Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Molecules ; 27(9)2022 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-35566317

RESUMEN

Excessive inflammatory reaction aggravates brain injury and hinders the recovery of neural function in nervous system diseases. Microglia, as the major players of neuroinflammation, control the progress of the disease. There is an urgent need for effective non-invasive therapy to treat neuroinflammation mediated by microglia. However, the lack of specificity of anti-inflammatory agents and insufficient drug dose penetrating into the brain lesion area are the main problems. Here, we evaluated a series of calixarenes and found that among them the self-assembling architecture of amphiphilic sulfonatocalix[8]arene (SC8A12C) had the most potent ability to suppress neuroinflammation in vitro and in vivo. Moreover, SC8A12C assemblies were internalized into microglia through macropinocytosis. In addition, after applying the SC8A12C assemblies to the exposed brain tissue, we observed that SC8A12C assemblies penetrated into the brain parenchyma and eliminated the inflammatory factor storm, thereby restoring neurobiological functions in a mouse model of traumatic brain injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Calixarenos , Animales , Lesiones Traumáticas del Encéfalo/patología , Calixarenos/farmacología , Calixarenos/uso terapéutico , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Microglía , Enfermedades Neuroinflamatorias
2.
J Nanobiotechnology ; 19(1): 451, 2021 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-34961540

RESUMEN

BACKGROUND: Hypoxia is a major contributor to global kidney diseases. Targeting hypoxia is a promising therapeutic option against both acute kidney injury and chronic kidney disease; however, an effective strategy that can achieve simultaneous targeted kidney hypoxia imaging and therapy has yet to be established. Herein, we fabricated a unique nano-sized hypoxia-sensitive coassembly (Pc/C5A@EVs) via molecular recognition and self-assembly, which is composed of the macrocyclic amphiphile C5A, the commercial dye sulfonated aluminum phthalocyanine (Pc) and mesenchymal stem cell-excreted extracellular vesicles (MSC-EVs). RESULTS: In murine models of unilateral or bilateral ischemia/reperfusion injury, MSC-EVs protected the Pc/C5A complex from immune metabolism, prolonged the circulation time of the complex, and specifically led Pc/C5A to hypoxic kidneys via surface integrin receptor α4ß1 and αLß2, where Pc/C5A released the near-infrared fluorescence of Pc and achieved enhanced hypoxia-sensitive imaging. Meanwhile, the coassembly significantly recovered kidney function by attenuating cell apoptosis, inhibiting the progression of renal fibrosis and reducing tubulointerstitial inflammation. Mechanistically, the Pc/C5A coassembly induced M1-to-M2 macrophage transition by inhibiting the HIF-1α expression in hypoxic renal tubular epithelial cells (TECs) and downstream NF-κB signaling pathway to exert their regenerative effects. CONCLUSION: This synergetic nanoscale coassembly with great translational potential provides a novel strategy for precise kidney hypoxia diagnosis and efficient kidney injury treatment. Furthermore, our strategy of coassembling exogenous macrocyclic receptors with endogenous cell-derived membranous structures may offer a functional platform to address multiple clinical needs.


Asunto(s)
Lesión Renal Aguda/diagnóstico por imagen , Lesión Renal Aguda/tratamiento farmacológico , Hipoxia de la Célula/efectos de los fármacos , Vesículas Extracelulares/química , Compuestos Macrocíclicos/química , Tensoactivos/química , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Calixarenos/química , Calixarenos/metabolismo , Calixarenos/farmacología , Calixarenos/uso terapéutico , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Indoles/química , Indoles/metabolismo , Indoles/farmacología , Indoles/uso terapéutico , Inflamación , Integrinas/metabolismo , Compuestos Macrocíclicos/metabolismo , Compuestos Macrocíclicos/farmacología , Compuestos Macrocíclicos/uso terapéutico , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , FN-kappa B/metabolismo , Compuestos Organometálicos/química , Compuestos Organometálicos/metabolismo , Compuestos Organometálicos/farmacología , Compuestos Organometálicos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Tensoactivos/metabolismo , Tensoactivos/farmacología , Tensoactivos/uso terapéutico
3.
Molecules ; 26(13)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209495

RESUMEN

Since cancer is a multifactorial disease with a high mortality rate, the study of new therapeutic strategies is one of the main objectives in modern research. Numerous chemotherapeutic agents, although widely used, have the disadvantage of being not very soluble in water or selective towards cancerous cells, with consequent side effects. Therefore, in recent years, a greater interest has emerged in innovative drug delivery systems (DDSs) such as calixarene, a third-generation supramolecular compound. Calixarene and its water-soluble derivatives show good biocompatibility and have low cytotoxicity. Thanks to their chemical-physical characteristics, calixarenes can be easily functionalized, and by itself can encapsulate host molecules forming nanostructures capable of releasing drugs in a controlled way. The encapsulation of anticancer drugs in a calixarene derivate improves their bioavailability and efficacy. Thus, the use of calixarenes as carriers of anticancer drugs could reduce their side effects and increase their affinity towards the target. This review summarizes the numerous research advances regarding the development of calixarene nanoparticles capable of encapsulating various anticancer drugs.


Asunto(s)
Antineoplásicos , Calixarenos , Portadores de Fármacos , Nanopartículas , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Calixarenos/química , Calixarenos/uso terapéutico , Portadores de Fármacos/química , Portadores de Fármacos/uso terapéutico , Humanos , Nanopartículas/química , Nanopartículas/uso terapéutico
4.
Cells ; 10(5)2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-34063063

RESUMEN

Although the overall survival rate of papillary or follicular thyroid cancers is good, anaplastic carcinomas and radio iodine refractory cancers remain a significant therapeutic challenge. Galectin-1 (Gal-1) is overexpressed in tumor cells and tumor-associated endothelial cells, and is broadly implicated in angiogenesis, cancer cell motility and invasion, and immune system escape. Our team has previously demonstrated a higher serum level of Gal-1 in patients with differentiated thyroid cancers versus healthy patients, and explored, by a knockdown strategy, the effect of Gal-1 silencing on cell proliferation and invasion in vitro, and on tumor and metastasis development in vivo. OTX008 is a calixarene derivative designed to bind the Gal-1 amphipathic ß-sheet conformation and has previously demonstrated anti-proliferative and anti-invasive properties in several cancer cell lines including colon, breast, head and neck, and prostate cancer lines. In the current work, the impacts of OTX008 were evaluated in six thyroid cancer cell lines, and significant inhibitions of proliferation, migration, and invasion were observed in all lines expressing high Gal-1 levels. In addition, the signaling pathways affected by this drug were examined using RPPA (reverse phase protein array) and phosphoprotein expression assays, and opposite regulation of eNos, PYK2, and HSP27 by OTX008 was detected by comparing the two anaplastic lines 8505c and CAL 62. Finally, the sensitive 8505c line was xenografted in nude mice, and 3 weeks of OTX008 treatment (5 mg/kg/day) demonstrated a significant reduction in tumor and lung metastasize sizes without side effects. Overall, OXT008 showed significant anti-cancer effects both in vitro and in vivo in thyroid cancer lines expressing Gal-1, supporting further investigation of the molecular mechanisms of the drug and future clinical trials in patients with anaplastic thyroid cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Calixarenos/uso terapéutico , Galectina 1/metabolismo , Neoplasias de la Tiroides/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Calixarenos/farmacología , Línea Celular Tumoral , Proliferación Celular , Quinasa 2 de Adhesión Focal/metabolismo , Galectina 1/antagonistas & inhibidores , Proteínas de Choque Térmico/metabolismo , Humanos , Masculino , Ratones , Chaperonas Moleculares/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Neoplasias de la Tiroides/metabolismo
5.
J Exp Clin Cancer Res ; 38(1): 423, 2019 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-31640796

RESUMEN

BACKGROUND: Galectins are beta-galactose specific binding proteins. In human cancers, including hepatocellular carcinoma (HCC), galectin-1 (Gal-1) is often found to be overexpressed. In order to combat the dismal diagnosis and death rates of HCC, gene silencing and targeted inhibition of Gal-1 was investigated for its improved therapeutic potential. METHODS: Cellular and secretory Gal-1 levels were analyzed using HCC clinical samples. The study of Gal-1 was carried by both knockdown and overexpression approaches. The stable clones were tested by in vitro assays and in vivo experiments. Mass spectrometry was used to identify downstream targets of Gal-1. The upstream regulator of Gal-1, microRNA-22 (miR-22) was characterized by functional assays. The therapeutic effect of inhibiting Gal-1 was also analyzed. RESULTS: Gal-1 overexpression was observed in HCC and correlated with aggressive clinicopathological features and poorer survival. The loss of Gal-1 resulted in hindered cell migration, invasion and anchorage independent growth. This was also observed in the animal models, in that when Gal-1 was knocked down, there were fewer lung metastases. Proteomic profiling of control and Gal-1 knockdown cells identified that the level of retention in endoplasmic reticulum 1 (RER1) was suppressed when Gal-1 level was reduced. The cell motility of Gal-1 knockdown cells was enhanced upon the rescue of RER1 expression. In HCC tissues, Gal-1 and RER1 expressions displayed a significant positive correlation. The upstream regulator of Gal-1, miR-22 was observed to be underexpressed in HCC tissues and negatively correlated with Gal-1. Silencing of miR-22 resulted in the upregulation of Gal-1 and enhanced cell growth, migration and invasion. However, such enhancement was abolished in cells treated with OTX008, an inhibitor of Gal-1. Combinational treatment of OTX008 and sorafenib significantly reduced tumor growth and size. CONCLUSIONS: Gal-1 overexpression was detected in HCC and this played a role in promoting tumorigenic processes and metastasis. The function of Gal-1 was found to be mediated through RER1. The correlations between miR-22, Gal-1 and RER1 expressions demonstrated the importance of miR-22 regulation on Gal-1/RER1 oncogenic activity. Lastly, the combinational treatment of OTX008 and sorafenib proved to be an improved therapeutic option compared to when administering sorafenib alone.


Asunto(s)
Calixarenos/uso terapéutico , Carcinoma Hepatocelular/genética , Galectina 1/efectos adversos , Neoplasias Hepáticas/genética , Sorafenib/uso terapéutico , Animales , Calixarenos/farmacología , Carcinoma Hepatocelular/patología , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Sorafenib/farmacología , Transfección
6.
ACS Appl Mater Interfaces ; 11(42): 38497-38502, 2019 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-31556585

RESUMEN

A charge-reversal amphiphilic pillar[5]arene, P5NH-DCA, bearing 10 charge-reversal headgroups is reported. It targets the cell membrane of cancer cells and selectively destroys the cancer cells by disrupting the membrane. In the acidic tumor microenvironment, the headgroup charge of P5NH-DCA reversed from negative to positive owing to hydrolysis of the acid-labile amide group. The hydrolyzed product bearing multiple positive charges can bind to the cell membrane and then disrupt the membrane of cancer cells with high efficiency. However, under the neutral microenvironment of healthy cells, the negatively charged P5NH-DCA remains stable and the cytotoxicity is considerably reduced. The strategy killing the cancer cells by membrane disruption may represent a new route of cancer chemotherapy.


Asunto(s)
Calixarenos/química , Membrana Celular/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Calixarenos/farmacología , Calixarenos/uso terapéutico , Línea Celular Tumoral , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Desnudos , Microscopía Confocal , Neoplasias/tratamiento farmacológico
7.
Nat Commun ; 10(1): 3546, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31391464

RESUMEN

Polyamines are essential for the growth of eukaryotic cells and can be dysregulated in tumors. Here we describe a strategy to deplete polyamines through host-guest encapsulation using a peptide-pillar[5]arene conjugate (P1P5A, P1 = RGDSK(N3)EEEE) as a supramolecular trap. The RGD in the peptide sequence allows the molecule to bind to integrin αvß3-overexpressing tumor cells. The negative charged glutamic acid residues enhance the inclusion affinities between the pillar[5]arene and cationic polyamines via electrostatic interactions and facilitate the solubility of the conjugate in aqueous media. The trap P1P5A efficiently encapsulates polyamines with association constants of 105-106 M-1. We show that P1P5A has a wide spectrum of antitumor activities, and induces apoptosis via affecting the polyamine biosynthetic pathway. Experiments in vivo show that P1P5A effectively inhibits the growth of breast adenocarcinoma xenografts in female nude mice. This work reveals an approach for suppressing tumor growth by using supramolecular macrocycles to trap polyamines in tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Poliaminas/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Vías Biosintéticas/efectos de los fármacos , Neoplasias de la Mama/patología , Calixarenos/química , Calixarenos/farmacología , Calixarenos/uso terapéutico , Cationes/química , Cationes/metabolismo , Femenino , Humanos , Integrina alfaVbeta3/metabolismo , Células MCF-7 , Ratones , Ratones Desnudos , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Poliaminas/química , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Int J Mol Sci ; 19(1)2018 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-29320431

RESUMEN

The incidence and mortality of cancer have increased over the past decades. Significant progress has been made in understanding the underpinnings of this disease and developing therapies. Despite this, cancer still remains a major therapeutic challenge. Current therapeutic research has targeted several aspects of the disease such as cancer development, growth, angiogenesis and metastases. Many molecular and cellular mechanisms remain unknown and current therapies have so far failed to meet their intended potential. Recent studies show that glycans, especially oligosaccharide chains, may play a role in carcinogenesis as recognition patterns for galectins. Galectins are members of the lectin family, which show high affinity for ß-galactosides. The galectin-glycan conjugate plays a fundamental role in metastasis, angiogenesis, tumor immunity, proliferation and apoptosis. Galectins' action is mediated by a structure containing at least one carbohydrate recognition domain (CRD). The potential prognostic value of galectins has been described in several neoplasms and helps clinicians predict disease outcome and determine therapeutic interventions. Currently, new therapeutic strategies involve the use of inhibitors such as competitive carbohydrates, small non-carbohydrate binding molecules and antibodies. This review outlines our current knowledge regarding the mechanism of action and potential therapy implications of galectins in cancer.


Asunto(s)
Galectinas/metabolismo , Neoplasias/tratamiento farmacológico , Calixarenos/metabolismo , Calixarenos/uso terapéutico , Ensayos Clínicos como Asunto , Galactosa/análogos & derivados , Galactosa/metabolismo , Galactosa/uso terapéutico , Galectinas/antagonistas & inhibidores , Humanos , Mananos , Neoplasias/patología , Pectinas/química , Pectinas/uso terapéutico , Péptidos/metabolismo , Péptidos/uso terapéutico , Polisacáridos/metabolismo , Polisacáridos/uso terapéutico , Tiogalactósidos/química , Tiogalactósidos/metabolismo , Tiogalactósidos/uso terapéutico
9.
Anticancer Res ; 36(10): 5053-5061, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27798864

RESUMEN

The goal of this study was to develop combinatorial application of two drugs currently either in active use as anticancer agents (rapamycin) or in clinical trials (OTX008) as a novel strategy to inhibit Harvey RAS (HRAS)-driven tumor progression. HRAS anchored to the plasma membrane shuttles from the lipid ordered (Lo) domain to the lipid ordered/lipid disordered border upon activation, and retention of HRAS at these sites requires galectin-1. We recently showed that genetically enforced Lo sequestration of HRAS inhibited mitogen-activated protein kinase (MAPK) signaling, but not phoshatidylinositol 3-kinase (PI3K) activation. Here we show that inhibition of galectin-1 with OTX008 sequestered HRAS in the Lo domain, blocked HRAS-mediated MAPK signaling, and attenuated HRAS-driven tumor progression in mice. HRAS-driven tumor growth was also attenuated by treatment with mammalian target of rapamycin (mTOR) inhibitor rapamycin, and this effect was further enhanced in tumors driven by Lo-sequestered HRAS. These drugs also revealed bidirectional cross-talk in HRAS pathways. Moreover, dual pathway inhibition with OTX008 and rapamycin resulted in nearly complete ablation of HRAS-driven tumor growth. These findings indicate that membrane microdomain sequestration of HRAS with galectin-1 inhibition, coupled with mTOR inhibition, may support a novel therapeutic approach to treat HRAS-mutant cancer.


Asunto(s)
Antineoplásicos/farmacología , Calixarenos/farmacología , Galectina 1/antagonistas & inhibidores , Neoplasias/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Sirolimus/farmacología , Animales , Antineoplásicos/uso terapéutico , Calixarenos/uso terapéutico , Femenino , Proteínas Fluorescentes Verdes/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Células 3T3 NIH , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
10.
Drug Des Devel Ther ; 9: 2831-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26082613

RESUMEN

Research on the therapeutic applications of calixarene derivatives is an emerging area of interest. The anticancer activity of various functionalized calixarenes has been reported by several research groups. Due to their superior geometric shape, calixarenes can accommodate drug molecules by forming inclusion complexes. Controlled release of anticancer drugs by calixarenes might help in targeted chemotherapy. This review summarizes the anticancer potential of the calixarenes and their drug loading properties. The potential use of calixarenes in chemoradiotherapy is also highlighted in brief.


Asunto(s)
Antineoplásicos/uso terapéutico , Calixarenos/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Calixarenos/administración & dosificación , Quimioradioterapia/métodos , Preparaciones de Acción Retardada , Humanos
11.
Eur J Med Chem ; 89: 279-95, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25462244

RESUMEN

Calixarene 0118 is a potent anti-angiogenic agent that effectively inhibited tumor growth in preclinical studies, and is currently being evaluated in a phase I clinical trial. We have designed two close mimetics of calixarene 0118 containing a terminal alkynyl-functional group, and developed an optimized semi-automated procedure for radiolabeling with 2-[(18)F]fluoroethylazide using click chemistry. Following semi-preparative HPLC purification and formulation, the lower-rim modified analog [(18)F]6 and the equatorially labeled [(18)F]13 were obtained in >97% radiochemical purity and overall decay-corrected isolated radiochemical yields of 18.7 ± 2.7% (n = 4) and 10.2 ± 5.0% (n = 4), respectively, in a total synthesis time of about 2 h. Preliminary in vivo studies in nude mice bearing human tumor xenografts revealed highest accumulation of both tracers in the liver, followed by spleen, kidney, lung and bone, with no substantial uptake in the tumor. Still, these first-in-class radiotracers are a valuable tool for pharmacokinetic profiling and improvement of calixarene-based anti-angiogenic therapeutics in the future, as similar radiolabeling strategies may be applied to other compounds in the calixarene series. The cold reference compounds of the radiotracers were characterized in terms of cytotoxicity and anti-proliferative effects on HUVEC cells and on MA148 human ovarian carcinoma cells, along with the respective precursors, a small series of 0118 analogs modified with short-chain linear alkyl substituents, and a PEG3-spaced calixarene dimer. While all of the new analogs proved at least equipotent to parent 0118, some of them inhibited HUVEC and MA148 cell growth almost 4- and 10-fold more effectively, rendering these analogs promising candidates for further evaluation in anti-angiogenic cancer therapy.


Asunto(s)
Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/uso terapéutico , Azidas/química , Calixarenos/síntesis química , Calixarenos/uso terapéutico , Radioisótopos de Flúor/química , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Animales , Calixarenos/química , Calixarenos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Química Clic , Reacción de Cicloadición , Diseño de Fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Radioquímica , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Bioconjug Chem ; 24(10): 1710-20, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24041198

RESUMEN

MUC1 protein overexpressed in human epithelial carcinoma is a target in development of novel anticancer vaccines. Multiple units of immunodominant B-cell epitope PDTRP MUC1 core sequence were conjugated to calix[4,8]arene platforms containing TLR2 ligand, to produce two novel anticancer self-adjuvant vaccine candidates. The immunogenicity of the synthetic constructs was investigated by immunization of mice in vivo. ELISA assay evidenced that the vaccine candidates stimulate anti MUC1 IgG antibody production (major for the octavalent construct) and no additive effect but a multivalency effect was observed when compared to an analogous monovalent. Octa- and tetravalent constructs lacking in PDTRP peptide moieties did not show anti MUC1 IgG antibody production in mice. The antibodies induced by the synthesized constructs are able to recognize the MUC1 structures present on MCF7 tumor cells. The results display that calixarenes are convenient platforms for building multicomponent self-adjuvant vaccine constructs promising as immunotherapeutic anticancer agents.


Asunto(s)
Adyuvantes Inmunológicos/química , Calixarenos/química , Vacunas contra el Cáncer/química , Epítopos Inmunodominantes/química , Mucina-1/química , Neoplasias/prevención & control , Adyuvantes Inmunológicos/uso terapéutico , Animales , Formación de Anticuerpos , Calixarenos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Femenino , Humanos , Epítopos Inmunodominantes/uso terapéutico , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Mucina-1/inmunología , Mucina-1/uso terapéutico , Neoplasias/inmunología , Oligopéptidos/química , Oligopéptidos/uso terapéutico , Receptor Toll-Like 2/química
13.
Invest New Drugs ; 31(5): 1142-50, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23392775

RESUMEN

Previously, we reported on the anti-tumor activities of two designed calix[4]arene-based topomimetics (PTX008 and PTX009) of the amphipathic, angiostatic peptide Anginex. Here, we chemically modified the hydrophobic and hydrophilic faces of PTX008 and PTX009, and discovered new calixarene compounds that are more potent, cytotoxic anti-tumor agents. One of them, PTX013, is particularly effective at inhibiting the growth of several human cancer cell lines, as well as drug resistant cancer cells. Mechanistically, PTX013 induces cell cycle arrest in sub-G1 and G0/G1 phases of e.g. SQ20B cells, a radio-resistant human head and neck carcinoma model. In the syngeneic B16F10 melanoma tumor mouse model, PTX013 (0.5 mg/Kg) inhibits tumor growth by about 50-fold better than parent PTX008. A preliminary pharmacodynamics study strongly suggests that PTX013 exhibits good in vivo exposure and a relatively long half-life. Overall, this research contributes to the discovery of novel therapeutics as potentially useful agents against cancer in the clinic.


Asunto(s)
Antineoplásicos/farmacología , Calixarenos/farmacología , Citotoxinas/farmacología , Animales , Antineoplásicos/uso terapéutico , Calixarenos/uso terapéutico , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citotoxinas/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Carga Tumoral/efectos de los fármacos
14.
Chem Commun (Camb) ; 47(21): 6063-5, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21519601

RESUMEN

A supramolecular drug delivery system has been developed via the self-assembly of a supramolecular amphiphilic polymer, which is constructed by the host-guest interaction of hydrophilic PEGylated calix[4]arene and hydrophobic photosensitizer chlorin e6. It provides a new strategy for the preparation of supramolecular polymeric micelles, and plays an important role in biological applications.


Asunto(s)
Calixarenos/química , Micelas , Fenoles/química , Porfirinas/química , Calixarenos/uso terapéutico , Clorofilidas , Femenino , Células HeLa , Humanos , Fenoles/uso terapéutico , Fotoquimioterapia , Polietilenglicoles/química , Porfirinas/uso terapéutico , Neoplasias del Cuello Uterino/tratamiento farmacológico
15.
Bioorg Med Chem Lett ; 20(15): 4412-5, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20605095

RESUMEN

Li-Fraumeni syndrome, a hereditary disorder characterized by familial clusters of early-onset multiple tumors, is caused by mutation of the TP53 gene, which encodes the p53 tumor suppressor protein. Mutation of Arg337 to histidine in the tetramerization domain of p53 is most frequently observed in Li-Fraumeni syndrome. This mutation is reported to destabilize the tetrameric structure of p53. We designed and synthesized calix[6]arene derivatives, which have six imidazole or pyrazole groups at the upper rim. In this study, we report, for the first time, the enhancement of the in vivo transcriptional activity of the most common Li-Fraumeni p53 mutant by imidazole-calix[6]arene through stabilization of the oligomer formation.


Asunto(s)
Calixarenos/química , Proteína p53 Supresora de Tumor/metabolismo , Calixarenos/uso terapéutico , Humanos , Síndrome de Li-Fraumeni/tratamiento farmacológico , Conformación Molecular , Mutación , Multimerización de Proteína , Estabilidad Proteica , Termodinámica , Transcripción Genética , Temperatura de Transición , Proteína p53 Supresora de Tumor/genética
16.
J Biol Inorg Chem ; 14(1): 101-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18810507

RESUMEN

Mononuclear 5-(4-pyridyl)-10,15,20-triphenylporphyrin and 5-(3-pyridyl)-10,15,20-triphenylporphyrin as well as tetranuclear 5,10,15,20-tetra(4-pyridyl)porphyrin (tetra-4-pp) and 5,10,15,20-tetra(3-pyridyl)porphyrin) (tetra-3-pp) arene ruthenium(II) derivatives (arene is C(6)H(5)Me or p-Pr(i)C(6)H(4)Me) were prepared and evaluated as potential dual photosensitizers and chemotherapeutics in human Me300 melanoma cells. In the absence of light, all tetranuclear complexes were cytotoxic (IC(50) < or = 20 microM), while the mononuclear derivatives were not (IC(50) > or = 100 microM). Kinetic studies of tritiated thymidine and tritiated leucine incorporations in cells exposed to a low concentration (5 microM) of tetranuclear p-cymene derivatives demonstrated a rapid inhibition of DNA synthesis, while protein synthesis was inhibited only later, suggesting arene ruthenium-DNA interactions as the initial cytotoxic process. All complexes exhibited phototoxicities toward melanoma cells when exposed to laser light of 652 nm. At low concentration (5 microM), LD(50) of the mononuclear derivatives was between 5 and 10 J/cm(2), while for the tetranuclear derivatives LD(50) was approximately 2.5 J/cm(2) for the [Ru(4)(eta(6)-arene)(4)(tetra-4-pp)Cl(8)] complexes and less than 0.5 J/cm(2) for the [Ru(4)(eta(6)-arene)(4)(tetra-3-pp)Cl(8)] complexes. Examination of cells under a fluorescence microscope revealed the [Ru(4)(eta(6)-arene)(4)(tetra-4-pp)Cl(8)] complexes as cytoplasmic aggregates, whereas the [Ru(4)(eta(6)-arene)(4)(tetra-3-pp)Cl(8)] complexes were homogenously dispersed in the cytoplasm. Thus, these complexes present a dual synergistic effect with good properties of both the arene ruthenium chemotherapeutics and the porphyrin photosensitizer.


Asunto(s)
Antineoplásicos/uso terapéutico , Calixarenos/uso terapéutico , Metaloporfirinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Rutenio/uso terapéutico , Antineoplásicos/síntesis química , Antineoplásicos/química , Calixarenos/química , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Luz , Metaloporfirinas/síntesis química , Metaloporfirinas/química , Estructura Molecular , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Rutenio/química , Factores de Tiempo , Células Tumorales Cultivadas
17.
Cancer Lett ; 265(2): 270-80, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18378392

RESUMEN

Combination of chemotherapeutic agents and angiogenesis inhibitors is now commonly employed in the clinic to treat cancer. Here, we used angiostatic agents anginex and 0118, in combination with the chemotherapeutic irofulven, to treat human ovarian tumor xenografts in mice. General linear mixed models were used to statistically analyze tumor growth curves. Overall, combination of a low, non-toxic dose of irofulven with either angiogenesis inhibitor was more effective at inhibiting tumor growth than any of the single agent therapies. For example, the anginex/irofulven and 0118/irofulven combinations inhibited tumor growth relative to controls by 92% (p<0.0001) and 96% (p<0.0001), respectively, with the 0118/irofulven combinations yielding 100% complete responses. This study suggests that combination therapy of 0118 or anginex and irofulven may be highly effective in the clinical setting.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Proteínas/uso terapéutico , Sesquiterpenos/uso terapéutico , Animales , Calixarenos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Péptidos , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Chem Soc Rev ; 36(2): 254-66, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17264928

RESUMEN

Multivalency is a powerful concept which explains the strong binding observed in biological systems and guides the design and synthesis of ligands for self-assembly and molecular recognition in Chemistry. The phenol-formaldehyde cyclic oligomers, called calixarenes, have been used as scaffolds for the synthesis of multivalent ligands thanks to the fact that they have a variable number of reactive positions for attaching the ligating functions, well defined conformational properties and, in some cases, cavities of molecular dimensions eventually able to encapsulate guest species. This tutorial review illustrates the fundamental aspects of multivalency and the properties of calixarene-based multivalent ligands in lectin binding and inhibition, DNA condensation and cell transfection, protein surface recognition, self-assembly, crystal engineering, and nanofabrication.


Asunto(s)
Calixarenos/química , Sitios de Unión , Biotecnología/métodos , Calixarenos/síntesis química , Calixarenos/uso terapéutico , Carbohidratos/química , Ligandos , Péptidos/química
19.
Infect Immun ; 72(11): 6318-23, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15501760

RESUMEN

Tuberculosis remains the leading cause of death among infectious diseases, accounting for more than two million deaths annually. The incidence of the disease is increasing globally, partially because of the resurgence of drug-resistant strains of Mycobacterium tuberculosis. Calixarenes are macrocyclic oligomers, some of which are able to modify the growth of M. tuberculosis in infected cells. Most experimental work has been carried out with Macrocyclon, also known as HOC 12.5EO. In this study, we demonstrate that Macrocyclon is effective in controlling M. tuberculosis infections, and we provide evidence that its effect is partially mediated by an l-arginine-dependent mechanism of macrophage activation that involves the activity of the inducible nitric oxide synthase. We also show that Macrocyclon is effective in athymic and major histocompatibility complex class II-/- mice and synthesized a number of structurally related calixarenes expressing significant antimycobacterial activity.


Asunto(s)
Calixarenos/farmacología , Activación de Macrófagos , Mycobacterium tuberculosis/efectos de los fármacos , Polietilenglicoles/farmacología , Tuberculosis Pulmonar/prevención & control , Animales , Arginina/metabolismo , Células de la Médula Ósea , Calixarenos/síntesis química , Calixarenos/química , Calixarenos/uso terapéutico , Células Cultivadas , Femenino , Pulmón/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos Peritoneales/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Mycobacterium tuberculosis/aislamiento & purificación , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Polietilenglicoles/química , Polietilenglicoles/uso terapéutico , Bazo/microbiología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...