Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Intervalo de año de publicación
1.
Lancet ; 393(10191): 2591-2598, 2019 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-31178155

RESUMEN

BACKGROUND: In 2013, the interim analysis of the Protocol for Herceptin as Adjuvant therapy with Reduced Exposure (PHARE) trial could not show that 6 months of adjuvant trastuzumab was non-inferior to 12 months. Here, we report the planned final analysis based on the prespecified number of occurring events. METHODS: PHARE is an open-label, phase 3, non-inferiority randomised trial of patients with HER2-positive early breast cancer comparing 6 months versus 12 months of trastuzumab treatment concomitant with or following standard neoadjuvant or adjuvant chemotherapy. The study was undertaken in 156 centres in France. Eligible patients were women aged 18 years or older with non-metastatic, operable, histologically confirmed adenocarcinoma of the breast and either positive axillary nodes or negative axillary nodes but a tumour of at least 10 mm. Participants must have received at least four cycles of a chemotherapy for this breast cancer and have started receiving adjuvant trastuzumab-treatment. Eligible patients were randomly assigned to either 6 months or 12 months of trastuzumab therapy duration between the third and sixth months of adjuvant trastuzumab. The randomisation was stratified by concomitant or sequential treatment with chemotherapy, oestrogen receptor status, and centre. The primary objective was non-inferiority in the intention-to-treat population in the 6-month group in terms of disease-free survival with a prespecified hazard margin of 1·15. This trial is registered with ClinicalTrials.gov, number NCT00381901. FINDINGS: 3384 patients were enrolled and randomly assigned to either 12 months (n=1691) or 6 months (n=1693) of adjuvant trastuzumab. One patient in the 12-month group and three patients in the 6-month group were excluded, so 1690 patients in each group were included in the intention-to-treat analysis. At a median follow-up of 7·5 years (IQR 5·3-8·8), 704 events relevant to disease-free survival were observed (345 [20·4%] in the 12-month group and 359 [21·2%] in the 6-month group). The adjusted hazard ratio for disease-free survival in the 12-month group versus the 6-month group was 1·08 (95% CI 0·93-1·25; p=0·39). The non-inferiority margin was included in the 95% CI. No differences in effects pertaining to trastuzumab duration were found in any of the subgroups. After the completion of trastuzumab treatment, rare adverse events occurred over time and the safety analysis remained similar to the previously published report. In particular, we found no change in the cardiac safety comparison, and only three additional cases in which the left ventricular ejection fraction decreased to less than 50% have been reported in the 12-month group. INTERPRETATION: The PHARE study did not show the non-inferiority of 6 months versus 12 months of adjuvant trastuzumab. Hence, adjuvant trastuzumab standard duration should remain 12 months. FUNDING: The French National Cancer Institute.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Trastuzumab/administración & dosificación , Adulto , Anciano , Antineoplásicos Inmunológicos/efectos adversos , Neoplasias de la Mama/metabolismo , Quimioterapia Adyuvante , Esquema de Medicación , Femenino , Francia , Humanos , Infusiones Intravenosas , Persona de Mediana Edad , Receptor ErbB-2/metabolismo , Análisis de Supervivencia , Trastuzumab/efectos adversos , Resultado del Tratamiento , Adulto Joven
2.
Breast Cancer Res Treat ; 166(2): 631-639, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28779219

RESUMEN

PURPOSE: This report compares the risk factors, the tumor phenotypes, and the BRCA1/BRCA2 genotype of early onset breast cancer (EOBC) patients between Southern Europe and North Africa. METHODS: Four hundred and fifty six women with invasive EOBC (≤40 years) were prospectively included from four centers in France (n = 270) and four centers in North Africa (Algeria, Egypt, Morocco, Tunisia; n = 186). Life style, tumor phenotype, familial history, BRCA1/BRCA2 genotype were compared between the two populations. RESULTS: We found an older age at menarche, a higher number of childbearing, a more frequent breastfeeding, a higher body mass index, a lower use of oral contraceptives in North African women compared to French women. TNM stage at diagnosis was higher in North African women than in French women. North African women had a lower incidence of triple negative and proliferative (Ki 67 index > 20%) tumors. There was a lower rate of BRCA1 mutation in North Africa (7 vs. 15%, P = 0.02). Three putative BRCA1/2 founder mutations were identified in North Africa. CONCLUSIONS: In EOBC, we found significant differences in risk factors, phenotype and a higher incidence of BRCA1 mutations in Southern Europe as compared to North Africa. The worst prognosis previously reported for EOBC in North Africa is more likely due to a higher stage at diagnosis than to a more aggressive phenotype, since triple negative tumors are more common in Southern Europe and advanced tumors in North Africa.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/patología , África del Norte , Edad de Inicio , Neoplasias de la Mama/genética , Femenino , Francia , Genotipo , Humanos , Estadificación de Neoplasias , Estudios Prospectivos , Factores de Riesgo
3.
Histopathology ; 70(7): 1089-1097, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28106924

RESUMEN

AIMS: Translocation renal cell carcinoma (tRCC) is a rare subtype of kidney tumour characterized by translocations involving the transcription factor TFE3 or TFEB. tRCC was introduced into the World Health Organization classification in 2004, but much is still unknown about the natural history, clinicopathological features and outcomes of the disease. The aim of this study was to describe the landscape of fusion transcript in a large single-institution series of fluorescence in-situ hybridization (FISH)-confirmed tRCCs and then to compare it to morphological and clinical data. METHODS AND RESULTS: Paired-end RNA sequencing was performed within a prospective database of the Department of Pathology, Centre Hospitalier Régional Universitaire (Lille, France). The diagnosis of tRCC was confirmed by FISH. Among a total of 1130 identified renal cell carcinomas, 21 cases (1.9%) showed rearrangement of the TFE3 (n = 20) or (TFEB) (n = 1) gene. Median patient age was 31 years (range = 15-47), and the female-to-male ratio was 6:1. Five different TFE3 fusion transcripts were identified; the most frequent TFE3 partners were PRCC (n = 4) and SFPQ (n = 4). The other partners involved were ASPCR1 (n = 1) and MED15 (n = 1) genes as well as a novel TFE3 partner, GRIPAP1. CONCLUSIONS: We identified a new fusion partner, GRIPAP1. The prognostic role of transcript type could not be determined because our number of cases was too small. Four patients (19%) died of the disease, all of which presented with a lymph node involvement at diagnosis. We confirm that tRCC can be an aggressive tumour, especially those of advanced clinical stage.


Asunto(s)
Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Proteínas de Fusión Oncogénica/genética , Adolescente , Adulto , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Carcinoma de Células Renales/epidemiología , Carcinoma de Células Renales/patología , Proteínas Portadoras/genética , Femenino , Humanos , Incidencia , Neoplasias Renales/epidemiología , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Adulto Joven
4.
Nat Rev Cancer ; 6(8): 645-51, 2006 08.
Artículo en Inglés | MEDLINE | ID: mdl-16862194

RESUMEN

The new French National Cancer Institute is unique in Europe in that it associates all aspects of cancer care, research and patient advocacy in one single umbrella body. It brings together all sectors of the cancer field in a patient-centred, multidisciplinary approach to the disease. By facilitating national, European and international collaborations, it aims to attack the disease with a strong and united front.


Asunto(s)
Academias e Institutos/organización & administración , Programas Nacionales de Salud/organización & administración , Neoplasias/terapia , Investigación Biomédica , Francia , Humanos , Defensa del Paciente , Atención Dirigida al Paciente
5.
Kidney Int ; 85(2): 457-70, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24067439

RESUMEN

Renal toxicity constitutes a dose-limiting side effect of anticancer therapies targeting vascular endothelial growth factor (VEGF). In order to study this further, we followed up 29 patients receiving this treatment, who experienced proteinuria, hypertension, and/or renal insufficiency. Eight developed minimal change nephropathy/focal segmental glomerulopathy (MCN/FSG)-like lesions and 13 developed thrombotic microangiopathy (TMA). Patients receiving receptor tyrosine kinase inhibitors (RTKIs) mainly developed MCN/FSG-like lesions, whereas TMA complicated anti-VEGF therapy. There were no mutations in factor H, factor I, or membrane cofactor protein of the complement alternative pathway, while plasma ADAMTS13 activity persisted and anti-ADAMTS13 antibodies were undetectable in patients with TMA. Glomerular VEGF expression was undetectable in TMA and decreased in MCN/FSG. Glomeruli from patients with TMA displayed a high abundance of RelA in endothelial cells and in the podocyte nuclei, but c-mip was not detected. Conversely, MCN/FSG-like lesions exhibited a high abundance of c-mip, whereas RelA was scarcely detected. RelA binds in vivo to the c-mip promoter and prevents its transcriptional activation, whereas RelA knockdown releases c-mip activation. The RTKI sorafenib inhibited RelA activity, which then promoted c-mip expression. Thus, our results suggest that c-mip and RelA define two distinct types of renal damage associated with VEGF-targeted therapies.


Asunto(s)
Inhibidores de la Angiogénesis/efectos adversos , Proteínas Portadoras/metabolismo , Enfermedades Renales/inducido químicamente , Glomérulos Renales/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/efectos adversos , Inhibidores de Proteínas Quinasas/efectos adversos , Factor de Transcripción ReIA/metabolismo , Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Adulto , Anciano , Animales , Secuencia de Bases , Sitios de Unión , Biomarcadores/metabolismo , Proteínas Portadoras/genética , Estudios de Casos y Controles , Línea Celular , Femenino , Regulación de la Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Glomeruloesclerosis Focal y Segmentaria/enzimología , Humanos , Hipertensión/inducido químicamente , Hipertensión/diagnóstico , Hipertensión/enzimología , Enfermedades Renales/diagnóstico , Enfermedades Renales/enzimología , Glomérulos Renales/enzimología , Glomérulos Renales/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Datos de Secuencia Molecular , Nefrosis Lipoidea/inducido químicamente , Nefrosis Lipoidea/diagnóstico , Nefrosis Lipoidea/enzimología , Niacinamida/efectos adversos , Valor Predictivo de las Pruebas , Regiones Promotoras Genéticas , Proteinuria/inducido químicamente , Proteinuria/diagnóstico , Proteinuria/enzimología , Insuficiencia Renal/inducido químicamente , Insuficiencia Renal/diagnóstico , Insuficiencia Renal/enzimología , Sorafenib , Microangiopatías Trombóticas/inducido químicamente , Microangiopatías Trombóticas/diagnóstico , Microangiopatías Trombóticas/enzimología , Factor de Transcripción ReIA/deficiencia , Factor de Transcripción ReIA/genética , Transcripción Genética , Transfección , Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
6.
BJU Int ; 113(4): 574-80, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23937547

RESUMEN

OBJECTIVE: To evaluate the impact of 'hereditary-like' status in upper tract urothelial carcinoma (UTUC) on the survival of patients who have undergone radical nephroureterectomy (RNU) and adjuvant chemotherapy. PATIENTS AND METHODS: A multicentre retrospective study was performed on all patients with high-risk UTUC who underwent RNU and adjuvant cisplatin-based chemotherapy. Using a patient risk identification tool, we distinguished tumours suspected to be hereditary from sporadic tumours and compared survival rates. RESULTS: A total of 112 patients with a median age of 67 years were included. Hereditary-like tumour status was detected in 35 patients (31.3%), while 77 patients (68.7%) had sporadic tumours. The median age was significantly younger in the hereditary-like tumour group (56.0 vs 69.8 years, P < 0.001). Overall survival (OS) after chemotherapy was significantly better in the group with hereditary-like tumours than in the group with sporadic tumours (5-year OS: 48.2 vs 32%; P = 0.008). The cancer-specific survival (CSS) rate was significantly better in the group with 'hereditary-like' tumours than in the group with sporadic tumours (5-year CSS: 58 vs 35%; P = 0.006). Although there was a trend in favour of the hereditary-like tumours, we observed no significant difference regarding progression-free survival (PFS) between the two groups (5-year PFS: 71 vs 52%; P = 0.07). CONCLUSION: Adjuvant chemotherapy after RNU improves survival outcomes in patients with hereditary-like UTUC compared with those with sporadic tumours.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Neoplasias Renales/cirugía , Nefrectomía/métodos , Uréter/cirugía , Neoplasias Ureterales/cirugía , Anciano , Quimioterapia Adyuvante , Supervivencia sin Enfermedad , Femenino , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento , Neoplasias Ureterales/tratamiento farmacológico , Neoplasias Ureterales/genética
7.
Lancet Oncol ; 14(8): 741-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23764181

RESUMEN

BACKGROUND: Since 2005, 12 months of adjuvant trastuzumab has been the standard treatment for patients with HER2-positive early-stage breast cancer. However, the optimum duration of treatment has been debated. We did a non-inferiority trial of a shorter exposure of 6 months versus the standard 12 months of trastuzumab for patients with early breast cancer. METHODS: We did an open-label, randomised, phase 3 trial in 156 centres in France. Patients with HER2-positive early breast cancer who had received at least four cycles of chemotherapy, had breast-axillary surgery, and had received up to 6 months of trastuzumab (administered by intravenous infusions over 30-90 min every 3 weeks; initial loading dose 8 mg/kg; 6 mg/kg thereafter) before randomisation were eligible. Patients were randomly assigned via central randomisation procedure with web-based software to continue trastuzumab for another 6 months (12 months total duration; control group) or to discontinue trastuzumab at 6 months (6 months total duration; experimental group). Randomisation was stratified by concomitant or sequential administration of trastuzumab with chemotherapy, oestrogen-receptor status, and centre using a minimisation algorithm. The primary endpoint was disease-free survival, with a prespecified non-inferiority margin of 1·15. Analyses were done in the intention-to-treat population. This study is registered at ClinicalTrials.gov, number NCT00381901. FINDINGS: 1691 patients were randomly assigned to receive 12 months of trastuzumab and 1693 to receive 6 months of trastuzumab; 1690 patients in each group were included in the intention-to-treat analyses. After a median follow-up of 42·5 months (IQR 30·1-51·6), 175 disease-free survival events were noted in the 12-month group and 219 in the 6-month group. 2-year disease-free survival was 93·8% (95% CI 92·6-94·9) in the 12-month group and 91·1% (89·7-92·4) in the 6-month group (hazard ratio 1·28, 95% CI 1·05-1·56; p=0·29). 119 (93%) of the 128 cardiac events (clinical or based on assessment of left ventricular ejection fraction) occurred while patients were receiving trastuzumab. Significantly more patients in the 12-month group experienced a cardiac event than did those in the 6-month group (96 [5·7%] of 1690 patients vs 32 [1·9%] of 1690 patients, p<0·0001). INTERPRETATION: After 3·5 years follow-up, we failed to show that 6 months of treatment with trastuzumab was non-inferior to 12 months of trastuzumab. Despite the higher rates of cardiac events, 12 months of adjuvant trastuzmab should remain the standard of care. FUNDING: French National Cancer Institute.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/cirugía , Mastectomía , Inhibidores de Proteínas Quinasas/administración & dosificación , Receptor ErbB-2/análisis , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/química , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/secundario , Quimioterapia Adyuvante , Supervivencia sin Enfermedad , Esquema de Medicación , Detección Precoz del Cáncer , Femenino , Francia , Cardiopatías/inducido químicamente , Humanos , Infusiones Intravenosas , Estimación de Kaplan-Meier , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Neoplasias Primarias Secundarias , Valor Predictivo de las Pruebas , Modelos de Riesgos Proporcionales , Inhibidores de Proteínas Quinasas/efectos adversos , Radioterapia Adyuvante , Receptor ErbB-2/genética , Factores de Tiempo , Trastuzumab , Resultado del Tratamiento , Adulto Joven
8.
Cancer ; 118(9): 2367-71, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21918962

RESUMEN

The costs of cancer care are rising, and spending on expensive innovative anticancer agents is likely to come under scrutiny as health care payers are confronted by the challenge of resource limits in the face of infinite demand. Indirect costs account for the major part of total attributable costs of cancer and are dominated by the cost of mortality in individuals of working age (who therefore do not contribute to economic productivity). Although cancer is a leading cause of morbidity and premature mortality, in 2007, it was estimated that cancer accounted for only around 6% of the total health care costs in Europe. It is estimated that cancer drug costs constitute around 12% of total direct cancer costs and 5% of the costs of all drugs. Countries vary in their uptake of novel anticancer agents. However, even in France--a leading nation for the use of these agents--the costs of innovative anticancer drugs accounted for <0.6% of total health care expenditure in 2006. Population-level data suggest that novel therapies have contributed (together with advances in screening and other aspects of care) to improvements in survival from cancer. If this is the case, then the potential reduction in the associated indirect costs could exceed the direct costs associated with the uptake of innovative drug therapies. Further research is required to establish the costs and benefits of novel agents in routine practice.


Asunto(s)
Antineoplásicos/economía , Costos de la Atención en Salud , Neoplasias/tratamiento farmacológico , Neoplasias/economía , Terapias en Investigación/economía , Europa (Continente) , Femenino , Francia , Humanos
9.
Lancet Oncol ; 12(10): 933-80, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21958503

RESUMEN

The burden of cancer is growing, and the disease is becoming a major economic expenditure for all developed countries. In 2008, the worldwide cost of cancer due to premature death and disability (not including direct medical costs) was estimated to be US$895 billion. This is not simply due to an increase in absolute numbers, but also the rate of increase of expenditure on cancer. What are the drivers and solutions to the so-called cancer-cost curve in developed countries? How are we going to afford to deliver high quality and equitable care? Here, expert opinion from health-care professionals, policy makers, and cancer survivors has been gathered to address the barriers and solutions to delivering affordable cancer care. Although many of the drivers and themes are specific to a particular field-eg, the huge development costs for cancer medicines-there is strong concordance running through each contribution. Several drivers of cost, such as over-use, rapid expansion, and shortening life cycles of cancer technologies (such as medicines and imaging modalities), and the lack of suitable clinical research and integrated health economic studies, have converged with more defensive medical practice, a less informed regulatory system, a lack of evidence-based sociopolitical debate, and a declining degree of fairness for all patients with cancer. Urgent solutions range from re-engineering of the macroeconomic basis of cancer costs (eg, value-based approaches to bend the cost curve and allow cost-saving technologies), greater education of policy makers, and an informed and transparent regulatory system. A radical shift in cancer policy is also required. Political toleration of unfairness in access to affordable cancer treatment is unacceptable. The cancer profession and industry should take responsibility and not accept a substandard evidence base and an ethos of very small benefit at whatever cost; rather, we need delivery of fair prices and real value from new technologies.


Asunto(s)
Prestación Integrada de Atención de Salud/economía , Costos de la Atención en Salud , Gastos en Salud , Accesibilidad a los Servicios de Salud/economía , Neoplasias/economía , Neoplasias/terapia , Australia , Ahorro de Costo , Análisis Costo-Beneficio , Prestación Integrada de Atención de Salud/legislación & jurisprudencia , Europa (Continente) , Costos de la Atención en Salud/legislación & jurisprudencia , Reforma de la Atención de Salud/economía , Gastos en Salud/legislación & jurisprudencia , Política de Salud/economía , Accesibilidad a los Servicios de Salud/legislación & jurisprudencia , Mal Uso de los Servicios de Salud/economía , Investigación sobre Servicios de Salud , Disparidades en Atención de Salud/economía , Humanos , Seguro de Salud/economía , Modelos Económicos , Neoplasias/diagnóstico , Factores Socioeconómicos , Estados Unidos
10.
Cancer ; 117(14): 3069-80, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21283982

RESUMEN

Physicians who are responsible for the delivery of systemic treatment in lung cancer should be aware of the potential risk of drug-induced pulmonary toxicity (DIPT), because such toxicity may develop in the context of a multifactorial clinical condition. First, most patients with lung cancer may suffer from other non-neoplastic, smoking-related lung diseases, such as emphysema and chronic obstructive lung disease, which may generate pathologic changes in lung parenchyma. In addition, lung cancer itself may worsen the respiratory function, inducing atelectasis and lymphangitic carcinomatosis. The superimposed iatrogenic damage may lead to respiratory failure and, sometimes, death. The risk of DIPT from chemotherapeutic agents has been widely examined in the past; and, currently, the potential for lung toxicity has been extended by the introduction of molecular targeted therapies. Because there are no univocal criteria with which to recognize DIPT, the diagnosis often is made by exclusion; consequently, it is hard to establish an early diagnosis. The objective of this review was to describe the major DIPTs associated with antineoplastic agents against nonsmall cell lung cancer to help physicians with this difficult diagnostic challenge.


Asunto(s)
Antineoplásicos/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Enfermedades Pulmonares/inducido químicamente , Neoplasias Pulmonares/tratamiento farmacológico , Humanos , Terapia Molecular Dirigida/efectos adversos
11.
Cancer ; 117(24): 5500-8, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21638278

RESUMEN

BACKGROUND: Urothelial carcinoma of the upper urinary tract (UUT-UC) was a rare, aggressive urologic cancer with a propensity for multifocality, local recurrence, and metastasis. High-risk patients had poor outcomes. Because of the rarity of these tumors, randomized clinical trials and data regarding adjuvant chemotherapy in locally advanced tumors are currently unavailable. Our objective was to assess the effect of adjuvant chemotherapy and the impact of potential prognostic factors on survival in high-risk, postsurgical UUT-UC patients. METHODS: Using a multi-institutional, international retrospective database, identified were 627 patients with high risk UUT-UCs (pT3N0, pT4N0 and/or N+ and/or M+) who underwent surgical removal. Only patients who received adjuvant chemotherapy were included. RESULTS: Overall, 140 patients (22.6%) with a median age of 67 years were included. The median follow-up was 22.5 months. The 5-year, overall survival for the entire cohort was 43%, the 5-year recurrence-free survival was 54%, and metastasis-free survival was 53% at 5 years. Positive surgical margins were an independent prognostic factor for recurrence (P = .06), cancer-specific mortality (P = .05), and overall mortality (P = .02) of any cause. Adjuvant chemotherapy was not linked with overall or cancer-specific survival in patients with high risk disease (adjuvant chemotherapy [n = 140] vs no treatment [n = 487]) (P >.5). CONCLUSIONS: Adjuvant postoperative chemotherapy did not offer any significant benefit to overall survival in our population. Additional data were necessary, and studies enrolling patients at high risk in clinical trials investigating neoadjuvant chemotherapy in conjunction with chemotherapy should have been highly encouraged.


Asunto(s)
Carcinoma de Células Transicionales/tratamiento farmacológico , Neoplasias Urológicas/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Transicionales/patología , Carcinoma de Células Transicionales/cirugía , Quimioterapia Adyuvante , Cisplatino/administración & dosificación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Pronóstico , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento , Neoplasias Urológicas/patología , Neoplasias Urológicas/cirugía
12.
Oncologist ; 16(12): 1684-97, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21976315

RESUMEN

Significant advances in the treatment of patients with breast cancer have been made in the past 10 years. The current systemic treatment of breast cancer is characterized by the discovery of multiple cancer targets leading to treatments that are more sophisticated and specific than conventional cytotoxic chemotherapy. Two classes of compounds that have helped improve clinical outcomes are small molecules and monoclonal antibodies targeting specific tyrosine kinase receptors. Many novel targets have been discovered, and parallel multiple approaches to anticancer therapy have recently emerged from the literature. One promising strategy is targeting the proangiogenic vascular endothelial growth factors (VEGFs), either by ligand sequestration (preventing VEGF receptor binding) or inhibiting downstream receptor signaling. Bevacizumab, a monoclonal antibody directed against VEGF, has been shown to improve the efficacy of taxanes in frontline treatment of patients with metastatic breast cancer. This review outlines the most promising breast cancer studies using bevacizumab combined with traditional cytotoxic agents in advanced breast cancer. In addition, we discuss the current indications reviewed by the Oncologic Drug Advisory Committee and define our vision of how the benefit of patient clinical trials should be measured.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Bevacizumab , Femenino , Humanos
13.
Genome Biol ; 22(1): 7, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397444

RESUMEN

BACKGROUND: Crosstalk between genetic, epigenetic, and immune alterations in upper tract urothelial carcinomas and their role in shaping muscle invasiveness and patient outcome are poorly understood. RESULTS: We perform an integrative genome- and methylome-wide profiling of diverse non-muscle-invasive and muscle-invasive upper tract urothelial carcinomas. In addition to mutations of FGFR3 and KDM6A, we identify ZFP36L1 as a novel, significantly mutated tumor suppressor gene. Overall, mutations of ZFP36 family genes (ZFP36, ZFP36L1, and ZFP36L2) are identified in 26.7% of cases, which display a high mutational load. Unsupervised DNA methylation subtype classification identifies two epi-clusters associated with distinct muscle-invasive status and patient outcome, namely, EpiC-low and EpiC-high. While the former is hypomethylated, immune-depleted, and enriched for FGFR3-mutated, the latter is hypermethylated, immune-infiltrated, and tightly associated with somatic mutations of SWI/SNF genes. CONCLUSIONS: Our study delineates for the first time the key role for convergence between genetic and epigenetic alterations in shaping clinicopathological and immune upper tract urothelial carcinoma features.


Asunto(s)
Carcinoma de Células Transicionales/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Vejiga Urinaria/genética , Factor 1 de Respuesta al Butirato/genética , Carcinoma de Células Transicionales/inmunología , Línea Celular Tumoral , Metilación de ADN , Epigenómica , Histona Demetilasas/genética , Humanos , Inmunidad , Mutación , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción/genética , Transcriptoma , Tristetraprolina/genética , Neoplasias de la Vejiga Urinaria/inmunología
14.
Sci Rep ; 10(1): 701, 2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31959902

RESUMEN

Sarcomatoid clear-cell renal cell carcinomas (sRCC) are associated with dismal prognosis. Genomic alterations associated with sarcomatoid dedifferentiation are poorly characterized. We sought to define the genomic landscape of sRCC and uncover potentially actionable therapeutic targets. We assessed the genomic landscape of sRCC using targeted panel sequencing including patients with microdissected sarcomatoid and epithelial components. Along with common genomic alterations associated with clear-cell histology, we found that Hippo was one of the most frequently altered pathways in these tumours. Hippo alterations were differentially enriched in sRCC compared to non-sRCC. Functional analysis showed that Hippo members mutations were associated with higher nuclear accumulation of YAP/TAZ, core effectors of the Hippo pathway. In a NF2-mutant sRCC model, YAP1 knockdown and NF2 reconstitution suppressed cell proliferation, tumour growth and invasion, both in vitro and in vivo. Overall, we show that Hippo pathway alterations are a feature of sRCC, and enable the exploration of the Hippo pathway as a novel potential therapeutic target.


Asunto(s)
Carcinoma de Células Renales/genética , Redes Reguladoras de Genes , Neoplasias Renales/genética , Mutación , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Predisposición Genética a la Enfermedad , Vía de Señalización Hippo , Humanos , Captura por Microdisección con Láser , Masculino , Ratones , Trasplante de Neoplasias , Neurofibromina 2/genética , Proteínas Serina-Treonina Quinasas/genética , Análisis de Secuencia de ADN/métodos , Transducción de Señal , Transactivadores/genética , Factores de Transcripción/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Regulación hacia Arriba , Proteínas Señalizadoras YAP
15.
Oncology ; 76(3): 170-2, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19212144

RESUMEN

A 58-year-old man with advanced renal cell carcinoma developed grade 3 proteinuria (8.5 g/24 h) without microscopic hematuria or renal insufficiency five days after temsirolimus infusion. Kidney biopsy revealed ischemic glomeruli and focal segmental glomerulosclerosis lesion. Proteinuria level decreased to 2.80 g per day two weeks after temsirolimus withdrawal. Clinicians should be aware to this complication.


Asunto(s)
Antineoplásicos/efectos adversos , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Sirolimus/análogos & derivados , Carcinoma de Células Renales/tratamiento farmacológico , Humanos , Neoplasias Renales/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Proteinuria/inducido químicamente , Sirolimus/efectos adversos
16.
Eur J Cancer ; 120: 132-139, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31522033

RESUMEN

BACKGROUND: Brivanib is a selective inhibitor of vascular endothelial growth factor and fibroblast growth factor (FGF) signalling. We performed a phase II randomised discontinuation trial of brivanib in 7 tumour types (soft-tissue sarcomas [STS], ovarian cancer, breast cancer, pancreatic cancer, non-small-cell lung cancer [NSCLC], gastric/esophageal cancer and transitional cell carcinoma [TCC]). PATIENTS AND METHODS: During a 12-week open-label lead-in period, patients received brivanib 800 mg daily and were evaluated for FGF2 status by immunohistochemistry. Patients with stable disease at week 12 were randomised to brivanib or placebo. A study steering committee evaluated week 12 response to determine if enrolment in a tumour type would continue. The primary objective was progression-free survival (PFS) for brivanib versus placebo in patients with FGF2-positive tumours. RESULTS: A total of 595 patients were treated, and stable disease was observed at the week 12 randomisation point in all tumour types. Closure decisions were made for breast cancer, pancreatic cancer, NSCLC, gastric cancer and TCC. Criteria for expansion were met for STS and ovarian cancer. In 53 randomised patients with STS and FGF2-positive tumours, the median PFS was 2.8 months for brivanib and 1.4 months for placebo (hazard ratio [HR]: 0.58, p = 0.08). For all randomised patients with sarcomas, the median PFS was 2.8 months (95% confidence interval [CI]: 1.4-4.0) for those treated with brivanib compared with 1.4 months (95% CI: 1.3-1.6) for placebo (HR = 0.64, 95% CI: 0.38-1.07; p = 0.09). In the 36 randomised patients with ovarian cancer and FGF2-positive tumours, the median PFS was 4.0 (95% CI: 2.6-4.2) months for brivanib and 2.0 months (95% CI: 1.2-2.7) for placebo (HR: 0.56, 95% CI: 0.26-1.22). For all randomised patients with ovarian cancer, the median PFS in those randomised to brivanib was 4.0 months (95% CI: 2.6-4.2) and was 2.0 months (95% CI: 1.2-2.7) in those randomised to placebo (HR = 0.54, 95% CI: 0.25-1.17; p = 0.11). CONCLUSION: Brivanib demonstrated activity in STS and ovarian cancer with an acceptable safety profile. FGF2 expression, as defined in the protocol, is not a predictive biomarker of the efficacy of brivanib.


Asunto(s)
Alanina/análogos & derivados , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Triazinas/uso terapéutico , Privación de Tratamiento/estadística & datos numéricos , Anciano , Alanina/uso terapéutico , Biomarcadores de Tumor/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/patología , Pronóstico , Tasa de Supervivencia
17.
Crit Rev Oncol Hematol ; 66(1): 21-30, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18206383

RESUMEN

The importance of the epidermal growth factor receptor (EGFR) axis in tumorigenesis and tumor progression makes it an attractive target for the development of anticancer therapies. Strategies aimed at inhibiting the EGFR pathway included different classes of compounds, with monoclonal antibodies and tyrosine kinase inhibitors being the most widely-investigated agents in colorectal cancer. Although anti-EGFR therapies are active in some patients, disease will become refractory to therapy in nearly all patients. Identification of specific markers likely to predict which patients will best respond to anti-EGFR therapy is a major challenge. While the occurrence of rash is associated with greater likelihood of response, EGFR staining by immunohistochemistry at baseline is not. Among biological predictors, some studies indicate that activated EGFR, EGFR amplification, absence of KRAS mutations, PTEN expression, and low VEGFR expression are implicated in response to anti-EGFR monoclonal antibodies. Moreover, germinal gene polymorphisms, such as dinucleotide repeats polymorphism or FcgammaR polymorphism, have been shown to be associated with response to anti-EGFR therapy. Since most available data come from retrospective studies, there is a need to validate these results in prospective trials.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Biomarcadores de Tumor , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Receptores ErbB/análisis , Receptores ErbB/genética , Receptores ErbB/fisiología , Amplificación de Genes , Genes ras , Humanos , Mutación , Fosfohidrolasa PTEN/fisiología , Polimorfismo Genético , Pronóstico , Transducción de Señal , Piel/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/fisiología
18.
Case Rep Gastroenterol ; 12(2): 532-539, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30283289

RESUMEN

We report a case of long-term survival with complete response of liver metastases within RAINBOW, a randomized, controlled trial of ramucirumab 8 mg/kg intravenously (days 1, 15) versus placebo, both plus paclitaxel 80 mg/m2 intravenously (days 1, 8, 15), every 4 weeks in patients with previously treated advanced gastroesophageal junction adenocarcinoma. A 64-year-old man with gastroesophageal junction adenocarcinoma and liver metastases received first-line folinic acid, 5-fluorouracil plus oxaliplatin (FOLFOX) following jejunostomy. On liver progression, he enrolled in RAINBOW (April 2012), receiving ramucirumab. In November 2013, positron emission tomography scan was consistent with complete metabolic response, confirmed by a follow-up scan in March 2016.

19.
Cell Rep ; 25(3): 811-821.e5, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332658

RESUMEN

Esthesioneuroblastoma (ENB) is a rare cancer of the olfactory mucosa, with no established molecular stratification to date. We report similarities of ENB with tumors arising in the neural crest and perform integrative analysis of these tumors. We propose a molecular-based subtype classification of ENB as basal or neural, both of which have distinct pathological, transcriptomic, proteomic, and immune features. Among the basal subtype, we uncovered an IDH2 R172 mutant-enriched subgroup (∼35%) harboring a CpG island methylator phenotype reminiscent of IDH2 mutant gliomas. Compared with the basal ENB methylome, the neural ENB methylome shows genome-wide reprogramming with loss of DNA methylation at the enhancers of axonal guidance genes. Our study reveals insights into the molecular pathogenesis of ENB and provides classification information of potential therapeutic relevance.


Asunto(s)
Biomarcadores de Tumor/análisis , Linaje de la Célula/genética , Metilación de ADN , Estesioneuroblastoma Olfatorio/genética , Variación Genética , Cavidad Nasal/metabolismo , Neoplasias Nasales/genética , Biología Computacional , Islas de CpG , Epigénesis Genética , Estesioneuroblastoma Olfatorio/clasificación , Estesioneuroblastoma Olfatorio/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Linfocitos Infiltrantes de Tumor , Masculino , Persona de Mediana Edad , Cavidad Nasal/patología , Neoplasias Nasales/clasificación , Neoplasias Nasales/metabolismo , Pronóstico , Proteoma/análisis , Tasa de Supervivencia , Transcriptoma
20.
Crit Rev Oncol Hematol ; 64(2): 115-28, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17702596

RESUMEN

Several models have been proposed to explain the mechanisms of endocrine resistance including aberrant growth-signaling pathways, and have led to the rational design of studies combining hormonotherapy with signal transduction inhibitors (STI) in advanced breast cancer. This article reviews the current status of these clinical trials. Preliminary results from the randomized controlled trials are rather disappointing. The mTOR inhibitor temsirolimus and the farnesyl transferase inhibitor tipifarnib combined with letrozole did not show any benefit compared to letrozole alone. As neoadjuvant therapy, gefinitib did not enhance the response rate induced by anastrozole. Interesting results were obtained with exemestane combined to celecoxib but should be further explored with adequate cardiac monitoring. Trastuzumab combined with anastrozole was more effective than anastrozole in terms of response rate and progression-free survival but not survival. Several controlled trials as first- or second-line therapy have started recently and over the next few years we should learn whether this approach will provide significant gains in efficacy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ensayos Clínicos como Asunto , Femenino , Antagonistas de Hormonas , Humanos , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA