Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Clin Cancer Res ; 13(8): 2512-8, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17438112

RESUMEN

PURPOSE: To optimally integrate epidermal growth factor receptor (EGFR) inhibitors into the clinical treatment of head and neck cancer, two important questions must be answered: (a) does EGFR inhibition add to the effects of radiochemotherapy, and (b) if so, which method of inhibiting EGFR is superior (an EGFR antibody versus a small molecule tyrosine kinase inhibitor)? We designed an in vivo study to address these questions. EXPERIMENTAL DESIGN: Nude mice with UMSCC-1 head and neck cancer xenografts received either single, double, or triple agent therapy with an EGFR inhibitor (either cetuximab or gefitinib), gemcitabine, and/or radiation for 3 weeks. Tumor volumes and animal weights were measured for up to 15 weeks. Immunoblotting and immunofluorescent staining were done on tumors treated with either cetuximab or gefitinib alone. RESULTS: The addition of an EGFR inhibitor significantly delayed the tumor volume doubling time, from a median of 40 days with radiochemotherapy (gemcitabine and radiation) alone, to 106 days with cetuximab and 66 days with gefitinib (both P < 0.005). Cetuximab resulted in significantly less weight loss than gefitinib. Immunoblot analysis and immunofluorescent staining of tumors show that although levels of phosphorylated AKT and extracellular signal-regulated kinase were decreased similarly in response to cetuximab or gefitinib, cetuximab caused prolonged suppression of pEGFR, pSTAT3, and Bcl(XL) compared with gefitinib. CONCLUSIONS: EGFR inhibition, particularly with cetuximab, improves the effectiveness of radiochemotherapy in this model of head and neck cancer. The correlation of response with prolonged suppression of EGFR, STAT3, and Bcl(XL) offers the possibility that these may be candidate biomarkers for response.


Asunto(s)
Antineoplásicos/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Cetuximab , Terapia Combinada , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Gefitinib , Ratones , Ratones Desnudos , Quinazolinas/uso terapéutico , Ratas , Trasplante Heterólogo , Gemcitabina
2.
Cancer Res ; 66(2): 981-8, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16424033

RESUMEN

Although the combination of gemcitabine and radiation produces a high frequency of complete responses in the treatment of locally advanced head and neck cancer, substantial toxicity suggests that an improvement in the therapeutic index is required. The purpose of this study was to determine if gefitinib could improve the efficacy of gemcitabine and if drug schedule is important. We hypothesized that gemcitabine followed by gefitinib would be superior to the opposite order because of both cell cycle and growth factor signaling interactions. Using UMSCC-1 cells in vitro, we confirmed that gefitinib arrested cells in G(1) and suppressed phospho-epidermal growth factor receptor (p(Y845)EGFR) and that gemcitabine arrested cells in S phase and stimulated p(Y845)EGFR. The schedule of gemcitabine followed by gefitinib caused arrest of cells in S phase. Gefitinib suppressed gemcitabine-mediated p(Y845)EGFR stimulation. This schedule caused decreased p(S473)AKT, increased poly(ADP-ribose) polymerase cleavage, and increased apoptosis compared with gemcitabine alone. The schedule of gefitinib followed by gemcitabine also caused suppression of p(Y845)EGFR but arrested cells in G(1). This schedule in which gefitinib was used first was associated with stable levels of p(S473)AKT and minimal poly(ADP-ribose) polymerase cleavage and apoptosis. These results were reflected in experiments in nude mice bearing UMSCC-1 xenografts, in which there was greater tumor regression and apoptosis when animals received gemcitabine followed by gefitinib during the first week of therapy. These findings suggest that the schedule of gemcitabine followed by gefitinib may increase the therapeutic index over gemcitabine alone and, combined with clinical data, encourage exploration of combination of gemcitabine, EGFR inhibitors, and radiation.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/patología , Desoxicitidina/análogos & derivados , Neoplasias de Cabeza y Cuello/patología , Quinazolinas/farmacología , Antibióticos Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Desoxicitidina/farmacología , Esquema de Medicación , Interacciones Farmacológicas , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/biosíntesis , Gefitinib , Humanos , Células Tumorales Cultivadas , Gemcitabina
3.
Cancer Res ; 66(24): 11554-9, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17178844

RESUMEN

Ataxia telangiectasia mutated (ATM) kinase plays a crucial role in the cellular response to DNA damage and in radiation resistance. Although much effort has focused on the relationship between ATM and other nuclear signal transducers, little is known about interactions between ATM and mitogenic signaling pathways. In this study, we show a novel relationship between ATM kinase and extracellular signal-regulated kinase 1/2 (ERK1/2), a key mitogenic stimulator. Activation of ATM by radiation down-regulates phospho-ERK1/2 and its downstream signaling via increased expression of mitogen-activated protein kinase phosphatase MKP-1 in both cell culture and tumor models. This dephosphorylation of ERK1/2 is independent of epidermal growth factor receptor (EGFR) activity and is associated with radioresistance. These findings show a new function for ATM in the control of mitogenic pathways affecting cell signaling and emphasize the key role of ATM in coordinating the cellular response to DNA damage.


Asunto(s)
Antígenos de Diferenciación/genética , Carcinoma de Células Escamosas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/efectos de la radiación , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Inmediatas-Precoces/efectos de la radiación , Glicoproteínas de Membrana/genética , Moléculas de Adhesión de Célula Nerviosa/genética , Fosfoproteínas Fosfatasas/metabolismo , Fosfoproteínas Fosfatasas/efectos de la radiación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas/efectos de la radiación , Receptores Inmunológicos/genética , Proteínas Supresoras de Tumor/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/radioterapia , Línea Celular Tumoral , Supervivencia Celular , Daño del ADN , Replicación del ADN , Fosfatasa 1 de Especificidad Dual , Activación Enzimática , Humanos , Ratones , Ratones Desnudos , Reacción en Cadena de la Polimerasa , Proteína Fosfatasa 1 , ARN Neoplásico/genética , ARN Neoplásico/aislamiento & purificación , Trasplante Heterólogo
4.
JCI Insight ; 3(4)2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29467321

RESUMEN

BACKGROUND: Constitutive activation of ERK1/2 occurs in various cancers, and its reactivation is a well-described resistance mechanism to MAPK inhibitors. ERK inhibitors may overcome the limitations of MAPK inhibitor blockade. The dual mechanism inhibitor SCH772984 has shown promising preclinical activity across various BRAFV600/RAS-mutant cancer cell lines and human cancer xenografts. METHODS: We have developed an orally bioavailable ERK inhibitor, MK-8353; conducted preclinical studies to demonstrate activity, pharmacodynamic endpoints, dosing, and schedule; completed a study in healthy volunteers (P07652); and subsequently performed a phase I clinical trial in patients with advanced solid tumors (MK-8353-001). In the P07652 study, MK-8353 was administered as a single dose in 10- to 400-mg dose cohorts, whereas in the MK-8353-001 study, MK-8353 was administered in 100- to 800-mg dose cohorts orally twice daily. Safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity were analyzed. RESULTS: MK-8353 exhibited comparable potency with SCH772984 across various preclinical cancer models. Forty-eight patients were enrolled in the P07652 study, and twenty-six patients were enrolled in the MK-8353-001 study. Adverse events included diarrhea (44%), fatigue (40%), nausea (32%), and rash (28%). Dose-limiting toxicity was observed in the 400-mg and 800-mg dose cohorts. Sufficient exposure to MK-8353 was noted that correlated with biological activity in preclinical data. Three of fifteen patients evaluable for treatment response in the MK-8353-001 study had partial response, all with BRAFV600-mutant melanomas. CONCLUSION: MK-8353 was well tolerated up to 400 mg twice daily and exhibited antitumor activity in patients with BRAFV600-mutant melanoma. However, antitumor activity was not particularly correlated with pharmacodynamic parameters. TRIAL REGISTRATION: ClinicalTrials.gov NCT01358331. FUNDING: Merck Sharp & Dohme Corp., a subsidiary of Merck & Co. Inc., and NIH (P01 CA168585 and R35 CA197633).


Asunto(s)
Indazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirrolidinas/farmacología , Triazoles/farmacología , Administración Oral , Adulto , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Diarrea/inducido químicamente , Diarrea/epidemiología , Perros , Relación Dosis-Respuesta a Droga , Erupciones por Medicamentos/epidemiología , Erupciones por Medicamentos/etiología , Evaluación Preclínica de Medicamentos , Fatiga/inducido químicamente , Fatiga/epidemiología , Femenino , Humanos , Indazoles/uso terapéutico , Masculino , Dosis Máxima Tolerada , Ratones , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Náusea/inducido químicamente , Náusea/epidemiología , Estadificación de Neoplasias , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/uso terapéutico , Pirrolidinas/uso terapéutico , Ratas , Triazoles/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
6.
Mol Oncol ; 8(2): 169-77, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24268915

RESUMEN

Mice heterozygous for mutations in the adenomatous polyposis coli gene (Apc(+/-) mice) develop intestinal neoplasia. Apc(+/-) tumor formation is thought to be dependent on cyclooxygenase 2 (COX2) expression; both pharmacologic COX2 inhibition and global Cox2 gene deletion reduce the number of intestinal tumors in Apc(+/-) mice. COX2 expression is reported in epithelial cells, fibroblasts, macrophages and endothelial cells of Apc(+/-) mouse polyps. However, the cell type(s) in which COX2 expression is required for Apc(+/-) tumor induction is not known. To address this question, we developed Apc(Min/+) mice in which the Cox2 gene is specifically deleted either in intestinal epithelial cells or in myeloid cells. There is no significant difference in intestinal polyp number between Apc(Min/+) mice with a targeted Cox2 gene deletion in myeloid cells and their control littermate Apc(Min/+) mice. In contrast, Apc(Min/+) mice with a targeted Cox2 deletion in intestinal epithelial cells have reduced intestinal tumorigenesis when compared to their littermate control Apc(Min/+) mice. However, two gender-specific effects are notable. First, female Apc(Min/+) mice developed more intestinal tumors than male Apc(Min/+) mice. Second, targeted intestinal epithelial cell Cox2 deletion decreased tumorigenesis in female, but not in male, Apc(Min/+) mice. Considered in the light of pharmacologic studies and studies with global Cox2 gene knockout mice, our data suggest that (i) intrinsic COX2 expression in intestinal epithelial cells plays a gender-specific role in tumor development in Apc(Min/+) mice, and (ii) COX2 expression in cell type(s) other than intestinal epithelial cells also modulates intestinal tumorigenesis in Apc(Min/+) mice, by a paracrine process.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon , Ciclooxigenasa 2 , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Mucosa Intestinal , Caracteres Sexuales , Poliposis Adenomatosa del Colon/enzimología , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Femenino , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Noqueados
7.
Radiat Oncol ; 5: 80, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20846422

RESUMEN

BACKGROUND: The purpose of this study was to determine the expected time to prostate specific antigen (PSA) normalization with or without neoadjuvant androgen deprivation (NAAD) therapy after treatment with intensity modulated radiotherapy (IMRT) for patients with clinically localized prostate cancer. METHODS: A retrospective cohort research design was used. A total of 133 patients with clinical stage T1c to T3b prostate cancer (2002 AJCC staging) treated in a community setting between January 2002 and July 2005 were reviewed for time to PSA normalization using 1 ng/mL and 2 ng/mL as criteria. All patients received IMRT as part of their management. Times to PSA normalization were calculated using the Kaplan-Meier method. Significance was assessed at p < 0.05. RESULTS: Fifty-six of the 133 patients received NAAD (42.1%). Thirty-one patients (23.8%) received radiation to a limited pelvic field followed by an IMRT boost, while 99 patients received IMRT alone (76.2%). The times to serum PSA normalization < 2 ng/mL when treated with or without NAAD were 298 ± 24 and 302 ± 33 days (mean ± SEM), respectively (p > 0.05), and 303 ± 24 and 405 ± 46 days, respectively, for PSA < 1 ng/mL (p < 0.05). Stage T1 and T2 tumors had significantly increased time to PSA normalization < 1 ng/mL in comparison to Stage T3 tumors. Also, higher Gleason scores were significantly correlated with a faster time to PSA normalization < 1 ng/mL. CONCLUSIONS: Use of NAAD in conjunction with IMRT leads to a significantly shortened time to normalization of serum PSA < 1 ng/mL in patients with clinically localized prostate cancer.


Asunto(s)
Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/radioterapia , Anciano , Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/efectos de la radiación , Estudios de Cohortes , Terapia Combinada , Humanos , Estimación de Kaplan-Meier , Masculino , Terapia Neoadyuvante , Estadificación de Neoplasias , Antígeno Prostático Específico/efectos de la radiación , Neoplasias de la Próstata/patología , Radioterapia de Intensidad Modulada , Estudios Retrospectivos
8.
Gastrointest Cancer Res ; 3(5): 191-6, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20084160

RESUMEN

The optimal adjuvant treatment for stage II colon cancer remains controversial. While chemotherapy is often recommended for high-risk stage II disease, many low-grade tumors with similar histopathologic features will recur and ultimately cause cancer-associated mortality. The development of molecular markers that predict clinical outcome or response to therapy in stage II colon cancer is an important tool that could give clinicians added information in discussions regarding the role of adjuvant chemotherapy. While many potential molecular biomarkers have been investigated, to date none have been validated in prospective clinical trials. Among the most promising molecular markers to be studied are microsatellite instability and 18q and 17p loss of heterozygosity, both of which are currently being evaluated as prognostic indicators in a large prospective clinical trial (Eastern Cooperative Oncology Group 5202). This review focuses on potential molecular biomarkers being evaluated for their prognostic value in stage II colon cancer and their potential role in clinical decision-making regarding the use of adjuvant chemotherapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA