM cells, viability and proliferation using CD38 staining, trypan blue exclusion assays and carboxy fluorescein succinimidyl ester (CFSE) staining, respectively. We observed significantly more CD38+ viable cells in 3D than in conventional culture (65% vs. 25%, p = 0.006) on day 3. CFSE staining showed no significant difference in cell proliferation between the two culture systems. Moreover, we found that PM cells in 3D culture are more STAT3 active by measure of pSTAT3 staining (66% vs. 10%, p = 0.008). Treatment of IL6, a STAT3 activator significantly increased CD38+ cell viability (41% to 68%, p = 0.021). In comparison, inhibition of STAT3 with Stattic significantly decreased PM cell viability in 3D culture (38% to 17% p = 0.010). Neither IL6 nor Stattic affected the PM cell viability in conventional culture. This study suggests that 3D culture can significantly improve the longevity of PM cells in-vitro, and STAT3 activation can further improve their viability.


Assuntos
Medula Óssea/patologia , Técnicas de Cultura de Células , Sobrevivência Celular , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/fisiopatologia , Fator de Transcrição STAT3/metabolismo , ADP-Ribosil Ciclase 1/biossíntese , Idoso , Proliferação de Células , Células Cultivadas , Óxidos S-Cíclicos/farmacologia , Feminino , Fluoresceínas/farmacologia , Humanos , Técnicas In Vitro , Leucócitos Mononucleares/citologia , Masculino , Glicoproteínas de Membrana/biossíntese , Pessoa de Meia-Idade , Succinimidas/farmacologia
7.
Mol Pharm ; 18(5): 1862-1894, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33787270

RESUMO

Albumin is an appealing carrier in nanomedicine because of its unique features. First, it is the most abundant protein in plasma, endowing high biocompatibility, biodegradability, nonimmunogenicity, and safety for its clinical application. Second, albumin chemical structure and conformation allows interaction with many different drugs, potentially protecting them from elimination and metabolism in vivo, thus improving their pharmacokinetic properties. Finally, albumin can interact with receptors overexpressed in many diseased tissues and cells, providing a unique feature for active targeting of the disease site without the addition of specific ligands to the nanocarrier. For this reason, albumin, characterized by an extended serum half-life of around 19 days, has the potential of promoting half-life extension and targeted delivery of drugs. Therefore, this article focuses on the importance of albumin as a nanodrug delivery carrier for hydrophobic drugs, taking advantage of the passive as well as active targeting potential of this nanocarrier. Particular attention is paid to the breakthrough NAB-Technology, with emphasis on the advantages of Nab-Paclitaxel (Abraxane), compared to the solvent-based formulations of Paclitaxel, i.e., CrEL-paclitaxel (Taxol) in a clinical setting. Finally, the role of albumin in carrying anticancer compounds is depicted, with a particular focus on the albumin-based formulations that are currently undergoing clinical trials. The article sheds light on the power of an endogenous substance, such as albumin, as a drug delivery system, signifies the importance of the drug vehicle in drug performance in the biological systems, and highlights the possible future trends in the use of this drug delivery system.


Assuntos
Antineoplásicos/administração & dosagem , Portadores de Fármacos/farmacocinética , Nanopartículas/química , Neoplasias/tratamento farmacológico , Albumina Sérica Humana/farmacocinética , Albuminas/administração & dosagem , Albuminas/química , Albuminas/farmacocinética , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Modelos Animais de Doenças , Portadores de Fármacos/química , Meia-Vida , Humanos , Interações Hidrofóbicas e Hidrofílicas , Paclitaxel/administração & dosagem , Paclitaxel/química , Paclitaxel/farmacocinética , Albumina Sérica Humana/química
8.
Pharm Res ; 38(1): 51-65, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33559046

RESUMO

PURPOSE: We have previously reported on a polymeric micellar formulation of Cyclosporine A (CyA) based on poly(ethylene oxide)-block-poly(ε-caprolactone) (PEO5K-b-PCL13K) capable of changing drug biodistribution and pharmacokinetic profile following intravenous administration. The objective of the present study was to explore the potential of this formulation in changing the tissue distribution and pharmacokinetics of the encapsulated CyA following oral administration making comparisons with Sandimmune®. METHODS: The in vitro CyA release and stability CyA-loaded PEO-b-PCL micelles (CyA-micelles) were evaluated in biorelevant media. The pharmacokinetics and tissue distribution of orally administered CyA-micelles or Sandimmune® and tissue distribution of traceable Cyanine-5.5 (Cy5.5)-conjugated PEO-b-PCL micelles were then investigated in healthy rats. RESULTS: CyA-micelles showed around 60-70% CyA release in simulated intestinal and gastric fluids within 24 h, while Sandimmune® released its entire CyA content in the simulated intestinal fluid. CyA-micelles and Sandimmune® showed similar pharmacokinetics, but different tissue distribution profile in rats. In particular, the calculated AUC for CyA-micelles was higher in liver, comparable in heart, and lower in spleen, lungs, and kidneys when compared to that for Sandimmune®. CONCLUSIONS: The results point to the influence of excipients in Sandimmune® on CyA disposition and more inert nature of PEO-b-PCL micelles in defining CyA biological interactions.


Assuntos
Ciclosporina/farmacocinética , Portadores de Fármacos/química , Poliésteres/química , Administração Oral , Animais , Ciclosporina/administração & dosagem , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Excipientes/química , Masculino , Micelas , Modelos Animais , Ratos , Distribuição Tecidual
9.
J Pharm Pharm Sci ; 24: 344-362, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34224665

RESUMO

PURPOSE: The ultimate goal of this study is to develop a novel delivery system for a new potent cytotoxic compound, CCI-001, with anti-b tubulin activity, so that the drug can be effectively administered and at the same time its harmful side effects can be reduced. METHODS: In the current study, CCI-001 was loaded into serum albumin (SA), using a modified desolvation method, generating CCI-001-SA nanoparticles. Both bovine and human SA were used for the encapsulation of this drug candidate. Optimum conditions for drug loading were achieved when already formed and crosslinked albumin nanoparticles were incubated overnight at 37°C with CCI-001 solutions. The CCI-001-loaded albumin nanoparticles were assessed for average particle diameter and polydispersity, zeta potential, drug loading, in vitro release, morphology and cell toxicity against SW620 and HCT116 colorectal cancer cells. RESULTS: The spherical nanoparticles obtained were negatively charged (~ -30 mV) and had an average diameter of ~ 130 nm, with a narrow size distribution. The in vitro release of CCI-001 from the albumin nanoparticles showed a sustained release pattern over 24 hours without any initial burst release, compared to the fast release of the free drug under experimental conditions. No difference between the SA from the two species in terms of CCI-001 loading was observed. However, a significant difference was observed between the release profiles of CCI-001 from drug-loaded HSA and drug-loaded BSA nanoparticles with HSA nanoparticles showing slower drug release (mean release time, MRT, values of 5.14 ± 0.33 h and 6.88 ± 0.15 h for BSA-NPs and HSA-NPs, respectively, P < 0.01). Cellular toxicity studies showed higher cytotoxicity for CCI-001-SA compared to the free drug (IC50s of 0.62 ± 0.31 nM vs 2.06 ± 0.29 nM in SW620 cells and 0.9 ± 0.1 nM vs 4.2 ± 0.2 nM in HCT116 cells, for CCI-001-HSA NPs and free drug, respectively). Therefore, despite the low drug content level in the HSA nanoparticles of CCI-001, the formulation provides relevant concentrations for further in vivo studies in animal models due to high drug potency. CONCLUSIONS: The data support the potential use of albumin as a nanocarrier for CCI-001 in biological systems.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Nanopartículas , Moduladores de Tubulina/farmacologia , Animais , Bovinos , Linhagem Celular Tumoral , Química Farmacêutica , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Células HCT116 , Humanos , Tamanho da Partícula , Soroalbumina Bovina/química , Albumina Sérica Humana/química , Moduladores de Tubulina/administração & dosagem , Moduladores de Tubulina/química
10.
Molecules ; 26(9)2021 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-34064416

RESUMO

The objective of this study was to synthesize and characterize a set of biodegradable block copolymers based on TPGS-block-poly(ε-caprolactone) (TPGS-b-PCL) and to assess their self-assembled structures as a nanodelivery system for paclitaxel (PAX). The conjugation of PCL to TPGS was hypothesized to increase the stability and the drug solubilization characteristics of TPGS micelles. TPGS-b-PCL copolymer with various PCL/TPGS ratios were synthesized via ring opening bulk polymerization of ε-caprolactone using TPGS, with different molecular weights of PEG (1-5 kDa), as initiators and stannous octoate as a catalyst. The synthesized copolymers were characterized using 1H NMR, GPC, FTIR, XRD, and DSC. Assembly of block copolymers was achieved via the cosolvent evaporation method. The self-assembled structures were characterized for their size, polydispersity, and CMC using dynamic light scattering (DLS) technique. The results from the spectroscopic and thermal analyses confirmed the successful synthesis of the copolymers. Only copolymers that consisted of TPGS with PEG molecular weights ≥ 2000 Da were able to self-assemble and form nanocarriers of ≤200 nm in diameter. Moreover, TPGS2000-b-PCL4000, TPGS3500-b-PCL7000, and TPGS5000-b-PCL15000 micelles enhanced the aqueous solubility of PAX from 0.3 µg/mL up to 88.4 ug/mL in TPGS5000-b-PCL15000. Of the abovementioned micellar formulations, TPGS5000-b-PCL15000 showed the slowest in vitro release of PAX. Specifically, the PAX-loaded TPGS5000-b-PCL15000 micellar formulation showed less than 10% drug release within the first 12 h, and around 36% cumulative drug release within 72 h compared to 61% and 100% PAX release, respectively, from the commercially available formulation (Ebetaxel®) at the same time points. Our results point to a great potential for TPGS-b-PCL micelles to efficiently solubilize and control the release of PAX.


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , Paclitaxel/farmacologia , Poliésteres/química , Vitamina E/química , Varredura Diferencial de Calorimetria , Cromatografia em Gel , Preparações de Ação Retardada , Liberação Controlada de Fármacos , Micelas , Nanopartículas/ultraestrutura , Tamanho da Partícula , Poliésteres/síntese química , Espectroscopia de Prótons por Ressonância Magnética , Solubilidade , Espectroscopia de Infravermelho com Transformada de Fourier , Vitamina E/síntese química , Água/química , Difração de Raios X
11.
Mol Pharm ; 17(4): 1377-1386, 2020 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-32023064

RESUMO

In this study, we tested whether the extent of drug presence in the heart contributes to the elevated cardiovascular risk of nonsteroidal anti-inflammatory drugs (NSAIDs). A fluorescently tagged nanoformulation of an NSAID with high cardiovascular (CV) risk (diclofenac) was developed as diclofenac ethyl ester (DFEE) encapsulated in traceable (cyanine-5.5-labeled) polymeric micelles (DFEE-TM) based on methoxypoly(ethylene oxide)-block-poly(ε-caprolactone)(PEO-b-PCL) (MW, 5000:3500 g/mol). Diclofenac pharmacokinetics and tissue distribution, as well as ex vivo near-infrared images of organs and whole bodies, were compared between healthy rats and rats with adjuvant arthritis (AA) following the administration of a single intravenous (iv) dose of DFEE-TM. Moreover, the biodistribution and antiarthritic activity of DFEE-TM were compared with those of free diclofenac (once-daily intraperitoneal, ip, 10 mg/kg for 7 days). The concentration ratios of cytochrome-P450-mediated cardiotoxic (20-hydroxyeicosatetraenoic acid) over cardioprotective (epoxyeicosatrienoic acids) metabolites of arachidonic acid (ArA) in the heart, kidneys, and plasma were measured as markers of cardiotoxicity. The nanocarrier was found in the joints of AA, but not in those of healthy rats. Both free diclofenac and DFEE-TM comparably controlled AA. Diclofenac delivery via PEO-b-PCL micelles reduced the accumulation of diclofenac in the heart of AA rats. Despite similar antiarthritic activity, the polymeric micellar formulation showed a reduction in the ratio of cardiotoxic-over-cardioprotective eicosanoids of ArA in the heart and plasma of AA rats. The results showed the positive effect of diclofenac prodrug nanodelivery in changing the normal biodistribution of diclofenac away from the heart, leading to lowered diclofenac-induced biomarkers of cardiotoxicity in the heart and plasma of AA rats.


Assuntos
Ácido Araquidônico/metabolismo , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Diclofenaco/efeitos adversos , Diclofenaco/farmacologia , Coração/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/farmacologia , Caproatos/química , Portadores de Fármacos/química , Ácidos Hidroxieicosatetraenoicos/química , Lactonas/química , Masculino , Micelas , Nanopartículas/química , Poliésteres/química , Polímeros/química , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
12.
Mol Pharm ; 17(5): 1470-1481, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32233491

RESUMO

Polymeric micellar nanoparticles represent versatile and biocompatible platforms for targeted drug delivery. However, tracking their biodistribution, stability, and clearance profile in vivo is challenging. The goal of this study was to prepare surface-modified micelles with peptide GE11 for targeting the epidermal growth factor receptor (EGFR). In vitro fluorescence studies demonstrated significantly higher internalization of GE11 micelles into EGFR-expressing HCT116 colon cancer cells versus EGFR-negative SW620 cells. Azo coupling chemistry of tyrosine residues in the peptide backbone with aryl diazonium salts was used to label the micelles with radionuclide 64Cu for positron emission tomography (PET) imaging. In vivo analysis of 64Cu-labeled micelles showed prolonged blood circulation and predominant hepatobiliary clearance. The biodistribution profile of EGFR-targeting GE11 micelles was compared with nontargeting HW12 micelles in HCT116 tumor-bearing mice. PET revealed increasing tumor-to-muscle ratios for both micelles over 48 h. Accumulation of GE11-containing micelles in HCT116 tumors was higher compared to HW12-decorated micelles. Our data suggest that the efficacy of image-guided therapies with micellar nanoparticles could be enhanced by active targeting, as demonstrated with cancer biomarker EGFR.


Assuntos
Neoplasias Colorretais/diagnóstico por imagem , Radioisótopos de Cobre/farmacocinética , Receptores ErbB/antagonistas & inibidores , Imagem Molecular/métodos , Peptídeos/metabolismo , Compostos Radiofarmacêuticos/síntese química , Animais , Linhagem Celular Tumoral , Humanos , Marcação por Isótopo , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Nanopartículas , Polímeros/metabolismo , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/farmacocinética
13.
Pharm Dev Technol ; 25(4): 492-509, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31903817

RESUMO

In this study, pH-triggered polymeric micelle comprising α-tocopherol (TOC) and heparin (HEP) was developed and loaded with docetaxel (DTX). The amphiphilic copolymer was synthesized by grafting TOC onto HEP backbone by a pH-cleavable bond. DTX-loaded micelles were characterized in terms of critical micelle concentration (CMC), particle size, zeta potential, entrapment efficiency (EE), pH-responsive behavior, and drug release. In vitro cytotoxicity of the micelles against breast cancer cells was investigated by MTT assay. The cellular uptake of coumarin-loaded micelles was also evaluated. Furthermore, the pharmacokinetics of DTX-loaded micelles was evaluated and compared with that of Taxotere®.HEP-CA-TOC copolymers showed low CMC values and high EE. At pH 7.4, the micelles remained stable in size and shape, whereas considerable changes in particle size and morphology were observed at pH 5.5. DTX-loaded micelles showed pH-dependent drug release profiles. Coumarin-loaded micelles showed higher cellular uptake than free coumarin. Therefore, the DTX-loaded micelles showed more toxicity against breast cancer cells than free DTX. A significant increase in T1/2 ß, AUC0-∞ and MRT was observed in DTX-loaded micelle treated group as compared to the group treated with Taxotere®.The results suggest that the pH-sensitive HEP-modified micelles could be promising for enhanced intracellular drug delivery of DTX for cancer treatment.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Preparações de Ação Retardada/química , Docetaxel/administração & dosagem , Heparina/análogos & derivados , alfa-Tocoferol/análogos & derivados , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Docetaxel/farmacocinética , Docetaxel/farmacologia , Liberação Controlada de Fármacos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Células MCF-7 , Camundongos , Micelas
14.
J Cell Mol Med ; 22(4): 2210-2219, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29397578

RESUMO

Activation of hepatic stellate cells (HSCs) is an integral component of the wound-healing process in liver injury/inflammation. However, uncontrolled activation of HSCs leads to constant secretion of collagen-rich extracellular matrix (ECM) proteins, resulting in liver fibrosis. The enhanced ECM synthesis/secretion demands an uninterrupted supply of intracellular energy; however, there is a paucity of data on the bioenergetics, particularly the mitochondrial (mito) metabolism of fibrogenic HSCs. Here, using human and rat HSCs in vitro, we show that the mito-respiration, mito-membrane potential (Δψm) and cellular 'bioenergetic signature' distinguish fibrogenic HSCs from normal, less-active HSCs. Ex vivo, HSCs from mouse and rat models of liver fibrosis further confirmed the altered 'bioenergetic signature' of fibrogenic HSCs. Importantly, the distinctive elevation in mito-Δψm sensitized fibrogenic HSCs for selective inhibition by mitotropic doxorubicin while normal, less-active HSCs and healthy human primary hepatocytes remained minimally affected if not, unaffected. Thus, the increased mito-Δψm may provide an opportunity to selectively target fibrogenic HSCs in liver fibrosis.


Assuntos
Doxorrubicina/farmacologia , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Mitocôndrias Hepáticas/metabolismo , Animais , Linhagem Celular , Metabolismo Energético , Células Estreladas do Fígado/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Análise do Fluxo Metabólico , Mitocôndrias Hepáticas/efeitos dos fármacos , Ratos
15.
Mol Pharm ; 15(6): 2316-2326, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29688721

RESUMO

There is increasing interest in developing and applying DNA repair inhibitors in cancer treatment to augment the efficacy of radiation and conventional genotoxic chemotherapy. However, targeting the inhibitor is required to avoid reducing the repair capacity of normal tissue. The aim of this study was to develop nanodelivery systems for the encapsulation of novel imidopiperidine-based inhibitors of the DNA 3'-phosphatase activity of polynucleotide kinase/phosphatase (PNKP), a DNA repair enzyme that plays a critical role in rejoining DNA single- and double-strand breaks. For this purpose, newly identified hit compounds with potent PNKP inhibitory activity, imidopiperidines A12B4C50 and A83B4C63 were encapsulated in polymeric micelles of different poly(ethylene oxide)- b-poly(ε-caprolactone) (PEO- b-PCL)-based structures. Our results showed efficient loading of A12B4C50 and A83B4C63 in PEO- b-PCLs with pendent carboxyl and benzyl carboxylate groups, respectively, and relatively slow release over 24 h. Both free and encapsulated inhibitors were able to sensitize HCT116 cells to radiation and the topoisomerase I poison, irinotecan. In addition, the encapsulated inhibitors were capable of inducing synthetic lethalilty in phosphatase and tensin homologue (PTEN)-deficient cells. We also established the validity of the peptide GE11 as a suitable ligand for active targeted delivery of nanoencapsulated drugs to colorectal cancer cells overexpressing epidermal growth factor receptor (EGFR). Our results show the potential of nanoencapsulated inhibitors of PNKP as either mono or combined therapeutic agents for colorectal cancer.


Assuntos
Neoplasias Colorretais/terapia , Enzimas Reparadoras do DNA/antagonistas & inibidores , Nanocápsulas/química , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Piperidinas/administração & dosagem , Mutações Sintéticas Letais/efeitos dos fármacos , Quimiorradioterapia/métodos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Enzimas Reparadoras do DNA/metabolismo , Composição de Medicamentos/métodos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Células HCT116 , Humanos , Irinotecano/farmacologia , Micelas , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Piperidinas/farmacologia , Radiação Ionizante
16.
Mol Pharm ; 15(3): 882-891, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29357260

RESUMO

Multidrug resistance (MDR) is the major obstacle for chemotherapy. In a previous study, we have successfully synthesized a novel doxorubicin (DOX) derivative modified by triphenylphosphonium (TPP) to realize mitochondrial delivery of DOX and showed the potential of this compound to overcome DOX resistance in MDA-MB-435/DOX cells. (1) To introduce specificity for DOX-TPP to cancer cells, here we report on the conjugation of DOX-TPP to hyaluronic acid (HA) by hydrazone bond with adipic acid dihydrazide (ADH) as the acid-responsive linker, producing HA- hydra-DOX-TPP nanoparticles. Hyaluronic acid (HA) is a natural water-soluble linear glycosaminoglycan, which was hypothesized to increase the accumulation of nanoparticles containing DOX-TPP in the mitochondria of tumor cells upon systemic administration, overcoming DOX resistance, in vivo. Our results showed HA- hydra-DOX-TPP to self-assemble to core/shell nanoparticles of good dispersibility and effective release of DOX-TPP from the HA- hydra-DOX-TPP conjugate in cancer cells, which was followed by enhanced DOX mitochondria accumulation. The HA- hydra-DOX-TPP nanoparticles also showed improved anticancer effects, better tumor cell apoptosis, and better safety profile compared to free DOX in MCF-7/ADR bearing mice.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/administração & dosagem , Mitocôndrias/metabolismo , Nanoconjugados/química , Animais , Antibióticos Antineoplásicos/química , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Doxorrubicina/química , Liberação Controlada de Fármacos , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/química , Concentração de Íons de Hidrogênio , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Compostos Organofosforados/administração & dosagem , Compostos Organofosforados/química , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Acta Pharmacol Sin ; 39(10): 1681-1692, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29849132

RESUMO

Breast cancer is the leading cause of cancer-related death for women, and multidrug resistance (MDR) is the major obstacle faced by chemotherapy for breast cancer. We have previously synthesized a doxorubicin (DOX) derivative by conjugating DOX with triphenylphosphonium (TPP) to achieve mitochondrial delivery, which induced higher cytotoxicity in drug-resistant breast cancer cells than DOX itself. Due to its amphiphilicity, TPP-DOX is difficult to physically entrap in nanocarriers. Thus, we linked it to hyaluronic acid (HA) by a novel ionic bond utilizing the specific bromide ion of TPP to form supra-molecular self-assembled structures (HA-ionic-TPP-DOX). The product was analyzed uisng 1H-NMR, 13C-NMR and mass spectrometry. The HA nanocarriers (HA-ionic-TPP-DOX) were shown to self-assemble into spherical nanoparticles, and sensitive to acidic pH in terms of morphology and drug release. Compared with free DOX, HA-ionic-TPP-DOX produced much greater intracellular DOX accumulation and mitochondrial localization, leading to increased ROS production, slightly decreased mitochondrial membrane potential, increased cytotoxicity in MCF-7/ADR cells and enhanced tumor targeting in vivo. In xenotransplant zebrafish model with the MCF-7/ADR cell line, both TPP-DOX and HA-ionic-TPP-DOX inhibited tumor cell proliferation without inducing significant side effects compared with free DOX. In addition, we observed a better anti-tumor effect of HA-ionic-TPP-DOX on MCF-7/ADR cells in zebrafish than that of TPP-DOX treatment. Furthermore, HA-ionic-DOX-TPP exhibited favorable biocompatibility and anti-tumor effects in MCF-7/ADR tumor-bearing nude mice in comparison with the effects of TPP-DOX and DOX, suggesting the potential of HA-ionic-TPP-DOX for the targeted delivery and controlled release of TPP-DOX, which can lead to the sensitization of resistant breast tumors.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/uso terapêutico , Portadores de Fármacos/química , Ácido Hialurônico/química , Mitocôndrias/metabolismo , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Doxorrubicina/química , Liberação Controlada de Fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oniocompostos/química , Compostos Organofosforados/química , Peixe-Zebra
18.
J Pharm Pharm Sci ; 21(1s): 192s-199s, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30139426

RESUMO

PURPOSE: The aim of this study was to assess the pharmacokinetics of methoxy poly(ethylene oxide)-block-poly(ε-caprolactone) (PEO-b-PCL) micellar formulation of cyclosporine A (CyA) following oral administration in rats making comparisons with its commercial microemulsion formulation, Neoral®. METHODS: PEO-b-PCL copolymer was synthesized and used to form micelles encapsulating CyA. The release of CyA from Neoral® and PEO-b-PCL as well as PEO-b-PCL degradation were assessed in simulated gastric fluid (SGF) and simulated intestinal fluid (SIF). Polymeric micellar CyA and Neoral® were administered by oral gavage to healthy Wistar rats. At predetermined intervals, rats (n=5 for each time point) were euthanized, samples of blood and plasma were collected and analyzed for CyA using an LC-MS/MS assay. Blood and plasma pharmacokinetic parameters of CyA in its polymeric micellar formulation were compared to those of Neoral®. RESULTS: Polymeric micelles of CyA showed < 15 and 10% increase in diameter in SGF and SIF, respectively, within 24 h. PEO-b-PCL showed signs of minimal degradation when incubated for > 8 h in SGF, but was stable in SIF. Drug release in both SGF and SIF was comparable between the two formulations except for significantly higher release of CyA in SIF only at 24 h time point from Neoral®. Following oral administration (10 mg/kg), the blood AUC0-∞ and tmax of CyA in the polymeric micellar formulation was comparable to that for Neoral®. However, the Cmax of CyA-loaded PEO-b-PCL micelles was significantly (p < 0.05) higher than that obtained with Neoral® (2.10 ± 0.41 versus 1.40 ± 0.25 µg/mL, respectively). CyA had higher blood-to-plasma concentration ratios in polymeric micelles compared to Neoral®, in vivo. CONCLUSION: Our results show that PEO-b-PCL micelles can serve as stable and good solubilizing carriers for oral delivery of CyA providing similar pharmacokinetic profile to that of Neoral®.


Assuntos
Ciclosporina/farmacocinética , Poliésteres/química , Administração Oral , Animais , Ciclosporina/administração & dosagem , Ciclosporina/química , Micelas , Estrutura Molecular , Ratos
19.
J Pharm Pharm Sci ; 21(1s): 103s-116s, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30011392

RESUMO

PURPOSE: Alberta Health Services (AHS) recommends the adoption of a new neonatal multi-trace element formulation containing zinc sulfate, copper sulfate, selenious acid and sodium iodide to be compounded internally in appropriate AHS pharmacies. The objective of this study was to assess the physicochemical stability of this formulation under commonly used storage conditions. METHOD: Three batches of trace element solution were compounded by University of Alberta Hospital pharmacy staff using sterile compounding procedures. Appropriate amount of zinc sulfate (500 mg/mL), copper sulfate (40 mg/mL), selenious acid (4 mg/mL), sodium iodide (2 mg/mL) and sterile water for injection were mixed. Samples from each batch were divided in individual vials and syringes for each time point and kept protected from light either at room temperature (15-30°C) or fridge (2-8°C). Vial samples were also kept at room temperature for 12 h and then transferred to fridge. Vial samples were analyzed at time 0, 12 h, and 1, 3, 7, 9, 30, 60, 90 days for their physical appearance and pH, then centrifuged and assessed for the soluble zinc (atomic absorption), copper (atomic absorption), selenium (ICP-MS) and iodine (HPLC and ICP-MS) concentrations. Syringe samples were tested at time 0 and 12 h for element concentrations. RESULTS: Under all storage conditions, when stored in vials, samples' appearance, pH and soluble zinc, copper and selenium concentrations stayed within the USP acceptable limits up to 90 days. Iodine concentration was within the permitted limits only up to 7 days. The USP recommended HPLC method of iodine analysis seemed inadequate for this preparation and needed modifications, through frequent washing of the column with KI (2 %) solution. Samples kept in syringes at room temperature, showed lower than permitted concentration of Zn at 12h in this study. CONCLUSION: The AHS neonatal multi-trace element formulation seem to be physio-chemically stable up to 7 days in all three storage conditions when kept in vials.  A decline in iodine concentration is seen after 7 days irrespective of storage conditions. This article is open to POST-PUBLICATION REVIEW. Registered readers (see "For Readers") may comment by clicking on ABSTRACT on the issue's contents page.


Assuntos
Atenção à Saúde , Oligoelementos/química , Alberta , Físico-Química , Estabilidade de Medicamentos , Humanos , Soluções , Temperatura
20.
J Pharm Pharm Sci ; 21(1): 143-159, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29789104

RESUMO

PURPOSE: Silibinin, is a natural compound, which has shown anticancer activity in various malignancies. In this study, we evaluated the anticancer effects of silibinin in B16-F10 melanoma cells and developed a novel thermoresponsive hydrogel for local delivery of this compound. METHOD: A thermoresponsive hydrogel loaded with silibinin was prepared using triblock copolymers of poly[(α-benzyl carboxylate-e-caprolactone)-co-(α-carboxyl-e-caprolactone)]ran-b-PEG-b-[(α-benzyl carboxylate-e-caprolactone) -co-(α-carboxyl-e-caprolactone)]ran (PCBCL-b-PEG-b-PCBCL), namely PolyGelTM, and compared with a Pluronic F-127 formulation of silibinin. Sol-gel transition temperature of hydrogels was measured by inverse flow method and modulated differential scanning calorimetry (MDSC). Silibinin loading efficiency was measured by HPLC. The MTT and clonogenic assays were used to assess the cytotoxicity and anti-proliferative effects of silibinin on B16-F10 melanoma cells. Flow cytotmetry was used to quantify the induced level of apoptosis and measure the intracellular level of activated STAT3 (pSTAT3) following silibinin treatment in B16.F10 cells. The effects of silibinin on the activation of oncogenic proteins were also evaluated by western blot. RESULTS: Silibinin inhibited cell proliferation (IC50 = 67 µM), provoked cell cycle arrest, induced apoptosis, suppressed key oncogenic pathways (i.e STAT3 and MEK/ERK), and enhanced the cytotoxic effects of doxorubicin in B16-F10 cells. Both PolyGelTM and Pluronic F-127 hydrogels were effective in loading silibinin. A lower drug release pattern within 24h, fitting first- order release kinetics, was observed for the release of silibinin from both gels compared to free drug.  PolyGelTM demonstrated enhanced percutaneous absorption of silibinin through increasing mouse skin intracellular lipid fluidity as documented by DSC of skin following PolyGelTM use. Silibinin loaded in PolyGel TM inhibited the growth of B16-F10 cells (IC50 = 30 µM) and effectively suppressed pSTAT3 activity in B16-F10 cells at 10 µM. CONCLUSION: Our results imply a great potential for PolyGel TM formulations of silibinin for local treatment of malignant melanoma. This article is open to POST-PUBLICATION REVIEW. Registered readers (see "For Readers") may comment by clicking on ABSTRACT on the issue's content page.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Caproatos/química , Lactonas/química , Melanoma/tratamento farmacológico , Polietilenoglicóis/química , Silibina/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Temperatura , Animais , Antineoplásicos Fitogênicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Hidrogéis/química , Hidrogéis/farmacologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Silibina/química , Neoplasias Cutâneas/patologia , Melanoma Maligno Cutâneo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA