Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38657228

RESUMEN

Disruption of DNA damage repair via impaired homologous recombination is characteristic of Ewing sarcoma (EWS) cells. We hypothesize that this disruption results in increased reliance on non-homologous end joining (NHEJ) to repair DNA damage. In this study, we investigated if pharmacological inhibition of the enzyme responsible for NHEJ, the DNA-PK holoenzyme, alters the response of EWS cells to genotoxic standard of care chemotherapy. We used analyses of cell viability and proliferation to investigate the effects of clinical DNA-PK inhibitors (DNA-PKi) in combination with six therapeutic or experimental agents for EWS. We performed calculations of synergy using the Loewe Additivity Model. Immunoblotting evaluated treatment effects on DNA-PK, DNA damage, and apoptosis. Flow cytometric analyses evaluated effects on cell cycle and fate. We used orthotopic xenograft models to interrogate tolerability, drug mechanism, and efficacy in vivo. DNA-PKi demonstrated on-target activity, reducing phosphorylated DNA-PK levels in EWS cells. DNA-PKi sensitized EWS cell lines to agents that function as topoisomerase 2 (TOP2) poisons and enhanced the DNA damage induced by TOP2 poisons. Nanomolar concentrations of single agent TOP2 poisons induced G2M arrest and little apoptotic response, while adding DNA-PKi mediated apoptosis. In vivo, the combination of AZD-7648 and etoposide had limited tolerability but resulted in enhanced DNA damage, apoptosis, and EWS tumor shrinkage. The combination of DNA-PKi with standard of care TOP2 poisons in EWS models is synergistic, enhances DNA damage and cell death, and may form the basis of a promising future therapeutic strategy for EWS.

2.
Clin Cancer Res ; 29(21): 4479-4491, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37616468

RESUMEN

PURPOSE: Deregulated metabolism in cancer cells represents a vulnerability that may be therapeutically exploited to benefit patients. One such target is nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the NAD+ salvage pathway. NAMPT is necessary for efficient NAD+ production and may be exploited in cells with increased metabolic demands. We have identified NAMPT as a dependency in rhabdomyosarcoma (RMS), a malignancy for which novel therapies are critically needed. Here we describe the effect of NAMPT inhibition on RMS proliferation and metabolism in vitro and in vivo. EXPERIMENTAL DESIGN: Assays of proliferation and cell death were used to determine the effects of pharmacologic NAMPT inhibition in a panel of ten molecularly diverse RMS cell lines. Mechanism of the clinical NAMPTi OT-82 was determined using measures of NAD+ and downstream NAD+-dependent functions, including energy metabolism. We used orthotopic xenograft models to examine tolerability, efficacy, and drug mechanism in vivo. RESULTS: Across all ten RMS cell lines, OT-82 depleted NAD+ and inhibited cell growth at concentrations ≤1 nmol/L. Significant impairment of glycolysis was a universal finding, with some cell lines also exhibiting diminished oxidative phosphorylation. Most cell lines experienced profound depletion of ATP with subsequent irreversible necrotic cell death. Importantly, loss of NAD and glycolytic activity were confirmed in orthotopic in vivo models, which exhibited complete tumor regressions with OT-82 treatment delivered on the clinical schedule. CONCLUSIONS: RMS is highly vulnerable to NAMPT inhibition. These findings underscore the need for further clinical study of this class of agents for this malignancy.


Asunto(s)
NAD , Rabdomiosarcoma , Humanos , NAD/metabolismo , Citocinas/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Pirazoles , Necrosis , Rabdomiosarcoma/tratamiento farmacológico , Línea Celular Tumoral
3.
Oncogenesis ; 9(9): 80, 2020 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-32908120

RESUMEN

NAMPT mediates the rate-limiting step of the NAD salvage pathway, which maintains cellular bioenergetics and provides a necessary substrate for functions essential to rapidly proliferating cancer cells. In this study, we evaluated the efficacy and mechanisms of action of OT-82, a novel, high-potency NAMPT inhibitor with a favorable toxicity profile, in preclinical models of Ewing sarcoma (EWS), an aggressive pediatric malignancy with previously reported selective sensitivity to NAMPT inhibition. We show that OT-82 decreased NAD concentration and impaired proliferation of EWS cells in a dose-dependent manner, with IC50 values in the single-digit nanomolar range. Notably, genetic depletion of NAMPT phenocopied pharmacological inhibition. On-target activity of OT-82 was confirmed with the addition of NMN, the product of NAMPT, which rescued NAD concentration and EWS cellular viability. Mechanistically, OT-82 treatment resulted in impaired DNA damage repair through loss of PARP activity, G2 cell-cycle arrest, and apoptosis in EWS cells. Additional consequences of OT-82 treatment included reduction of glycolytic and mitochondrial activity. In vivo, OT-82 impaired tumor growth and prolonged survival in mice bearing EWS xenografts. Importantly, antitumor effect correlated with pharmacodynamic markers of target engagement. Furthermore, combining low-dose OT-82 with low doses of agents augmenting DNA damage demonstrated enhanced antitumor activity in vitro and in vivo. Thus, OT-82 treatment represents a potential novel targeted approach for the clinical treatment of EWS.

4.
Cancer Res ; 80(14): 3046-3056, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32354737

RESUMEN

Rhabdomyosarcoma is the most common childhood soft-tissue sarcoma, yet patients with metastatic or recurrent disease continue to do poorly, indicating a need for new treatments. The SRC family tyrosine kinase YES1 is upregulated in rhabdomyosarcoma and is necessary for growth, but clinical trials using single agent dasatinib, a SRC family kinase inhibitor, have failed in sarcomas. YAP1 (YES-associated protein) is highly expressed in rhabdomyosarcoma, driving growth and survival when the upstream Hippo tumor suppressor pathway is silenced, but efforts to pharmacologically inhibit YAP1 have been unsuccessful. Here we demonstrate that treatment of rhabdomyosarcoma with DNA methyltransferase inhibitor (DNMTi) upregulates Hippo activators RASSF1 and RASSF5 by promoter demethylation, activating canonical Hippo signaling and increasing inactivation of YAP1 by phosphorylation. Treatment with DNMTi decreased rhabdomyosarcoma cell growth and increased apoptosis and differentiation, an effect partially rescued by expression of constitutively active YAP (S127A), suggesting the effects of DNMTi treatment are, in part, due to Hippo-dependent inhibition of YAP1. In addition, YES1 and YAP1 interacted in the nucleus of rhabdomyosarcoma cells, and genetic or pharmacologic suppression of YES1 resulted in cytoplasmic retention of YAP1 and decreased YAP1 target gene expression, suggesting YES1 regulates YAP1 in a Hippo-independent manner. Combined treatment with DNMTi and dasatinib targeted both Hippo-dependent and Hippo-independent regulation of YAP1, ablating rhabdomyosarcoma cell growth in vitro and trending toward decreased tumor growth in vivo. These results show that the mechanisms regulating YAP1 in rhabdomyosarcoma can be inhibited by combinatorial therapy of DNMTi and dasatinib, laying the groundwork for future clinical investigations. SIGNIFICANCE: This study elucidates the signaling pathways that regulate the oncogenic protein YAP1 and identifies a combination therapy to target these pathways in the childhood tumor rhabdomyosarcoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Azacitidina/análogos & derivados , Terapia Molecular Dirigida , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Rabdomiosarcoma/tratamiento farmacológico , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis , Azacitidina/farmacología , Proliferación Celular , Niño , Femenino , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Humanos , Ratones , Ratones SCID , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP
5.
Cancer Res ; 79(19): 5060-5073, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31431459

RESUMEN

Altered cellular metabolism, including an increased dependence on aerobic glycolysis, is a hallmark of cancer. Despite the fact that this observation was first made nearly a century ago, effective therapeutic targeting of glycolysis in cancer has remained elusive. One potentially promising approach involves targeting the glycolytic enzyme lactate dehydrogenase (LDH), which is overexpressed and plays a critical role in several cancers. Here, we used a novel class of LDH inhibitors to demonstrate, for the first time, that Ewing sarcoma cells are exquisitely sensitive to inhibition of LDH. EWS-FLI1, the oncogenic driver of Ewing sarcoma, regulated LDH A (LDHA) expression. Genetic depletion of LDHA inhibited proliferation of Ewing sarcoma cells and induced apoptosis, phenocopying pharmacologic inhibition of LDH. LDH inhibitors affected Ewing sarcoma cell viability both in vitro and in vivo by reducing glycolysis. Intravenous administration of LDH inhibitors resulted in the greatest intratumoral drug accumulation, inducing tumor cell death and reducing tumor growth. The major dose-limiting toxicity observed was hemolysis, indicating that a narrow therapeutic window exists for these compounds. Taken together, these data suggest that targeting glycolysis through inhibition of LDH should be further investigated as a potential therapeutic approach for cancers such as Ewing sarcoma that exhibit oncogene-dependent expression of LDH and increased glycolysis. SIGNIFICANCE: LDHA is a pharmacologically tractable EWS-FLI1 transcriptional target that regulates the glycolytic dependence of Ewing sarcoma.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Glucólisis/efectos de los fármacos , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Sarcoma de Ewing/patología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Ratones SCID , Sarcoma de Ewing/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Transl Oncol ; 9(6): 540-547, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27835791

RESUMEN

To determine what alternative pathways may act as mechanisms of bypass resistance to type 1 insulin-like growth factor receptor (IGF-1R) blockade in rhabdomyosarcoma (RMS), we compared expression of receptor tyrosine kinase activity in a number of IGF-1R antibody-resistant and -sensitive RMS cell lines. We found that platelet-derived growth factor receptor ß (PDGFR-ß) activity was upregulated in three xenograft-derived IGF-1R antibody-resistant cell lines that arose from a highly sensitive fusion-positive RMS cell line (Rh41). Furthermore, we identified four additional fusion-negative RMS cell lines that similarly upregulated PDGFR-ß activity when selected for IGF-1R antibody resistance in vitro. In the seven cell lines described, we observed enhanced growth inhibition when cells were treated with dual IGF-1R and PDGFR-ß inhibition in vitro. In vivo studies have confirmed the enhanced effect of targeting IGF-1R and PDGFR-ß in several mouse xenograft models of fusion-negative RMS. These findings suggest that PDGFR-ß acts as a bypass resistance pathway to IGF-1R inhibition in a subset of RMS. Therapy co-targeting these receptors may be a promising new strategy in RMS care.

7.
Neoplasia ; 17(4): 358-66, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25925378

RESUMEN

The insulin-like growth factor 1 receptor (IGF-1R) has surfaced as a significant target in multiple solid cancers due to its fundamental roles in pro-survival and anti-apoptotic signaling. However, development of resistance to IGF-1R blockade represents a significant hindrance and limits treatment efficacy in the clinic. In this study, we identified acquired resistance to IGF-1R blockade with R1507, an antibody against IGF-1R, and with BMS-754807, a small molecular inhibitor of IGF-1R/insulin receptor (IR). We showed that treatment with an IGF-IR antibody, R1507, or an IR/IGF-IR kinase inhibitor, BMS-754807, was associated with increased activation of YES/SRC family tyrosine kinase (SFK) in rhabdomyosarcoma (RMS). Combining anti-IGF-1R agents with SFK inhibitors resulted in blockade of IGF-1R inhibition-induced activation of YES/SFK and displayed advantageous antitumor activity in vitro and in vivo. Our data provide evidence that IGF-1R blockade results in activation of the YES/SRC family kinase bypass resistance pathway in vitro and in vivo. This may be of particular clinical relevance since both Yes and IGF components are overexpressed in RMS. Increased YES/SFK activation might serve as a clinical biomarker for predicting tumor resistance to IGF-1R inhibition. Dual inhibition of IGF-1R and SFK may have a broader and enhanced clinical benefit for patients with RMS.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-yes/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Receptor de Insulina/antagonistas & inhibidores , Receptor de Insulina/metabolismo , Triazinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/metabolismo
8.
Clin Cancer Res ; 20(5): 1190-203, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24277455

RESUMEN

PURPOSE: The goal of this study is to optimize the activity of trabectedin for Ewing sarcoma by developing a molecularly targeted combination therapy. EXPERIMENTAL DESIGN: We have recently shown that trabectedin interferes with the activity of EWS-FLI1 in Ewing sarcoma cells. In this report, we build on this work to develop a trabectedin-based combination therapy with improved EWS-FLI1 suppression that also targets the drug-associated DNA damage to Ewing sarcoma cells. RESULTS: We demonstrate by siRNA experiments that EWS-FLI1 drives the expression of the Werner syndrome protein (WRN) in Ewing sarcoma cells. Because WRN-deficient cells are known to be hypersensitive to camptothecins, we utilize trabectedin to block EWS-FLI1 activity, suppress WRN expression, and selectively sensitize Ewing sarcoma cells to the DNA-damaging effects of SN38. We show that trabectedin and SN38 are synergistic, demonstrate an increase in DNA double-strand breaks, an accumulation of cells in S-phase and a low picomolar IC50. In addition, SN38 cooperates with trabectedin to augment the suppression of EWS-FLI1 downstream targets, leading to an improved therapeutic index in vivo. These effects translate into the marked regression of two Ewing sarcoma xenografts at a fraction of the dose of camptothecin used in other xenograft studies. CONCLUSIONS: These results provide the basis and rationale for translating this drug combination to the clinic. In addition, the study highlights an approach that utilizes a targeted agent to interfere with an oncogenic transcription factor and then exploits the resulting changes in gene expression to develop a molecularly targeted combination therapy.


Asunto(s)
Antineoplásicos/farmacología , Camptotecina/análogos & derivados , Daño del ADN/efectos de los fármacos , Dioxoles/farmacología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteína Proto-Oncogénica c-fli-1/antagonistas & inhibidores , Proteína EWS de Unión a ARN/antagonistas & inhibidores , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Tetrahidroisoquinolinas/farmacología , Animales , Camptotecina/farmacología , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Irinotecán , Ratones , Proteínas de Fusión Oncogénica/genética , Fenotipo , Proteína Proto-Oncogénica c-fli-1/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteína EWS de Unión a ARN/genética , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/mortalidad , Sarcoma de Ewing/patología , Trabectedina , Helicasa del Síndrome de Werner , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Res ; 72(22): 5889-99, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23002205

RESUMEN

We identified Bub1b as an essential element for the growth and survival of rhabdomyosarcoma (RMS) cells using a bar-coded, tetracycline-inducible short hairpin RNA (shRNA) library screen. Knockdown of Bub1b resulted in suppression of tumor growth in vivo, including the regression of established tumors. The mechanism by which this occurs is via postmitotic endoreduplication checkpoint and mitotic catastrophe. Furthermore, using a chromatin immunoprecipitation assay, we found that Bub1b is a direct transcriptional target of Forkhead Box M1 (FoxM1). Suppression of FoxM1 either by shRNA or the inhibitor siomycin A resulted in reduction of Bub1b expression and inhibition of cell growth and survival. These results show the important role of the Bub1b/FoxM1 pathway in RMS and provide potential therapeutic targets.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Rabdomiosarcoma/metabolismo , Animales , Proteínas de Ciclo Celular , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Código de Barras del ADN Taxonómico/métodos , Femenino , Proteína Forkhead Box M1 , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Ratones SCID , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/genética , Sistema del Grupo Sanguíneo Rh-Hr/biosíntesis , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Transducción de Señal , Trasplante Heterólogo
10.
J Natl Cancer Inst ; 103(12): 962-78, 2011 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-21653923

RESUMEN

BACKGROUND: Chromosomal translocations generating oncogenic transcription factors are the hallmark of a variety of tumors, including many sarcomas. Ewing sarcoma family of tumors (ESFTs) are characterized by the t(11;22)(q24;q12) translocation that generates the Ewing sarcoma breakpoint region 1 and Friend leukemia virus integration 1 (EWS-FLI1) fusion transcription factor responsible for the highly malignant phenotype of this tumor. Although continued expression of EWS-FLI1 is believed to be critical for ESFT cell survival, a clinically effective small-molecule inhibitor remains elusive likely because EWS-FLI1 is a transcription factor and therefore widely felt to be "undruggable." METHODS: We developed a high-throughput screen to evaluate more than 50 000 compounds for inhibition of EWS-FLI1 activity in TC32 ESFT cells. We used a TC32 cell-based luciferase reporter screen using the EWS-FLI1 downstream target NR0B1 promoter and a gene signature secondary screen to sort and prioritize the compounds. We characterized the lead compound, mithramycin, based on its ability to inhibit EWS-FLI1 activity in vitro using microarray expression profiling, quantitative reverse transcription-polymerase chain reaction, and immunoblot analysis, and in vivo using immunohistochemistry. We studied the impact of this inhibition on cell viability in vitro and on tumor growth in ESFT xenograft models in vivo (n = 15-20 mice per group). All statistical tests were two-sided. RESULTS: Mithramycin inhibited expression of EWS-FLI1 downstream targets at the mRNA and protein levels and decreased the growth of ESFT cells at half maximal inhibitory concentrations between 10 (95% confidence interval [CI] = 8 to 13 nM) and 15 nM (95% CI = 13 to 19 nM). Mithramycin suppressed the growth of two different ESFT xenograft tumors and prolonged the survival of ESFT xenograft-bearing mice by causing a decrease in mean tumor volume. For example, in the TC32 xenograft model, on day 15 of treatment, the mean tumor volume for the mithramycin-treated mice was approximately 3% of the tumor volume observed in the control mice (mithramycin vs control: 69 vs 2388 mm(3), difference = 2319 mm(3), 95% CI = 1766 to 2872 mm(3), P < .001). CONCLUSION: Mithramycin inhibits EWS-FLI1 activity and demonstrates ESFT antitumor activity both in vitro and in vivo.


Asunto(s)
Antineoplásicos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Plicamicina/farmacología , Proteína Proto-Oncogénica c-fli-1/efectos de los fármacos , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/efectos de los fármacos , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Inmunohistoquímica , Ratones , Microscopía Confocal , Análisis por Matrices de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Ewing/tratamiento farmacológico , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Translocación Genética/efectos de los fármacos , Trasplante Heterólogo
11.
Neoplasia ; 13(2): 145-53, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21403840

RESUMEN

ET-743 (trabectedin; Yondelis) is approved in Europe for the treatment of soft tissue sarcomas. Emerging phase 1 and 2 clinical data have shown high response rates in myxoid liposarcoma in part owing to the inhibition of the FUS-CHOP transcription factor. In this report, we show that modulation of specific oncogenic transcription factors by ET-743 may extend to other tumor types. We demonstrate that, among a panel of pediatric sarcomas, Ewing sarcoma family of tumors (ESFTs) cell lines bearing the EWS-FLI1 transcription factor are the most sensitive to treatment with ET-743 compared with osteosarcoma, rhabdomyosarcoma, and synovial sarcoma. We show that ET-743 reverses a gene signature of induced downstream targets of EWS-FLI1 in two different ESFT cell lines (P = .001). In addition, ET-743 directly suppresses the promoter activity of a known EWS-FLI1 downstream target NR0B1 luciferase reporter construct without changing the activity of a constitutively active control in ESFT cells. Furthermore, the effect is specific to EWS-FLI1, as forced expression of EWS-FLI1 in a cell type that normally lacks this fusion protein, HT1080 cells, induces the same NR0B1 promoter, but this activation is completely blocked by ET-743 treatment. Finally, we used gene set enrichment analysis to confirm that other mechanisms of ET-743 are active in ESFT cells. These results suggest a particular role for ET-743 in the treatment of translocation-positive tumors. In addition, the modulation of EWS-FLI1 makes it a novel targeting agent for ESFT and suggests that further development of this compound for the treatment of ESFT is warranted.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Dioxoles/uso terapéutico , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteína Proto-Oncogénica c-fli-1/antagonistas & inhibidores , Proteína EWS de Unión a ARN/antagonistas & inhibidores , Sarcoma de Ewing/tratamiento farmacológico , Tetrahidroisoquinolinas/uso terapéutico , Línea Celular Tumoral , Niño , Receptor Nuclear Huérfano DAX-1/efectos de los fármacos , Receptor Nuclear Huérfano DAX-1/genética , Europa (Continente) , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Proteína FUS de Unión a ARN/efectos de los fármacos , Sarcoma/tratamiento farmacológico , Trabectedina , Factor de Transcripción CHOP/efectos de los fármacos
12.
J Clin Invest ; 119(11): 3395-407, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19809159

RESUMEN

Rhabdomyosarcoma (RMS) is a childhood cancer originating from skeletal muscle, and patient survival is poor in the presence of metastatic disease. Few determinants that regulate metastasis development have been identified. The receptor tyrosine kinase FGFR4 is highly expressed in RMS tissue, suggesting a role in tumorigenesis, although its functional importance has not been defined. Here, we report the identification of mutations in FGFR4 in human RMS tumors that lead to its activation and present evidence that it functions as an oncogene in RMS. Higher FGFR4 expression in RMS tumors was associated with advanced-stage cancer and poor survival, while FGFR4 knockdown in a human RMS cell line reduced tumor growth and experimental lung metastases when the cells were transplanted into mice. Moreover, 6 FGFR4 tyrosine kinase domain mutations were found among 7 of 94 (7.5%) primary human RMS tumors. The mutants K535 and E550 increased autophosphorylation, Stat3 signaling, tumor proliferation, and metastatic potential when expressed in a murine RMS cell line. These mutants also transformed NIH 3T3 cells and led to an enhanced metastatic phenotype. Finally, murine RMS cell lines expressing the K535 and E550 FGFR4 mutants were substantially more susceptible to apoptosis in the presence of a pharmacologic FGFR inhibitor than the control cell lines expressing the empty vector or wild-type FGFR4. Together, our results demonstrate that mutationally activated FGFR4 acts as an oncogene, and these are what we believe to be the first known mutations in a receptor tyrosine kinase in RMS. These findings support the potential therapeutic targeting of FGFR4 in RMS.


Asunto(s)
Mutación/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Rabdomiosarcoma/fisiopatología , Animales , Ciclo Celular , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular , Replicación del ADN , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/fisiopatología , Neoplasias Pulmonares/secundario , Ratones , Modelos Moleculares , Metástasis de la Neoplasia , Fosforilación , Estructura Terciaria de Proteína , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química , Rabdomiosarcoma/mortalidad , Factor de Transcripción STAT3/metabolismo , Trasplante Heterólogo
13.
Clin Exp Metastasis ; 25(3): 201-11, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18071913

RESUMEN

Metastasis continues to be the leading cause of mortality for patients with cancer. High expression of the chemokine receptor CXCR4 correlates with poor prognosis in many cancers, including osteosarcoma and melanoma. CXCL12, the ligand for CXCR4, is expressed at high levels in the lung and lymph node, which are the primary sites to which these tumors metastasize respectively. These findings suggest that therapy aimed at disruption of this specific receptor/ligand complex may lead to a decrease in metastases. CTCE-9908, a small peptide CXCR4 antagonist was utilized in two murine metastasis models to test this hypothesis. Treatment of osteosarcoma cells in vitro with CTCE-9908 led to the following changes: decreased adhesion, decreased migration, decreased invasion, and decreased growth rate. Following tail vein injection of osteosarcoma cells, mice that were treated with CTCE-9908 had a 50% reduction in the number of gross metastatic lung nodules and a marked decrease in micro-metastatic disease. Similar findings were observed following injection of melanoma cells and treatment with CTCE-9908. However, these results could only be consistently reproduced when the cells were pre-treated with the inhibitor. A novel ex vivo luciferase assay showed decreased numbers of cells in the lung immediately after injection into mice, when treated with CTCE-9908, suggesting the importance of interactions between the receptor and the ligand. Our findings show that inhibition of the CXCR4/CXCL12 pathway decreases metastatic disease in two murine tumor models and expands on previous reports to describe potential mechanisms of action.


Asunto(s)
Quimiocina CXCL12/antagonistas & inhibidores , Neoplasias Pulmonares/prevención & control , Melanoma/prevención & control , Osteosarcoma/prevención & control , Péptidos/uso terapéutico , Receptores CXCR4/antagonistas & inhibidores , Animales , Western Blotting , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Movimiento Celular , Proliferación Celular , Quimiocina CXCL12/metabolismo , Citoesqueleto , Femenino , Neoplasias Pulmonares/secundario , Metástasis Linfática , Melanoma/secundario , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Osteosarcoma/secundario , Fragmentos de Péptidos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
14.
Cancer Res ; 67(7): 3431-40, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17409454

RESUMEN

Mapping of protein signaling networks within tumors can identify new targets for therapy and provide a means to stratify patients for individualized therapy. Despite advances in combination chemotherapy, the overall survival for childhood rhabdomyosarcoma remains approximately 60%. A critical goal is to identify functionally important protein signaling defects associated with treatment failure for the 40% nonresponder cohort. Here, we show, by phosphoproteomic network analysis of microdissected tumor cells, that interlinked components of the Akt/mammalian target of rapamycin (mTOR) pathway exhibited increased levels of phosphorylation for tumors of patients with short-term survival. Specimens (n = 59) were obtained from the Children's Oncology Group Intergroup Rhabdomyosarcoma Study (IRS) IV, D9502 and D9803, with 12-year follow-up. High phosphorylation levels were associated with poor overall and poor disease-free survival: Akt Ser(473) (overall survival P < 0.001, recurrence-free survival P < 0.0009), 4EBP1 Thr(37/46) (overall survival P < 0.0110, recurrence-free survival P < 0.0106), eIF4G Ser(1108) (overall survival P < 0.0017, recurrence-free survival P < 0.0072), and p70S6 Thr(389) (overall survival P < 0.0085, recurrence-free survival P < 0.0296). Moreover, the findings support an altered interrelationship between the insulin receptor substrate (IRS-1) and Akt/mTOR pathway proteins (P < 0.0027) for tumors from patients with poor survival. The functional significance of this pathway was tested using CCI-779 in a mouse xenograft model. CCI-779 suppressed phosphorylation of mTOR downstream proteins and greatly reduced the growth of two different rhabdomyosarcoma (RD embryonal P = 0.00008; Rh30 alveolar P = 0.0002) cell lines compared with controls. These results suggest that phosphoprotein mapping of the Akt/mTOR pathway should be studied further as a means to select patients to receive mTOR/IRS pathway inhibitors before administration of chemotherapy.


Asunto(s)
Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Rabdomiosarcoma/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Humanos , Lactante , Proteínas Sustrato del Receptor de Insulina , Masculino , Ratones , Ratones SCID , Fosfoproteínas/metabolismo , Fosforilación , Rabdomiosarcoma/tratamiento farmacológico , Transducción de Señal , Tasa de Supervivencia , Serina-Treonina Quinasas TOR , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nat Med ; 10(2): 182-6, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14704791

RESUMEN

Metastatic cancers, once established, are the primary cause of mortality associated with cancer. Previously, we used a genomic approach to identify metastasis-associated genes in cancer. From this genomic data, we selected ezrin for further study based on its role in physically and functionally connecting the actin cytoskeleton to the cell membrane. In a mouse model of osteosarcoma, a highly metastatic pediatric cancer, we found ezrin to be necessary for metastasis. By imaging metastatic cells in the lungs of mice, we showed that ezrin expression provided an early survival advantage for cancer cells that reached the lung. AKT and MAPK phosphorylation and activity were reduced when ezrin protein was suppressed. Ezrin-mediated early metastatic survival was partially dependent on activation of MAPK, but not AKT. To define the relevance of ezrin in the biology of metastasis, beyond the founding mouse model, we examined ezrin expression in dogs that naturally developed osteosarcoma. High ezrin expression in dog tumors was associated with early development of metastases. Consistent with this data, we found a significant association between high ezrin expression and poor outcome in pediatric osteosarcoma patients.


Asunto(s)
Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Metástasis de la Neoplasia , Osteosarcoma/metabolismo , Osteosarcoma/secundario , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas , Animales , Línea Celular Tumoral , Niño , Proteínas del Citoesqueleto , Perros , Humanos , Neoplasias Pulmonares/secundario , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteosarcoma/patología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal/fisiología , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...