Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biomacromolecules ; 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38869359

RESUMEN

Acute myeloid leukemia (AML) is often associated with poor prognosis and survival. Small molecule inhibitors, though widening the treatment landscape, have limited monotherapy efficacy. The combination therapy, however, shows suboptimal clinical outcomes due to low bioavailability, overlapping systemic toxicity and drug resistance. Here, we report that CXCR4-mediated codelivery of the BCL-2 inhibitor venetoclax (VEN) and the FLT3 inhibitor sorafenib (SOR) via T22 peptide-tagged disulfide cross-linked polymeric micelles (TM) achieves synergistic treatment of FLT3-ITD AML. TM-VS with a VEN/SOR weight ratio of 1/4 and T22 peptide density of 20% exhibited an extraordinary inhibitory effect on CXCR4-overexpressing MV4-11 AML cells. TM-VS at a VEN/SOR dosage of 2.5/10 mg/kg remarkably reduced leukemia burden, prolonged mouse survival, and impeded bone loss in orthotopic MV4-11-bearing mice, outperforming the nontargeted M-VS and oral administration of free VEN/SOR. CXCR4-mediated codelivery of BCL-2 and FLT3 inhibitors has emerged as a prospective clinical treatment for FLT3-ITD AML.

2.
Biomacromolecules ; 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38907698

RESUMEN

Supramolecular delivery systems with the prolonged circulation, the potential for diverse functionalization, and few toxin-related limitations have been extensively studied. For the present study, we constructed a linear polyglycerol-shelled polymersome attached with the anti-HER-2-antibody trastuzumab. We then covalently loaded the anticancer drug DM1 in the polymersome via dynamic disulfide bonding. The resulted trastuzumab-polymersome-DM1 (Tra-PS-DM1) exhibits a mean size of 95.3 nm and remarkable drug loading efficiency % of 99.3%. In addition to its superior stability, we observed the rapid release of DM1 in a controlled manner under reductive conditions. Compared to the native polymersomes, Tra-PS-DM1 has shown greatly improved cellular uptake and significantly reduced IC50 up to 17-fold among HER-2-positive cancer cells. Moreover, Tra-PS-DM1 demonstrated superb growth inhibition of HER-2-positive tumoroids; specifically, BT474 tumoroids shrunk up to 62% after 12 h treatment. With exceptional stability and targetability, the PG-shelled Tra-PS-DM1 appears as an attractive approach for HER-2-positive tumor treatment.

3.
Med Sci Monit ; 30: e942855, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38755961

RESUMEN

BACKGROUND Nurses in the Intensive Care Unit (ICU) play a critical role in recognizing patients who are at risk of deterioration by conducting continual assessments and taking suitable measures in response to changing health status. The validity of the cluster nursing intervention has been studied previously, but its use among ICU patients with tracheal intubation and extubation has not been examined. This study assessed the effectiveness of cluster nursing intervention in ICU patients with tracheal intubation and extubation. MATERIAL AND METHODS In this retrospective study, 80 patients on mechanical ventilation in the ICU ward were randomly assigned to control and intervention groups (40 patients each). The control group received the routine nursing mode, while the intervention group was given 5 sessions of cluster nursing intervention. Tracheal intubation and extubation-associated complications, blood gas analysis, patient nursing satisfaction, and changes in patients' negative emotions were compared before and after the intervention. RESULTS After the nursing intervention, the levels of PaO2 were higher, while PaCO2 levels were lower in the intervention group compared to the control group (P<0.05). Importantly, anxiety and depression scores in the intervention group were lower than in the control group (P<0.05). Moreover, the overall incidence of complications in the intervention group was lower than in the control group, whereas patient satisfaction with nursing services was higher (P<0.05). CONCLUSIONS Cluster nursing intervention can effectively reduce the incidence of complications and improve patients’ physiological and psychological conditions. Moreover, it enhances patient satisfaction with nursing services, thus improving patients' clinical symptoms.


Asunto(s)
Extubación Traqueal , Unidades de Cuidados Intensivos , Intubación Intratraqueal , Humanos , Masculino , Femenino , Intubación Intratraqueal/efectos adversos , Intubación Intratraqueal/métodos , Persona de Mediana Edad , Extubación Traqueal/métodos , Estudios Retrospectivos , Anciano , Respiración Artificial/efectos adversos , Respiración Artificial/métodos , Ansiedad , Adulto , Satisfacción del Paciente , Depresión , Bienestar Psicológico
4.
Biomacromolecules ; 24(11): 5371-5380, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37801632

RESUMEN

Multiple myeloma (MM) is the second most common hematological malignancy. For relapsed and refractory MM, a proteasome inhibitor, carfilzomib (CFZ), has become one of the few clinical options. CFZ suffers, nevertheless, metabolic instability and poor bioavailability and may induce severe cardiovascular and renal adverse events. Here, we report that daratumumab (Dar)-decorated polypeptide micelles (Dar-PMs) mediate the targeted delivery of CFZ to CD38-positive MM, effectively boosting its anti-MM efficacy. CFZ-loaded Dar-PMs (Dar-PMs-CFZ) exhibited an average diameter of ca. 80 nm and Dar density-dependent cell endocytosis and anti-MM activity, in which over 6-fold greater inhibitory effect to LP-1 and MM.1S MM cells than nontargeted PMs-CFZ control was achieved at a Dar density of 3.2 (Dar3.2-PMs-CFZ). Interestingly, Dar3.2-PMs-CFZ markedly enhanced the growth inhibition of orthotopic LP-1 MM in mice and significantly extended the median survival time compared with PMs-CFZ and free CFZ (95 days vs 60 and 54 days, respectively). In line with its high MM targetability and anti-MM efficacy, Dar3.2-PMs-CFZ revealed little toxic effects and effectively prevented osteolytic lesions. The antibody-targeted nanodelivery of a proteasome inhibitor appears to be an appealing strategy to treat multiple myeloma.


Asunto(s)
Antineoplásicos , Mieloma Múltiple , Nanopartículas , Animales , Ratones , Inhibidores de Proteasoma/efectos adversos , Antineoplásicos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Nanopartículas/uso terapéutico
5.
Biomacromolecules ; 24(11): 5353-5363, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37871289

RESUMEN

The silencing of disease-causing genes with small interfering RNA (siRNA) offers a particularly effective therapeutic strategy for different disorders; however, its clinical efficacy relies on the development of nontoxic and tissue-specific delivery vehicles. Herein, we report that bioresponsive chimaeric polymersomes (BCP) with short poly(ethylenimine) as inner shell mediate highly efficacious, sustained, and liver-specific siRNA transfection in vivo. BCP exhibited remarkable encapsulation efficiencies of siRNA (95-100%) at siRNA-feeding contents of 15-25 wt %, to afford stable, small-sized (55-64 nm), and neutral-charged BCP-siRNA. siApoB-Loaded BCP (BCP-siApoB) outperformed lipofectamine counterparts and silenced 93% of ApoB mRNA in HepG2 cells at 50 nM siApoB without inducing cytotoxicity. Intriguingly, the in vivo studies using wild-type C57BL/6 mice revealed that BCP-siApoB preferentially accumulated in the liver, and a single dose of 4.5 mg/kg achieved over 90% downregulation of ApoB mRNA for at least 10 days. The systemic administration of BCP-siApoB at 4.5 mg/kg every 2 weeks or 1.5 mg/kg weekly in diet-induced obese mice could also achieve up to 80% silencing of ApoB mRNA. The liver specificity and silencing efficacy of BCP-siApoB could further be improved by decorating it with the trivalent N-acetylgalactosamine (TriGalNAc) ligand. These bioresponsive and liver-specific chimaeric polymersomes provide an enabling technology for siRNA therapy of various liver-related diseases.


Asunto(s)
Apolipoproteínas B , Hígado , Animales , Ratones , ARN Interferente Pequeño/genética , Ratones Endogámicos C57BL , Apolipoproteínas B/genética , Transfección , ARN Mensajero
6.
Biomacromolecules ; 23(1): 100-111, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-34913340

RESUMEN

Targeted nanomedicines particularly armed with monoclonal antibodies are considered to be the most promising advanced chemotherapy for malignant cancers; however, their development is hindered by their instability and drug leakage problems. Herein, we constructed a robust cetuximab-polymersome-mertansine nanodrug (C-P-DM1) for highly potent and targeted therapy of epidermal growth factor receptor (EGFR)-positive solid tumors. C-P-DM1 with a tailored cetuximab surface density of 2 per P-DM1 exhibited a size of ca. 60 nm, high stability with minimum DM1 leakage, glutathione-triggered release of native DM1, and 6.0-11.3-fold stronger cytotoxicity in EGFR-positive human breast (MDA-MB-231), lung (A549), and liver (SMMC-7721) cancer cells (IC50 = 27.1-135.5 nM) than P-DM1 control. Notably, intravenous injection of C-P-DM1 effectively repressed subcutaneous MDA-MB-231 breast cancer and orthotopic A549-Luc lung carcinoma in mice without inducing toxic effects. Strikingly, intratumoral injection of C-P-DM1 completely cured 60% of mice bearing breast tumor without recurrence. This robust cetuximab-polymersome-mertansine nanodrug provides a promising new strategy for targeted treatment of EGFR-positive solid malignancies.


Asunto(s)
Neoplasias de la Mama , Cetuximab , Maitansina , Nanopartículas , Animales , Anticuerpos Monoclonales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cetuximab/farmacología , Receptores ErbB/metabolismo , Femenino , Humanos , Maitansina/farmacología , Ratones , Nanopartículas/metabolismo , Nanopartículas/uso terapéutico
7.
Biomacromolecules ; 23(7): 2989-2998, 2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35758844

RESUMEN

The incorporation of a phenylboronic acid group has appeared as an attractive strategy to build smart drug delivery systems. Here, we report novel synthesis of phenylboronic acid-functionalized copolypeptides based on an l-boronophenylalanine N-carboxyanhydride (BPA-NCA) monomer and their application for robust co-encapsulation and responsive release of dual anticancer drugs. By employing different poly(ethylene glycol) (PEG) initiators and copolymerizing with varying NCA monomers, linear and star PEG-poly(l-boronophenylalanine) copolymers (PEG-PBPA, star-PEG-PBPA), PEG-poly(l-tyrosine-co-l-boronophenylalanine) [PEG-P(Tyr-co-BPA)], PEG-poly(l-lysine-co-l-boronophenylalanine) [PEG-P(Lys-co-BPA)], and PEG-poly(ß-benzyl-l-aspartate-co-l-boronophenylalanine) [PEG-P(BLA-co-BPA)] were obtained with controlled compositions. Interestingly, PEG-PBPA self-assembled into uniform micellar nanoparticles that mediated robust co-encapsulation and hydrogen peroxide (H2O2) and acid-responsive release of dual antitumor drugs, curcumin (Cur) and sorafenib tosylate (Sor). These dual drug-loaded nanoparticles (PBN-Cur/Sor) exhibited a greatly enhanced anticancer effect toward U87 MG-luciferase glioblastoma cells. The facile synthesis of phenylboronic acid-functionalized copolypeptides from BPA coupled with their robust drug loading and responsive drug release behaviors make them interesting for construction of smart cancer nanomedicines.


Asunto(s)
Antineoplásicos , Curcumina , Nanopartículas , Antineoplásicos/química , Antineoplásicos/farmacología , Ácidos Borónicos , Curcumina/química , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Peróxido de Hidrógeno , Micelas , Nanopartículas/química , Polietilenglicoles/química
8.
Biomacromolecules ; 23(1): 377-387, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-34913676

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common malignancy in children. Although intensive chemotherapy greatly improved the survival rate, it is often accompanied by severe and lifelong side effects as a result of weak ALL selectivity. The intensive and poorly selective chemotherapy is also detrimental to patients' immune system. There is an urgent need to develop more selective and less toxic chemotherapy for ALL. Here, we report daratumumab-polymersome-vincristine (DP-VCR) as a CD38-directed nanotherapy for ALL. DP-VCR showed selective uptake in CD38-positive 697 and Nalm-6-Luc ALL cells and potent anti-ALL activity with an IC50 as low as 0.06 nM VCR, which was 13.7-fold more potent than free VCR. In contrast, no toxicity to human peripheral blood mononuclear cells was detected for DP-VCR even at 108.3 nM VCR. The apoptotic assays confirmed a high selectivity of DP-VCR to CD38-positive ALL cells. DP-VCR exhibited superior treatment of both 697 and Nalm-6-Luc orthotopic ALL models to all controls, as revealed by significant survival benefit and marked reduction of leukemia burden in bone marrow, blood, spleen, and liver. Importantly, DP-VCR induced few side effects. DP-VCR emerges as a safe and potent nanotherapy for CD38-positive ALL.


Asunto(s)
Leucocitos Mononucleares , Leucemia-Linfoma Linfoblástico de Células Precursoras , Recuento de Células , Niño , Humanos , Leucocitos Mononucleares/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Vincristina/farmacología , Vincristina/uso terapéutico
9.
Biomacromolecules ; 22(11): 4814-4822, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34677048

RESUMEN

Targeted molecular therapy, for example, with sorafenib (SF) is considered as a new and potent strategy for glioblastoma (GBM) that remains hard to treat today. Several clinical trials with SF, as monotherapy or combination therapy with current treatments, have not met the clinical endpoints, likely as a result of the blood-brain barrier (BBB) and inferior GBM delivery. Here, we designed and explored small, smart, and LDLR-specific micelles to load SF (LDLR-mSF) and to improve SF therapy of GBM by enhancing BBB penetration, GBM accumulation, and cell uptake. LDLR-mSF with 2.5% ApoE peptide functionality based on poly(ethylene glycol)-poly(ε-caprolactone-co-dithiolane trimethylene carbonate)-mefenamate exhibited nearly quantitative SF loading, small size (24 nm), high colloidal stability, and glutathione-activated SF release. The in vitro and in vivo studies certified that LDLR-mSF greatly enhanced BBB permeability and U-87 MG cell uptake and caused 10.6- and 12.9-fold stronger anti-GBM activity and 6.0- and 2.5-fold higher GBM accumulation compared with free SF and non-LDLR mSF controls, respectively. The treatment of an orthotopic human GBM tumor model revealed that LDLR-mSF at a safe dosage of 15 mg of SF/kg significantly retarded tumor progression and improved the survival rate by inducing tumor cell apoptosis and inhibiting tumor angiogenesis. These small, smart, and LDLR-specific micelles provide a potential solution to enhance targeted molecular therapy of GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Barrera Hematoencefálica , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Humanos , Micelas , Sorafenib/farmacología
10.
Biomacromolecules ; 22(6): 2625-2640, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34076415

RESUMEN

In this paper, we present well-defined dPGS-SS-PCL/PLGA/PLA micellar systems demonstrating excellent capabilities as a drug delivery platform in light of high stability and precise in vitro and in vivo drug release combined with active targetability to tumors. These six amphiphilic block copolymers were each targeted in two different molecular weights (8 or 16 kDa) and characterized using 1H NMR, gel permeation chromatography (GPC), and elemental analysis. The block copolymer micelles showed monodispersed size distributions of 81-187 nm, strong negative charges between -52 and -41 mV, and low critical micelle concentrations (CMCs) of up to 1.13-3.58 mg/L (134-527 nM). The serum stability was determined as 94% after 24 h. The drug-loading efficiency for Sunitinib ranges from 38 to 83% (8-17 wt %). The release was selectively triggered by glutathione (GSH) and lipase, reaching 85% after 5 days, while only 20% leaching was observed under physiological conditions. Both the in vitro and in vivo studies showed sustained release of Sunitinib over 1 week. CCK-8 assays on HeLa lines demonstrated the high cell compatibility (1 mg/mL, 94% cell viability, 48 h) and the high cancer cell toxicity of Sunitinib-loaded micelles (IC50 2.5 µg/mL). By in vivo fluorescence imaging studies on HT-29 tumor-bearing mice, the targetability of dPGS7.8-SS-PCL7.8 enabled substantial accumulation in tumor tissue compared to nonsulfated dPG3.9-SS-PCL7.8. As a proof of concept, Sunitinib-loaded dPGS-SS-poly(ester) micelles improved the antitumor efficacy of the chemotherapeutic. A tenfold lower dosage of loaded Sunitinib led to an even higher tumor growth inhibition compared to the free drug, as demonstrated in a HeLa human cervical tumor-bearing mice model. No toxicity for the organism was observed, confirming the good biocompatibility of the system.


Asunto(s)
Micelas , Neoplasias , Animales , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Ésteres , Glicerol , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Polietilenglicoles , Sulfatos
11.
Angew Chem Int Ed Engl ; 60(32): 17629-17637, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34036695

RESUMEN

Biodegradable nanostructures displaying aggregation-induced emission (AIE) are desirable from a biomedical point of view, due to the advantageous features of loading capacity, emission brightness, and fluorescence stability. Herein, biodegradable polymers comprising poly (ethylene glycol)-block-poly(caprolactone-gradient-trimethylene carbonate) (PEG-P(CLgTMC)), with tetraphenylethylene pyridinium-TMC (PAIE) side chains have been developed, which self-assembled into well-defined polymersomes. The resultant AIEgenic polymersomes are intrinsically fluorescent delivery vehicles. The presence of the pyridinium moiety endows the polymersomes with mitochondrial targeting ability, which improves the efficiency of co-encapsulated photosensitizers and improves therapeutic index against cancer cells both in vitro and in vivo. This contribution showcases the ability to engineer AIEgenic polymersomes with structure inherent fluorescence and targeting capacity for enhanced photodynamic therapy.


Asunto(s)
Antineoplásicos/farmacología , Plásticos Biodegradables/farmacología , Colorantes Fluorescentes/farmacología , Fármacos Fotosensibilizantes/farmacología , Poliésteres/farmacología , Polietilenglicoles/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/efectos de la radiación , Compuestos de Bencilideno/síntesis química , Compuestos de Bencilideno/farmacología , Compuestos de Bencilideno/efectos de la radiación , Plásticos Biodegradables/síntesis química , Plásticos Biodegradables/efectos de la radiación , Compuestos de Boro/síntesis química , Compuestos de Boro/farmacología , Compuestos de Boro/efectos de la radiación , Línea Celular Tumoral , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/efectos de la radiación , Humanos , Luz , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/efectos de la radiación , Poliésteres/síntesis química , Poliésteres/efectos de la radiación , Polietilenglicoles/síntesis química , Polietilenglicoles/efectos de la radiación , Compuestos de Piridinio/síntesis química , Compuestos de Piridinio/farmacología , Compuestos de Piridinio/efectos de la radiación
12.
Biomacromolecules ; 21(12): 5119-5127, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33174734

RESUMEN

Antiangiogenic therapy with bevacizumab while being interesting for metastatic triple-negative breast cancer (mTNBC) is restrained by tumor hypoxia elevation and cancer stem cell enrichment. Here, we find that neuropilin-1 (NRP-1)-targeted delivery of nucleus accumbens-associated protein-1 (NAC-1) siRNA mediated by tLyP-1 peptide-functionalized chimaeric polymersomes (tLyP-1-Ps) effectively sensitizes antiangiogenic therapy of mTNBC in vivo. tLyP-1-Ps showed good encapsulation (up to 14.4 wt. %) of siNAC-1, giving robust tLyP-1-Ps-siNAC-1 nanoformulation with a defined size of 48.5 nm (PDI = 0.13) and a surface charge of -9.2 mV, and mediated efficient cytoplasmic transportation of siNAC-1 in MDA-MB-231 TNBC cells, resulting in significant silencing of NAC-1 mRNA and the corresponding oncoprotein. Transwell invasion and wound healing assays revealed that tLyP-1-Ps-siNAC-1 potently inhibited MDA-MB-231 cell invasion and migration. Intriguingly, tLyP-1-Ps-siNAC-1 was shown to markedly improve the bevacizumab therapy of mTNBC, significantly curbing lung metastasis and prolonging the survival time of the MDA-MB-231 metastatic model. The combination of targeted NAC-1 gene silencing and antiangiogenic therapy appears to be an innovative treatment for mTNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Inhibidores de la Angiogénesis/farmacología , Línea Celular Tumoral , Humanos , Neuropilinas , ARN Interferente Pequeño/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética
13.
Biomacromolecules ; 21(6): 2049-2059, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32338875

RESUMEN

Carfilzomib (CFZ) is a second-generation proteasome inhibitor approved for treating relapsed/refractory multiple myeloma (MM). The clinical formulation utilizing sulfobutylether-ß-cyclodextrin to solubilize CFZ (Captisol, CFZ-CD) shows, however, short circulation time, lack of cell selectivity, and unmet antitumor efficacy. Here, we designed and prepared A6 peptide (sequence: KPSSPPEE)-tagged core-disulfide-cross-linked biodegradable micelles (A6-PMs) for targeted CFZ therapy of CD44-overexpressing LP-1 human MM in vivo. A6-PMs had a small size of about 40 nm and stable CFZ encapsulation. CFZ-loaded micelles (CFZ-A6-PMs) showed a glutathione-triggered drug release profile with negligible drug leakage under physiological conditions. CFZ-A6-PMs displayed good proteasome activity inhibition and more potent apoptotic activity than CFZ-CD and nontargeted CFZ-PMs toward LP-1 MM cells in vitro. The in vivo fluorescence images revealed that Cy5-labeled A6-PMs induced much higher tumor accumulation than the nontargeted Cy5-labeled PMs control. The systemic administration of CFZ-A6-PMs to subcutaneous LP-1 xenografts in mice brought about notably more potent tumor suppression, higher survival rate and lower systemic toxicities than clinically used CFZ-CD formulation. These A6-tagged core-disulfide-cross-linked micelles appear interesting for targeted delivery of proteasome inhibitors to CD44+ MM.


Asunto(s)
Antineoplásicos , Mieloma Múltiple , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Disulfuros , Ratones , Micelas , Mieloma Múltiple/tratamiento farmacológico , Oligopéptidos , Fragmentos de Péptidos , Inhibidores de Proteasoma/uso terapéutico , Activador de Plasminógeno de Tipo Uroquinasa
14.
Biomacromolecules ; 21(1): 104-113, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31532629

RESUMEN

Metastasis is responsible for >90% of the deaths of breast cancer patients in the clinic. Here, we report on cross-linked multifunctional hyaluronic acid nanoparticles carrying docetaxel (DTX-CMHN) for enhanced suppression of highly metastatic 4T1 breast tumors in vivo. DTX-CMHN was formed from a single and all-natural hyaluronic acid-g-polytyrosine-lipoic acid conjugate (HA-g-PTyr-LA; HA, 20 kDa; PTyr, 2.2 kDa), and the size of DTX-CMHN increased from 69 to 78 to 96 nm as the increasing degree of substitution (DS) of PTyr increased from 4 to 11 to 15, respectively. Robust encapsulation of DTX was obtained when DS ≥ 11. DTX-CMHN while steady in a nonreducing environment was destabilized under 10 mM glutathione releasing ∼90% of the DTX within 24 h. It is noteworthy that DTX-CMHN exhibited better antitumor, antimigration, and anti-invasion activity in CD44-overexpressed 4T1-Luc breast cancer cells than free DTX. Interestingly, DTX-CMHN displayed a long elimination half-life of 5.75 h, in contrast to half-lives of 2.11 and 0.75 h for its non-cross-linked counterpart (DTX-MHN) and free DTX, respectively. In vivo therapeutic studies showed significantly better inhibition of primary 4T1-Luc tumor growth and lung metastasis and lower toxicity of DTX-CMHN compared with that of free DTX. These multifunctional nanoformulations based on a single and all-natural hyaluronic acid conjugate emerge as a potential nanoplatform for targeted treatment of CD44-positive metastatic tumors.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Receptores de Hialuranos , Nanopartículas/química , Animales , Antineoplásicos/farmacocinética , Neoplasias de la Mama/patología , Docetaxel/administración & dosificación , Docetaxel/farmacocinética , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Ácido Hialurónico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Terapia Molecular Dirigida/métodos , Nanomedicina/métodos , Nanopartículas/administración & dosificación , Ácido Tióctico/química , Distribución Tisular , Tirosina/química , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Biomacromolecules ; 21(2): 716-724, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-31809037

RESUMEN

Sorafenib (SF) is an FDA-approved molecular-targeted drug for treating hepatocellular carcinoma (HCC). SF, however, suffers from poor water solubility, low bioavailability, dose-limiting side effects, and possible drug resistance. Here, we report on apolipoprotein E peptide-decorated disulfide-cross-linked micellar SF (ApoE-Ms-SF) as a targeted and intelligent formulation for HCC therapy. ApoE-Ms-SF was prepared with a good SF loading of 7.0 wt %, small size (37 nm), high stability, and reduction-triggered drug release from poly(ethylene glycol)-b-poly(ε-caprolactone-co-dithiolane trimethylene carbonate)-mefenamate (PEG-P(CL-DTC)-MA) and ApoE-modified ApoE-PEG-P(CL-DTC) block copolymers. MTT assays in low-density lipoprotein receptors (LDLRs) overexpressing SMMC-7721 human liver cancer cells showed ApoE density-dependent antitumor potency of ApoE-Ms-SF, in which 7.5% ApoE led to the best antitumor effect (IC50: 8.5 vs 23.3 µg/mL for free SF). Confocal studies, flow cytometry, western blot, and apoptotic assays illustrated clearly a more efficient uptake of ApoE-Ms than nontargeted Ms by SMMC-7721 cells as well as lower phosphorylated extracellular signal-regulated kinase protein level and better cell apoptosis caused by ApoE-Ms-SF compared with Ms-SF and free SF. ApoE-Ms-SF revealed a long circulation time (elimination half-life = 6.8 h). DiR-loaded ApoE-Ms showed a significantly higher accumulation in SMMC-7721 tumor than the nontargeted counterpart. The therapeutic outcomes in the orthotopic SMMC-7721 tumor models demonstrated that ApoE-Ms-SF reduced SF-associated side effects and brought about enhanced angiogenesis inhibition and tumor apoptosis compared to free SF and Ms-SF controls, leading to a better treatment of HCC.


Asunto(s)
Antineoplásicos/metabolismo , Apolipoproteínas E/metabolismo , Carcinoma Hepatocelular/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Hepáticas/metabolismo , Micelas , Sorafenib/metabolismo , Animales , Antineoplásicos/administración & dosificación , Apolipoproteínas E/administración & dosificación , Carcinoma Hepatocelular/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Reactivos de Enlaces Cruzados/administración & dosificación , Reactivos de Enlaces Cruzados/metabolismo , Disulfuros/administración & dosificación , Disulfuros/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Sorafenib/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
16.
Small ; 15(45): e1902577, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31539202

RESUMEN

Future healthcare requires development of novel theranostic agents that are capable of not only enhancing diagnosis and monitoring therapeutic responses but also augmenting therapeutic outcomes. Here, a versatile and stable nanoagent is reported based on poly(ethylene glycol)-b-poly(l-thyroxine) (PEG-PThy) block copolypeptide for enhanced single photon emission computed tomography/computed tomography (SPECT/CT) dual-modality imaging and targeted tumor radiotherapy in vivo. PEG-PThy acquired by polymerization of l-thyroxine-N-carboxyanhydride (Thy-NCA) displays a controlled Mn , high iodine content of ≈49.2 wt%, and can spontaneously form 65 nm-sized nanoparticles (PThyN). In contrast to clinically used contrast agents like iohexol and iodixanol, PThyN reveals iso-osmolality, low viscosity, and long circulation time. While PThyN exhibits comparable in vitro CT attenuation efficacy to iohexol, it greatly enhances in vivo CT imaging of vascular systems and soft tissues. PThyN allows for surface decoration with the cRGD peptide achieving enhanced CT imaging of subcutaneous B16F10 melanoma and orthotopic A549 lung tumor. Taking advantages of a facile iodine exchange reaction, 125 I-labeled PThyN enables SPECT/CT imaging of tumors and monitoring of PThyN biodistribution in vivo. Besides, 131 I-labeled and cRGD-functionalized PThyN displays remarkable growth inhibition of the B16F10 tumor in mice (tumor inhibition rate > 89%). These poly(l-thyroxine) nanoparticles provide a unique and versatile theranostic platform for varying diseases.


Asunto(s)
Péptidos/química , Polietilenglicoles/química , Radioterapia/métodos , Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Medios de Contraste/química , Humanos , Nanomedicina Teranóstica/métodos
17.
Small ; 15(38): e1901849, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31379132

RESUMEN

Engineering biodegradable nanostructures with precise morphological characteristics is a key objective in nanomedicine. In particular, asymmetric (i.e., nonspherical) nanoparticles are desirable due to the advantageous effects of shape in a biomedical context. Using molecular engineering, it is possible to program unique morphological features into the self-assembly of block copolymers (BCPs). However, the criteria of biocompatibility and scalability limit progress due to the prevalence of nondegradable components and the use of toxic solvents during fabrication. To address this shortfall, a robust strategy for the fabrication of morphologically asymmetric nanoworms, comprising biodegradable BCPs, has been developed. Modular BCPs comprising poly (ethylene glycol)-block-poly(caprolactone-gradient-trimethylene carbonate) (PEG-PCLgTMC), with a terminal chain of quaternary ammonium-TMC (PTMC-Q), undergo self-assembly via direct hydration into well-defined nanostructures. By controlling the solution ionic strength during hydration, particle morphology switches from spherical micelles to nanoworms (of varying aspect ratio). This ionically-induced switch is driven by modulation of chain packing with salts screening interchain repulsions, leading to micelle elongation. Nanoworms can be loaded with cytotoxic cargo (e.g., doxorubicin) at high efficiency, preferentially interact with cancer cells, and increase tumor penetration. This work showcases the ability to program assembly of BCPs and the potential of asymmetric nanosystems in anticancer drug delivery.


Asunto(s)
Caproatos/química , Sistemas de Liberación de Medicamentos/métodos , Lactonas/química , Nanomedicina/métodos , Nanoestructuras/química , Micelas , Polímeros/química
18.
Crit Rev Biotechnol ; 39(4): 451-468, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30929528

RESUMEN

Coronary artery disease is a leading cause of death in developed nations. As the disease progresses, myocardial infarction can occur leaving areas of dead tissue in the heart. To compensate, the body initiates its own repair/regenerative response in an attempt to restore function to the heart. These efforts serve as inspiration to researchers who attempt to capitalize on the natural regenerative processes to further augment repair. Thus far, researchers are exploiting these repair mechanisms in the functionalization of soft materials using a variety of growth factor-, ligand- and peptide-incorporating approaches. The goal of functionalizing soft materials is to best promote and direct the regenerative responses that are needed to restore the heart. This review summarizes the opportunities for the use of functionalized soft materials for cardiac repair and regeneration, and some of the different strategies being developed.


Asunto(s)
Materiales Biocompatibles/uso terapéutico , Enfermedad de la Arteria Coronaria/terapia , Ingeniería de Tejidos/tendencias , Andamios del Tejido , Corazón , Humanos , Ligandos , Medicina Regenerativa/tendencias
19.
Mol Pharm ; 16(8): 3711-3719, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31299161

RESUMEN

The unbiased cytotoxicity and blood-brain barrier (BBB) impermeability render common chemotherapeutics nonviable for treating glioblastoma (GBM) patients. Although rigosertib (RGS), a RAS effector protein inhibitor, has shown low toxicity to healthy cells and high efficacy toward various cancer cells by inactivating PI3K-Akt, it hardly overcomes the BBB barricade. Here, we report that RGS loaded in apolipoprotein E derived peptide (ApoE)-targeted chimaeric polymersomes (ApoE-CP) is safe and highly potent against human GBM in vivo. ApoE-CP exhibited stable loading of RGS in its lumen, giving RGS nanoformulations (ApoE-CP-RGS) with a size of 60 nm and reduction-triggered drug release behavior. Notably, ApoE-CP-RGS induction markedly enhanced the G2/M cell cycle arrest and inhibitory effect in U-87 MG glioblastoma cells compared with the nontargeted CP-RGS and free RGS. The therapeutic outcomes in orthotopic U-87 MG GBM models demonstrated that ApoE-CP-RGS brought about effective GBM inhibition, greatly prolonged survival time, and depleted adverse effects. Rigosertib formulated in ApoE-targeted chimaeric polymersomes has emerged as a novel, highly specific, efficacious, and nontoxic treatment for glioblastoma.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Portadores de Fármacos/química , Glioblastoma/tratamiento farmacológico , Glicina/análogos & derivados , Sulfonas/administración & dosificación , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Apolipoproteínas E/química , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Liberación de Fármacos , Femenino , Glioblastoma/patología , Glicina/administración & dosificación , Glicina/efectos adversos , Glicina/farmacocinética , Humanos , Ratones , Nanopartículas/química , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/metabolismo , Fragmentos de Péptidos/química , Polímeros/química , Sulfonas/efectos adversos , Sulfonas/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Biomacromolecules ; 20(1): 184-191, 2019 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-30289705

RESUMEN

Low cell selectivity and uptake coupled with endosomal entrapment pose critical hurdles for intracellular delivery and clinical translation of therapeutic proteins. Herein, we report that smart polymersomes dually functionalized with cRGD and fusogenic GALA peptides (cRGD/GALA-Ps) enable ανß3-specific and high-efficiency cytosolic delivery of cytochrome C (CC), a model apoptotic protein, to A549 human lung cancer cells. cRGD/GALA-Ps was prepared with 20 mol % cRGD and varying GALA contents from 2 to 4 to 6 mol % via coassembly of PEG- b-poly(trimethylene carbonate- co-dithiolane trimethylene carbonate)-spermine (PEG- b-P(TMC- co-DTC)-spermine), cRGD-PEG- b-P(TMC- co-DTC), and maleimide-PEG- b-P(TMC- co-DTC) and postmodification using GALA-SH (sequence: CWEAALAEALAEALAEHLAEALAEALEALAA). cRGD/GALA-Ps loaded with ∼13 wt % CC displayed a small size of about 65 nm and fast glutathione-triggered protein release. Interestingly, cRGD/GALA-Ps maintained a similar targeting ability to cRGD-Ps in ανß3-positive A549 lung cancer cells, while markedly enhanced cytosolic release of FITC-labeled CC, as revealed by confocal microscopy. MTT assays exhibited that CC-loaded cRGD/GALA-Ps was significantly more potent than CC-loaded cRGD-Ps, in which cell viabilities of 76.2, 51.0, 29.6, and 35.5% were discerned for cRGD/GALA-Ps with 0, 2, 4, and 6 mol % GALA, respectively, at 15.4 µM CC. Apoptosis assays corroborated that cRGD/GALA-Ps-CC with 4 mol % GALA induced better apoptosis of A549 cells than cRGD-Ps-CC (cell apoptosis: 36.4 vs 14.4%). These results highlight that dual-functionalization of polymersomes with targeting and fusogenic peptides provides an appealing strategy for cytosolic protein delivery.


Asunto(s)
Apoptosis , Liberación de Fármacos , Oligopéptidos/química , Péptidos/química , Polímeros de Estímulo Receptivo/química , Línea Celular Tumoral , Citocromos c/administración & dosificación , Citocromos c/química , Dioxanos/química , Glutatión/química , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA