Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Biol Chem ; 294(20): 8311-8322, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30804215

RESUMEN

Screening of cellular activity for inhibitors of histone lysine modifiers is most frequently performed indirectly by analyzing changes in the total levels of histone marks targeted by lysine methylases/demethylases. However, inhibition of histone lysine modifiers often leads to local rather than total changes in histone marks. Also, because histone modifications can be modulated by more than one cellular enzyme, it is not always clear whether changes in histone marks are a direct or indirect consequence of the inhibitor treatment applied. Direct assessment of target occupation can provide a useful tool to quantify the fraction of an epigenetic modifier that is bound to a pharmacological inhibitor (target engagement). Here, we developed and used a novel chemoprobe-based immunoassay to quantify target engagement in cells. Quantification of the fraction of free KDM1A was made possible, in an immune-based assay, by coupling a biotinylated chemoprobe to a warhead capable of selectively and irreversibly binding to the free active form of KDM1A. The results obtained confirmed that this approach is able to determine the degree of target engagement in a dose-dependent manner. Furthermore, the assay can be also used on tissue extracts to analyze the in vivo pharmacokinetics and pharmacodynamics relationship of KDM1A inhibitors, as has been exemplified with ORY-1001 (iadademstat), a potent and irreversible inhibitor of KDM1A. The principle of this assay may be applied to other targets, and the KDM1A probe may be employed in chemoproteomic analyses.


Asunto(s)
Inhibidores Enzimáticos , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Humanos , Células THP-1
2.
Pharmaceutics ; 14(7)2022 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-35890315

RESUMEN

Lysine specific demethylase 1 (LSD1; also known as KDM1A), is an epigenetic modulator that modifies the histone methylation status. KDM1A forms a part of protein complexes that regulate the expression of genes involved in the onset and progression of diseases such as cancer, central nervous system (CNS) disorders, viral infections, and others. Vafidemstat (ORY-2001) is a clinical stage inhibitor of KDM1A in development for the treatment of neurodegenerative and psychiatric diseases. However, the role of ORY-2001 targeting KDM1A in neuroinflammation remains to be explored. Here, we investigated the effect of ORY-2001 on immune-mediated and virus-induced encephalomyelitis, two experimental models of multiple sclerosis and neuronal damage. Oral administration of ORY-2001 ameliorated clinical signs, reduced lymphocyte egress and infiltration of immune cells into the spinal cord, and prevented demyelination. Interestingly, ORY-2001 was more effective and/or faster acting than a sphingosine 1-phosphate receptor antagonist in the effector phase of the disease and reduced the inflammatory gene expression signature characteristic ofEAE in the CNS of mice more potently. In addition, ORY-2001 induced gene expression changes concordant with a potential neuroprotective function in the brain and spinal cord and reduced neuronal glutamate excitotoxicity-derived damage in explants. These results pointed to ORY-2001 as a promising CNS epigenetic drug able to target neuroinflammatory and neurodegenerative diseases and provided preclinical support for the subsequent design of early-stage clinical trials.

3.
Clin Transl Sci ; 13(2): 419-429, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31729169

RESUMEN

Reliably predicting in vivo efficacy from in vitro data would facilitate drug development by reducing animal usage and guiding drug dosing in human clinical trials. However, such prediction remains challenging. Here, we built a quantitative pharmacokinetic/pharmacodynamic (PK/PD) mathematical model capable of predicting in vivo efficacy in animal xenograft models of tumor growth while trained almost exclusively on in vitro cell culture data sets. We studied a chemical inhibitor of LSD1 (ORY-1001), a lysine-specific histone demethylase enzyme with epigenetic function, and drug-induced regulation of target engagement, biomarker levels, and tumor cell growth across multiple doses administered in a pulsed and continuous fashion. A PK model of unbound plasma drug concentration was linked to the in vitro PD model, which enabled the prediction of in vivo tumor growth dynamics across a range of drug doses and regimens. Remarkably, only a change in a single parameter-the one controlling intrinsic cell/tumor growth in the absence of drug-was needed to scale the PD model from the in vitro to in vivo setting. These findings create a framework for using in vitro data to predict in vivo drug efficacy with clear benefits to reducing animal usage while enabling the collection of dense time course and dose response data in a highly controlled in vitro environment.


Asunto(s)
Antineoplásicos/farmacología , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Conjuntos de Datos como Asunto , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/metabolismo , Humanos , Ratones , Neoplasias/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Cell ; 33(3): 495-511.e12, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29502954

RESUMEN

The lysine-specific demethylase KDM1A is a key regulator of stem cell potential in acute myeloid leukemia (AML). ORY-1001 is a highly potent and selective KDM1A inhibitor that induces H3K4me2 accumulation on KDM1A target genes, blast differentiation, and reduction of leukemic stem cell capacity in AML. ORY-1001 exhibits potent synergy with standard-of-care drugs and selective epigenetic inhibitors, reduces growth of an AML xenograft model, and extends survival in a mouse PDX (patient-derived xenograft) model of T cell acute leukemia. Surrogate pharmacodynamic biomarkers developed based on expression changes in leukemia cell lines were translated to samples from patients treated with ORY-1001. ORY-1001 is a selective KDM1A inhibitor in clinical trials and is currently being evaluated in patients with leukemia and solid tumors.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Histona Demetilasas/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Modelos Animales de Enfermedad , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/genética , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Células Madre/efectos de los fármacos , Células Madre/metabolismo
6.
Nat Commun ; 8: 14797, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28300085

RESUMEN

The docking protein p140Cap negatively regulates tumour cell features. Its relevance on breast cancer patient survival, as well as its ability to counteract relevant cancer signalling pathways, are not fully understood. Here we report that in patients with ERBB2-amplified breast cancer, a p140Cap-positive status associates with a significantly lower probability of developing a distant event, and a clear difference in survival. p140Cap dampens ERBB2-positive tumour cell progression, impairing tumour onset and growth in the NeuT mouse model, and counteracting epithelial mesenchymal transition, resulting in decreased metastasis formation. One major mechanism is the ability of p140Cap to interfere with ERBB2-dependent activation of Rac GTPase-controlled circuitries. Our findings point to a specific role of p140Cap in curbing the aggressiveness of ERBB2-amplified breast cancers and suggest that, due to its ability to impinge on specific molecular pathways, p140Cap may represent a predictive biomarker of response to targeted anti-ERBB2 therapies.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Neoplasias de la Mama/metabolismo , Receptor ErbB-2/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Endogámicos BALB C , Ratones Transgénicos , Metástasis de la Neoplasia , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Receptor ErbB-2/genética , Proteínas de Unión al GTP rac/genética
7.
Radiother Oncol ; 119(2): 300-5, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27247056

RESUMEN

AIMS: We have previously shown in a phase I trial that nelfinavir (NFV) is safe with chemoradiation in PDAC with good signs for efficacy. Reverse translationally, we aimed to test the influence of PSCs on nelfinavir mediated radiosensitization to PDAC preclinically, because PDAC is very rich in desmoplasia and PSCs are known to mediate radioresistance. METHODS: In a direct co-culture model of several PDAC cell lines with PSC we performed clonogenic assays +/- nelfinavir. This was repeated exposing cells to hypoxic conditions. In xenograft PDAC tumors we tested radiation +/- nelfinavir +/- PSC. RESULTS: NFV sensitized both, PDAC only and PDAC cocultured with PSC (PDAC: Panc-1, MiaPaCa-2, PSN-1). In Panc-1 and PSN-1 this effect was larger +PSC compared to -PSC. Human pancreatic stellate cells (hPSC) were also sensitized by NFV which reduced p-FAK levels in hPSC, an effect that we previously found to sensitize specifically PDAC/PSC coculture. Contrarily, LY294002 reduced p-Akt in PSC (hPSC and LTC-14) but had no impact on PSC radiation survival. In vitro, nelfinavir sensitized Panc-1 and PSN-1 under normoxic and hypoxic conditions. In PSN-1 xenografts, +PSC led to faster tumor regrowth after radiation vs -PSC. The regrowth delay effect of nelfinavir after radiation was dramatically larger +PSC vs -PSC (time to reach 250mm(3) 183% vs 22%). CONCLUSION: NFV mediated radiosensitization in PDAC with stroma is partly mediated by p-FAK inhibition (Chen et al., 2013). In vitro, NFV sensitizes both normoxic and hypoxic PDAC +/- PSC to a roughly similar extent. The dramatic increased effect of xenograft regrowth inhibition by nelfinavir in tumors with PSC is attributed to vascular normalization (Brunner et al., 2014) rather than direct modification of hypoxia as shown by the tumor regrowth after gemcitabine with NFV.


Asunto(s)
Inhibidores de la Proteasa del VIH/farmacología , Nelfinavir/farmacología , Neoplasias Pancreáticas/radioterapia , Células Estrelladas Pancreáticas/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/patología
8.
Oncoimmunology ; 4(9): e1027473, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26405599

RESUMEN

Pancreatic stellate cells (PSCs) are key components of pancreatic ductal adenocarcinoma (PDAC). We recently demonstrated that IP-10/CXCL10 is highly expressed by PSCs in the presence of pancreatic cancer cells (PCCs) and its expression correlates with infiltration by regulatory T cells (Tregs) and poor survival. Thus, stromal cells in pancreatic cancer can promote immunosuppression and tumor progression, through the expression of IP-10.

9.
Epigenomics ; 7(4): 609-26, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26111032

RESUMEN

Histone methylation and demethylation are important processes associated with the regulation of gene transcription, and alterations in histone methylation status have been linked to a large number of human diseases. Initially thought to be an irreversible process, histone methylation is now known to be reversed by two families of proteins containing over 30 members that act to remove methyl groups from specific lysine residues present in the tails of histone H3 and histone H4. A rapidly growing number of reports have implicated the FAD-dependent lysine specific demethylase (KDM1) family in cancer, and several small-molecule inhibitors are in development for the treatment of cancer. An additional role has emerged for KDM1 in brain function, offering additional opportunities for the development of novel therapeutic strategies in neurodegenerative disease. A decade after the identification of KDM1A as a histone demethylase, the first selective inhibitors have now reached the clinic.


Asunto(s)
Epigénesis Genética , Histona Demetilasas/genética , Neoplasias/genética , Enfermedades Neurodegenerativas/genética , Secuencia de Aminoácidos , Animales , Terapia Genética , Histona Demetilasas/química , Histona Demetilasas/metabolismo , Humanos , Datos de Secuencia Molecular , Neoplasias/terapia , Enfermedades Neurodegenerativas/terapia
10.
Cancer Lett ; 343(2): 147-55, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24141189

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterised by an abundant stromal response also known as a desmoplastic reaction. Pancreatic Stellate Cells have been identified as playing a key role in pancreatic cancer desmoplasia. There is accumulating evidence that the stroma contributes to tumour progression and to the low therapeutic response of PDAC patients. In this review we described the main actors of the desmoplastic reaction within PDAC and novel therapeutic approaches that are being tested to block the detrimental function of the stroma.


Asunto(s)
Sistemas de Liberación de Medicamentos , Neoplasias Pancreáticas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Compartimento Celular/efectos de los fármacos , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Células del Estroma/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
11.
Radiother Oncol ; 111(2): 243-51, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24780634

RESUMEN

BACKGROUND AND PURPOSE: Progression of pancreatic ductal adenocarcinoma (PDAC) is promoted by desmoplasia induced by pancreatic stellate cells (PSC). Contributory to this progression is epithelial mesenchymal transition (EMT), which shares many characteristics with the cancer stem cell (CSC) hypothesis. We investigated the role of these processes on the radioresponse and tumorigenicity of pancreatic cancer cells. MATERIALS AND METHODS: We used an in vitro sphere model and in vivo xenograft model to examine the role of PSC in EMT and CSC processes. RESULTS: We demonstrated that PSC enhanced the CSC phenotype and radioresistance of pancreatic cancer cells. Furthermore, the expression of several EMT and CSC markers supported enhanced processes in our models and that translated into remarkable in vivo tumorigenicity. Multi-dose TGFß neutralizing antibody inhibited the EMT and CSC processes, sensitized cells to radiation and reduced in vivo tumorigenicity. A proteomic screen identified multiple novel factors that were regulated by PSC in pancreatic cells. CONCLUSION: These results are critical in highlighting the role of PSC in tumor progression and radioresistance by manipulating the EMT and CSC processes. TGFß and the novel factors identified are important targets for better therapeutic outcome in response to PSC mediated mechanisms.


Asunto(s)
Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Transición Epitelial-Mesenquimal/fisiología , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/fisiología , Tolerancia a Radiación/fisiología , Adenocarcinoma/radioterapia , Anticuerpos Neutralizantes/farmacología , Biomarcadores de Tumor/análisis , Carcinoma Ductal Pancreático/radioterapia , Supervivencia Celular/fisiología , Supervivencia Celular/efectos de la radiación , Fibroblastos/patología , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Neoplasias Pancreáticas/radioterapia , Fenotipo , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas
12.
Oncotarget ; 5(22): 11064-80, 2014 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-25415223

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by an abundant desmoplastic reaction driven by pancreatic stellate cells (PSCs) that contributes to tumor progression. Here we sought to characterize the interactions between pancreatic cancer cells (PCCs) and PSCs that affect the inflammatory and immune response in pancreatic tumors. Conditioned media from mono- and cocultures of PSCs and PCCs were assayed for expression of cytokines and growth factors. IP-10/CXCL10 was the most highly induced chemokine in coculture of PSCs and PCCs. Its expression was induced in the PSCs by PCCs. IP-10 was elevated in human PDAC specimens, and positively correlated with high stroma content. Furthermore, gene expression of IP-10 and its receptor CXCR3 were significantly associated with the intratumoral presence of regulatory T cells (Tregs). In an independent cohort of 48 patients with resectable pancreatic ductal adenocarcinoma, high IP-10 expression levels correlated with decreased median overall survival. Finally, IP-10 stimulated the ex vivo recruitment of CXCR3+ effector T cells as well as CXCR3+ Tregs derived from patients with PDAC. Our findings suggest that, in pancreatic cancer, CXCR3+ Tregs can be recruited by IP-10 expressed by PSCs in the tumor stroma, leading to immunosuppressive and tumor-promoting effects.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Quimiocina CXCL10/biosíntesis , Neoplasias Pancreáticas/inmunología , Linfocitos T Reguladores/inmunología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Células HEK293 , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/inmunología , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Células del Estroma/patología , Análisis de Supervivencia , Linfocitos T Reguladores/patología
13.
Cancer Res ; 71(10): 3453-8, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21558392

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a strong desmoplastic reaction where the stromal compartment often accounts for more than half of the tumor volume. Pancreatic stellate cells (PSC) are a central mediator of desmoplasia. There is increasing evidence that desmoplasia is contributing to the poor therapeutic response of PDAC. We show that PSCs promote radioprotection and stimulate proliferation in pancreatic cancer cells (PCC) in direct coculture. Our in vivo studies show PSC-dependent radioprotection in response to a single dose and to fractionated radiation. Abrogating ß1-integrin signaling abolishes the PSC-mediated radioprotection in PCCs. Furthermore, this effect is independent of PI3K (phosphoinositide 3-kinase) but dependent on FAK. Taken together, we show for the first time that PSCs promote radioprotection of PCCs in a ß1-integrin-dependent manner.


Asunto(s)
Integrina beta1/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Células Estrelladas Pancreáticas/citología , Animales , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Femenino , Humanos , Ratones , Ratones Desnudos , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
14.
Cancer Biol Ther ; 11(12): 1028-35, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21558789

RESUMEN

The concept of cancer stem cells is generally accepted in different malignancies. We have previously shown that the MDA-MB231 breast cancer cells were more radiation resistant when sorted for the two stem cell markers CD24 and ESA. In this study, we examined a possible mechanism that might underlie this phenotype by looking at cell cycle profile and the effect this has on DNA repair pathways. The cell cycle profile showed that there were more CD24(-) ESA(+) sorted MDA-MB231 cells in the S- and G(2)-phases compared with the unsorted cells, 60 and 38% respectively. Cyclin D and E protein levels supported the cell cycle profile and highlighted the possible involvement of homologous recombination (HR) repair in the radioresistant phenotype. To further support this, CD24(-) ESA(+) sorted MDA-MB231 cells demonstrated statistically significant more RAD51 and less γ-H2AX foci 2 h post 4Gy ionising radiation, compared with the unsorted population. Inhibition of the HR pathway effectively sterilised the CD24(- ) ESA(+) sorted MDA-MB231 cells but had no effect on the unsorted cells or MDA468 control breast cancer cell line. Although the changes we saw were specific to MDA-MB231, these results merit further investigation and can be crucial in identifying a mechanism responsible for cancer stem cells treatment resistance in primary tumors.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Células Madre Neoplásicas/patología , Recombinación Genética/genética , Benzofuranos/farmacología , Ciclo Celular , Línea Celular Tumoral , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Células Madre Neoplásicas/citología , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Recombinación Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA