Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
2.
Clin Cancer Res ; 22(20): 5087-5096, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27154914

RESUMEN

PURPOSE: c-Src has been shown to play a pivotal role in breast cancer progression, metastasis, and angiogenesis. In the clinic, however, the limited efficacy and high toxicity of existing c-Src inhibitors have tempered the enthusiasm for targeting c-Src. We developed a novel c-Src inhibitor (UM-164) that specifically binds the DFG-out inactive conformation of its target kinases. We hypothesized that binding the inactive kinase conformation would lead to improved pharmacologic outcomes by altering the noncatalytic functions of the targeted kinases. EXPERIMENTAL DESIGN: We have analyzed the anti-triple-negative breast cancer (TNBC) activity of UM-164 in a comprehensive manner that includes in vitro cell proliferation, migration, and invasion assays (including a novel patient-derived xenograft cell line, VARI-068), along with in vivo TNBC xenografts. RESULTS: We demonstrate that UM-164 binds the inactive kinase conformation of c-Src. Kinome-wide profiling of UM-164 identified that Src and p38 kinase families were potently inhibited by UM-164. We further demonstrate that dual c-Src/p38 inhibition is superior to mono-inhibition of c-Src or p38 alone. We demonstrate that UM-164 alters the cell localization of c-Src in TNBC cells. In xenograft models of TNBC, UM-164 resulted in a significant decrease of tumor growth compared with controls, with limited in vivo toxicity. CONCLUSIONS: In contrast with c-Src kinase inhibitors used in the clinic (1, 2), we demonstrate in vivo efficacy in xenograft models of TNBC. Our results suggest that the dual activity drug UM-164 is a promising lead compound for developing the first targeted therapeutic strategy against TNBC. Clin Cancer Res; 22(20); 5087-96. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Familia-src Quinasas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión/fisiología , Proteína Tirosina Quinasa CSK , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dasatinib/farmacología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/patología , Unión Proteica/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Integr Cancer Ther ; 8(3): 242-53, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19815594

RESUMEN

PURPOSE: Pomegranate fruit extracts (PFEs) possess polyphenolic and other compounds with antiproliferative, pro-apoptotic and anti-inflammatory effects in prostate, lung, and other cancers. Because nuclear transcription factor-kB (NF-kB) is known to regulate cell survival, proliferation, tumorigenesis, and inflammation, it was postulated that PFEs may exert anticancer effects at least in part by modulating NF-kB activity. EXPERIMENTAL DESIGN: The authors investigated the effect of a novel, defined PFE consisting of both fermented juice and seed oil on the NF-kB pathway, which is constitutively active in aggressive breast cancer cell lines. The effects of the PFE on NF-kB-regulated cellular processes such as cell survival, proliferation, and invasion were also examined. RESULTS: Analytical characterization of the bioactive components of the PFE revealed active constituents, mainly ellagitannins and phenolic acids in the aqueous PFE and conjugated octadecatrienoic acids in the lipid PFE derived from seeds.The aqueous PFE dose-dependently inhibited NF-kB-dependent reporter gene expression associated with proliferation, invasion, and motility in aggressive breast cancer phenotypes while decreasing RhoC and RhoA protein expression. CONCLUSION: Inhibition of motility and invasion by PFEs, coincident with suppressed RhoC and RhoA protein expression, suggests a role for these defined extracts in lowering the metastatic potential of aggressive breast cancer species.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Frutas/química , Lythraceae/química , Extractos Vegetales/farmacología , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dinoprostona/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Leucotrieno B4/metabolismo , FN-kappa B/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Invasividad Neoplásica/patología , Fitoterapia , Factor de Transcripción ReIA/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Proteína rhoC de Unión a GTP
4.
Cancer Res ; 66(19): 9379-84, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17018591

RESUMEN

Over 70% of patients with head and neck squamous cell carcinoma (HNSCC) present with locoregionally advanced stage III and IV disease. In spite of aggressive therapy, locoregional disease recurs in 60% and metastatic disease develops in 15% to 25% of patients causing a major decline in quality and length of life. Therefore, there is a need to identify and understand genes that are responsible for inducing an aggressive HNSCC phenotype. Evidence has shown that protein kinase C (PKC) epsilon is a transforming oncogene and may play a role in HNSCC progression. In this study, we determine the downstream signaling pathway mediated by PKC epsilon to promote an aggressive HNSCC phenotype. RNA interference knockdown of PKC epsilon in UMSCC11A and UMSCC36, two highly invasive and motile HNSCC cell lines with elevated endogenous PKC epsilon levels, resulted in cells that were significantly less invasive and motile than the small interfering RNA-scrambled control transfectants; 51 +/- 5% (P < 0.006) and 49 +/- 3% (P < 0.010) inhibition in invasion and 69 +/- 1% (P < 0.0005) and 66 +/- 3% (P < 0.0001) inhibition in motility, respectively. PKC epsilon-deficient UMSCC11A clones had reduced levels of active and serine-phosphorylated RhoA and RhoC. Moreover, constitutive active RhoA completely rescued the invasion and motility defect, whereas constitutive active RhoC completely rescued the invasion and partially rescued the motility defect of PKC epsilon-deficient UMSCC11A clones. These results indicate that RhoA and RhoC are downstream of PKC epsilon and critical for PKC epsilon-mediated cell invasion and motility. Our study shows, for the first time, that PKC epsilon is involved in a coordinated regulation of RhoA and RhoC activation, possibly through direct post-translational phosphorylation.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Neoplasias de Cabeza y Cuello/enzimología , Invasividad Neoplásica/fisiopatología , Proteínas de Neoplasias/fisiología , Proteína Quinasa C-epsilon/fisiología , Proteínas de Unión al GTP rho/fisiología , Proteína de Unión al GTP rhoA/fisiología , Carcinoma de Células Escamosas/patología , Movimiento Celular , Activación Enzimática , Neoplasias de Cabeza y Cuello/patología , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Fosforilación , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/farmacología , Transducción de Señal , Proteína rhoC de Unión a GTP
5.
Cancer Res ; 65(18): 8366-71, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16166314

RESUMEN

Tumor metastasis is the major cause of morbidity and mortality in patients with breast cancer. It is critical to identify metastasis enabling genes and understand how they are responsible for inducing specific aspects of the metastatic phenotype to allow for improved clinical detection and management. Protein kinase C epsilon (PKC epsilon), a member of a family of serine/threonine protein kinases, is a transforming oncogene that has been reported to be involved in cell invasion and motility. In this study, we investigated the role of PKC epsilon in breast cancer development and progression. High-density tissue microarray analysis showed that PKC epsilon protein was detected in 73.6% (106 of 144) of primary tumors from invasive ductal breast cancer patients. Increasing PKC epsilon staining intensity was associated with high histologic grade (P = 0.0206), positive Her2/neu receptor status (P = 0.0419), and negative estrogen (P = 0.0026) and progesterone receptor status (P = 0.0008). Kaplan-Meier analyses showed that PKC epsilon was significantly associated with poorer disease-free and overall survival (log-rank, P = 0.0478 and P = 0.0414, respectively). RNA interference of PKC epsilon in MDA-MB231 cells, an aggressive breast cancer cell line with elevated PKC epsilon levels, resulted in a cell phenotype that was significantly less proliferative, invasive, and motile than the parental or the control RNA interference transfectants. Moreover, in vivo tumor growth of small interfering RNA-PKC epsilon MDA-MB231 clones was retarded by a striking 87% (P < 0.05) and incidence of lung metastases was inhibited by 83% (P < 0.02). PKC epsilon-deficient clones were found to have lower RhoC GTPase protein levels and activation. Taken together, these results revealed that PKC epsilon plays a critical and causative role in promoting an aggressive metastatic breast cancer phenotype and as a target for anticancer therapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/enzimología , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo , Animales , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Carcinoma Ductal de Mama/enzimología , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patología , Carcinoma Ductal de Mama/terapia , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Humanos , Ratones , Ratones Desnudos , Valor Predictivo de las Pruebas , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras , Proteínas de Unión al GTP rho/metabolismo , Proteína rhoC de Unión a GTP
6.
Mol Cancer Res ; 1(10): 701-6, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12939395

RESUMEN

Tetrathiomolybdate (TM), a specific copper chelator, has been shown to be a potent antiangiogenic and antimetastatic compound possibly through suppression of the NFkappaB signaling cascade. To further delineate the molecular mechanism of the anticancer effect of TM, we investigated whether TM has antineoplastic activity in the setting of genetic NFkappaB inhibition. In this study, SUM149 inflammatory breast carcinoma cells were transfected with a dominant-negative IkappaBalpha (S32AS36A) expression vector. Similar to TM-treated SUM149 cells, SUM149-IkappaBalphaMut clones secreted lower amounts of proangiogenic mediators, vascular endothelial growth factor, interleukin-1alpha, and interleukin-8 and exhibited a less invasive and motile phenotype. The reduction in the angiogenic and metastatic potential of SUM149-IkappaBalphaMut clones was not further affected by TM in vitro. SUM149-IkappaBalphaMut xenografts grew substantially slower and had less lung metastasis than SUM149 and SUM149-empty vector xenografts. The growth and metastatic potential of SUM149 and SUM149-empty tumors was significantly inhibited with systemic TM treatment, whereas TM had no further antitumor effect on the SUM149-IkappaBalphaMut tumors. Additionally, nuclear proteins isolated from TM-treated SUM149 tumors had lower NFkappaB binding activity, while AP1 and SP1 binding activities were unchanged. Taken together, these results strongly support that suppression of NFkappaB is the major mechanism used by TM to inhibit angiogenesis and metastasis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Mama/patología , Molibdeno/farmacología , FN-kappa B/metabolismo , Metástasis de la Neoplasia/prevención & control , Neovascularización Patológica/prevención & control , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , División Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Inhibidor NF-kappaB alfa , FN-kappa B/genética , Transcripción Genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Ther ; 2(7): 617-22, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12883034

RESUMEN

Constitutive activation of nuclear factor kappaB is implicated to be a critical survival mechanism used by carcinoma cells to escape apoptosis. Tetrathiomolybdate (TM), a novel copper chelator, exhibits potent antiangiogenic properties, in part, through suppression of the nuclear factor kappaB signaling cascade. In this study, we determined whether TM enhances doxorubicin-induced apoptosis in SUM149 inflammatory breast carcinoma cells. Apoptosis was not observed in these cells after TM treatment. Moreover, SUM149 cells were relatively resistant to doxorubicin-induced apoptosis ranging from 9.9 +/- 2.9% to 21.5 +/- 2.0% apoptotic cells for 0.1 to 2.5 micro M doxorubicin treatment. A greater-than-additive increase (33.8 +/- 4.6%, 57.5 +/- 5.2%, or 83.7 +/- 1.0% apoptosis with TM and 0.1, 0.5, or 2.5 micro M doxorubicin, respectively) in apoptosis was observed in cells treated with the combination therapy of TM and doxorubicin. In SUM149 xenografts, TM and doxorubicin significantly retarded tumor growth in comparison with either agent administered alone (P < 0.03). Tumor cell apoptosis in the combination therapy-treated mice was 113.3 +/- 20% greater than that in TM-treated mice and 52.4 +/- 14.3% greater than that in doxorubicin-treated mice. These results suggest that TM may enhance the rate of pathological complete response when used in combination with an anthracycline in neoadjuvant therapy of breast carcinoma.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antibióticos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Doxorrubicina/uso terapéutico , Neoplasias Mamarias Experimentales/prevención & control , Molibdeno/uso terapéutico , Adenosina Trifosfatasas/antagonistas & inhibidores , Animales , Núcleo Celular/metabolismo , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Masculino , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Células Tumorales Cultivadas/trasplante
8.
Clin Exp Metastasis ; 19(4): 301-11, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12090470

RESUMEN

Inflammatory breast cancer (IBC) is the most lethal form of locally advanced breast cancer known. IBC carries a guarded prognosis primarily due to rapid onset of disease, typically within six months, and the propensity of tumor emboli to invade the dermal lymphatics and spread systemically. Although the clinical manifestations of IBC have been well documented, until recently little was known about the genetic mechanisms underlying the disease. In a comprehensive study aimed at identifying the molecular mechanisms responsible for the unique IBC phenotype, our laboratory identified overexpression of RhoC GTPase in over 90% of IBC tumors in contrast to 36% of stage-matched non-IBC tumors. We also demonstrated that overexpression of RhoC GTPase in human mammary epithelial (HME) cells nearly recapitulated the IBC phenotype with regards to invasion, motility and angiogenesis. In the current study we sought to delineate which signaling pathways were responsible for each aspect of the IBC phenotype. Using well-established inhibitors to the mitogen activated protein kinase (MAPK) and phosphatidylinositol-3 kinase (PI3K) pathways. We found that activation of the MAPK pathway was responsible for motility, invasion and production of angiogenic factors. In contrast, growth under anchorage independent conditions was dependent on the PI3K pathway.


Asunto(s)
Adenocarcinoma/patología , Toxinas Botulínicas , Neoplasias de la Mama/patología , GTP Fosfohidrolasas/fisiología , Sistema de Señalización de MAP Quinasas , Proteínas de Neoplasias/fisiología , Proteínas de Unión al GTP rho/fisiología , ADP Ribosa Transferasas/farmacología , Adenocarcinoma/enzimología , Neoplasias de la Mama/enzimología , Cromonas/farmacología , Factores de Crecimiento Endotelial/biosíntesis , Factores de Crecimiento Endotelial/genética , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Femenino , GTP Fosfohidrolasas/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación , Linfocinas/biosíntesis , Linfocinas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Morfolinas/farmacología , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/antagonistas & inhibidores , Neovascularización Patológica/enzimología , Fenotipo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transfección , Células Tumorales Cultivadas/enzimología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteína rhoC de Unión a GTP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA