Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Cancer Ther ; 22(10): 1154-1165, 2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37486983

RESUMEN

AZD5153, a reversible, bivalent inhibitor of the bromodomain and extraterminal family protein BRD4, has preclinical activity in multiple tumors. This first-in-human, phase I study investigated AZD5153 alone or with olaparib in patients with relapsed/refractory solid tumors or lymphoma. Adults with relapsed tumors intolerant of, or refractory to, prior therapies received escalating doses of oral AZD5153 once daily or twice daily continuously (21-day cycles), or AZD5153 once daily/twice daily continuously or intermittently plus olaparib 300 mg twice daily, until disease progression or unacceptable toxicity. Between June 30, 2017 and April 19, 2021, 34 patients received monotherapy and 15 received combination therapy. Dose-limiting toxicities were thrombocytopenia/platelet count decreased (n = 4/n = 2) and diarrhea (n = 1). The recommended phase II doses (RP2D) were AZD5153 30 mg once daily or 15 mg twice daily (monotherapy) and 10 mg once daily (intermittent schedule) with olaparib. With AZD5153 monotherapy, common treatment-emergent adverse events (TEAE) included fatigue (38.2%), thrombocytopenia, and diarrhea (each 32.4%); common grade ≥ 3 TEAEs were thrombocytopenia (14.7%) and anemia (8.8%). With the combination, common TEAEs included nausea (66.7%) and fatigue (53.3%); the most common grade ≥ 3 TEAE was thrombocytopenia (26.7%). AZD5153 had dose-dependent pharmacokinetics, with minimal accumulation, and demonstrated dose-dependent modulation of peripheral biomarkers, including upregulation of HEXIM1. One patient with metastatic pancreatic cancer receiving combination treatment had a partial response lasting 4.2 months. These results show AZD5153 was tolerable as monotherapy and in combination at the RP2Ds; common toxicities were fatigue, hematologic AEs, and gastrointestinal AEs. Strong evidence of peripheral target engagement was observed.


Asunto(s)
Antineoplásicos , Linfoma , Neoplasias , Trombocitopenia , Adulto , Humanos , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Proteínas de Ciclo Celular , Diarrea/inducido químicamente , Fatiga/inducido químicamente , Fatiga/tratamiento farmacológico , Linfoma/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Proteínas Nucleares , Proteínas de Unión al ARN , Trombocitopenia/inducido químicamente , Factores de Transcripción
2.
Cancer Res ; 81(7): 1667-1680, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33558336

RESUMEN

Insights into oncogenesis derived from cancer susceptibility loci (SNP) hold the potential to facilitate better cancer management and treatment through precision oncology. However, therapeutic insights have thus far been limited by our current lack of understanding regarding both interactions of these loci with somatic cancer driver mutations and their influence on tumorigenesis. For example, although both germline and somatic genetic variation to the p53 tumor suppressor pathway are known to promote tumorigenesis, little is known about the extent to which such variants cooperate to alter pathway activity. Here we hypothesize that cancer risk-associated germline variants interact with somatic TP53 mutational status to modify cancer risk, progression, and response to therapy. Focusing on a cancer risk SNP (rs78378222) with a well-documented ability to directly influence p53 activity as well as integration of germline datasets relating to cancer susceptibility with tumor data capturing somatically-acquired genetic variation provided supportive evidence for this hypothesis. Integration of germline and somatic genetic data enabled identification of a novel entry point for therapeutic manipulation of p53 activities. A cluster of cancer risk SNPs resulted in increased expression of prosurvival p53 target gene KITLG and attenuation of p53-mediated responses to genotoxic therapies, which were reversed by pharmacologic inhibition of the prosurvival c-KIT signal. Together, our results offer evidence of how cancer susceptibility SNPs can interact with cancer driver genes to affect cancer progression and identify novel combinatorial therapies. SIGNIFICANCE: These results offer evidence of how cancer susceptibility SNPs can interact with cancer driver genes to affect cancer progression and present novel therapeutic targets.


Asunto(s)
Resistencia a Antineoplásicos/genética , Neoplasias/genética , Neoplasias/patología , Proteína p53 Supresora de Tumor/genética , Animales , Antineoplásicos/uso terapéutico , Biomarcadores Farmacológicos/metabolismo , Carcinogénesis/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Mutación de Línea Germinal/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación Missense , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Polimorfismo de Nucleótido Simple/fisiología , Pronóstico , Factores de Riesgo , Transducción de Señal/genética , Resultado del Tratamiento
3.
J Natl Cancer Inst ; 110(8): 914-917, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29529211

RESUMEN

Heat shock proteins (HSPs) are molecular chaperones that maintain proteins in their correct conformation to ensure stability and protect carcinoma cells from apoptosis. HSP90 inhibitors (HSP90i) block multiple targets simultaneously, and despite responses in a selected population, no HSP90i have yet been approved. We present a patient with a lung tumor with an exceptional response to cisplatin/gemcitabine in combination with HSP90i, which nowadays continues with HSP90i maintenance after three years. Whole-exome sequencing of the lung tumor unveiled a BRCA1/2 deficiency mutational signature, and mutation analysis confirmed a germline BRCA1 mutation. The striking efficacy of HSP90i plus chemotherapy vs chemotherapy alone was reproduced in a patient-derived xenograft (PDX) model from a breast cancer patient with a BRCA1 mutation (mean tumor volume [SD], No. of tumors: vehicle 8.38 [7.07] mm3, n = 3; HSP90i 4.18 [1.93] mm3, n = 5; cisplatin plus gemcitabine 3.31 [1.95] mm3, n = 5; cisplatin plus gemcitabine plus HSP90i 0.065 [0.076] mm3, n = 6). This case and the PDX demonstrate the efficacy for therapeutic inhibition of HSP90 in a BRCA-mutated patient, opening a new potential avenue for better identifying patients who might benefit most from HSP90i.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteína BRCA1/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Mutación de Línea Germinal , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Isoxazoles/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Resorcinoles/administración & dosificación , Adulto , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/secundario , Cisplatino/administración & dosificación , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Quimioterapia de Mantención , Resultado del Tratamiento , Gemcitabina
4.
JAMA Oncol ; 4(3): 351-357, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29327044

RESUMEN

IMPORTANCE: Derived neutrophils/(leukocytes minus neutrophils) ratio (dNLR) and lactate dehydrogenase (LDH) level have been correlated with immune checkpoint inhibitor (ICI) outcomes in patients with melanoma. OBJECTIVE: To determine whether pretreatment dNLR and LDH are associated with resistance to ICIs in patients with advanced non-small cell lung cancer (NSCLC). DESIGN, SETTING, AND PARTICIPANTS: Multicenter retrospective study with a test (n = 161) and a validation set (n = 305) treated with programmed death 1/programmed death ligand 1 (PD-1/PD-L1) inhibitors in 8 European centers, and a control cohort (n = 162) treated with chemotherapy only. Complete blood cell counts, LDH, and albumin levels were measured before ICI treatment. A lung immune prognostic index (LIPI) based on dNLR greater than 3 and LDH greater than upper limit of normal (ULN) was developed, characterizing 3 groups (good, 0 factors; intermediate, 1 factor; poor, 2 factors). MAIN OUTCOMES AND MEASURES: The primary end point was overall survival (OS). Secondary end points were progression-free survival (PFS) and disease control rate (DCR). RESULTS: In the pooled ICI cohort (N = 466), 301 patients (65%) were male, 422 (90%) were current or former smokers, and 401 (87%) had performance status of 1 or less; median age at diagnosis was 62 (range, 29-86) years; 270 (58%) had adenocarcinoma and 159 (34%) had squamous histologic subtype. Among 129 patients with PD-L1 data, 96 (74%) had PD-L1 of at least 1% by immunohistochemical analysis, and 33 (26%) had negative results. In the test cohort, median PFS and OS were 3 (95% CI, 2-4) and 10 (95% CI, 8-13) months, respectively. A dNLR greater than 3 and LDH greater than ULN were independently associated with OS (hazard ratio [HR] 2.22; 95% CI, 1.23-4.01 and HR, 2.51; 95% CI, 1.32-4.76, respectively). Median OS for poor, intermediate, and good LIPI was 3 months (95% CI, 1 month to not reached [NR]), 10 months (95% CI, 8 months to NR), and 34 months (95% CI, 17 months to NR), respectively, and median PFS was 2.0 (95% CI, 1.7-4.0), 3.7 (95% CI, 3.0-4.8), and 6.3 (95% CI, 5.0-8.0) months (both P < .001). Disease control rate was also correlated with dNLR greater than 3 and LDH greater than ULN. Results were reproducible in the ICI validation cohort for OS, PFS, and DCR, but were nonsignificant in the chemotherapy cohort. CONCLUSIONS AND RELEVANCE: Pretreatment LIPI, combining dNLR greater than 3 and LDH greater than ULN, was correlated with worse outcomes for ICI, but not for chemotherapy, suggesting that LIPI can serve as a potentially useful tool when selecting ICI treatment, raising the hypothesis that the LIPI might be useful for identifying patients unlikely to benefit from treatment with an ICI.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Pulmón/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Progresión de la Enfermedad , Femenino , Indicadores de Salud , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
5.
Pancreas ; 46(7): 913-920, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28697132

RESUMEN

OBJECTIVES: Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer. Overexpression of Yes associated protein 1 (YAP1), a downstream target of Hippo pathway, implicated in regulation of cell growth and apoptosis, has been reported in several human tumor types. The objective of this study was to investigate YAP1 expression in patients with PDAC and its prognostic values. METHODS: We evaluated YAP1 expression in 64 PDAC and 15 chronic pancreatitis (CP) cases and its related pancreatic intraepithelial neoplasia (PanIN) lesions and in 5 control subjects. Yes associated protein 1 expression was determined by immunohistochemistry. Association of YAP1 with clinicopathologic features in PDAC, disease-free survival, and overall survival was analyzed. RESULTS: We found a higher positive rate of nuclear expression of YAP1 in PDAC than in CP (P = 0.000) and lower expression of YAP1 in PanIN lesions in CP in contrast with expression in PanIN lesions in PDAC. Nuclear overexpression of YAP1 in PDAC is associated with hepatic metastasis (P = 0.0280) and is a prognostic factor (P = 0.0320), as well as surgical margin involvement (P = 0.0013) and tumoral stage (P = 0.0109). CONCLUSIONS: Overexpression of YAP1 may occur as a part of tumorigenesis of PDAC. Yes associated protein 1 is an independent prognostic marker for overall survival of PDAC and associated with liver metastasis, being a potential therapeutic target.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Biomarcadores de Tumor/biosíntesis , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Pancreáticas/metabolismo , Fosfoproteínas/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/patología , Femenino , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Análisis Multivariante , Neoplasias Pancreáticas/patología , Pronóstico , Análisis de Supervivencia , Factores de Transcripción , Proteínas Señalizadoras YAP
6.
Nat Rev Cancer ; 16(4): 251-65, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27009395

RESUMEN

Decades of research have shown that mutations in the p53 stress response pathway affect the incidence of diverse cancers more than mutations in other pathways. However, most evidence is limited to somatic mutations and rare inherited mutations. Using newly abundant genomic data, we demonstrate that commonly inherited genetic variants in the p53 pathway also affect the incidence of a broad range of cancers more than variants in other pathways. The cancer-associated single nucleotide polymorphisms (SNPs) of the p53 pathway have strikingly similar genetic characteristics to well-studied p53 pathway cancer-causing somatic mutations. Our results enable insights into p53-mediated tumour suppression in humans and into p53 pathway-based cancer surveillance and treatment strategies.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Neoplasias/genética , Polimorfismo de Nucleótido Simple/genética , Proteína p53 Supresora de Tumor/genética , Genoma Humano , Humanos , Mutación
7.
Semin Oncol ; 42(6): 775-87, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26615125

RESUMEN

Advances in tumor biology and human genetics that have identified driver events and critical dependencies, together with the development of drugs for specific targets, hold promise for an era of personalized oncology treatment. The efficacy of targeted therapies in patient populations selected for treatment on the basis of the molecular features of their tumors is shifting the current practice, and molecular profiling of patients to support clinical decision-making is becoming part of the everyday clinical practice in oncology. Actionable biomarkers are those that allow matching patients to specific therapies. Currently, there is a strong need to have accurate and reliable means of identifying these biomarkers. With the evolution of biotechnology and the advent of next-generation sequencing, platforms for molecular analysis have quickly evolved, allowing a high degree of multiplexing that aims to transform the use of targeted therapies in oncology. This article aims to summarize the different biomarker applications and discuss the current challenges along with the major technologies presently used in clinical practice.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Neoplasias/genética , Medicina de Precisión/métodos , Biología Computacional/métodos , Marcadores Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Patología Molecular/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Cell Rep ; 8(4): 983-90, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25131200

RESUMEN

A rare germline duplication upstream of the bone morphogenetic protein antagonist GREM1 causes a Mendelian-dominant predisposition to colorectal cancer (CRC). The underlying disease mechanism is strong, ectopic GREM1 overexpression in the intestinal epithelium. Here, we confirm that a common GREM1 polymorphism, rs16969681, is also associated with CRC susceptibility, conferring ∼20% differential risk in the general population. We hypothesized the underlying cause to be moderate differences in GREM1 expression. We showed that rs16969681 lies in a region of active chromatin with allele- and tissue-specific enhancer activity. The CRC high-risk allele was associated with stronger gene expression, and higher Grem1 mRNA levels increased the intestinal tumor burden in Apc(Min) mice. The intestine-specific transcription factor CDX2 and Wnt effector TCF7L2 bound near rs16969681, with significantly higher affinity for the risk allele, and CDX2 overexpression in CDX2/GREM1-negative cells caused re-expression of GREM1. rs16969681 influences CRC risk through effects on Wnt-driven GREM1 expression in colorectal tumors.


Asunto(s)
Neoplasias del Colon/genética , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Animales , Secuencia de Bases , Factor de Transcripción CDX2 , Línea Celular Tumoral , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Ratones Transgénicos , Especificidad de Órganos , Polimorfismo de Nucleótido Simple , Riesgo
9.
Cell ; 155(2): 410-22, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-24120139

RESUMEN

The ability of p53 to regulate transcription is crucial for tumor suppression and implies that inherited polymorphisms in functional p53-binding sites could influence cancer. Here, we identify a polymorphic p53 responsive element and demonstrate its influence on cancer risk using genome-wide data sets of cancer susceptibility loci, genetic variation, p53 occupancy, and p53-binding sites. We uncover a single-nucleotide polymorphism (SNP) in a functional p53-binding site and establish its influence on the ability of p53 to bind to and regulate transcription of the KITLG gene. The SNP resides in KITLG and associates with one of the largest risks identified among cancer genome-wide association studies. We establish that the SNP has undergone positive selection throughout evolution, signifying a selective benefit, but go on to show that similar SNPs are rare in the genome due to negative selection, indicating that polymorphisms in p53-binding sites are primarily detrimental to humans.


Asunto(s)
Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple , Elementos de Respuesta , Factor de Células Madre/genética , Neoplasias Testiculares/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proliferación Celular , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Selección Genética , Transcripción Genética
11.
Cold Spring Harb Perspect Biol ; 2(5): a001032, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20452958

RESUMEN

The p53 tumor suppressor pathway is central both in reducing cancer frequency in vertebrates and in mediating the response of commonly used cancer therapies. This article aims to summarize and discuss a large body of evidence suggesting that the p53 pathway harbors functional inherited single-nucleotide polymorphisms (SNPs) that affect p53 signaling in cells, resulting in differences in cancer risk and clinical outcome in humans. The insights gained through these studies into how the functional p53 pathway SNPs could help in the tailoring of cancer therapies to the individual are discussed. Moreover, recent work is discussed that suggests that many more functional p53 pathway SNPs are yet to be fully characterized and that a thorough analysis of the functional human genetics of this important tumor suppressor pathway is required.


Asunto(s)
Polimorfismo de Nucleótido Simple , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-mdm2/genética
12.
J Gastrointest Surg ; 13(7): 1351-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19415401

RESUMEN

BACKGROUND: Patients with Systemic Lupus Erythematosus (SLE) that present with acute abdominal pain (AAP) represent a challenge for the general surgeon. The purpose of this study was to identify the major causes of AAP among these patients and to define the role of disease activity scores and the APACHE II score in identifying patients with an increased perioperative risk. METHODS: We conducted a prospective study of patients admitted to the ER with AAP and SLE in an 11-year period. Demographic, diagnostic, and treatment data were recorded. Systemic lupus erythematosus disease activity index (SLEDAI), systemic lupus international collaboration clinics damage index (SLICC/DI), and APACHE II Score were analyzed. The main outcome variables were morbidity and mortality within 30 days of admission. RESULTS: Seventy-three patients were included. Ninety-three percent were female. Most common causes of AAP were: pancreatitis (29%), intestinal ischemia (16%), gallbladder disease (15%), and appendicitis (14%). Most causes of AAP in patients with LES were not related to the disease. APACHE II score > 12 was statistically associated with the diagnosis of intestinal ischemia compared to other causes. No relationship was observed between SLEDAI and outcome. Furthermore, this index did not have impact on diagnosis or decision making. Overall morbidity was 57% and overall mortality 11%. On multivariate analysis, only APACHE II > 12 was associated with mortality (P = 0.0001). CONCLUSION: This is one of the largest series of AAP and SLE. Most common causes of AAP were pancreatitis and intestinal ischemia. APACHE II score in patients with intestinal ischemia was higher than those with serositis; further studies are needed to examine whether this score may help to differentiate these etiologies when CT findings are inconclusive. APACHE II score was the most important factor associated with mortality. Furthermore, a prompt diagnosis and an appropriate surgical management are essential in order to improve patient outcome.


Asunto(s)
Dolor Abdominal/diagnóstico , Dolor Abdominal/epidemiología , Causas de Muerte , Lupus Eritematoso Sistémico/diagnóstico , Lupus Eritematoso Sistémico/epidemiología , APACHE , Dolor Abdominal/terapia , Enfermedad Aguda , Adolescente , Adulto , Distribución por Edad , Anciano , Análisis de Varianza , Estudios de Cohortes , Comorbilidad , Diagnóstico Precoz , Servicio de Urgencia en Hospital , Femenino , Humanos , Incidencia , Lupus Eritematoso Sistémico/terapia , Masculino , Persona de Mediana Edad , Análisis Multivariante , Probabilidad , Estudios Prospectivos , Índice de Severidad de la Enfermedad , Distribución por Sexo , Análisis de Supervivencia , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...