Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros










Intervalo de año de publicación
2.
J Control Release ; 352: 861-878, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36397636

RESUMEN

Cancer, infectious diseases, and metabolic and hereditary genetic disorders are a global health burden affecting millions of people, with contemporary treatments offering limited relief. Antisense technology treats diseases by targeting their causal agents using its ability to alter or inhibit endogenous or malfunctioning genes. Nine antisense oligonucleotide (ASO) drugs that represent four different chemical classes have been approved for the treatment of rare diseases, including nusinersen, the first new oligonucleotide-based drug. Advances in medicinal chemistry, understanding the molecular pathways, and the availability of vast genetic data have resulted in enormous improvements in the therapeutic performance of ASO drugs; however, their susceptibility to degradation in the circulation, rapid renal clearance, and immunostimulatory adverse effects greatly limit their clinical applications. An increasing number of ASO-based therapeutics is being tested in clinical trials. Improvements to the delivery of ASO drugs could potentially change the therapeutic landscape for many conditions in the near future. This review describes the technological advances and developments in drug delivery systems pertaining to ASO therapeutics.


Asunto(s)
Sistema de Administración de Fármacos con Nanopartículas , Oligonucleótidos Antisentido , Humanos , Oligonucleótidos Antisentido/uso terapéutico , Química Farmacéutica , Sistemas de Liberación de Medicamentos , Inmunización
3.
Nanotheranostics ; 6(2): 215-229, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34976596

RESUMEN

Rationale: The treatment of microvascular obstruction (MVO) using ultrasound-targeted LNP cavitation (UTC) therapy mechanically relieves the physical obstruction in the microcirculation but does not specifically target the associated inflammatory milieu. Electrophilic fatty acid nitroalkene derivatives (nitro-fatty acids), that display pleiotropic anti-inflammatory signaling and transcriptional regulatory actions, offer strong therapeutic potential but lack a means of rapid targeted delivery. The objective of this study was to develop nitro-fatty acid-containing lipid nanoparticles (LNP) that retain the mechanical efficacy of standard LNP and can rapidly target delivery of a tissue-protective payload that reduces inflammation and improves vascular function following ischemia-reperfusion. Methods: The stability and acoustic behavior of nitro-fatty acid LNP (NO2-FA-LNP) were characterized by HPLC-MS/MS and ultra-high-speed microscopy. The LNP were then used in a rat hindlimb model of ischemia-reperfusion injury with ultrasound-targeted cavitation. Results: Intravenous administration of NO2-FA-LNP followed by ultrasound-targeted LNP cavitation (UTC) in both healthy rat hindlimb and following ischemia-reperfusion injury showed enhanced NO2-FA tissue delivery and microvascular perfusion. In addition, vascular inflammatory mediator expression and lipid peroxidation were decreased in tissues following ischemia-reperfusion revealed NO2-FA-LNP protected against inflammatory injury. Conclusions: Vascular targeting of NO2-FA-LNP with UTC offers a rapid method of focal anti-inflammatory therapy at sites of ischemia-reperfusion injury.


Asunto(s)
Nanopartículas , Espectrometría de Masas en Tándem , Animales , Ácidos Grasos/metabolismo , Isquemia , Liposomas , Ratas , Reperfusión
4.
Biotechnol J ; 16(2): e1900408, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32702191

RESUMEN

Nucleic acid-based therapies are promising therapeutics for the treatment of several systemic disorders, and they offer an exciting opportunity to address emerging biological challenges. The scope of nucleic acid-based therapeutics in the treatment of multiple disease states including cancers has been widened by recent progress in Ribonucleic acids (RNA) biology. However, cascades of systemic and intracellular barriers, including rapid degradation, renal clearance, and poor cellular uptake, hinder the clinical effectiveness of nucleic acid-based therapies. These barriers can be circumvented by utilizing advanced smart nanocarriers that efficiently deliver and release the encapsulated nucleic acids into the target tissues. This review describes the current status of clinical trials on nucleic acid-based therapeutics and highlights representative examples that provide an overview on the current and emerging trends in nucleic acid-based therapies. A better understanding of the design of advanced nanocarriers is essential to promote the translation of therapeutic nucleic acids into a clinical reality.


Asunto(s)
Neoplasias , Humanos , Nanoestructuras , Neoplasias/tratamiento farmacológico , Ácidos Nucleicos , ARN
5.
PLoS One ; 15(11): e0242264, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33206698

RESUMEN

Signal transducer and activator of transcription-3 (STAT3) is an oncogenic transcription factor implicated in carcinogenesis, tumor progression, and drug resistance in head and neck squamous cell carcinoma (HNSCC). A decoy oligonucleotide targeting STAT3 offers a promising anti-tumor strategy, but achieving targeted tumor delivery of the decoy with systemic administration poses a significant challenge. We previously showed the potential for STAT3 decoy-loaded microbubbles, in conjunction with ultrasound targeted microbubble cavitation (UTMC), to decrease tumor growth in murine squamous cell carcinoma. As a next step towards clinical translation, we sought to determine the anti-tumor efficacy of our STAT3 decoy delivery platform against human HNSCC and the effect of higher STAT3 decoy microbubble loading on tumor cell inhibition. STAT3 decoy was loaded on cationic lipid microbubbles (STAT3-MB) or loaded on liposome-conjugated lipid microbubbles to form STAT3-loaded liposome-microbubble complexes (STAT3-LPX). UTMC treatment efficacy with these two formulations was evaluated in vitro using viability and apoptosis assays in CAL33 (human HNSCC) cells. Anti-cancer efficacy in vivo was performed in a CAL33 tumor murine xenograft model. UTMC with STAT3-MB caused significantly lower CAL33 cell viability compared to UTMC with STAT3-LPX (56.8±8.4% vs 84.5±8.8%, respectively, p<0.05). In vivo, UTMC with STAT3-MB had strong anti-tumor effects, with significantly less tumor burden and greater survival compared to that of UTMC with microbubbles loaded with a mutant control decoy and untreated control groups (p<0.05). UTMC with STAT3 decoy-loaded microbubbles significantly decreases human HNSSC tumor progression. These data set the stage for clinical translation of our microbubble platform as an imaged-guided, targeted delivery strategy for STAT3 decoy, or other nucleotide-based therapeutics, in human cancer treatment.


Asunto(s)
Microburbujas , Oligonucleótidos/uso terapéutico , Factor de Transcripción STAT3/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Terapia por Ultrasonido/métodos , Animales , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Humanos , Liposomas , Ratones , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
6.
Acta Biomater ; 101: 531-543, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31706039

RESUMEN

Herein, we developed a multifunctional nanoplatform based on the nanoassembly of gold nanoparticles (GNP) conjugated with lonidamine (LND) and aptamer AS1411 (AS-LAGN) as an effective cancer treatment. Conjugating AS1411 aptamer on the surface of the nanoparticle significantly improved particle accumulation in cancer cells via specific affinity toward the nucleolin receptors. In vitro study clearly revealed that laser irradiation-based hyperthermia effect enhanced the chemotherapeutic effects of LND. Combinational treatment modalities revealed significant apoptosis with higher cell killing effect due to increased ROS production and inhibition of cell migration. GNP's ability to convert the excited state photon energy into thermal heat enabled synergistic photothermal/chemotherapy with improved therapeutic efficacy in animal models. Moreover, immunohistochemistry staining assays confirmed the ability of AS-LAGN to induce cellular apoptosis/necrosis and ablation in tumor tissues, without causing evident damages to the surrounding healthy tissues. Altogether, this AS-LAGN nanoplatform could be a promising strategy for mitochondria-based cancer treatment. STATEMENT OF SIGNIFICANCE: We have designed a facile biodegradable multifunctional nanocarrier system to target the mitochondria, the major "power house" of the cancer cells. We have constructed a multifunctional nanoassembly of protein coronated gold nanoparticles (GNP) conjugated with lonidamine (LND) and aptamer AS1411 (AS-LAGN) as an effective combination of phototherapy with chemotherapy for cancer treatment. The LND was conjugated with albumin which was in turn conjugated to GNP via redox-liable disulfide linkage to generate oxidative stress and ROS to kill cancer cells. GNP's ability to convert the excited state photon energy into thermal heat enabled synergistic photothermal/chemotherapy with improved therapeutic efficacy in animal models. Consistently, AS-LAGN showed enhanced antitumor efficacy in xenograft tumor model with remarkable tumor regression property.


Asunto(s)
Albúminas/química , Antineoplásicos/farmacología , Oro/química , Indazoles/química , Nanopartículas del Metal/química , Terapia Fototérmica , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Endocitosis/efectos de los fármacos , Humanos , Nanopartículas del Metal/ultraestructura , Ratones Endogámicos BALB C , Ratones Desnudos , Especies Reactivas de Oxígeno/metabolismo , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
7.
ACS Biomater Sci Eng ; 5(10): 5159-5168, 2019 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33455222

RESUMEN

Metformin (MET) is a common treatment for type II diabetes. Here, we demonstrate the anticancer activity of a polymeric metformin derivative. We successfully synthesized the polypeptide (poly-l-lysine [PLL]) derivative of metformin (LysMET) and demonstrated its capacity as an anticancer therapeutic and gene carrier. miRNA-320a was loaded into the cationic LysMET and enveloped in a lipid bilayer, and a MUC1-specific aptamer was conjugated to the surface (A-Lipo@mLysMET). The LysMET-containing guanidine moiety was more tolerable than the secondary amine-containing PLL. LysMET showed similar efficacy to MET in the induction of HT-29 tumor suppression, indicating the importance of the biguanide moiety. The synergistic effect of miRNA-320a and LysMET treatment significantly decreased cell viability compared with LysMET treatment alone, which was attributed to the role of miRNA in the ß-catenin pathway. A-Lipo@mLysMET showed excellent antitumor efficacy and significantly reduced the tumor burden in all groups. AMPKα phosphorylation was markedly increased by LysMET compared with the control, with significant inhibition of the mTOR pathway. The TUNEL assay showed that apoptosis was the main mechanism responsible for cancer cell death and that A-Lipo@mLysMET resulted in the highest proportion of TUNEL-positive cells (∼36%). No noticeable organ damage was observed after treatment with either LysMET or A-Lipo@mLysMET, confirming the excellent safety profile of guanide-modified polymers. Overall, we demonstrated the feasibility of LysMET for the effective control of tumor progression as well as its dual role, as both a drug and a gene carrier.

8.
Int J Pharm ; 548(1): 92-103, 2018 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-29959089

RESUMEN

This study reports a new strategy for in situ fabrication of plasmonic hollow silver-gold nanoshell (with resonance tuned to NIR region) encased in the hollow mesoporous silica as an efficient platform to efficiently and precisely regulate the release of 5-fluorouracil (anticancer drug) for prostate cancer therapy and photothermal therapy. The mesopores were capped with thermosensitive phase-change material lauric acid, which allowed for remote, precise, and spatiotemporal control of drug release via external heating or photothermal heating of plasmonic silver-gold nanoshell via NIR laser irradiation. The system was nanometric, monodispersed, and showed negative surface charge. The nanocarrier showed better pH stability and thermodynamic stability compared to dense silica-coated gold nanoshells. The drug release could be triggered remotely by applying low powered continuous wave NIR laser (λ = 808 nm). The nanocarrier showed improved internalization by cancer cells, which was further enhanced by laser irradiation. High powered laser directly killed the cancer cells via photothermal effect in the region irradiated. Thus, this system fabricated by novel synthetic strategy provided efficient chemo- and phototherapy.


Asunto(s)
Sistemas de Liberación de Medicamentos , Oro , Nanocáscaras , Dióxido de Silicio , Plata , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Fluorouracilo/administración & dosificación , Fluorouracilo/química , Oro/administración & dosificación , Oro/química , Humanos , Rayos Infrarrojos , Rayos Láser , Ácidos Láuricos/administración & dosificación , Ácidos Láuricos/química , Nanocáscaras/administración & dosificación , Nanocáscaras/química , Fototerapia , Porosidad , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/química , Plata/administración & dosificación , Plata/química
9.
Colloids Surf B Biointerfaces ; 165: 56-66, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29453086

RESUMEN

The major goal of cancer chemotherapy is to maximize the therapeutic efficacy of anticancer drugs, while minimizing their associated side effects. Celastrol (CST), which is extracted from the traditional Chinese medicinal plant Tripterygium wilfordii, has been reported to exhibit significant anticancer effects in various in vitro and in vivo cancer models. Nanoparticulate drug delivery systems could be employed to preserve and enhance the pharmacological effects of CST in cancer cells. Among these, mesoporous silica nanoparticles (MSNs) are one of the most promising drug delivery systems. MSNs possess the capability of passive accumulation within solid tumors, and could efficiently transport anticancer drugs to such tumors in a site-specific manner. In this study, PEGylated polyaminoacid-capped CST-loaded MSN (CMSN-PEG) showed controlled in vitro drug release behavior, and exhibited high in vitro cytotoxicity in different cancer cells. Compared to treatment with free CST, treatment with CMSN-PEG resulted in the increased expression of the apoptosis protein HIF-1α and proteins corresponding to mitochondrial apoptosis pathway. Importantly, CMSN-PEG remarkably reduced tumor burden with no toxicity to healthy cells in the SCC7 tumor-bearing xenograft model. Our results clearly demonstrate a promising potential of CMSN-PEG for the treatment of solid tumors.


Asunto(s)
Aminoácidos/química , Sistemas de Liberación de Medicamentos , Mitocondrias/metabolismo , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Polietilenglicoles/química , Dióxido de Silicio/química , Triterpenos/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Femenino , Disulfuro de Glutatión/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Nanopartículas/ultraestructura , Neoplasias/patología , Triterpenos Pentacíclicos , Polietilenglicoles/síntesis química , Dióxido de Silicio/síntesis química , Triterpenos/farmacología
10.
Arch Pharm Res ; 41(2): 111-129, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29214601

RESUMEN

The development of novel drug delivery systems based on well-defined polymer therapeutics has led to significant improvements in the treatment of multiple disorders. Advances in material chemistry, nanotechnology, and nanomedicine have revolutionized the practices of drug delivery. Stimulus-responsive material-based nanosized drug delivery systems have remarkable properties that allow them to circumvent biological barriers and achieve targeted intracellular drug delivery. Specifically, the development of novel nanocarrier-based therapeutics is the need of the hour in managing complex diseases. In this review, we have briefly described the fundamentals of drug targeting to diseased tissues, physiological barriers in the human body, and the mechanisms/modes of drug-loaded carrier systems. To that end, this review serves as a comprehensive overview of the recent developments in stimulus-responsive drug delivery systems, with focus on their potential applications and impact on the future of drug delivery.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/tendencias , Nanomedicina/tendencias , Nanopartículas/administración & dosificación , Animales , Antineoplásicos/metabolismo , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Humanos , Nanomedicina/métodos , Nanopartículas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
11.
Colloids Surf B Biointerfaces ; 160: 73-83, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28917152

RESUMEN

Pancreatic cancer has extremely poor prognosis with an 85% mortality rate that results from aggressive and asymptomatic growth, high metastatic potential, and rapid development of resistance to already ineffective chemotherapy. In this study, plasmonic hollow gold nanoshells (GNS) coated with PEGylated thermosensitive lipids were prepared as an efficient platform to ratiometrically co-deliver two drugs, bortezomib and gemcitabine (GNS-L/GB), for combinational chemotherapy and photothermal therapy of pancreatic cancer. Bortezomib was loaded within the lipid bilayers, while gemcitabine was loaded into the hydrophilic interior of the porous GNS via an ammonium sulfate-driven pH gradient method. Physicochemical characterizations and biological studies of GNS-L/GB were performed, with the latter using cytotoxicity assays, cellular uptake and apoptosis assays, live/dead assays, and western blot analysis of pancreatic cancer cell lines (MIA PaCa-2 and PANC-1). The nanoshells showed remotely controllable drug release when exposed to near-infrared laser for site-specific delivery. GNS-L/GB showed synergistic cytotoxicity and improved internalization by cancer cells. High-powered near-infrared continuous wave laser (λ=808nm) effectively killed cancer cells via the photothermal effect of GNS-L/GB, irrespective of cell type in a power density-, time-, and GNS dose-dependent manner. These results suggest that this method can provide a novel approach to achieve synergistic combinational chemotherapy and photothermal therapy, even with resistant pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Oro/química , Lípidos/química , Nanocáscaras/química , Neoplasias Pancreáticas/tratamiento farmacológico , Fototerapia/métodos , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Bortezomib/administración & dosificación , Bortezomib/química , Línea Celular Tumoral , Terapia Combinada , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos/efectos de la radiación , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Rayos Infrarrojos , Nanocáscaras/ultraestructura , Polietilenglicoles/química , Temperatura , Gemcitabina
12.
Drug Deliv ; 24(1): 1262-1272, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28891336

RESUMEN

Since breast cancer is one of the most lethal malignancies, targeted strategies are urgently needed. In this study, we report the enhanced therapeutic efficacy of docetaxel (DTX) when combined with polyunsaturated fatty acids (PUFA) for effective treatment of multi-resistant breast cancers. Folic acid (FA)-conjugated PUFA-based lipid nanoparticles (FA-PLN/DTX) was developed. The physicochemical properties, in vitro uptake, in vitro cytotoxicity, and in vivo anticancer activity of FA-PLN/DTX were evaluated. FA-PLN/DTX could efficiently target and treat human breast tumor xenografts in vivo. They showed high payload carrying capacity with controlled release characteristics and selective endocytic uptake in folate receptor-overexpressing MCF-7 and MDA-MB-231 cells. PUFA synergistically improved the anticancer efficacy of DTX in both tested cancer cell lines by inducing a G2/M phase arrest and cell apoptosis. Combination of PUFA and DTX remarkably downregulated the expression levels of pro-apoptotic and anti-apoptotic markers, and blocked the phosphorylation of AKT signaling pathways. Compared to DTX alone, FA-PLN/DTX showed superior antitumor efficacy, with no signs of toxic effects in cancer xenograft animal models. We propose that PUFA could improve the therapeutic efficacy of anticancer agents in cancer therapy. Further studies are necessary to fully understand these findings and achieve clinical translation.


Asunto(s)
Nanoestructuras , Animales , Antineoplásicos , Línea Celular Tumoral , Docetaxel , Ácidos Grasos Insaturados , Humanos , Taxoides
13.
Acta Biomater ; 63: 135-149, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28890258

RESUMEN

In this study, we developed pH and redox-responsive crosslinked polypeptide-based combination micelles for enhanced chemotherapeutic efficacy and minimized side effects. The stability and drug release properties of the polypeptide micelles were efficiency balanced by the corona-crosslinking of the triblock copolymer, poly(ethylene glycol)-b-poly(aspartic acid)-b-poly(tyrosine) (PEG-b-pAsp-b-pTyr) with coordinated redox and pH dual-sensitivity by introducing disulfide crosslinkages. Because of the crosslinking of the middle shell of the triblock polypeptide micelles, their robust structure was maintained in strong destabilization conditions and exhibited excellent stability. GSH concentrations were significantly higher in tumor tissue than in normal tissue, which formed the basis for our design. Drug release was elevated under redox and low acidic conditions. Furthermore, crosslinked micelles showed a superior anticancer effect compared to that of non-crosslinked micelles. Incorporation of docetaxel (DTX) and lonidamine (LND) in crosslinked polypeptide micelles increased the intracellular reactive oxygen species (ROS) level and oxidative stress and caused damage to intracellular components that resulted in greater apoptosis of cancer cells than when DTX or LND was used alone. The combination of DTX and LND in crosslinked micelles exhibited efficacious inhibition of tumor growth with an excellent safety profile compared to that reported for drug cocktail combinations and non-crosslinked micelles. Overall, redox/pH-responsive polypeptide micelles could be an interesting platform for efficient chemotherapy. STATEMENT OF SIGNIFICANCE: We have synthesized a biodegradable polypeptide block copolymer to construct a facile pH and redox-responsive polymeric micelle asan advanced therapeutic system for cancer therapy. We have designed a corona-crosslinked triblock copolymer (poly (ethylene glycol)-b-poly(aspartic acid)-b-poly(tyrosine) (PEG-b-pAsp-b-pTyr)) micelles co-loaded with docetaxel and lonidamine (cl-M/DL). The corona of triblock polymer was crosslinked to maintain its structural integrity in the physiological environment. The mitochondrial targeting LND is expected to generate ROS, oxidative stress and thereby synergize the chemotherapeutic efficacy of DTX in killing cancer cells. Consistently, cl-M/DL exhibited excellent antitumor efficacy in xenograft tumor model with remarkable tumor regression. Overall, we demonstrated the construction of bioreducible nanosystem for the effective synergistic delivery of DTX/LND in tumor tissues towards cancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Materiales Biocompatibles/química , Reactivos de Enlaces Cruzados/química , Micelas , Neoplasias/tratamiento farmacológico , Péptidos/uso terapéutico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Rastreo Diferencial de Calorimetría , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Glutatión/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones Desnudos , Neoplasias/patología , Oxidación-Reducción , Péptidos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Espectroscopía Infrarroja por Transformada de Fourier , Distribución Tisular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Carbohydr Polym ; 173: 57-66, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28732901

RESUMEN

In the present study, a unique strategy was developed to develop nanocarriers containing multiple therapeutics with controlled release characteristics. In this study, we demonstrated the synthesis of dextran sulfate-doxorubicin (DS-DOX) and alginate-cisplatin (AL-CIS) polymer-drug complexes to produce a transferrin ligand-conjugated liposome. The targeted nanoparticles (TL-DDAC) were nano-sized and spherical. The targeted liposome exhibited a specific receptor-mediated endocytic uptake in cancer cells. The enhanced cellular uptake of TL-DDAC resulted in a significantly better anticancer effect in resistant and sensitive breast cancer cells compared to that of the free drugs. Specifically, DOX and CIS at a molar ratio of 1:1 exhibited better therapeutic performance compared to that of other combinations. The combination of an anthracycline-based topoisomerase II inhibitor (DOX) and a platinum compound (CIS) resulted in significantly higher cell apoptosis (early and late) in both types of cancer cells. In conclusion, treatment with DS-DOX and AL-CIS based combination liposomes modified with transferrin (TL-DDAC) was an effective cancer treatment strategy. Further investigation in clinically relevant animal models is warranted to prove the therapeutic efficacy of this unique strategy.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Liposomas , Polisacáridos/química , Alginatos/química , Apoptosis , Bromelaínas/química , Línea Celular Tumoral , Cisplatino/administración & dosificación , Doxorrubicina/administración & dosificación , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Humanos , Nanopartículas , Neoplasias
15.
J Control Release ; 258: 226-253, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28472638

RESUMEN

This review focuses on the smart chemistry that has been utilized in developing polymer-based drug delivery systems over the past 10years. We provide a comprehensive overview of the different functional moieties and reducible linkages exploited in these systems, and outline their design, synthesis, and application from a therapeutic efficacy viewpoint. Furthermore, we highlight the next generation nanomedicine strategies based on this novel chemistry.


Asunto(s)
Preparaciones de Acción Retardada/química , Sistemas de Liberación de Medicamentos/métodos , Nanoestructuras/química , Polímeros/química , Animales , Técnicas de Química Sintética/métodos , Preparaciones de Acción Retardada/síntesis química , Humanos , Nanomedicina/métodos , Nanotecnología/métodos , Polímeros/síntesis química
16.
ACS Appl Mater Interfaces ; 9(11): 9280-9290, 2017 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-28240860

RESUMEN

Metastasis of cancers accounts for almost all cancer-related deaths. In this study, we report a PEGylated nanostructured platform for coadministration of doxorubicin (DOX) and imatinib (IMT) intended to effectively inhibit metastatic tumors. The DOX and IMT coloaded nanostructured system (DOX/IMT-N) is characterized by an excellent encapsulation potential for both drugs and shows sequential and sustained drug release in vitro. DOX/IMT-N significantly inhibited the in vitro proliferation of MDA-MB-231 and SK-MEL-28 cells. The inhibitory effect on in vitro proliferation of the cells was significantly greater than the effect of free DOX, DOX/IMT cocktail, or the nanostructured system housing DOX only (DOX-N). DOX/IMT-N remarkably enhanced cellular drug uptake, resulting in enhanced apoptosis, caused by significant increases in the expression levels of apoptotic marker proteins. Intravenous administration of DOX/IMT-N to MBA-MB-231 xenograft tumor-bearing mice resulted in significantly improved inhibition of tumor progression compared to that with DOX, DOX/IMT, or DOX-N. Therefore, the nanostructured DOX/IMT-N system could potentially aid in overcoming drug resistance in metastatic tumors and improve the effectiveness of metastatic tumor therapeutics.


Asunto(s)
Nanoestructuras , Animales , Línea Celular Tumoral , Doxorrubicina , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Mesilato de Imatinib , Ratones
17.
Oncotarget ; 8(9): 14925-14940, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-28122339

RESUMEN

In this study, a transferrin-anchored albumin nanoplatform with PEGylated lipid bilayers (Tf-L-APVN) was developed for the targeted co-delivery of paclitaxel and vorinostat in solid tumors. Tf-L-APVN exhibited a sequential and controlled release profile of paclitaxel and vorinostat, with an accelerated release pattern at acidic pH. At cellular levels, Tf-L-APVN significantly enhanced the synergistic effects of paclitaxel and vorinostat on the proliferation of MCF-7, MDA-MB-231, and HepG2 cancer cells. Vorinostat could significantly enhance the cytotoxic potential of paclitaxel, induce marked cell apoptosis, alter cell cycle patterns, and inhibit the migratory capacity of cancer cells. In addition, Tf-L-APVN showed prolonged circulation in the blood and maintained an effective ratio of 1:1 (for paclitaxel and vorinostat) throughout the study period. In HepG2 tumor-bearing mice, Tf-L-APVN displayed excellent antitumor efficacy and the combination of paclitaxel and vorinostat significantly inhibited the tumor growth. Taken together, dual drug-loaded Tf receptor-targeted nanomedicine holds great potential in chemotherapy of solid tumors.


Asunto(s)
Adenoma de Células Hepáticas/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Lípidos/química , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas/administración & dosificación , Paclitaxel/farmacología , Adenoma de Células Hepáticas/metabolismo , Adenoma de Células Hepáticas/patología , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Ácidos Hidroxámicos/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Nanopartículas/química , Células Tumorales Cultivadas , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Int J Pharm ; 519(1-2): 11-21, 2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-28069389

RESUMEN

Although protein-bound paclitaxel (PTX, Abraxane®) has been established as a standard PTX-based therapy against multiple cancers, its clinical success is limited by unfavorable pharmacokinetics, suboptimal biodistribution, and acute toxicities. In the present study, we aimed to apply the principles of a layer-by-layer (LbL) technique to improve the poor colloidal stability and pharmacokinetic pattern of nanoparticle albumin-bound paclitaxel (nab-PTX). LbL-based nab-PTX was successfully fabricated by the alternate deposition of polyarginine (pARG) and poly(ethylene glycol)-block-poly (L-aspartic acid) (PEG-b-PLD) onto an albumin conjugate. The presence of protective entanglement by polyamino acids prevented the dissociation of nab-PTX and improved its colloidal stability even at a 100-fold dilution. The combined effect of high nanoparticle internalization and controlled release of PTX from LbL-nab-PTX increased its cytotoxicity in MCF-7 and MDA-MB-231 breast cancer cells. LbL-nab-PTX consistently induced apoptosis in approximately 52% and 22% of MCF-7 and MDA-MB-231 cancer cells, respectively. LbL assembly of polypeptides effectively prevented exposure of PTX to the systemic environment and thereby inhibited drug-induced hemolysis. Most importantly, LbL assembly of polypeptides to nab-PTX effectively increased the blood circulation potential of PTX and improved therapeutic efficacy via a significantly higher area under the curve (AUC)0-∞. We report for the first time the application of LbL functional architectures for improving the systemic performance of nab-PTX with a view toward its clinical translation for cancer therapy.


Asunto(s)
Paclitaxel Unido a Albúmina/química , Nanopartículas/química , Paclitaxel Unido a Albúmina/metabolismo , Paclitaxel Unido a Albúmina/farmacología , Animales , Ácido Aspártico/química , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Masculino , Polietilenglicoles/química , Ratas , Ratas Sprague-Dawley , Distribución Tisular/efectos de los fármacos
19.
Colloids Surf B Biointerfaces ; 150: 393-401, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27825759

RESUMEN

In this study, a core-shell type polypeptide-based lipid nanocapsule was developed to enhance anticancer efficacy of erlotinib in non-small cell lung cancers. Mean particle size of PEGylated polypeptide-lipid nanocapsules (PLN) for erlotinib (ERL) delivery was ∼200nm with an effective surface charge of -20mV. Protective PEGylated polypeptide layer acted as a molecular fence and effectively controlled the diffusion of erlotinib from the lipid nanocapsule core, whereas pH-responsiveness of poly(L-aspartic acid) accelerated the release of erlotinib in acidic conditions. Blank lipid nanocapsules showed excellent biocompatibility. ERL-loaded PLN (ERL-PLN) showed dose-dependent cytotoxicity in NCI-H358 and HCC-827 lung cancer cells. ERL-PLN treatment resulted in a superior tumor regression profile in a xenograft tumor model, compared to free ERL and control, suggesting high therapeutic efficacy. ERL-PLN-treated mice showed 5- and 2-fold smaller tumor volume compared to control and free ERL groups, respectively. Based on these results, PLN provide a promising drug delivery approach for lung cancer therapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Clorhidrato de Erlotinib/administración & dosificación , Lípidos/química , Neoplasias Pulmonares/tratamiento farmacológico , Péptidos/química , Polietilenglicoles/química , Animales , Línea Celular Tumoral , Supervivencia Celular , Difusión , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Clorhidrato de Erlotinib/química , Humanos , Hidrodinámica , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Endogámicos BALB C , Nanocápsulas/química , Nanopartículas/química , Trasplante de Neoplasias , Tamaño de la Partícula , Propiedades de Superficie
20.
Acta Biomater ; 48: 131-143, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27794477

RESUMEN

In this study, we report a facile method to construct a bioactive (poly(phenylalanine)-b-poly(l-histidine)-b-poly(ethylene glycol) polypeptide nanoconstruct to co-load doxorubicin (DOX) and quercetin (QUR) (DQ-NV). The smart pH-sensitive nanovehicle was fabricated with precisely tailored drug-to-carrier ratio that resulted in accelerated, sequential drug release. As a result of ratiometric loading, QUR could significantly enhance the cytotoxic potential of DOX, induced marked cell apoptosis; change cell cycle patterns, inhibit the migratory capacity of sensitive and resistant cancer cells. In particular, pro-oxidant QUR from DQ-NV remarkably reduced the GSH/GSSG ratio, indicating high oxidative stress and damage to cellular components. DQ-NV induced tumor shrinkage more effectively than the single drugs in mice carrying subcutaneous SCC-7 xenografts. DQ-NV consistently induced high expression of caspase-3 and PARP and low expression of Ki67 and CD31 immunomarkers. In summary, we demonstrate the development of a robust polypeptide-based intracellular nanovehicle for synergistic delivery of DOX/QUR in cancer chemotherapy. STATEMENT OF SIGNIFICANCE: In this study, we report a facile method to construct bioactive and biodegradable polypeptide nanovehicles as an advanced platform technology for application in cancer therapy. We designed a robust (poly(phenylalanine)-b-poly(l-histidine)-b-poly(ethylene glycol) nanoconstruct to co-load doxorubicin (DOX) and quercetin (QUR) (DQ-NV). The conformational changes of the histidine block at tumor pH resulted in accelerated, sequential drug release. QUR could significantly enhance the cytotoxic potential of DOX, induce marked cell apoptosis, change cell cycle patterns, and inhibit the migratory capacity of sensitive and resistant cancer cells. DQ-NV induced tumor shrinkage more effectively than the single drugs and the 2-drug cocktail in tumor xenografts. In summary, we demonstrate the development of an intracellular nanovehicle for synergistic delivery of DOX/QUR in cancer chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Microambiente Celular/efectos de los fármacos , Portadores de Fármacos/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Ingeniería de Tejidos/métodos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Doxorrubicina/farmacología , Sinergismo Farmacológico , Endocitosis/efectos de los fármacos , Citometría de Flujo , Hidrodinámica , Concentración de Iones de Hidrógeno , Inmunohistoquímica , Ratones , Neoplasias/patología , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Péptidos , Quercetina , Bibliotecas de Moléculas Pequeñas/farmacología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...