Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cancer Res Commun ; 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38669053

RESUMEN

INTRODUCTION: Chemotherapy can potentially enhance the activity of immune checkpoint inhibitors by promoting immune priming. The phase 1b/2 JAVELIN Chemotherapy Medley trial evaluated first-line avelumab + concurrent chemotherapy in patients with advanced urothelial carcinoma or nonsmall cell lung cancer (NSCLC). PATIENTS AND METHODS: Avelumab 800 mg or 1200 mg was administered continuously every 3 weeks (Q3W) with standard doses of cisplatin + gemcitabine in patients with urothelial carcinoma, or carboplatin + pemetrexed in patients with nonsquamous NSCLC. Dual primary endpoints were dose-limiting toxicity (DLT; phase 1b) and confirmed objective response (phase 1b/2). RESULTS: In phase 1b, urothelial carcinoma and NSCLC cohorts received avelumab 800 mg (n=13 and n=6, respectively) or 1200 mg (n=6 each) + chemotherapy. In evaluable patients with urothelial carcinoma treated with avelumab 800 mg or 1200 mg + chemotherapy, DLT occurred in 1/12 (8.3%) and 1/6 (16.7%), respectively; no DLT occurred in the NSCLC cohort. In phase 2, 35 additional patients with urothelial carcinoma received avelumab 1200 mg + chemotherapy. Across all treated patients, safety profiles were similar irrespective of avelumab dose. Objective response rates (95% confidence internal) with avelumab 800 mg or 1200 mg + chemotherapy, respectively, across phase 1b/2, were 53.8% (25.1-80.8) and 39.0% (24.2-55.5) in urothelial carcinoma, and 50.0% (11.8-88.2) and 33.3% (4.3-77.7) in NSCLC. CONCLUSIONS: Preliminary efficacy and safety findings with avelumab + chemotherapy in urothelial carcinoma and NSCLC were consistent with previous studies of similar combination regimens. Conclusions about clinical activity are limited by small patient numbers. CLINICALTRIALS: gov identifier, NCT03317496.

2.
Urol Oncol ; 41(12): 461-475, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37968169

RESUMEN

Transurethral resection of bladder tumor followed by intravesical Bacillus Calmette-Guérin (BCG) is the standard of care in high-risk, non-muscle-invasive bladder cancer (NMIBC). Although many patients respond, recurrence and progression are common. In addition, patients may be unable to receive induction + maintenance due to intolerance or supply issues. Therefore, alternative treatment options are urgently required. Programmed cell death (ligand) 1 (PD-[L]1) inhibitors show clinical benefit in phase 1/2 trials in BCG-unresponsive NMIBC patients. This review presents the status of PD-(L)1 inhibition in high-risk NMIBC and discusses future directions. PubMed and Google scholar were searched for articles relating to NMIBC immunotherapy and ClinicalTrials.gov for planned and ongoing clinical trials. Preclinical and early clinical studies show that BCG upregulates PD-L1 expression in bladder cancer cells and, when combined with a PD-(L)1 inhibitor, a potent antitumor response is activated. Based on this mechanism, several PD-(L)1 inhibitors are in phase 3 trials in BCG-naïve, high-risk NMIBC in combination with BCG. Whereas PD-(L)1 inhibitors are well characterized in patients with advanced malignancies, the impact of immune-related adverse events (irAE) on the benefit/risk ratio in NMIBC should be determined. Alternative routes to intravenous administration, like subcutaneous and intravesical administration, may facilitate adherence and access. The outcomes of combination of PD-(L)1 inhibitors and BCG in NMIBC are highly anticipated. There will be a need to address treatment resources, optimal management of irAEs and education and training related to use of this therapy in clinical practice.


Asunto(s)
Neoplasias Vesicales sin Invasión Muscular , Neoplasias de la Vejiga Urinaria , Humanos , Adyuvantes Inmunológicos/uso terapéutico , Vacuna BCG/farmacología , Vacuna BCG/uso terapéutico , Neoplasias de la Vejiga Urinaria/patología , Medición de Riesgo , Administración Intravesical , Invasividad Neoplásica , Recurrencia Local de Neoplasia/tratamiento farmacológico
3.
JAMA Oncol ; 9(1): 40-50, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36394849

RESUMEN

Importance: Preclinical data suggest that poly(ADP-ribose) polymerase (PARP) inhibitors have synergistic activity when combined with immune checkpoint inhibitors (ICIs); however, it is unknown which tumor types or molecular subtypes may benefit from this combination. Objective: To investigate responses associated with the combination of avelumab and talazoparib in different tumor types and/or molecular subtypes. Design, Setting, and Participants: In this phase 1b and 2 basket nonrandomized controlled trial, patients with advanced solid tumors were enrolled in the following cohorts: non-small cell lung cancer (NSCLC); DNA damage response (DDR)-positive NSCLC; triple-negative breast cancer (TNBC); hormone receptor-positive, human epidermal growth factor receptor 2 (ERBB2)-negative, DDR-positive breast cancer; recurrent, platinum-sensitive ovarian cancer (OC); recurrent, platinum-sensitive, BRCA1/2-altered OC; urothelial cancer; metastatic castration-resistant prostate cancer (mCRPC); DDR-positive mCRPC; and BRCA1/2- or ATM-altered solid tumors. Data were analyzed between June 17, 2021, and August 6, 2021. Interventions: All patients in phases 1b and 2 received avelumab plus talazoparib. Main Outcomes and Measures: The phase 1b primary end point was dose-limiting toxic effects. The phase 2 primary end point was objective response, measured as objective response rate (ORR). Secondary end points included safety, time to response, duration of response (DOR), progression-free survival, time to prostate-specific antigen progression and PSA response of 50% or greater (for mCRPC), cancer antigen 125 response (for OC), pharmacokinetics, immunogenicity, and biomarkers. Results: A total of 223 patients (mean [SD] age, 63.2 [11.0] years; 117 [52.5%] men) were treated, including 12 patients in phase 1b and 211 patients in phase 2. The recommended phase 2 dose was avelumab 800 mg every 2 weeks plus talazoparib 1 mg once daily. In phase 2, the ORR was 18.2% (95% CI, 5.2%-40.3%) in patients with TNBC; 34.8% (95% CI, 16.4%-57.3%) in patients with HR-positive, ERBB2-negative, and DDR-positive BC; and 63.6% (95% CI, 30.8%-89.1%) in patients with platinum-sensitive, BRCA1/2-altered OC. Responses occurred more frequently in patients with BRCA1/2-altered tumors. Durable responses were observed in patients with TNBC (median [range] DOR, 11.1 [3.4-20.4] months); HR-positive, ERBB2-negative, and DDR-positive BC (median [range] DOR, 15.7 [3.9 to ≥20.6] months); and BRCA1/2-altered OC (median DOR not reached; range, 5.6 to ≥18.4 months). The most common grade 3 or greater treatment-related adverse events were anemia (75 patients [33.6%]), thrombocytopenia (48 patients [21.5%]), and neutropenia (31 patients [13.9%]). Conclusions and Relevance: This nonrandomized controlled trial found that ORRs for avelumab plus talazoparib were comparable with those with PARP inhibitor or ICI monotherapy. Prolonged DOR in patients with TNBC; HR-positive, ERBB2-negative, and DDR-positive BC; and BRCA1/2-altered OC warrant further investigation in randomized clinical trials. These data highlight the importance of prospective patient selection in future studies of ICI and PARP-inhibitor combinations. Trial Registration: ClinicalTrials.gov Identifier: NCT03330405.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Mama Triple Negativas , Masculino , Humanos , Persona de Mediana Edad , Femenino , Inhibidores de Poli(ADP-Ribosa) Polimerasas/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Estudios Prospectivos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Inmunoterapia
4.
Urol Oncol ; 39(10): 642-663, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34167873

RESUMEN

AIM: This review article summarizes the current clinical practice guidelines around disease definitions and risk stratifications, and the treatment of non-muscle-invasive bladder cancer (NMIBC). Recently completed and ongoing clinical trials of novel and investigational therapies in Bacillus Calmette-Guérin (BCG)-naïve, BCG-recurrent, and BCG-unresponsive patient populations are also described, e.g., those involving immune checkpoint inhibitors, targeted therapies, other chemotherapy regimens, vaccines, and viral- or bacterial-based treatments. Finally, a brief overview of enhanced cystoscopy and drug delivery systems for the diagnosis and treatment of NMIBC is provided. BACKGROUND: A global shortage of access to BCG is affecting the management of BCG-naïve and BCG-recurrent/unresponsive NMIBC; hence, there is an urgent need to assist patients and urologists to enhance the treatment of this disease. METHODS: Searches of ClinicalTrials.gov, PubMed, and Google Scholar were conducted. Published guidance and conference proceedings from major congresses were reviewed. CONCLUSION: Treatment strategies for NMIBC are generally consistent across guidelines. Several novel therapies have demonstrated promising antitumor activity in clinical trials, including in high-risk or BCG-unresponsive disease. The detection, diagnosis, surveillance, and treatment of NMIBC have also been improved through enhanced disease detection.


Asunto(s)
Vacuna BCG/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Administración Intravesical , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Masculino
5.
Clin Cancer Res ; 24(8): 1816-1823, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29549159

RESUMEN

Purpose: Utomilumab (PF-05082566) is an agonistic mAb that engages the immune costimulatory molecule 4-1BB/CD137. In this first-in-human, phase I, open-label, multicenter, multiple-dose study (NCT01307267) we evaluated safety, tolerability, pharmacokinetics, preliminary clinical activity, and pharmacodynamics of single-agent utomilumab in patients with advanced malignancies.Experimental Design: Dose escalation was based on a standard 3+3 design for doses of utomilumab from 0.006 to 0.3 mg/kg every 4 weeks and a time-to-event continual reassessment method for utomilumab 0.6 to 10 mg/kg every 4 weeks. The primary study endpoint was dose-limiting toxicity (DLT) in the first two cycles.Results: Utomilumab demonstrated a well-tolerated safety profile (N = 55). None of the patients experienced a DLT at the dose levels evaluated. The most common treatment-related adverse events were fatigue, pyrexia, decreased appetite, dizziness, and rash (<10% of patients). Only one (1.8%) patient experienced a grade 3-4 treatment-related adverse event (fatigue), and no clinically relevant elevations in transaminases were noted. Utomilumab demonstrated linear pharmacokinetics at doses ranging from 0.006 to 10 mg/kg, with similar safety and pharmacokinetics in anti-drug antibody (ADA)-negative and ADA-positive patients. The overall objective response rate was 3.8% (95% CI, 0.5%-13.0%) in patients with solid tumors and 13.3% in patients with Merkel cell carcinoma, including a complete response and a partial response. Circulating biomarkers support 4-1BB/CD137 engagement by utomilumab and suggest that circulating lymphocyte levels may influence probability of clinical benefit.Conclusions: The favorable safety profile and preliminary antitumor activity demonstrated by utomilumab warrant further evaluation in patients with advanced malignancies. Clin Cancer Res; 24(8); 1816-23. ©2018 AACR.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Inmunoglobulina G/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/farmacocinética , Biomarcadores de Tumor , Terapia Combinada , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/efectos adversos , Persona de Mediana Edad , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias/diagnóstico , Neoplasias/mortalidad , Resultado del Tratamiento , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/sangre
6.
Ann Surg Oncol ; 25(3): 768-775, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28887726

RESUMEN

BACKGROUND: Desmoid fibromatosis is a fibroblastic neoplasm driven by aberrations within the WNT pathway, exhibiting mutations in ß-catenin or APC. We review the long-term follow-up of patients in a phase I study treated with an oral gamma secretase inhibitor, PF-03084014. METHODS: PF-03084014 was administered orally at doses ranging from 20 to 330 mg twice daily. Tumor assessments were performed using computed tomography/magnetic resonance imaging (CT/MRI) within 4 weeks of study entry, and every other cycle through cycle 9. After cycle 9, patients were evaluated as clinically indicated. RESULTS: Seven patients with desmoid fibromatosis were treated between December 2009 and December 2016 at the University of Colorado. Five patients (71.4%, 95% confidence interval [CI] 29.0-96.3%) achieved a partial response (PR), with a mean time to achieving response of 11.9 months (95% CI 2.5-21.4 months). All patients who achieved a PR continue to maintain responses between 47.9 and 73+ months. Four patients stopped treatment yet remain free of progression between 11 and 53+ months. One patient had PFS of 42+ months, with a 17% decrease in the target lesion. A biopsy performed at the end of the study showed decreased tumoral cellularity compared with previous biopsies. Effective treatment doses ranged from 80 to 330 mg administered orally twice daily. CONCLUSIONS: PF-03084014 was effective in treating desmoid tumors, with an objective response rate of 71.4% (95% CI 29.0-96.3%) in this small cohort of patients. PF-03084014 exhibits promising activity, even at relatively low doses (80 mg twice daily), with high tolerability leading to prolonged disease control even after therapy discontinuation.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Fibromatosis Agresiva/tratamiento farmacológico , Tetrahidronaftalenos/administración & dosificación , Valina/análogos & derivados , Administración Oral , Fibromatosis Agresiva/enzimología , Fibromatosis Agresiva/patología , Estudios de Seguimiento , Humanos , Pronóstico , Estudios Retrospectivos , Factores de Tiempo , Valina/administración & dosificación
7.
Oncotarget ; 8(2): 2320-2328, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27906684

RESUMEN

BACKGROUND: The NOTCH signaling pathway may be involved in the survival of stem cell-like tumor-initiating cells and contribute to tumor growth. In this phase Ib, open-label, multicenter study (NCT01876251), we evaluated PF-03084014, a selective gamma-secretase inhibitor in patients with advanced triple-negative breast cancer. METHODS: The dose-finding part was based on a 2×3 matrix design using the modified toxicity probability interval method. Oral PF-03084014 was administered twice daily continuously in combination with intravenous docetaxel given on day 1 of each 21-day cycle. Primary endpoint was first-cycle dose-limiting toxicity (DLT) for the dose-finding part and 6-month progression-free survival (PFS) for the expansion cohort treated at the maximum tolerated dose (MTD). Secondary endpoints included safety, objective response, and pharmacokinetics of the combination. RESULTS AND CONCLUSIONS: The MTD was estimated to be PF-03084014 100 mg twice daily / docetaxel 75 mg/m2. At this dose level, combination treatment was generally well tolerated (one DLT, grade 3 diarrhea, among eight DLT-evaluable patients). The most common all-grade, treatment-related adverse events reported in all patients (N = 29) were neutropenia (90%), fatigue (79%), nausea (72%), leukopenia (69%), diarrhea (59%), alopecia (55%), anemia (55%), and vomiting (48%). No effect was observed on the pharmacokinetics of docetaxel when administered in combination with PF-03084014. Four (16%) of 25 response-evaluable patients achieved a confirmed partial response; nine (36%) patients had stable disease, including five patients with unconfirmed partial response. In the expansion cohort, median PFS was 4.1 (95% CI 1.3-8.1) months (6-month PFS rate 17.1% [95% CI 0.8-52.6%]).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Taxoides/administración & dosificación , Tetrahidronaftalenos/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Valina/análogos & derivados , Adulto , Anciano , Progresión de la Enfermedad , Docetaxel , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neutropenia/inducido químicamente , Neutropenia/epidemiología , Taxoides/efectos adversos , Tetrahidronaftalenos/efectos adversos , Neoplasias de la Mama Triple Negativas/epidemiología , Neoplasias de la Mama Triple Negativas/patología , Valina/administración & dosificación , Valina/efectos adversos , Vómitos/inducido químicamente , Vómitos/epidemiología
8.
Clin Cancer Res ; 21(1): 60-7, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25231399

RESUMEN

PURPOSE: To estimate the maximum tolerated dose (MTD) for continuous oral administration of the γ-secretase inhibitor PF-03084014, determine the recommended phase II dose (RP2D), and evaluate safety and preliminary activity in patients with advanced solid tumors. EXPERIMENTAL DESIGN: This open-label, phase I study consisted of a dose-finding portion based on a 3+3 design, followed by an expansion cohort. PF-03084014 was administered orally, twice daily (BID) for 21 continuous days. Tested doses ranged from 20 to 330 mg BID. In the expansion cohort, patients were to receive the estimated MTD or a lower dose of PF-03084014. RESULTS: A total of 64 patients received treatment. The MTD was estimated to be 220 mg BID. The RP2D was determined to be 150 mg BID, based on the better safety profile versus the 220-mg BID dose, given comparable NOTCH-related target inhibition. The most common treatment-related adverse events were diarrhea, nausea, fatigue, hypophosphatemia, vomiting, rash, and decreased appetite, which were generally mild to moderate in severity. One patient with advanced thyroid cancer had a complete response, and five of seven response-evaluable patients with desmoid tumor achieved a partial response (71.4% objective response rate). Tumor responses were mostly durable, ranging from 1.74+ to 24+ months. PF-03084014 demonstrated a generally dose-dependent pharmacokinetic profile at doses ranging from 20 to 330 mg BID. Consistent downmodulation of NOTCH-related HES4 gene expression was observed in peripheral blood from all evaluable patients. CONCLUSION: Further development of PF-03084014 for the treatment of patients with advanced solid tumors is warranted and currently under evaluation.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Neoplasias/tratamiento farmacológico , Tetrahidronaftalenos/administración & dosificación , Valina/análogos & derivados , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacocinética , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/patología , Tetrahidronaftalenos/efectos adversos , Tetrahidronaftalenos/farmacocinética , Resultado del Tratamiento , Valina/administración & dosificación , Valina/efectos adversos , Valina/farmacocinética
9.
Clin Cancer Res ; 21(5): 1044-51, 2015 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-25388167

RESUMEN

PURPOSE: To estimate the maximum tolerated dose (MTD) of single-agent PF-04449913, and to evaluate safety, tolerability, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity in patients with advanced tumors. EXPERIMENTAL DESIGN: A 3+3 design was used in this open-label, multicenter, phase I study and dose escalation/de-escalation applied until identification of the MTD. PF-04449913 was orally administered once daily in continuous 28-day treatment cycles. The starting dose was 80 mg. RESULTS: A total of 23 patients were enrolled; 19 were evaluable for first-cycle dose-limiting toxicity (DLT). The first-cycle DLT rate at the 640 mg dose level was 33.3%, and the MTD was estimated to be 320 mg once daily. The recommended phase II dose was not determined. PF-04449913 was generally well tolerated at doses of 80 to 320 mg once daily. The most common treatment-related adverse events (AE) were grade 1-2 dysgeusia, fatigue, decreased appetite, nausea, dizziness, dehydration, and diarrhea. Treatment-related grade 3 AEs only occurred in patients receiving PF-04449913 640 mg once daily. No treatment-related grade 4-5 AEs were reported. Pharmacokinetic analysis indicated a generally dose-proportional kinetics with biphasic elimination, supporting once-daily dosing. PF-04449913 modulated hedgehog signaling at the dose levels tested, as demonstrated by >80% downregulation of GLI1 expression in the skin of treated patients. Eight patients (34.8%) achieved stable disease; none had complete or partial response. Three patients with disease progression at enrollment had prolonged disease stabilization (≥6 months). CONCLUSIONS: The results obtained in this study support further evaluation of PF-04449913 in patients with advanced solid tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencimidazoles/uso terapéutico , Proteínas Hedgehog/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Compuestos de Fenilurea/uso terapéutico , Administración Oral , Adulto , Anciano , Antineoplásicos/farmacología , Bencimidazoles/farmacología , Femenino , Expresión Génica , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/diagnóstico , Neoplasias/genética , Proteínas Oncogénicas/genética , Compuestos de Fenilurea/farmacología , Transactivadores/genética , Resultado del Tratamiento , Proteína con Dedos de Zinc GLI1
10.
BMC Cancer ; 14: 166, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24606768

RESUMEN

BACKGROUND: This phase II study evaluated the efficacy and safety/tolerability of sunitinib plus trastuzumab in patients with HER2-positive advanced breast cancer (ABC). METHODS: Eligible patients received sunitinib 37.5 mg/day and trastuzumab administered either weekly (loading, 4 mg/kg; then weekly 2 mg/kg) or 3-weekly (loading, 8 mg/kg; then 3-weekly 6 mg/kg). Prior trastuzumab and/or lapatinib treatment were permitted. The primary endpoint was objective response rate (ORR). RESULTS: Sixty patients were enrolled and evaluable for safety; 57 were evaluable for efficacy. The majority of patients (58%) had received no prior chemotherapy in the metastatic setting. The ORR was 37%; the clinical benefit rate (CBR; percent objective response plus stable disease ≥ 24 weeks) was 56%. Among patients who were treatment-naïve or had received only adjuvant therapy, the ORR was 44% and the CBR was 59%. Overall, median overall survival had not been reached and the 1-year survival rate was 91%. The majority of adverse events (AEs) were mild to moderate in severity. Forty percent of patients experienced AEs related to measured left ventricular ejection fraction (LVEF) declines, which occurred more frequently in patients who had received prior anthracycline treatment. Ten percent of patients exhibited symptoms related to LVEF declines. One patient died on study from cardiogenic shock. Antitumor response and several safety parameters appeared to correlate with sunitinib exposure. CONCLUSIONS: Sunitinib plus trastuzumab demonstrated antitumor activity in patients with HER2-positive ABC, particularly those who were treatment-naïve or had only received prior adjuvant treatment. Sunitinib plus trastuzumab had acceptable safety and tolerability in patients with HER2-positive ABC who had not received prior anthracycline therapy. TRIAL REGISTRATION: NCT00243503.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Femenino , Humanos , Indoles/administración & dosificación , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pirroles/administración & dosificación , Sunitinib , Trastuzumab , Resultado del Tratamiento
11.
Breast ; 22(5): 650-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23958375

RESUMEN

PURPOSE: This randomized, open-label phase II study compared the efficacy of sunitinib monotherapy with that of single-agent standard-of-care (SOC) chemotherapy in patients with previously treated advanced triple-negative breast cancer (TNBC). METHODS: Patients with advanced TNBC, relapsed after anthracycline- and taxane-based chemotherapy, were randomized to receive either sunitinib (37.5 mg/day) or the investigator's choice of SOC therapy. Progression-free survival was the primary endpoint. RESULTS: Median progression-free survival was 2.0 months with sunitinib and 2.7 months with SOC chemotherapy (one-sided P = 0.888). Median overall survival was not prolonged with sunitinib (9.4 months) compared with SOC chemotherapy (10.5 months; one-sided P = 0.839). The objective response rate was 3% with sunitinib and 7% with SOC chemotherapy (one-sided P = 0.962). CONCLUSIONS: Sunitinib monotherapy did not improve efficacy compared with SOC chemotherapy in patients with previously treated advanced TNBC, for which identification of effective treatments and therapeutic targets remains an urgent need. TRIAL REGISTRATION: NCT00246571.


Asunto(s)
Antineoplásicos/uso terapéutico , Indoles/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Pirroles/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antraciclinas/uso terapéutico , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Capecitabina , Quimioterapia Adyuvante , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Supervivencia sin Enfermedad , Docetaxel , Fatiga/inducido químicamente , Femenino , Fluorouracilo/análogos & derivados , Fluorouracilo/uso terapéutico , Síndrome Mano-Pie/etiología , Humanos , Indoles/efectos adversos , Indoles/farmacocinética , Persona de Mediana Edad , Metástasis de la Neoplasia , Neutropenia/inducido químicamente , Paclitaxel/uso terapéutico , Pirroles/efectos adversos , Pirroles/farmacocinética , Sunitinib , Tasa de Supervivencia , Taxoides/uso terapéutico , Trombocitopenia/inducido químicamente , Neoplasias de la Mama Triple Negativas/patología , Gemcitabina
12.
Eur J Cancer ; 44(17): 2668-79, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18783938

RESUMEN

The high mobility group HMGA1 protein belongs to a family of architectural factors that play a role in chromosomal organisation and gene transcription regulation. HMGA1 overexpression represents a common feature of human malignant tumours and is causally associated with neoplastic transformation and metastatic progression. Recently, HMGA1 expression has been correlated with the presence of chromosomal rearrangements and suggested to promote genomic instability. Here, we report a novel interaction between HMGA1 protein and the ataxia-telangiectasia mutated (ATM) kinase, the major key player in the cellular response to DNA damage caused by several agents such as ionising radiation (IR). We identified an SQ motif on HMGA1, which is effectively phosphorylated by ATM in vitro and in vivo. Interestingly, confocal microscopy revealed that HMGA1 colocalises with the activated form of ATM (ATM S1981p). Moreover, HMGA1 ectopic expression decreases cell survival following exposure to IR as assessed by clonogenic survival in MCF-7 cells, further supporting the hypothesis that HMGA1 might act as a downstream target of the ATM pathway in response to DNA damage.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína HMGA1a/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/fisiología , Daño del ADN/fisiología , Proteína HMGA1a/efectos de la radiación , Humanos , Fosforilación , Radiación Ionizante
13.
Proc Natl Acad Sci U S A ; 105(30): 10547-52, 2008 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-18650378

RESUMEN

Gene therapy for beta-thalassemia requires stable transfer of a beta-globin gene into hematopoietic stem cells (HSCs) and high and regulated hemoglobin expression in the erythroblastic progeny. We developed an erythroid-specific lentiviral vector driving the expression of the human beta-globin gene from a minimal promoter/enhancer element containing two hypersensitive sites from the beta-globin locus control region. Transplantation of transduced HSCs into thalassemic mice leads to stable and long-term correction of anemia with all red blood cells expressing the transgene. A frequency of 30-50% of transduced HSCs, harboring an average vector copy number per cell of 1, was sufficient to fully correct the thalassemic phenotype. In the mouse model of Cooley's anemia transplantation of transduced cells rescues lethality, leading to either a normal or a thalassemia intermedia phenotype. We show that genetically corrected erythroblasts undergo in vivo selection with preferential survival of progenitors harboring proviral integrations in genome sites more favorable to high levels of vector-derived expression. These data provide a rationale for a gene therapy approach to beta-thalassemia based on partially myeloablative transplantation protocols.


Asunto(s)
Eritroblastos/metabolismo , Regulación de la Expresión Génica , Terapia Genética/métodos , Globinas/metabolismo , Células Madre Hematopoyéticas/citología , Talasemia beta/genética , Talasemia beta/terapia , Animales , Trasplante de Células , Clonación Molecular , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos C57BL , Fenotipo , Células Madre/citología
14.
Cancer Res ; 67(16): 7738-45, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699778

RESUMEN

ARLTS1 is a newly characterized tumor suppressor gene located at chromosome 13q14.3 and involved in the pathogenesis of various types of tumors: two single-nucleotide polymorphisms, one of them responsible for protein truncation, were found statistically associated with familial malignancies, whereas DNA hypermethylation and genomic deletions have been identified as a mechanism of ARLTS1 down-regulation in sporadic cancers. We found that in a large portion of lung carcinomas (37%) and in all analyzed lung cancer cell lines, ARLTS1 is strongly down-regulated due to DNA methylation in its promoter region. After its restoration by adenoviral transduction, ARLTS1-negative A549 and H1299 cells underwent apoptosis and inhibition of cell growth. Furthermore, ARLTS1 reexpression significantly reduced the ability of A549 and H1299 to form tumors in nude mice. Finally, we identified approximately 650 transcripts differentially expressed after restoration of ARLTS1 expression in A549 cells, suggesting that various pathways involved in cell survival, proliferation, signaling, and development mediate the effects of wild-type ARLTS1 in a lung cancer system.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Genes Supresores de Tumor , Neoplasias Pulmonares/genética , Factores de Ribosilacion-ADP/biosíntesis , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Secuencia Conservada , Metilación de ADN , Regulación hacia Abajo , Terapia Genética/métodos , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Alineación de Secuencia
15.
Clin Cancer Res ; 10(22): 7607-12, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15569992

RESUMEN

The fragile histidine triad (FHIT) gene located on chromosome 3p14.2 is frequently deleted in human tumors. We have previously reported deletions at the FHIT locus in 50% of bladder carcinoma derived cell lines and reduced expression in 61% of primary transitional carcinomas of the urinary bladder. To additionally investigate the role of FHIT alterations in the development of bladder cancer, we used heterozygous and nullizygous Fhit-deficient mice in a chemically induced carcinogenesis model. Results showed that 8 of 28 (28%) and 6 of 13 (46%) of the Fhit -/- and +/-, respectively, versus 2 of 25 (8%) Fhit +/+ mice developed invasive carcinoma after treatment with N-butyl-N-(4-hydroxybutyl) nitrosamine. To explore the possibility of a FHIT-based gene therapy for bladder cancer, we studied the effects of restored Fhit protein expression on cell proliferation, cell kinetics, and tumorigenicity in BALB/c nude mice, with human SW780 Fhit-null transitional carcinoma derived cells. In vitro transduction of SW780 Fhit-negative cells with adenoviral-FHIT inhibited cell growth, increased apoptotic cell population, and suppressed s.c. tumor growth in nude mice. These findings suggest the important role of Fhit in bladder cancer development and support the effort to additionally investigate a FHIT-based gene therapy.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Carcinoma de Células Transicionales/genética , Proteínas de Neoplasias/genética , Neoplasias de la Vejiga Urinaria/genética , Adenoviridae/genética , Animales , Butilhidroxibutilnitrosamina/farmacología , Línea Celular , Línea Celular Tumoral , Femenino , Citometría de Flujo , Terapia Genética/métodos , Genotipo , Heterocigoto , Humanos , Immunoblotting , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factores de Tiempo
16.
Cancer Gene Ther ; 11(9): 633-41, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15272314

RESUMEN

Pancreatic carcinoma is one of the most aggressive tumors, and, being refractory to conventional therapies, is an excellent target for new therapeutic approaches. Based on our previous finding of high HMGA1 expression in pancreatic cancer cells compared to normal pancreatic tissue, we evaluated whether suppression of HMGA1 protein expression could be a treatment option for patients affected by pancreatic cancer. Here we report that HMGA1 proteins are overexpressed in pancreatic carcinoma cell lines, and their downregulation through an adenovirus carrying the HMGA1 gene in an antisense orientation (Ad Yas-GFP) results in the death of three human pancreatic carcinoma cell lines (PANC1, Hs766T and PSN1). Pretreatment of PANC1 and PSN1 cells with Ad Yas-GFP suppressed and reduced, respectively, their ability to form xenograft tumors in nude mice. To further verify the role of HMGA1 in pancreatic tumorigenesis, we used a HMGA1 antisense phosphorothioate oligodeoxynucleotide (ODN); its addition induced a decrease in HMGA1 protein levels and a significant reduction of the proliferation rate of PANC1-, Hs766T- and PSN1-treated cells. Therefore, suppression of HMGA1 protein synthesis by an HMGA1 antisense approach seems to be a feasible treatment strategy in pancreatic carcinomas.


Asunto(s)
Apoptosis , Proliferación Celular/efectos de los fármacos , Proteína HMGA1a/antagonistas & inhibidores , Modelos Animales , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Animales , Regulación hacia Abajo , Proteínas Fluorescentes Verdes/metabolismo , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Oligonucleótidos Antisentido/farmacología , Neoplasias Pancreáticas/genética , Trasplante Heterólogo , Células Tumorales Cultivadas
17.
Carcinogenesis ; 25(11): 2107-14, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15231692

RESUMEN

DEP-1/HPTPeta, a receptor-type protein tyrosine phosphatase, is a candidate tumor suppressor gene because its expression was blocked in rat and human thyroid transformed cells, and its restoration reverted their neoplastic phenotype. In addition, loss of DEP-1/HPTPeta heterozygosity has been described in mammary, lung and colon primary tumors. We now show that DEP-1/HPTPeta is drastically reduced in several cell lines originating from human epithelial pancreatic carcinomas compared with normal pancreatic tissue. We also show that the infection of AsPC1 and PSN1 cells with a recombinant adenovirus carrying r-PTPeta cDNA (the rat homolog of DEP-1/HPTPeta) inhibits their proliferation. Flow cytometric analysis of the infected cells demonstrated that restoration of r-PTPeta activity disrupts their cell cycle and leads to apoptosis. Finally, the growth of PSN1 xenograft tumors was blocked by the intratumoral injection of a recombinant adeno-associated virus carrying r-PTPeta. The data suggest that restoration of DEP-1/HPTPeta expression could be a useful tool for the gene therapy of human pancreatic cancers.


Asunto(s)
Genes Supresores de Tumor , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Animales , División Celular , Línea Celular Tumoral , Cartilla de ADN , Exones/genética , Genotipo , Humanos , Cinética , Neoplasias Pancreáticas/genética , Reacción en Cadena de la Polimerasa , Ratas , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
18.
Clin Cancer Res ; 10(7): 2459-65, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15073125

RESUMEN

PURPOSE: WWOX (WW domain containing oxidoreductase) is a tumor suppressor gene that maps to the common fragile site FRA16D. We showed previously that WWOX is frequently altered in human lung and esophageal cancers. The purpose of this study was to delineate more precisely the role of WWOX in pancreatic carcinogenesis. EXPERIMENTAL DESIGN: We analyzed 15 paired pancreatic adenocarcinoma samples and 9 pancreatic cancer cell lines for WWOX alterations. Colony assay and cell cycle analysis were also performed to evaluate the role of the WWOX as a tumor suppressor gene. RESULTS: Loss of heterozygosity at the WWOX locus was observed in 4 primary tumors (27%). Methylation analysis showed that site-specific promoter hypermethylation was detected in 2 cell lines (22%) and treatment with the demethylating agent 5-aza-2'-deoxycytidine demonstrated an increase in the expression of WWOX. In addition, 2 primary tumor samples (13%) showed promoter hypermethylation including the position of site-specific methylation. Transcripts missing WWOX exons were detected in 4 cell lines (44%) and in 2 tumor samples (13%). Real-time reverse transcription PCR revealed a significant reduction of WWOX expression in all of the cell lines and in 6 primary tumors (40%). Western blot analysis showed a significant reduction of the WWOX protein in all of the cell lines. Furthermore, transfection with WWOX inhibited colony formation of pancreatic cancer cell lines by triggering apoptosis. CONCLUSION: These results indicate that the WWOX gene may play an important role in pancreatic tumor development.


Asunto(s)
Azacitidina/análogos & derivados , Genes Supresores de Tumor , Oxidorreductasas/fisiología , Neoplasias Pancreáticas/genética , Adenocarcinoma/metabolismo , Alelos , Antimetabolitos Antineoplásicos/farmacología , Apoptosis , Azacitidina/farmacología , Western Blotting , Ciclo Celular , Línea Celular Tumoral , Separación Celular , Transformación Celular Neoplásica , Metilación de ADN , Análisis Mutacional de ADN , ADN Complementario/metabolismo , Decitabina , Exones , Citometría de Flujo , Humanos , Pérdida de Heterocigocidad , Oxidorreductasas/genética , Neoplasias Pancreáticas/metabolismo , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Proteínas Supresoras de Tumor , Oxidorreductasa que Contiene Dominios WW
19.
Clin Cancer Res ; 10(8): 2720-4, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15102676

RESUMEN

PURPOSE: Parkin, a gene mutated in autosomal recessive juvenile Parkinsonism and mapped to the common fragile site FRA6E on human chromosome 6q25-q27, is associated with a frequent loss of heterozygosity and altered expression in breast and ovarian carcinomas. In addition, homozygous deletions of exon 2 creating deleterious truncations of the Parkin transcript were observed in the lung adenocarcinoma cell lines Calu-3 and H-1573, suggesting that the loss of this locus and the resulting changes in its expression are involved in the development of these tumors. EXPERIMENTAL DESIGN: We examined 20 paired normal and non-small cell lung cancer samples for the presence of Parkin alterations in the coding sequence and changes in gene expression. We also restored gene expression in the Parkin-deficient lung carcinoma cell line H460 by use of a recombinant lentivirus containing the wild-type Parkin cDNA. RESULTS: Loss of heterozygosity analysis identified a common region of loss in the Parkin/FRA6E locus with the highest frequency for the intragenic marker D6S1599 (45%), and semi-quantitative reverse transcription-PCR revealed reduced expression in 3 of 9 (33%) lung tumors. Although we did not observe any in vitro changes in cell proliferation or cell cycle, ectopic Parkin expression had the ability to reduce in vivo tumorigenicity in nude mice. CONCLUSION: These data suggest that Parkin is a tumor suppressor gene whose inactivation may play an important role in non-small cell lung cancer tumorigenesis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Ciclo Celular , División Celular , Línea Celular Tumoral , Metilación de ADN , Análisis Mutacional de ADN , ADN Complementario/metabolismo , Exones , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Homocigoto , Humanos , Lentivirus/genética , Pérdida de Heterocigocidad , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Repeticiones de Microsatélite , Modelos Genéticos , Mutación , Trasplante de Neoplasias , Polimorfismo Genético , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
20.
Proc Natl Acad Sci U S A ; 100(20): 11517-22, 2003 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-14504409

RESUMEN

Frequent allelic loss at human chromosome 11q23-q24 occurs in a wide variety of cancers, suggesting that this region may harbor a tumor suppressor gene. By constructing a physical map of the LOH11CR2 minimal region of loss on 11q23-q24 associated with lung and breast carcinomas, we were able to clone a hereditary translocation, t(11;12)(q23;q24), in a patient with early-onset breast cancer and family history of cancer. The breakpoint was found within 6 kb of the BCSC-1 candidate tumor suppressor gene located in the LOH11CR2 region whereas additional loss of heterozygosity (LOH) analysis in breast and ovarian tumors, including that of the patient with the t(11;12)(q23;q24), implicated the BCSC-1 locus as the primary target of deletion. Northern analysis of the BCSC-1 mRNA revealed a lack of expression in 33 of 41 (80%) tumor cell lines, and its ectopic expression led to the suppression of colony formation in vitro and tumorigenicity in vivo. These data suggest that BCSC-1 may exert a tumor suppressor activity and is a likely target of the LOH observed on 11q23-q24 in cancer.


Asunto(s)
Cromosomas Humanos Par 11 , Genes Supresores de Tumor , Animales , Cromosomas Humanos Par 12 , Femenino , Humanos , Cariotipificación , Pérdida de Heterocigocidad , Masculino , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Mutación , Linaje , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA