Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Breast Cancer Res Treat ; 199(1): 13-23, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36913051

RESUMEN

PURPOSE: Dysregulation of the PI3K pathway is one of the most common events in breast cancer. Here we investigate the activity of the PI3K inhibitor MEN1611 at both molecular and phenotypic levels by dissecting and comparing its profile and efficacy in HER2 + breast cancer models with other PI3K inhibitors. METHODS: Models with different genetic backgrounds were used to investigate the pharmacological profile of MEN1611 against other PI3K inhibitors. In vitro studies evaluated cell viability, PI3K signaling, and cell death upon treatment with MEN1611. In vivo efficacy of the compound was investigated in cell line- and patient-derived xenografts models. RESULTS: Consistent with its biochemical selectivity, MEN1611 demonstrated lower cytotoxic activity in a p110δ-driven cellular model when compared to taselisib, and higher cytotoxic activity in the p110ß-driven cellular model when compared to alpelisib. Moreover, MEN1611 selectively decreased the p110α protein levels in PIK3CA mutated breast cancer cells in a concentration- and proteasome-dependent manner. In vivo, MEN1611 monotherapy showed significant and durable antitumor activity in several trastuzumab-resistant PIK3CA-mutant HER2 + PDX models. The combination of trastuzumab and MEN1611 significantly improved the efficacy compared to single agent treatment. CONCLUSIONS: The profile of MEN1611 and its antitumoral activity suggest an improved profile as compared to pan-inhibitors, which are limited by a less than ideal safety profile, and isoform selective molecules, which may potentially promote development of resistance mechanisms. The compelling antitumor activity in combination with trastuzumab in HER2 + trastuzumab-resistant, PIK3CA mutated breast cancer models is at the basis of the ongoing B-Precise clinical trial (NCT03767335).


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Trastuzumab/farmacología , Trastuzumab/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor ErbB-2/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Fosfatidilinositol 3-Quinasa Clase I/genética
2.
Bioorg Med Chem ; 23(17): 5725-33, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26233797

RESUMEN

The canonical Wnt signaling pathway plays a fundamental role in embryonic as well as in adult development. Consequently, dysregulation of the pathway has been linked to a wide spectrum of pathological conditions. In a program aimed at the identification of small molecule inhibitors of the canonical Wnt pathway we identified a series of 2-aminopyrimidine derivatives which specifically inhibited the pathway with minimal or no sign of cellular toxicity. The hit molecules 1 and 2 showed promising inhibitory activity with IC50 values of approximately 10 µM, but low solubility and metabolic stability. During the early stage of the hit series exploration, the pyrimidine core was variously decorated to obtain active compounds with a better physico-chemical profile. In particular, compound 13 showed Wnt inhibition activity comparable to hit molecules 1 and 2, with improved physico-chemical properties. Therefore, this series of compounds may be considered a promising starting point for the design of novel small molecule inhibitors of the canonical Wnt pathway.


Asunto(s)
Pirimidinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo , Humanos , Estructura Molecular , Pirimidinas/metabolismo , Relación Estructura-Actividad , Vía de Señalización Wnt/genética
3.
Front Oncol ; 13: 1283951, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38033496

RESUMEN

Background: Lung cancer remains the leading cause of cancer-related death worldwide. Targeted therapies with tyrosine kinase inhibitors (TKIs) result in improvement in survival for non-small cell lung cancer (NSCLC) with activating mutations of the epidermal growth factor receptor (EGFR). Unfortunately, most patients who initially respond to EGFR-TKI ultimately develop resistance to therapy, resulting in cancer progression and relapse. Combination therapy is today a common strategy for the treatment of tumors to increase the success rate, improve the outcome and survival of patients, and avoid the selection of resistant cancer cells through the activation of compensatory pathways. In NSCLC, the phosphoinositide-3-kinase/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway has been heavily implicated in both tumorigenesis and the progression of disease. Objectives: In this study, we investigated the efficacy of a PI3K δ-sparing inhibitor, MEN1611, in models of NSCLC sensitive and resistant to EGFR inhibitors (erlotinib and gefitinib) with a wild-type PIK3CA gene. Methods: We performed functional, biochemical, and immunohistochemistry studies. Results: We demonstrated good efficacy of MEN1611 in NSCLC devoid of PIK3CA gene mutations but with constitutive activation of the PI3K/AKT pathway and its synergistic effect with gefitinib both in vitro and in vivo. Conclusions: Overall, this preclinical study indicates that the inhibitor could be a candidate for the treatment of NSCLC with an erlotinib/gefitinib-resistant phenotype and constitutive activation of the PI3K/AKT pathway, a phenotype mimicked by our model system.

4.
J Immunother ; 42(4): 97-109, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30865026

RESUMEN

Adoptive transfer of T lymphocytes (ACT) engineered with T-cell receptors (TCRs) of known antitumor specificity is an effective therapeutic strategy. However, a major constraint of ACT is the unpredictable interference of the endogenous TCR α and ß chains in pairing of the transduced TCR. This effect reduces the efficacy of the genetically modified primary T cells and carries the risk of generating novel TCR reactivities with unintended functional consequences. Here, we show a powerful approach to overcome these limitations. We engineered TCR α and ß chains with mutations encompassing a conserved motif (FXXXFXXS) required to stabilize the pairing of immunoglobulin heavy chain transmembrane domains. Molecular modeling supported the preferential pairing of mutated TCR and impaired pairing between mutated and wild-type TCRs. Expression of the mutated TCR was similar to wild type and conferred the expected specificity. Fluorescence resonance energy transfer analysis in mouse splenocytes transduced with mutated or wild-type TCRs showed a higher proximity of the former over the latter. Importantly, we show that mutated TCRs effectively outcompete endogenous TCRs and improve in vitro antitumor cytotoxicity when expressed in ex vivo isolated human T cells. This approach should contribute to improving current protocols of anticancer immunetherapy protocols.


Asunto(s)
Dominios Proteicos/genética , Dominios y Motivos de Interacción de Proteínas , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transducción Genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Expresión Génica , Terapia Genética , Vectores Genéticos , Humanos , Inmunoterapia Adoptiva , Membrana Dobles de Lípidos/química , Ratones , Modelos Moleculares , Mutagénesis , Conformación Proteica , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores Quiméricos de Antígenos/química , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Relación Estructura-Actividad , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología
5.
Mol Cancer Ther ; 18(9): 1533-1543, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31227646

RESUMEN

CD205 is a type I transmembrane glycoprotein and is a member of the C-type lectin receptor family. Analysis by mass spectrometry revealed that CD205 was robustly expressed and highly prevalent in a variety of solid malignancies from different histotypes. IHC confirmed the increased expression of CD205 in pancreatic, bladder, and triple-negative breast cancer (TNBC) compared with that in the corresponding normal tissues. Using immunofluorescence microscopy, rapid internalization of the CD205 antigen was observed. These results supported the development of MEN1309/OBT076, a fully humanized CD205-targeting mAb conjugated to DM4, a potent maytansinoid derivate, via a cleavable N-succinimidyl-4-(2-pyridyldithio) butanoate linker. MEN1309/OBT076 was characterized in vitro for target binding affinity, mechanism of action, and cytotoxic activity against a panel of cancer cell lines. MEN1309/OBT076 displayed selective and potent cytotoxic effects against tumor cells exhibiting strong and low to moderate CD205 expression. In vivo, MEN1309/OBT076 showed potent antitumor activity resulting in durable responses and complete tumor regressions in many TNBC, pancreatic, and bladder cancer cell line-derived and patient-derived xenograft models, independent of antigen expression levels. Finally, the pharmacokinetics and pharmacodynamic profile of MEN1309/OBT076 was characterized in pancreatic tumor-bearing mice, demonstrating that the serum level of antibody-drug conjugate (ADC) achieved through dosing was consistent with the kinetics of its antitumor activity. Overall, our data demonstrate that MEN1309/OBT076 is a novel and selective ADC with potent activity against CD205-positive tumors. These data supported the clinical development of MEN1309/OBT076, and further evaluation of this ADC is currently ongoing in the first-in-human SHUTTLE clinical trial.


Asunto(s)
Inmunoconjugados/farmacología , Lectinas Tipo C/antagonistas & inhibidores , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Receptores de Superficie Celular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Antígenos CD/metabolismo , Células CHO , Línea Celular Tumoral , Cricetulus , Femenino , Células HEK293 , Células HT29 , Humanos , Inmunoconjugados/química , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Células MCF-7 , Maitansina/química , Maitansina/farmacología , Ratones , Ratones Desnudos , Ratones SCID , Antígenos de Histocompatibilidad Menor/inmunología , Antígenos de Histocompatibilidad Menor/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo
6.
Recenti Prog Med ; 110(2): 68-74, 2019 Feb.
Artículo en Italiano | MEDLINE | ID: mdl-30843531

RESUMEN

Smart Specialization Strategy (S3) of Lazio defines smart specialization strategies to bring out the excellence of the territory with prospects of success on the global market. Chemical-pharmaceutical, biomedical and biotechnological field is one of the 7 sectors considered of greatest interest for the S3. Key engine of biotechnology development are biological materials and associated data, stored in biobanks. However, to ensure that the research and product development carried out with that resources gives statistically significant and reproducible results, it is essential that they are collected, manipulated and stored using standardized and traced methods. Implementation of the recent published standard ISO 20387- "Biotechnology-Biobanking-General requirements for biobanking" is bridging biobanks toward to storage and distribution of qualified biological material only. Human biobanks are also an essential part of the assistance and care of the citizen and constitute an unavoidable cost of the regional health system. However, biobanks organization, rationalization of their territorial distribution, completion of the process of recognition and regional accreditation, parallel to the implementation of the offer of remunerated services for biobanking, can turn the cost of the necessary preservation of the samples, into an opportunity of territorial development. The paper describes the necessity, shared by a working group represented by several Lazio biobanks, of including biobank activities in the virtuous circle designed by the S3,concretizing the framework prefigured by the S3 document on infrastructures for research, innovation and technology transfer. To allow inclusion of biobank activities in the virtuous circle, we underline the need to quickly start the process of recognition of the territorial research biobanks, to implement at regional level the process of optimization and rationalization of the management of biological samples, in accordance with the international harmonization standards and with the territorial indications of sustainability.


Asunto(s)
Bancos de Muestras Biológicas/organización & administración , Investigación Biomédica/organización & administración , Biotecnología/organización & administración , Bancos de Muestras Biológicas/normas , Investigación Biomédica/normas , Biotecnología/normas , Humanos , Italia , Manejo de Especímenes/normas
7.
Proteomics ; 8(11): 2165-7, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18452228

RESUMEN

DigesTip is a new device for in-solution protein digestion, based on a patent pending technology, able to immobilize enzymes (trypsin, in this case) on a solid surface, keeping their activity preserved. DigesTip is a standard pipette tip, usable both by human and by robots. Its main performances are: very short digestion time (1 min) and usability with low protein sample concentrations (5 microg/mL). DigesTip obtains a clear signal in MS measurements and its usage rules out several preparative steps.


Asunto(s)
Espectrometría de Masas/métodos , Proteómica/métodos , Biotecnología/métodos , Diseño de Equipo , Humanos , Concentración de Iones de Hidrógeno , Industrias , Cinética , Espectrometría de Masas/instrumentación , Péptidos/química , Proteínas/química , Albúmina Sérica/química , Temperatura , Factores de Tiempo , Tripsina/química
8.
Mol Cell Biol ; 25(4): 1379-88, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15684389

RESUMEN

Metastatic disease is a significant contributor to cancer patient mortality. We previously reported that the Kinase Suppressor of Ras1 (KSR1) scaffold protein for the Erk mitogen-activated protein kinase pathway coimmunoprecipitated the metastasis suppressor protein Nm23-H1. We now hypothesize that altered expression levels of Nm23-H1 influence the binding properties, stability, and function of the KSR1 scaffold. Increased coimmunoprecipitation of Hsp90 with KSR1 was observed in either stable or transient transfectants of nm23-H1 in MDA-MB-435 human breast carcinoma cells. Similar trends were also observed in the cytoplasmic and nuclear fractions of cells. Cells expressing high levels of Nm23-H1 exhibited increased KSR1 degradation in the presence of either cycloheximide or an Hsp90-directed drug currently in clinical trial, 17-allylamino-17-demethoxygeldanamycin (17-AAG). In agreement with KSR1 degradation data, high-Nm23-H1-expression cells were preferentially inhibited in anchorage-independent colonization assays by 17-AAG. KSR1 scaffold binding patterns are dynamic in both the cytoplasmic and nuclear compartments, modulated by metastasis suppressor expression. Metastasis suppressor expression levels can impact traditional signaling pathways, such as the Erk pathway, resulting in altered tumor cell sensitivity to cancer therapeutics.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Nucleósido-Difosfato Quinasa/metabolismo , Proteínas Quinasas/metabolismo , Rifabutina/análogos & derivados , Transducción de Señal/fisiología , Benzoquinonas , Neoplasias de la Mama/metabolismo , Fraccionamiento Celular , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Cicloheximida/farmacología , Citoplasma/metabolismo , Femenino , Genes Supresores de Tumor , Humanos , Lactamas Macrocíclicas , Nucleósido Difosfato Quinasas NM23 , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Desnaturalización Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Rifabutina/farmacología , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
9.
Eur J Med Chem ; 95: 526-45, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25847770

RESUMEN

Wnt signaling pathway plays a critical role in numerous cellular processes, including tumor initiation, proliferation, invasion/infiltration, metastasis formation and resistance to chemotherapy. In a drug discovery project aimed at the identification of inhibitors of the canonical Wnt pathway, we selected a series of quinazoline 2,4-diones as starting point for the therapeutic treatment of glioblastoma multiforme. Despite of poor physico-chemical properties of hit compound 1, our medicinal chemistry effort allowed the discovery and characterization of lead compound 33 (SEN461), with improved ADME profile, good bioavailability and active in vitro and in vivo in glioblastoma, gastric and sarcoma tumors.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Quinazolinas/química , Quinazolinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Concentración 50 Inhibidora , Masculino , Ratones , Quinazolinas/metabolismo , Quinazolinas/farmacocinética , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Clin Exp Metastasis ; 20(1): 3-10, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12650601

RESUMEN

The first metastasis suppressor gene identified was nm23. Transfection of nm23 into metastatic cell lines resulted in the inhibition of metastasis, but not primary tumor size in vivo. Using in vitro assays, nm23 overexpression resulted in reduced anchorage-independent colonization in response to TGF-beta, reduced invasion and motility in response to multiple factors, and increased differentiation. We hypothesize that the mechanism of action of Nm23 in metastasis suppression involves diminished signal transduction downstream of a particular receptor. Candidate biochemical mechanisms are identified and discussed herein.


Asunto(s)
Genes Supresores de Tumor , Proteínas de Unión al GTP Monoméricas/genética , Metástasis de la Neoplasia/genética , Nucleósido-Difosfato Quinasa , Transducción de Señal/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular , Histidina Quinasa , Humanos , Modelos Biológicos , Nucleósido Difosfato Quinasas NM23 , Proteínas Quinasas/metabolismo , Relación Estructura-Actividad , Transfección
11.
Cancer Lett ; 190(1): 1-12, 2003 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-12536071

RESUMEN

Intensive investigation of protein tyrosine, serine and threonine phosphorylation has lead to advances in signal transduction research and cancer treatment. This feature summarizes research on mammalian proteins exhibiting histidine phosphorylation. Histidine kinases are well known in prokaryotic and lower eukaryotic systems where they form the 'two-component' signal transduction system. The relative invisibility of histidine phosphorylation in mammalian cells may result from technical obstacles such as its acid lability, which precludes detection in electrophoretic systems, amino acid sequencing, etc. Emerging data have identified mammalian histidine kinases for the kinase suppressor of ras, a scaffold molecule for the Map kinase pathway, as well as histone H4, aldolase C and the beta-subunit of heterotrimeric G proteins. Additional mammalian proteins of interest to signal transduction and cancer research exhibit histidine phosphorylation, including P-selectin, annexin I and the 20S proteasome. Other candidate histidine phosphorylated proteins are identified. These data suggest the existence of another series of phosphorylation patterns in signal transduction.


Asunto(s)
Histidina/metabolismo , Neoplasias/metabolismo , Nucleósido-Difosfato Quinasa , Proteínas Quinasas/fisiología , Transducción de Señal , Animales , Anexina A1/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Histidina Quinasa , Humanos , Modelos Biológicos , Modelos Químicos , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multienzimáticos/metabolismo , Nucleósido Difosfato Quinasas NM23 , Metástasis de la Neoplasia , Selectina-P/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal , Factores de Transcripción/metabolismo
12.
Clin Breast Cancer ; 4(1): 51-62, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12744759

RESUMEN

Metastatic disease remains a significant contributor to morbidity and mortality in patients with breast cancer. An improved molecular and biochemical understanding of the metastatic process is expected to fuel the development of new therapeutic approaches. The suppression of tumor metastasis, despite tumor cell expression of oncogenes and metastasis-promoting events, has become a diverse and fruitful field of investigation. Although many genetic events promote metastasis, several genes show relatively reduced expression levels in metastatic tumor cells in mouse model systems and in aggressive human tumors. Re-expression of a metastasis-suppressor gene in a metastatic tumor cell line results in a significant reduction in metastatic behavior in vivo with no effect on tumorigenicity. The known metastasis-suppressor gene products nm23, KAI1, mitogen-activated protein kinase kinase 4, breast cancer metastasis suppressor-1, KiSS1, RHOGDI2, CRSP3, and vitamin D3-upregulated protein/thioredoxin interacting protein exhibit unexpected biochemical functions that have shed new light on signaling events that are important in metastasis. Most metastasis suppressors function at the translationally important stage of outgrowth of micrometastatic tumor cells at a distant site. We hypothesize that elevation of metastasis suppressor gene expression in micrometastatic tumor cells in the adjuvant high-risk population of patients with breast cancer will halt metastatic colonization and have a clinical benefit. DNA methylation inhibitors have shown limited promise in increasing metastasis-suppressor gene expression, and ligands of the nuclear hormone receptor family are currently under investigation in vitro and in vivo. Clinical testing of agents that increase metastasis-suppressor gene expression is expected to require tailored trial designs.


Asunto(s)
Neoplasias de la Mama/genética , Genes Supresores de Tumor , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , División Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia/genética
13.
PLoS One ; 9(5): e97847, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24842792

RESUMEN

Sarcomas are mesenchymal tumors showing high molecular heterogeneity, reflected at the histological level by the existence of more than fifty different subtypes. Genetic and epigenetic evidences link aberrant activation of the Wnt signaling to growth and progression of human sarcomas. This phenomenon, mainly accomplished by autocrine loop activity, is sustained by gene amplification, over-expression of Wnt ligands and co-receptors or epigenetic silencing of endogenous Wnt antagonists. We previously showed that pharmacological inhibition of Wnt signaling mediated by Axin stabilization produced in vitro and in vivo antitumor activity in glioblastoma tumors. Here, we report that targeting different sarcoma cell lines with the Wnt inhibitor/Axin stabilizer SEN461 produces a less transformed phenotype, as supported by modulation of anchorage-independent growth in vitro. At the molecular level, SEN461 treatment enhanced the stability of the scaffold protein Axin1, a key negative regulator of the Wnt signaling with tumor suppressor function, resulting in downstream effects coherent with inhibition of canonical Wnt signaling. Genetic phenocopy of small molecule Axin stabilization, through Axin1 over-expression, coherently resulted in strong impairment of soft-agar growth. Importantly, sarcoma growth inhibition through pharmacological Axin stabilization was also observed in a xenograft model in vivo in female CD-1 nude mice. Our findings suggest the usefulness of Wnt inhibitors with Axin stabilization activity as a potentialyl clinical relevant strategy for certain types of sarcomas.


Asunto(s)
Antineoplásicos/farmacología , Proteína Axina/metabolismo , Sarcoma/fisiopatología , Vía de Señalización Wnt/fisiología , Animales , Línea Celular Tumoral , Cartilla de ADN/genética , Femenino , Técnica del Anticuerpo Fluorescente , Vectores Genéticos/genética , Humanos , Immunoblotting , Lentivirus , Ratones , Ratones Desnudos , Microscopía Confocal , Plásmidos/genética , Estabilidad Proteica/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Sarcoma/tratamiento farmacológico
14.
Mol Cancer Ther ; 12(7): 1180-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23619303

RESUMEN

Glioblastoma multiforme (GBM) is the most common and prognostically unfavorable form of brain tumor. The aggressive and highly invasive phenotype of these tumors makes them among the most anatomically damaging human cancers with a median survival of less than 1 year. Although canonical Wnt pathway activation in cancers has been historically linked to the presence of mutations involving key components of the pathway (APC, ß-catenin, or Axin proteins), an increasing number of studies suggest that elevated Wnt signaling in GBM is initiated by several alternative mechanisms that are involved in different steps of the disease. Therefore, inhibition of Wnt signaling may represent a therapeutically relevant approach for GBM treatment. After the selection of a GBM cell model responsive to Wnt inhibition, we set out to develop a screening approach for the identification of compounds capable of modulating canonical Wnt signaling and associated proliferative responses in GBM cells. Here, we show that the small molecule SEN461 inhibits the canonical Wnt signaling pathway in GBM cells, with relevant effects at both molecular and phenotypic levels in vitro and in vivo. These include SEN461-induced Axin stabilization, increased ß-catenin phosphorylation/degradation, and inhibition of anchorage-independent growth of human GBM cell lines and patient-derived primary tumor cells in vitro. Moreover, in vivo administration of SEN461 antagonized Wnt signaling in Xenopus embryos and reduced tumor growth in a GBM xenograft model. These data represent the first demonstration that small-molecule-mediated inhibition of Wnt signaling may be a potential approach for GBM therapeutics.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glioblastoma/patología , Células HEK293 , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Ratones , Ratones Desnudos , Pronóstico , Transducción de Señal , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto , Xenopus
15.
ChemMedChem ; 4(6): 923-33, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19434656

RESUMEN

Confidence in mechanism: Creating a more holistic understanding of disease pathophysiology and an early confidence in the mechanism under investigation could help facilitate the selection of not only the most appropriate targets but also the best mechanisms for disease intervention and how to select and optimise the best compounds. Drug target and candidate selection are two of the key decision points within the drug discovery process for which all companies use certain selection criteria to make decisions on which targets to accept into their discovery pipelines and which compounds will pass into development. These steps not only help define the overall productivity of every company but they are also decisions taken without full predictive knowledge of the risks that lie ahead or how best to manage them. In particular, the process of selecting new targets does not normally involve full evaluation of the risk(s) in the mechanism under investigation (the modulation of the target), which may result in an inability to fully connect in vitro and animal model results to the disease (clinical) setting. The resulting poor progression statistics of many compounds in the clinic is at least partially the result of a lack of understanding of disease pathophysiology. Notably, the lack of efficacy is still a major reason for failure in the clinic.1 Creating a more holistic understanding of disease pathophysiology and an early confidence in the mechanism under investigation could help facilitate the selection of not only the most appropriate targets but also the best mechanisms for disease intervention and how to select and optimise the best compounds.


Asunto(s)
Química Farmacéutica , Descubrimiento de Drogas , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Química Farmacéutica/métodos , Química Farmacéutica/tendencias , Modelos Animales , Modelos Químicos , Proyectos de Investigación , Medición de Riesgo
16.
J Natl Cancer Inst ; 97(9): 632-42, 2005 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-15870434

RESUMEN

BACKGROUND: Reestablishment of metastasis suppressor gene expression may constitute a therapeutic strategy for high-risk breast cancer patients. We previously showed that medroxyprogesterone acetate (MPA), a progestin that has been tested as treatment for advanced breast cancer, elevates expression of the Nm23-H1 metastasis suppressor gene in hormone receptor-negative metastatic human breast carcinoma cell lines in vitro via a glucocorticoid receptor-based mechanism. Here, we tested whether MPA treatment inhibits metastatic colonization of a hormone receptor-negative breast cancer cell line in vivo. METHODS: We tested the soft-agar colony-forming efficiency of untransfected MDA-MB-231T human breast carcinoma cells and MDA-MB-231T cells transfected with antisense Nm23-H1 in the presence and absence of MPA. Pharmacokinetic studies were used to establish dose and injection schedules that led to MPA serum levels in mice similar to those achievable in humans. For in vivo studies, nude mice were injected intravenously with MDA-MB-231T cells. After 4 weeks, mice were randomized to control or MPA arms. Endpoints included incidence, number, and size of gross pulmonary metastases; Nm23-H1 protein expression in gross metastases; and side effects. All statistical tests were two-sided. RESULTS: MPA reduced colony formation of MDA-MB-231T cells by 40%-50% but had no effect on colony formation of Nm23-H1 antisense transfectants. Metastases developed in 100% (95% confidence interval [CI] = 78% to 100% and 77% to 100%, respectively) of control mice injected with MDA-MB-231T cells. In two independent experiments, only 73% (95% CI = 45% to 92%) and 64% (95% CI = 35% to 87%) of mice injected with 2 mg of MPA developed metastases. Mice injected with 2 mg of MPA showed reductions in the mean numbers, per mouse, of all metastases and of large (>3 mm) metastases (P = .04 and .013, respectively). Nm23-H1 was expressed at high levels in 43% of pulmonary metastases in MPA-treated mice but only 13% of metastases in untreated mice. Mice receiving at least 1-mg doses of MPA gained more weight than control-treated mice but exhibited no bone density alterations or abnormal mammary fat pad histology. CONCLUSION: Our preclinical results show that MPA appears to elevate Nm23-H1 metastasis suppressor gene expression, thereby reducing metastatic colonization. The data suggest a new use for an old agent in a molecularly defined subset of breast cancer patients.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor/efectos de los fármacos , Acetato de Medroxiprogesterona/farmacología , Nucleósido-Difosfato Quinasa/efectos de los fármacos , Nucleósido-Difosfato Quinasa/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/sangre , Línea Celular Tumoral , ADN sin Sentido , Femenino , Humanos , Inmunohistoquímica , Ratones , Nucleósido Difosfato Quinasas NM23 , Nucleósido-Difosfato Quinasa/sangre , Transfección
17.
J Bioenerg Biomembr ; 35(1): 73-9, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12848344

RESUMEN

Cancer metastasis is a significant contributor to breast cancer patient morbidity and mortality. To develop new anti-metastatic therapies, we need to understand the biological and biochemical mechanisms of metastasis. Toward these efforts, we and others have studied metastasis suppressor genes, which halt metastasis in vivo without affecting primary tumor growth. The first metastasis suppressor gene confirmed was nm23, also known as NDP kinase. Using in vitro assays, nm23 overexpression resulted in reduced anchorage-independent colonization in response to TGF-beta, reduced invasion and motility in response to multiple factors, and increased differentiation. We hypothesize that the mechanism of action of Nm23 in metastasis suppression involves diminished signal transduction, downstream of a particular receptor. We hypothesize that a histidine protein kinase activity of Nm23 underlies its suppression of metastasis, and identify candidate substrates. This review also discusses therapeutic options on the basis of reexpression of metastasis suppressors.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Metástasis de la Neoplasia/genética , Nucleósido-Difosfato Quinasa , Biosíntesis de Proteínas/genética , Proteínas/metabolismo , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Carcinoma in Situ/genética , Carcinoma Ductal de Mama/enzimología , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/secundario , Carcinoma Ductal de Mama/terapia , Femenino , Humanos , Metástasis Linfática , Nucleósido Difosfato Quinasas NM23 , Metástasis de la Neoplasia/terapia
18.
J Biol Chem ; 277(35): 32389-99, 2002 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-12105213

RESUMEN

The metastasis-suppressive activity of Nm23-H1 was previously correlated with its in vitro histidine protein kinase activity, but physiological substrates have not been identified. We hypothesized that proteins that interact with histidine kinases throughout evolution may represent partners for Nm23-H1 and focused on the interaction of Arabidopsis "two-component" histidine kinase ERS with CTR1. A mammalian homolog of CTR1 was previously reported to be c-Raf; we now report that CTR1 also exhibits homology to the kinase suppressor of Ras (KSR), a scaffold protein for the mitogen-activated protein kinase (MAPK) cascade. Nm23-H1 co-immunoprecipitated KSR from lysates of transiently transfected 293T cells and at endogenous protein expression levels in MDA-MB-435 breast carcinoma cells. Autophosphorylated recombinant Nm23-H1 phosphorylated KSR in vitro. Phosphoamino acid analysis identified serine as the major target, and two peaks of Nm23-H1 phosphorylation were identified upon high performance liquid chromatography analysis of KSR tryptic peptides. Using site-directed mutagenesis, we found that Nm23-H1 phosphorylated KSR serine 392, a 14-3-3-binding site, as well as serine 434 when serine 392 was mutated. Phosphorylated MAPK but not total MAPK levels were reduced in an nm23-H1 transfectant of MDA-MB-435 cells. The data identify a complex in vitro histidine-to-serine protein kinase pathway, which may contribute to signal transduction and metastasis.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Nucleósido-Difosfato Quinasa , Proteínas Quinasas/metabolismo , Factores de Transcripción/metabolismo , Proteínas ras/metabolismo , Alanina , Sustitución de Aminoácidos , Antígenos de Neoplasias/metabolismo , Secuencia de Bases , Neoplasias de la Mama , Línea Celular , Cartilla de ADN , Femenino , Histidina Quinasa , Humanos , Cinética , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Mutagénesis Sitio-Dirigida , Nucleósido Difosfato Quinasas NM23 , Metástasis de la Neoplasia/prevención & control , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serina , Especificidad por Sustrato , Factores de Transcripción/química , Factores de Transcripción/genética , Transfección , Células Tumorales Cultivadas , Proteínas ras/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA