Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
Molecules ; 26(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070457

RESUMEN

Cisplatin and derivatives are highly effective in the treatment of a wide range of cancer types; however, these metallodrugs display low selectivity, leading to severe side effects. Additionally, their administration often results in the development of chemoresistance, which ultimately results in therapeutic failure. This scenario triggered the study of other transition metals with innovative pharmacological profiles as alternatives to platinum, ruthenium- (e.g., KP1339 and NAMI-A) and gold-based (e.g., Auranofin) complexes being among the most advanced in terms of clinical evaluation. Concerning the importance of improving the in vivo selectivity of metal complexes and the current relevance of ruthenium and gold metals, this review article aims to survey the main research efforts made in the past few years toward the design and biological evaluation of target-specific ruthenium and gold complexes. Herein, we give an overview of the inorganic and organometallic molecules conjugated to different biomolecules for targeting membrane proteins, namely cell adhesion molecules, G-protein coupled receptors, and growth factor receptors. Complexes that recognize the progesterone receptors or other targets involved in metabolic pathways such as glucose transporters are discussed as well. Finally, we describe some complexes aimed at recognizing cell organelles or compartments, mitochondria being the most explored. The few complexes addressing targeted gene therapy are also presented and discussed.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Compuestos de Oro/farmacología , Compuestos de Rutenio/farmacología , Antineoplásicos/administración & dosificación , Moléculas de Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Complejos de Coordinación/administración & dosificación , Compuestos de Oro/administración & dosificación , Humanos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Factores de Crecimiento/efectos de los fármacos , Compuestos de Rutenio/administración & dosificación
2.
Nat Biomed Eng ; 4(4): 463-475, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31685999

RESUMEN

Growth factors can stimulate tissue regeneration, but the side effects and low effectiveness associated with suboptimal delivery systems have impeded their use in translational regenerative medicine. Physiologically, growth factor interactions with the extracellular matrix control their bioavailability and spatiotemporal cellular signalling. Growth factor signalling is also controlled at the cell surface level via binding to heparan sulfate proteoglycans, such as syndecans. Here we show that vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor-BB (PDGF-BB) that were engineered to have a syndecan-binding sequence trigger sustained low-intensity signalling (tonic signalling) and reduce the desensitization of growth factor receptors. We also show in mouse models that tonic signalling leads to superior morphogenetic activity, with syndecan-binding growth factors inducing greater bone regeneration and wound repair than wild-type growth factors, as well as reduced tumour growth (associated with PDGF-BB delivery) and vascular permeability (triggered by VEGF-A). Tonic signalling via syndecan binding may also enhance the regenerative capacity of other growth factors.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/farmacología , Transducción de Señal/efectos de los fármacos , Sindecanos/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Becaplermina/metabolismo , Regeneración Ósea/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microfluídica , Modelos Animales , Neuropilina-1 , Receptores de Factores de Crecimiento/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Rev. cuba. endocrinol ; 27(3): 69-79, sep.-dic. 2016. ilus, tab
Artículo en Español | LILACS | ID: biblio-830470

RESUMEN

Antecedentes: la metformina, fármaco económico y seguro, ha demostrado mejorar el pronóstico de varios tipos de cánceres. Objetivo: revisar los aspectos más relevantes de la relación entre la diabetes mellitus, la metformina y el cáncer. Desarrollo: la diabetes mellitus, en especial la tipo 2, se relaciona con algunos cánceres (mama, hígado, páncreas, ginecológico, vejiga, colon y recto), y en el sexo masculino, aumenta su recurrencia y la mortalidad. Los mecanismos responsables de esta relación no están del todo esclarecidos. La insulina y el factor de crecimiento similar a la insulina en un estado de hiperinsulinismo e insulinorresistencia, pudieran desempeñar un papel fundamental en el desarrollo de cáncer, así como otros factores de riesgo comunes a la diabetes mellitus y al cáncer (alimentación no saludable, sedentarismo, adicciones, edad, sexo, etnia y raza). La proteína liver kinase B1 se ha identificado como una proteína supresora tumoral, y al unirse con la metformina interrumpe el complejo 1 de la cadena respiratoria mitocondrial, y conduce a la disminución de la síntesis de trifosfato de adenosin, y al aumento del cociente proteína activada por mitógenos-trifosfato de adenosin en el espacio intracelular. Los quimioterápicos, esteroides y antiandrógenos, pueden afectar negativamente el metabolismo hidrocarbonado. Algunas drogas antihiperglucemiantes se han relacionado a cánceres específicos, aunque las evidencias son pobres, indirectas y controversiales. Conclusiones: la metformina pudiera utilizarse en la prevención y el tratamiento de algunos cánceres, y reducir su recurrencia y la mortalidad. Parece existir una relación entre cáncer y la diabetes mellitus, aunque muchos aspectos quedan por dilucidar, como el papel desempeñado por los fármacos anticancerígenos y antihiperglucemiantes utilizados en ambas entidades(AU)


Background: metformin, a safe inexpensive drug, has proved to improve the prognosis of several types of cancer. Objective: to review the most relevant aspects of the relationship among diabetes mellitus, metformin and cancer. Development: diabetes mellitus, particularly type 2, is related to some kinds of cancer (breast, liver, pancreas, gynecological, gallbladder, colon and rectum), and its recurrence and mortality increase in men. The mechanism behind this relationship is not fully clarified. Insulin and insulin-like growth factor under hyperinsulinism and insulin resistance conditions may play a fundamental role in developing cancer as well as other common risk factors for diabetes mellitus and cancer (unhealthy feeding, sedentary lifestyle, addictions, age, sex, ethnic group and race). Liver kinase B1 protein has been identified as tumor suppressor protein which binds the metformin to impair the mitochondrial respiratory chain complex I and leads to reduction of adenosine triphosphate synthesis and to the increase of mytogen-activated protein-adenosine triphosphate quotient in the intracellular space. Chemotherapeutic, steroid and anti-androgen drugs may negatively affect the hydrocarbon metabolism. Some antihyperglycemic drugs have been related to specific cancers, although the evidence is still poor, indirect and controversial. Conclusions: metformin may be used to prevent and treat some types of cancer and to reduce recurrence and mortality. There seems to be some relationship between cancer and diabetes mellitus, even when many aspects remain to be ascertained such as the role played by anticancer and antihyperglycemic drugs intended to treat both diseases(AU)


Asunto(s)
Humanos , Masculino , Diabetes Mellitus/tratamiento farmacológico , Metformina/uso terapéutico , Neoplasias/tratamiento farmacológico , Receptores de Factores de Crecimiento/efectos de los fármacos , Anticarcinógenos/administración & dosificación , Metformina/efectos adversos , Neoplasias/prevención & control , Factores de Riesgo
4.
Lancet ; 384(9942): 545-55, 2014 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-24954676

RESUMEN

Several important advances have been made in our understanding of the pathways that lead to cell dysfunction and death in Parkinson's disease and Huntington's disease. These advances have been informed by both direct analysis of the post-mortem brain and by study of the biological consequences of the genetic causes of these diseases. Some of the pathways that have been implicated so far include mitochondrial dysfunction, oxidative stress, kinase pathways, calcium dysregulation, inflammation, protein handling, and prion-like processes. Intriguingly, these pathways seem to be important in the pathogenesis of both diseases and have led to the identification of molecular targets for candidate interventions designed to slow or reverse their course. We review some recent advances that underlie putative therapies for neuroprotection in Parkinson's disease and Huntington's disease, and potential targets that might be exploited in the future. Although we will need to overcome important hurdles, especially in terms of clinical trial design, we propose several target pathways that merit further study. In Parkinson's disease, these targets include agents that might improve mitochondrial function or increase degradation of defective mitochondria, kinase inhibitors, calcium channel blockers, and approaches that interfere with the misfolding, templating, and transmission of α-synuclein. In Huntington's disease, strategies might also be directed at mitochondrial bioenergetics and turnover, the prevention of protein dysregulation, disruption of the interaction between huntingtin and p53 or huntingtin-interacting protein 1 to reduce apoptosis, and interference with expression of mutant huntingtin at both the nucleic acid and protein levels.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Humanos , Enfermedad de Huntington/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Int J Immunopathol Pharmacol ; 26(3): 621-32, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24067459

RESUMEN

Osteosarcoma is the most common primary malignant tumour of the bone. Although new therapies continue to be reported, osteosarcoma-related morbidity and mortality remain high. Modern medicine has greatly increased knowledge of the physiopathology of this neoplasm. Novel targets for drug development may be identified through an understanding of the normal molecular processes that are deeply modified in pathological conditions. The aim of the present study is to investigate, by immunohistochemistry, the localisation of different growth factors and of the proliferative marker Ki-67 in order to determine whether these factors are involved in the transformation of osteogenic cells and in the development of human osteosarcoma. We observed a general positivity for NGF - TrKA - NT3 - TrKC - VEGF in the cytoplasm of neoplastic cells and a strong expression for NT4 in the nuclear compartment. TGF-beta was strongly expressed in the extracellular matrix and vascular endothelium. BDNF and TrKB showed a strong immunolabeling in the extracellular matrix. Ki-67/MIB-1 was moderately expressed in the nucleus of neoplastic cells. We believe that these growth factors may be considered potential therapeutic targets in the treatment of osteosarcoma, although proof of this hypothesis requires further investigation.


Asunto(s)
Neoplasias Óseas/metabolismo , Proliferación Celular , Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Osteosarcoma/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Antineoplásicos/uso terapéutico , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Terapia Molecular Dirigida , Osteosarcoma/irrigación sanguínea , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Receptores de Factores de Crecimiento/efectos de los fármacos , Transducción de Señal
6.
Artículo en Inglés | MEDLINE | ID: mdl-23714450

RESUMEN

Adjuvant therapy with antiestrogens targeting estrogen receptor α (ER) signaling prevents disease recurrence in many patients with early-stage ER+ breast cancer. However, a significant number of cases exhibit de novo or acquired endocrine resistance. While other clinical subtypes of breast cancer (HER2+, triple-negative) have disproportionately higher rates of mortality, ER+ breast cancer is responsible for at least as many deaths because it is the most common subtype. Therefore, identifying mechanisms that drive endocrine resistance is a high clinical priority. A large body of experimental evidence indicates that oncogenic signaling pathways underlie endocrine resistance, including growth factor receptor tyrosine kinases (HER2, epidermal growth factor receptor [EGFR], fibroblast growth factor receptor 1/2 [FGFR], insulin-like growth factor-1 receptor [IGF-1R]/ insulin receptor [InsR]), PI3K/AKT/ mTOR, MAPK/ERK, Src, CDK4/CDK6, and ER itself. Combined targeting of ER and such pathways may be the most effective means to combat antiestrogen resistance, and clinical trials testing such strategies show promising results. Herein, we discuss pathways associated with endocrine resistance, biomarkers that may be useful to predict response to targeted agents, and avenues for further exploration to identify strategies for the treatment of patients with endocrine-resistant disease.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Moduladores de los Receptores de Estrógeno/farmacología , Estrógenos , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Progesterona , Transducción de Señal/efectos de los fármacos , Antineoplásicos Hormonales/uso terapéutico , Inhibidores de la Aromatasa/farmacología , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Ciclo Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/fisiología , Resistencia a Antineoplásicos/fisiología , Moduladores de los Receptores de Estrógeno/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/fisiología , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/fisiopatología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor ErbB-2/fisiología , Receptores de Estrógenos/efectos de los fármacos , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/fisiología , Receptores de Progesterona/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico
7.
Am J Physiol Regul Integr Comp Physiol ; 304(11): R966-79, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23552497

RESUMEN

Reptile embryos tolerate large decreases in the concentration of ambient oxygen. However, we do not fully understand the mechanisms that underlie embryonic cardiovascular short- or long-term responses to hypoxia in most species. We therefore measured cardiac growth and function in snapping turtle embryos incubated under normoxic (N21; 21% O2) or chronic hypoxic conditions (H10; 10% O2). We determined heart rate (fH) and mean arterial pressure (Pm) in acute normoxic (21% O2) and acute hypoxic (10% O2) conditions, as well as embryonic responses to cholinergic, adrenergic, and ganglionic pharmacological blockade. Compared with N21 embryos, chronic H10 embryos had smaller bodies and relatively larger hearts and were hypotensive, tachycardic, and following autonomic neural blockade showed reduced intrinsic fH at 90% of incubation. Unlike other reptile embryos, cholinergic and ganglionic receptor blockade both increased fH. ß-Adrenergic receptor blockade with propranolol decreased fH, and α-adrenergic blockade with phentolamine decreased Pm. We also measured cardiac mRNA expression. Cholinergic tone was reduced in H10 embryos, but cholinergic receptor (Chrm2) mRNA levels were unchanged. However, expression of adrenergic receptor mRNA (Adrb1, Adra1a, Adra2c) and growth factor mRNA (Igf1, Igf2, Igf2r, Pdgfb) was lowered in H10 embryos. Hypoxia altered the balance between cholinergic receptors, α-adrenoreceptor and ß-adrenoreceptor function, which was reflected in altered intrinsic fH and adrenergic receptor mRNA levels. This is the first study to link gene expression with morphological and cardioregulatory plasticity in a developing reptile embryo.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Fenómenos Fisiológicos Cardiovasculares , Embrión no Mamífero/fisiología , Regulación de la Expresión Génica/fisiología , Hipoxia/fisiopatología , Tortugas/fisiología , Antagonistas Adrenérgicos alfa/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Membrana Corioalantoides/fisiología , Enfermedad Crónica , ADN Complementario/biosíntesis , ADN Complementario/genética , Desarrollo Embrionario/fisiología , Bloqueadores Ganglionares/farmacología , Técnicas In Vitro , Sistema Nervioso Parasimpático/fisiología , Parasimpatolíticos/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Receptores de Neurotransmisores/genética , Sistema Nervioso Simpático/fisiología
8.
Invest Ophthalmol Vis Sci ; 53(12): 7560-6, 2012 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23074208

RESUMEN

PURPOSE: Exogenous vasoactive intestinal peptide (VIP) down-regulates pro-inflammatory but up-regulates anti-inflammatory cytokines, growth factors (GFs) and Toll-like receptors promoting healing in experimental Pseudomonas aeruginosa (P. aeruginosa) keratitis. Whether VIP is required for GF or GF receptor (R) expression in normal and infected corneas is unknown and is the purpose of this study. METHODS: VIP knockout ((-/-)) and wild-type (WT) C57BL/6 (B6) mice were infected and tested using PCR array, real-time RT-PCR, ELISA, and immunostaining. VIP antagonist treatment studies also were done using B6 and BALB/c mice. RESULTS: Infected corneas of VIP(-/-) versus WT B6 mice perforated earlier (2 vs. 5 days postinfection [p.i.]), and array data showed that GFs were differentially changed between groups. RT-PCR revealed that the infected cornea of VIP(-/-) versus WT mice expressed higher mRNA levels of epidermal growth factor (EGF) and hepatocyte growth factor (HGF), reduced FGF, EGFR, and HGFR, with no difference in FGFR; differences between groups were not seen in normal cornea. Immunostaining for GF and GFR in the normal cornea of VIP(-/-) versus WT mice was similar. However, at 1 day p.i., VIP(-/-) versus WT mice had more intense EGF and HGF, similar FGFR, and reduced FGF, EGFR, and HGFR staining. VIP antagonist treatment decreased protein levels for GFR at 5 days p.i. in both B6 and BALB/c mice, with no significant changes in normal cornea. CONCLUSIONS: The data showed that endogenous VIP is not requisite for GF or GFR expression in the normal cornea but, after infection, its absence or reduction is critical for their regulation.


Asunto(s)
Córnea/metabolismo , Infecciones Bacterianas del Ojo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Queratitis/metabolismo , ARN Mensajero/genética , Receptores de Factores de Crecimiento/genética , Péptido Intestinal Vasoactivo/uso terapéutico , Animales , Recuento de Colonia Microbiana , Córnea/microbiología , Córnea/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Infecciones Bacterianas del Ojo/genética , Infecciones Bacterianas del Ojo/patología , Femenino , Queratitis/genética , Queratitis/patología , Ratones , Ratones Endogámicos BALB C , Fármacos Neuroprotectores/uso terapéutico , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/aislamiento & purificación , ARN Mensajero/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptores Toll-Like/biosíntesis , Receptores Toll-Like/genética
9.
J Natl Cancer Inst ; 103(8): 645-61, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21464397

RESUMEN

BACKGROUND: Ionizing radiation (IR) is effectively used in cancer therapy. However, in subsets of patients, a few radioresistant cancer cells survive and cause disease relapse with metastatic progression. The MET oncogene encodes the hepatocyte growth factor (HGF) receptor and is known to drive "invasive growth", a regenerative and prosurvival program unduly activated in metastasis. METHODS: Human tumor cell lines (MDA-MB-231, MDA-MB-435S, U251) were subjected to therapeutic doses of IR. MET mRNA, and protein expression and signal transduction were compared in treated and untreated cells, and the involvement of the DNA-damage sensor ataxia telangiectasia mutated (ATM) and the transcription factor nuclear factor kappa B (NF-κB) in activating MET transcription were analyzed by immunoblotting, chromatin immunoprecipitation, and use of NF-κB silencing RNA (siRNA). Cell invasiveness was measured in wound healing and transwell assays, and cell survival was measured in viability and clonogenic assays. MET was inhibited by siRNA or small-molecule kinase inhibitors (PHA665752 or JNJ-38877605). Combinations of MET-targeted therapy and radiotherapy were assessed in MDA-MB-231 and U251 xenografts (n = 5-6 mice per group). All P values were from two-sided tests. RESULTS: After irradiation, MET expression in cell lines was increased up to fivefold via activation of ATM and NF-κB. MET overexpression increased ligand-independent MET phosphorylation and signal transduction, and rendered cells more sensitive to HGF. Irradiated cells became more invasive via a MET-dependent mechanism that was further enhanced in the presence of HGF. MET silencing by siRNA or inhibition of its kinase activity by treatment with PHA665752 or JNJ-38877605 counteracted radiation-induced invasiveness, promoted apoptosis, and prevented cells from resuming proliferation after irradiation in vitro. Treatment with MET inhibitors enhanced the efficacy of IR to stop the growth of or to induce the regression of xenografts (eg, at day 13, U251 xenografts, mean volume increase relative to mean tumor volume at day 0: vehicle = 438%, 5 Gy IR = 151%, 5 Gy IR + JNJ-38877605 = 76%; difference, IR vs JNJ-38877604 + IR = 75%, 95% CI = 59% to 91%, P = .01). CONCLUSION: IR induces overexpression and activity of the MET oncogene through the ATM-NF-κB signaling pathway; MET, in turn, promotes cell invasion and protects cells from apoptosis, thus supporting radioresistance. Drugs targeting MET increase tumor cell radiosensitivity and prevent radiation-induced invasiveness.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN/efectos de la radiación , Proteínas de Unión al ADN/metabolismo , FN-kappa B/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/efectos de la radiación , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de la radiación , Proteínas de la Ataxia Telangiectasia Mutada , Northern Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Movimiento Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/efectos de la radiación , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Etiquetado Corte-Fin in Situ , Indoles/farmacología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/efectos de la radiación , Invasividad Neoplásica/prevención & control , Neoplasias/patología , Neoplasias/radioterapia , Fosforilación/efectos de la radiación , Reacción en Cadena de la Polimerasa , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/efectos de la radiación , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/efectos de la radiación , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Tolerancia a Radiación , Radiación Ionizante , Fármacos Sensibilizantes a Radiaciones/farmacología , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/efectos de la radiación , Sulfonas/farmacología , Transcripción Genética/efectos de la radiación , Trasplante Heterólogo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/efectos de la radiación , Regulación hacia Arriba/efectos de la radiación
10.
Cancer Treat Rev ; 36 Suppl 3: S80-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21129616

RESUMEN

Breast cancer is not considered anymore a unique disease. Microarray gene expression analysis led to the identification of 4 major breast cancer "intrinsic" subtypes, including hormone receptor (HR)-positive luminal A and B, human epidermal growth receptor 2 (HER2)-positive and basal-like breast cancer (BLBC). These subtypes have distinct phenotypes, molecular profiles, clinical behaviour and response to therapy, with the BLBC carrying the worst outcome. Microarray analysis is not feasible in routine practice and therefore oncologists rely on a simpler immunohistochemical (IHC) classification to identify relevant breast cancer subtypes. Triple negative breast cancer (TNBC) is defined by the absence of oestrogen receptor, progesterone receptor and HER2 expression at IHC analysis. TNBC is strictly related to BLBC and, given the lack of common therapeutic targets, represent a major challenge for breast oncologist. In this review we will summarize the updated knowledge on TNBC, with emphasis on its current treatment and on the new therapeutic options under development.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias de la Mama/clasificación , Femenino , Predicción , Humanos , Receptor ErbB-2/efectos de los fármacos , Receptores de Estrógenos/efectos de los fármacos , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/metabolismo , Receptores de Progesterona/efectos de los fármacos
11.
J Biol Chem ; 285(34): 26335-40, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20554517

RESUMEN

Met, the tyrosine kinase receptor for the hepatocyte growth factor is a prominent regulator of cancer cell invasiveness and has emerged as a promising therapeutic target. Binding of the anti-Met monoclonal antibody DN30 to its epitope induces the proteolytic cleavage of Met, thereby impairing the invasive growth of tumors. The molecular mechanism controlling this therapeutic shedding process has so far been unknown. Here, we report that A Disintegrin And Metalloproteinase (ADAM)-10, but not ADAM-17, is required for DN30-induced Met shedding. Knockdown of ADAM-10 in different tumor cell lines or abrogation of its proteolytic activity by natural or synthetic inhibitors abolished Met down-regulation on the cell surface as well as reduction of Met activation. Moreover, hepatocyte growth factor-induced tumor cell migration and invasion were impaired upon ADAM-10 knockdown. Thus, the therapeutic effect of DN30 involves ADAM-10-dependent Met shedding, linking for the first time a specific metalloprotease to target therapy against a receptor tyrosine kinase.


Asunto(s)
Proteínas ADAM/fisiología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Anticuerpos/uso terapéutico , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Proteína ADAM10 , Anticuerpos/farmacología , Línea Celular Tumoral , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/prevención & control , Proteínas Proto-Oncogénicas c-met/efectos de los fármacos , Receptores de Factores de Crecimiento/efectos de los fármacos
12.
Curr Pharm Des ; 16(20): 2184-93, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20459386

RESUMEN

Even though treatment of several types of solid tumors has improved in the past few years with the introduction of molecular targeted agents in the therapeutic armamentarium of the medical oncologist, response rates to these agents are generally modest. Increasing evidence is now revealing that genetic factors are affecting patients' response to these therapeutic agents as well as the frequency and intensity of toxic reactions. Importantly, pharmacogenetic analysis is now required for the administration of several molecular targeted agents in clinical practice. For the vast majority of these agents, however, data remain purely experimental. Herein, we provide an overview of the genetic changes (mutations and polymorphisms) that have been associated with response to treatment with anticancer molecular targeted agents. Special emphasis is given on molecules (monoclonal antibodies and tyrosine kinase inhibitors) that target critical mediators in the epidermal growth factor receptor (EGFR), the human epidermal growth factor receptor 2 (HER2/ERBB2/NEU) and the vascular endothelial growth factor receptor (VEGFR) pathways. The true clinical utility of these applications remains to be proven in future prospective, randomized clinical trials in large patient cohorts of all different ethnic backgrounds.


Asunto(s)
Terapia Molecular Dirigida/tendencias , Proteínas de Neoplasias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Farmacogenética/métodos , Medicina de Precisión/métodos , Receptores de Factores de Crecimiento/efectos de los fármacos , Quimioterapia/métodos , Receptores ErbB/efectos de los fármacos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Mutación/efectos de los fármacos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Polimorfismo Genético/efectos de los fármacos , Receptor ErbB-2/efectos de los fármacos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Vet Pathol ; 47(1): 77-97, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20080487

RESUMEN

Growth factors are low molecular peptides active in the stimulation of cell proliferation and in the regulation of embryonic development and cellular differentiation. Significant progress has been made in developing effective strategies to treat human malignancies with new chemical compounds based on a rationale directed against various components of signaling pathways. Many of these drugs target a growth factor receptor--for instance, in the form of monoclonal antibodies or inhibitors of tyrosine kinases, such as monoclonal antibodies against epidermal growth factor receptors used in treating certain types of breast cancer. Imatinib mesylate [Gleevec]) is an excellent example of mediators of signal transduction, such as tyrosine kinases. Growth factors proper are used to ameliorate various and sometimes fatal side effects of cytotoxic and/or myelosuppressive chemotherapy. Basic characteristics of several growth families are discussed with therapeutic modalities based on growth factor activity or, more often, inhibition of such activity.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Neoplasias/fisiopatología , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Factor de Crecimiento Epidérmico/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Neoplasias/tratamiento farmacológico , Factor de Crecimiento Derivado de Plaquetas/fisiología , Progranulinas , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Receptores de Factores de Crecimiento/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Somatomedinas/fisiología , Factor de Crecimiento Transformador beta/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
14.
Stem Cells ; 27(10): 2527-38, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19609936

RESUMEN

Skeletal muscle repair and regeneration requires the activity of satellite cells, a population of myogenic stem cells scattered throughout the tissue and activated to proliferate and differentiate in response to myotrauma or disease. While it seems likely that satellite cells would need to navigate local muscle tissue to reach damaged areas, relatively little data on such motility exist, and most studies have been with immortalized cell lines. We find that primary satellite cells are significantly more motile than myoblast cell lines, and that adhesion to laminin promotes primary cell motility more than fourfold over other substrates. Using timelapse videomicroscopy to assess satellite cell motility on single living myofibers, we have identified a requirement for the laminin-binding integrin alpha 7 beta 1 in satellite cell motility, as well as a role for hepatocyte growth factor in promoting directional persistence. The extensive migratory behavior of satellite cells resident on muscle fibers suggests caution when determining, based on fixed specimens, whether adjacent cells are daughters from the same mother cell. We also observed more persistent long-term contact between individual satellite cells than has been previously supposed, potential cell-cell attractive and repulsive interactions, and migration between host myofibers. Based on such activity, we assayed for expression of "pathfinding" cues, and found that satellite cells express multiple guidance ligands and receptors. Together, these data suggest that satellite cell migration in vivo may be more extensive than currently thought, and could be regulated by combinations of signals, including adhesive haptotaxis, soluble factors, and guidance cues.


Asunto(s)
Movimiento Celular/fisiología , Imagenología Tridimensional/métodos , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/citología , Animales , Antígenos CD/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Linaje de la Célula/fisiología , Células Cultivadas , Quimiotaxis/fisiología , Señales (Psicología) , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Cadenas alfa de Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Laminina/metabolismo , Laminina/farmacología , Ratones , Microscopía por Video/métodos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/metabolismo , Células Satélite del Músculo Esquelético/metabolismo
15.
Oral Oncol ; 45(9): 747-51, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19442565

RESUMEN

Overall survival for patients with squamous cell carcinoma of the head and neck (SCCHN) has not improved appreciably over the past few decades. Novel therapeutic approaches, such as immunotherapy, are under clinical investigation since the standard treatments are toxic and have not successfully controlled this disease with sufficiently high success rates. Cancer immunotherapy describes various techniques to expand and activate the immune system to control tumor growth in vivo, and clinical evaluation has so far demonstrated low toxicity. Immunotherapy appears to have the most applicability in settings of minimal residual disease and to reduce distant metastases after other therapeutic interventions, and its potential clinical value is now receiving intensive evaluation. Emerging forms of SCCHN immunotherapy involve both the use of monoclonal antibodies (mAb) that target growth factor receptors where immune activation appears to contribute to tumor cell lysis, as well as various forms of active vaccination strategies which activate and direct the patient's cellular immunity against the tumor. This article reviews immunotherapeutic strategies currently in clinical trials or under development for patients with SCCHN.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeza y Cuello/terapia , Inmunoterapia/métodos , Anticuerpos Monoclonales/uso terapéutico , Carcinoma de Células Escamosas/inmunología , Células Dendríticas/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Humanos , Factores Inmunológicos/uso terapéutico , Péptidos/uso terapéutico , Receptores de Factores de Crecimiento/efectos de los fármacos , Resultado del Tratamiento , Vacunas de ADN/uso terapéutico
16.
J Neurooncol ; 92(2): 137-47, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19043776

RESUMEN

In this study, we investigated the protein expression of platelet-derived growth factor receptor (PDGFR), insulin like growth factor-1 receptor (IGF-1R), phosphatidylinositol 3-kinase (PI3-K) and extracellular signal-regulated kinase (ERK1/2) in five primary glioblastoma (GB), with a view to their possible use as therapeutic targets. Our results demonstrated that appreciable levels of these proteins could be detected in the analysed GB cell lines, except for a low level of PDGFR and ERK1/2 expression in one GB cell line. The small molecule inhibitors towards IGF-1R, PDGFR, PI3-K and ERK1/2 respectively, have only modest or no anti-tumour activity on GB cells and therefore their combination with other therapy modalities was analysed. The interaction between small inhibitors and radiation was mostly additive or sub-additive; synergistic interaction was found in five of forty analysed combinations. Our results showed that GB cells are in general resistant to treatment and illustrate the difficulties in predicting the treatment response in malignant gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/fisiología , Antineoplásicos/farmacología , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Citometría de Flujo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/efectos de la radiación , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/efectos de la radiación , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/efectos de la radiación , Receptor IGF Tipo 1/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/efectos de la radiación , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/efectos de la radiación , Receptores del Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
17.
J Gastroenterol ; 43(12): 905-11, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19107333

RESUMEN

Current medical interventions for pancreatic cancer are insufficient. Recent molecular investigations have elucidated complex genetic mechanisms of cancer that especially involve multiple signal transduction pathways; this enables us to develop molecular medicines targeting specific genetic molecules in the pathways. These molecular medicines seem to promise clues for curing cancers, including pancreatic cancer. This review describes current knowledge and perspectives regarding the development of molecular medicines for pancreatic cancer by focusing on growth factor receptor systems and three major signal transduction pathways: the RAS-MAPK, PI3K-AKT-mTOR, and hedgehog pathways.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Ensayos Clínicos como Asunto , Humanos , Neoplasias Pancreáticas/genética , Receptores de Factores de Crecimiento/efectos de los fármacos , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/efectos de los fármacos
18.
World J Gastroenterol ; 14(46): 7021-32, 2008 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-19084910

RESUMEN

The limited efficacy of cytotoxic therapy for advanced biliary tract and gallbladder cancers emphasizes the need for novel and more effective medical treatment options. A better understanding of the specific biological features of these neoplasms led to the development of new targeted therapies, which take the abundant expression of several growth factors and cognate tyrosine kinase receptors into account. This review will briefly summarize the status and future perspectives of antiangiogenic and growth factor receptor-based pharmacological approaches for the treatment of biliary tract and gallbladder cancers. In view of multiple novel targeted approaches, the rationale for innovative therapies, such as combinations of growth factor (receptor)-targeting agents with cytotoxic drugs or with other novel anticancer drugs will be highlighted.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias del Sistema Biliar/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/tendencias , Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Citotoxinas/uso terapéutico , Humanos , Receptores de Factores de Crecimiento/efectos de los fármacos
19.
Neurosurg Focus ; 23(4): E10, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17961034

RESUMEN

In this article, the authors review the research supporting the use of calcium channel antagonists (CCAs) in the treatment of recurrent or unresectable meningiomas. Calcium channel antagonists (for example, diltiazem and verapamil) are known to augment the effects of chemotherapy drugs (for example, vincristine) in multiple cancers. Although it was initially thought that this occurred by interference with calcium-dependent secondary messenger systems, it appears that other mechanisms account for this effect. The authors' initial work in this field was based on the then-emerging data that meningiomas are receptor positive for growth factor receptors (for example, platelet-derived growth factor [PDGF]), which are known to trigger calcium-dependent secondary messenger pathways. In fact, they were able to show that CCAs block the growth stimulatory effects of multiple growth factors, including PDGF, in vitro and augment the growth inhibitory effects of hydroxyurea and RU486 (mifepristone). The authors have shown similar in vivo growth inhibition by these agents. In addition, diltiazem- and verapamil-treated meningiomas are less vascular and smaller, with decreased cell proliferation and increased apoptosis. The use of CCAs is attractive as an adjunct treatment for unresectable or recurrent meningiomas because they are safe drugs with well-known side effect profiles that lend themselves to long-term chronic therapy.


Asunto(s)
Bloqueadores de los Canales de Calcio/uso terapéutico , Neoplasias Meníngeas/tratamiento farmacológico , Meningioma/tratamiento farmacológico , Antineoplásicos/administración & dosificación , Bloqueadores de los Canales de Calcio/farmacología , Proliferación Celular/efectos de los fármacos , Quimioterapia Combinada , Humanos , Neoplasias Meníngeas/patología , Meningioma/patología , Receptores de Factores de Crecimiento/efectos de los fármacos
20.
Pharmacotherapy ; 27(8): 1125-44, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17655513

RESUMEN

Renal cell cancer (RCC) is a relatively uncommon malignancy, with 51,190 cases expected to be diagnosed in 2007. Localized disease is curable by surgery; however, locally advanced or metastatic disease is not curable in most cases and, until recently, had a limited response to drug treatment. Historically, biologic response modifiers or immunomodulating agents were tested in clinical trials based on observations that some cases of RCC can spontaneously regress. High-dose aldesleukin is approved by the United States Food and Drug Administration as a treatment for advanced RCC; however, the drug is associated with a high frequency of severe adverse effects. Responses have been observed with low-dose aldesleukin and interferon alfa, but with little effect on overall survival. Sorafenib and sunitinib are novel therapies that target growth factor receptors known to be activated by the hypoxia-inducible factor and the Ras-Raf/MEK/ERK pathways. These pathways are important in the pathophysiology of RCC. Sorafenib and sunitinib have shown antitumor activity as first- and second-line therapy in patients with cytokine-refractory metastatic RCC who have clear-cell histology. Although complete responses are not common, both drugs promote disease stabilization and increase progression-free survival. This information suggests that disease stabilization may be an important determinant for response in RCC and possibly other cancers. Sorafenib and sunitinib are generally well tolerated and are considered first- and second-line treatment options for patients with advanced clear cell RCC. In addition, sorafenib and sunitinib have shown promising results in initial clinical trials evaluating antitumor activity in patients who are refractory to other antiangiogenic therapy. The most common toxicities with both sorafenib and sunitinib are hand-foot syndrome, rash, fatigue, hypertension, and diarrhea. Research is directed toward defining the optimal use of these new agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencenosulfonatos/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Indoles/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Piridinas/uso terapéutico , Pirroles/uso terapéutico , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Bencenosulfonatos/efectos adversos , Bencenosulfonatos/farmacocinética , Carcinoma de Células Renales/fisiopatología , Ensayos Clínicos como Asunto , Sistemas de Liberación de Medicamentos , Interacciones Farmacológicas , Humanos , Indoles/efectos adversos , Indoles/farmacocinética , Neoplasias Renales/fisiopatología , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Piridinas/efectos adversos , Piridinas/farmacocinética , Pirroles/efectos adversos , Pirroles/farmacocinética , Receptores de Factores de Crecimiento/efectos de los fármacos , Sorafenib , Sunitinib
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...