Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Pancreas ; 44(2): 181-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25479584

RESUMEN

OBJECTIVE: The aim of this study was to evaluate efficacy and safety of everolimus in patients with pancreatic neuroendocrine tumors (pNET) by prior chemotherapy use in the RAD001 in Advanced Neuroendocrine Tumors, Third Trial (RADIANT-3). METHODS: Patients with advanced, progressive, low- or intermediate-grade pNET were prospectively stratified by prior chemotherapy use and World Health Organization performance status and were randomly assigned (1:1) to everolimus 10 mg/d (n = 207) or placebo (n = 203). RESULTS: Of the 410 patients, 204 (50%) were naive to chemotherapy (chemonaive). Baseline characteristics were similar for patients with or without prior chemotherapy. Everolimus significantly prolonged median progression-free survival regardless of prior chemotherapy use (prior chemotherapy: 11.0 vs 3.2 months; hazard ratio, 0.34; 95% confidence interval, 0.25-0.48; P < 0.0001) (chemonaive: 11.4 vs 5.4 months; hazard ratio, 0.42; 95% confidence interval, 0.29-0.60; P < 0.0001). Stable disease was the best overall response in 73% of everolimus-treated patients (151/207). The most common drug-related adverse events included stomatitis (60%-69%), rash (47%-50%), and diarrhea (34%). CONCLUSIONS: As more treatment options become available, it is important to consider the goals of treatment and to identify patients who would potentially benefit from a specific therapy. Findings from this planned subgroup analysis suggest the potential for first-line use of everolimus in patients with advanced pNET.


Asunto(s)
Antineoplásicos/uso terapéutico , Everolimus/uso terapéutico , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Método Doble Ciego , Everolimus/efectos adversos , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/mortalidad , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Selección de Paciente , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos
2.
PLoS One ; 9(12): e114264, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25493562

RESUMEN

OBJECTIVE: The optimal sequencing of targeted therapies for metastatic renal cell carcinoma (mRCC) is unknown. Observational studies with a variety of designs have reported differing results. The objective of this study is to systematically summarize and interpret the published real-world evidence comparing sequential treatment for mRCC. METHODS: A search was conducted in Medline and Embase (2009-2013), and conference proceedings from American Society of Clinical Oncology (ASCO), ASCO Genitourinary Cancers Symposium (ASCO-GU), and European Society for Medical Oncology (ESMO) (2011-2013). We systematically reviewed observational studies comparing second-line mRCC treatment with mammalian target of rapamycin inhibitors (mTORi) versus vascular endothelial growth factor (VEGF) tyrosine kinase inhibitors (TKI). Studies were evaluated for 1) use of a retrospective cohort design after initiation of second-line therapy, 2) adjustment for patient characteristics, and 3) use of data from multiple centers. Meta-analyses were conducted for comparisons of overall survival (OS) and progression-free survival (PFS). RESULTS: Ten studies reported OS and exhibited significant heterogeneity in estimated second-line treatment effects (I2 = 68%; P = 0.001). Four of these were adjusted, multicenter, retrospective cohort studies, and these showed no evidence of heterogeneity (I2 = 0%; P = 0.61) and a significant association between second-line mTORi (>75% everolimus) and longer OS compared to VEGF TKI (>60% sorafenib) (HR = 0.82, 95% CI: 0.68 to 0.98) in a meta-analysis. Seven studies comparing PFS showed significant heterogeneity overall and among the adjusted, multicenter, retrospective cohort studies. Real-world observational data for axitinib outcomes was limited at the time of this study. CONCLUSIONS: Real-world studies employed different designs and reported heterogeneous results comparing the effectiveness of second-line mTORi and VEGF TKI in the treatment of mRCC. Within the subset of adjusted, multicenter observational studies, second-line use of mTORi was associated with significantly prolonged survival compared with second-line use of VEGF TKI.


Asunto(s)
Carcinoma de Células Renales/terapia , Neoplasias Renales/terapia , Carcinoma de Células Renales/patología , Humanos , Neoplasias Renales/patología , Metástasis de la Neoplasia , Análisis de Supervivencia
3.
Eur Urol ; 64(6): 994-1002, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23219086

RESUMEN

BACKGROUND: The phase 3 RECORD-1 study demonstrated clinical benefit of everolimus over placebo (median progression-free survival: 4.9 mo compared with 1.9 mo, p<0.001) in treatment-resistant patients with metastatic renal cell carcinoma (mRCC). However, the Response Evaluation Criteria in Solid Tumors (RECIST) objective response rate was low. OBJECTIVE: To explore the potential role of tumor burden response to everolimus in predicting patient survival. DESIGN, SETTING, AND PARTICIPANTS: RECORD-1 patients with at least two tumor assessments (baseline and weeks 2-14) were included (n=246). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: A multivariate Cox proportional hazard model was used to assess the impact of various prognostic factors on overall survival (OS). Components of RECIST progression were explored using univariate Cox regression. RESULTS AND LIMITATIONS: The baseline sum of longest tumor diameters (SLD) and progression at weeks 2-14 were prognostic factors of OS by multivariate analysis. Univariate analysis at weeks 2-14 demonstrated that growth of nontarget lesions and appearance of new lesions were predictive of OS (p<0.001). This retrospective analysis used data from one arm of one trial; patients in the placebo arm were excluded because of confounding effects when they crossed over to everolimus. CONCLUSIONS: This analysis identified baseline SLD as a predictive factor of OS, and the appearance of a new lesion or progression of a nontarget lesion at first assessment after baseline also affects OS in patients with mRCC treated with everolimus.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/mortalidad , Inmunosupresores/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/mortalidad , Sirolimus/análogos & derivados , Carga Tumoral/efectos de los fármacos , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/secundario , Everolimus , Femenino , Humanos , Neoplasias Renales/patología , Masculino , Metástasis de la Neoplasia , Pronóstico , Estudios Retrospectivos , Sirolimus/uso terapéutico , Tasa de Supervivencia
4.
J Clin Endocrinol Metab ; 94(6): 2085-91, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19293268

RESUMEN

BACKGROUND: The "complex of myxomas, spotty skin pigmentation, and endocrine overactivity," or "Carney complex" (CNC), is caused by inactivating mutations of the regulatory subunit type 1A of the cAMP-dependent protein kinase (PRKAR1A) gene and as yet unknown defect(s) in other gene(s). Delineation of a genotype-phenotype correlation for CNC patients is essential for understanding PRKAR1A function and providing counseling and preventive care. METHODS: A transatlantic consortium studied the molecular genotype and clinical phenotype of 353 patients (221 females and 132 males, age 34 +/- 19 yr) who carried a germline PRKAR1A mutation or were diagnosed with CNC and/or primary pigmented nodular adrenocortical disease. RESULTS: A total of 258 patients (73%) carried 80 different PRKAR1A mutations; 114 (62%) of the index cases had a PRKAR1A mutation. Most PRKAR1A mutations (82%) led to lack of detectable mutant protein (nonexpressed mutations) because of nonsense mRNA mediated decay. Patients with a PRKAR1A mutation were more likely to have pigmented skin lesions, myxomas, and thyroid and gonadal tumors; they also presented earlier with these tumors. Primary pigmented nodular adrenocortical disease occurred earlier, was more frequent in females, and was the only manifestation of CNC with a gender predilection. Mutations located in exons were more often associated with acromegaly, myxomas, lentigines, and schwannomas, whereas the frequent c.491-492delTG mutation was commonly associated with lentigines, cardiac myxomas, and thyroid tumors. Overall, nonexpressed PRKAR1A mutations were associated with less severe disease. CONCLUSION: CNC is genetically and clinically heterogeneous. Certain tumors are more frequent, with specific mutations providing some genotype-phenotype correlation for PRKAR1A mutations.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Mutación , Adolescente , Enfermedades de la Corteza Suprarrenal/complicaciones , Enfermedades de la Corteza Suprarrenal/genética , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación/fisiología , Mixoma/complicaciones , Mixoma/genética , Fenotipo , Adulto Joven
5.
Eur J Hum Genet ; 16(1): 79-88, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17667967

RESUMEN

Gastrointestinal stromal tumors (GISTs) may be caused by germline mutations of the KIT and platelet-derived growth factor receptor-alpha (PDGFRA) genes and treated by Imatinib mesylate (STI571) or other protein tyrosine kinase inhibitors. However, not all GISTs harbor these genetic defects and several do not respond to STI571 suggesting that other molecular mechanisms may be implicated in GIST pathogenesis. In a subset of patients with GISTs, the lesions are associated with paragangliomas; the condition is familial and transmitted as an autosomal-dominant trait. We investigated 11 patients with the dyad of 'paraganglioma and gastric stromal sarcoma'; in eight (from seven unrelated families), the GISTs were caused by germline mutations of the genes encoding subunits B, C, or D (the SDHB, SDHC and SDHD genes, respectively). In this report, we present the molecular effects of these mutations on these genes and the clinical information on the patients. We conclude that succinate dehydrogenase deficiency may be the cause of a subgroup of GISTs and this offers a therapeutic target for GISTs that may not respond to STI571 and its analogs.


Asunto(s)
Tumores del Estroma Gastrointestinal/enzimología , Tumores del Estroma Gastrointestinal/genética , Mutación de Línea Germinal , Proteínas Hierro-Azufre/genética , Proteínas de la Membrana/genética , Síndromes Neoplásicos Hereditarios/enzimología , Síndromes Neoplásicos Hereditarios/genética , Paraganglioma/enzimología , Paraganglioma/genética , Succinato Deshidrogenasa/genética , Adolescente , Adulto , Alelos , Antineoplásicos/uso terapéutico , Secuencia de Bases , Benzamidas , Niño , Cartilla de ADN/genética , ADN de Neoplasias/genética , Femenino , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/patología , Genes Dominantes , Heterocigoto , Humanos , Mesilato de Imatinib , Pérdida de Heterocigocidad , Masculino , Síndromes Neoplásicos Hereditarios/tratamiento farmacológico , Síndromes Neoplásicos Hereditarios/patología , Paraganglioma/tratamiento farmacológico , Paraganglioma/patología , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico
6.
Am J Surg Pathol ; 31(11): 1764-75, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18059235

RESUMEN

Pigmented epithelioid melanocytoma (PEM) is a recently described entity comprising most cases previously described as "animal-type melanoma" and epithelioid blue nevus (EBN) occurring in patients with the multiple neoplasia syndrome Carney complex (CNC). Mutations of the protein kinase A regulatory subunit type 1alpha (R1alpha) (coded by the PRKAR1A gene) are found in more than half of CNC patients. In this study, we investigated whether PEM and EBN are related at the molecular level, and whether changes in the PRKAR1A gene status and the expression of the R1alpha protein may be involved in the pathogenesis of PEM and other melanocytic lesions. Histologic analysis of hematoxylin and eosin-stained sections and immunohistochemistry (IHC) with R1alpha antibody were performed on 34 sporadic PEMs, 8 CNC-associated PEMs from patients with known PRKAR1A mutations, 297 benign and malignant melanocytic tumors (127 conventional sections of 10 compound nevi, 10 Spitz nevi, 5 deep-penetrating nevi, 5 blue nevi, 6 cellular blue nevi, 2 malignant blue nevi, 3 lentigo maligna, and 86 melanomas of various types); in addition, 170 tissue microarray sections consisting of 35 benign nevi, 60 primary melanomas, and 75 metastatic melanomas, and 5 equine dermal melanomas, were examined. Histologic diagnoses were based on preexisting pathologic reports and were confirmed for this study. DNA studies [loss of heterozygosity (LOH) for the 17q22-24 locus and the PRKAR1A gene sequencing] were performed on 60 melanomas and 7 PEMs. IHC showed that R1alpha was expressed in all but one core from tissue microarrays (169/170), and in all 127 melanocytic lesions evaluated in conventional sections. By contrast, R1alpha was not expressed in the 8 EBN from patients with CNC and PRKAR1A mutations. Expression of R1alpha was lost in 28 of 34 PEMs (82%). R1alpha was expressed in the 5 equine melanomas studied. DNA studies correlated with IHC findings: there were no PRKAR1A mutations in any of the melanomas studied and the rate of LOH for 17q22-24 was less than 7%; 5 of the 7 PEMs showed extensive 17q22-24 LOH but no PRKAR1A mutations. The results support the concept that PEM is a distinct melanocytic tumor occurring in a sporadic setting and in the context of CNC. They also suggest that PEM differs from melanomas in equine melanotic disease, further arguing that the term animal-type melanoma may be a misnomer for this group of lesions. Loss of expression of R1alpha offers a useful diagnostic test that helps to distinguish PEM from lesions that mimic it histologically.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/análisis , Enfermedades de los Caballos/enzimología , Melanocitos/enzimología , Melanoma/enzimología , Neoplasias Primarias Múltiples/enzimología , Nevo Azul/enzimología , Nevo de Células Epitelioides y Fusiformes/enzimología , Neoplasias Cutáneas/enzimología , Animales , Cromosomas Humanos Par 17 , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Diagnóstico Diferencial , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Enfermedades de los Caballos/genética , Enfermedades de los Caballos/patología , Caballos , Humanos , Inmunohistoquímica , Pérdida de Heterocigocidad , Melanocitos/patología , Melanoma/patología , Melanoma/veterinaria , Mutación , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Primarias Múltiples/genética , Neoplasias Primarias Múltiples/patología , Nevo Azul/clasificación , Nevo Azul/genética , Nevo Azul/patología , Nevo de Células Epitelioides y Fusiformes/clasificación , Nevo de Células Epitelioides y Fusiformes/genética , Nevo de Células Epitelioides y Fusiformes/patología , Neoplasias Cutáneas/clasificación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/veterinaria , Terminología como Asunto , Análisis de Matrices Tisulares
7.
J Clin Endocrinol Metab ; 92(8): 2938-43, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17535989

RESUMEN

CONTEXT: Carney triad (CT) describes the association of paragangliomas (PGLs) with gastrointestinal stromal tumors (GISTs) and pulmonary chondromas. Inactivating mutations of the mitochondrial complex II succinate dehydrogenase (SDH) enzyme subunits SDHB, SDHC, and SDHD are found in PGLs, gain-of-function mutations of c-kit (KIT), and platelet-derived growth factor receptor A (PDGFRA) in GISTs. OBJECTIVE: Our objective was to investigate the possibility that patients with CT and/or their tumors may harbor mutations of the SDHB, SDHC, SDHD, KIT, and PDGFRA genes and identify any other genetic alterations in CT tumors. DESIGN: Three males and 34 females with CT were studied retrospectively. We sequenced the stated genes and performed comparative genomic hybridization on a total of 41 tumors. RESULTS: No patient had coding sequence mutations of the investigated genes. Comparative genomic hybridization revealed a number of DNA copy number changes: losses dominated among benign lesions, there were an equal number of gains and losses in malignant lesions, and the average number of alterations in malignant tumors was higher compared with benign lesions. The most frequent and greatest contiguous change was 1q12-q21 deletion, a region that harbors the SDHC gene. Another frequent change was loss of 1p. Allelic losses of 1p and 1q were confirmed by fluorescent in situ hybridization and loss-of-heterozygosity studies. CONCLUSIONS: We conclude that CT is not due to SDH-inactivating or KIT- and PDGFRA-activating mutations. GISTs and PGLs in CT are associated with chromosome 1 and other changes that appear to participate in tumor progression and point to their common genetic cause.


Asunto(s)
Cromosomas Humanos Par 1/genética , Neoplasias Gastrointestinales/genética , Tumores del Estroma Gastrointestinal/genética , Mutación de Línea Germinal/genética , Neoplasias Pulmonares/genética , Paraganglioma/genética , Adolescente , Adulto , Niño , ADN de Neoplasias/genética , Femenino , Dosificación de Gen , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Pérdida de Heterocigocidad , Masculino , Hibridación de Ácido Nucleico , Estudios Retrospectivos , Succinato Deshidrogenasa/genética , Células Tumorales Cultivadas
8.
Clin Endocrinol (Oxf) ; 67(2): 168-74, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17466001

RESUMEN

OBJECTIVE: ACTH resistance is a feature of several human syndromes with known genetic causes, including familial glucocorticoid deficiency (types 1 and 2) and triple A syndrome. However, many patients with ACTH resistance lack an identifiable genetic aetiology. The human homolog of the Acd gene, mutated in a mouse model of adrenal insufficiency, was sequenced in 25 patients with a clinical diagnosis of familial glucocorticoid deficiency or triple A syndrome. DESIGN: A 3.4 kilobase genomic fragment containing the entire ACD gene was analysed for mutations in all 25 patients. SETTING: Samples were obtained by three investigators from different institutions. PATIENTS: The primary cohort consisted of 25 unrelated patients, primarily of European or Middle Eastern descent, with a clinical diagnosis of either familial glucocorticoid deficiency (FGD) or triple A syndrome. Patients lacked mutations in other genes known to cause ACTH resistance, including AAAS for patients diagnosed with triple A syndrome and MC2R and MRAP for patients diagnosed with familial glucocorticoid deficiency. Thirty-five additional patients with adrenal disease phenotypes were added to form an expanded cohort of 60 patients. MEASUREMENTS: Identification of DNA sequence changes in the ACD gene in the primary cohort and analysis of putative ACD haplotypes in the expanded cohort. RESULTS: No disease-causing mutations were found, but several novel single nucleotide polymorphisms (SNPs) and two putative haplotypes were identified. The overall frequency of SNPs in ACD is low compared to other gene families. CONCLUSIONS: No mutations were identified in ACD in this collection of patients with ACTH resistance phenotypes. However, the newly identified SNPs in ACD should be more closely examined for possible links to disease.


Asunto(s)
Hormona Adrenocorticotrópica/metabolismo , Errores Innatos del Metabolismo/genética , Polimorfismo Genético , Proteínas de Unión a Telómeros/genética , Insuficiencia Suprarrenal/genética , Secuencia de Bases , Preescolar , Análisis Mutacional de ADN , Acalasia del Esófago/genética , Femenino , Glucocorticoides/deficiencia , Haplotipos , Humanos , Lactante , Recién Nacido , Masculino , Errores Innatos del Metabolismo/metabolismo , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Complejo Shelterina , Síndrome
9.
J Clin Endocrinol Metab ; 91(9): 3626-32, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16772351

RESUMEN

CONTEXT: Primary adrenocortical hyperplasias leading to Cushing syndrome include primary pigmented nodular adrenocortical disease and ACTH-independent macronodular adrenal hyperplasia (AIMAH). Inactivating mutations of the 17q22-24-located PRKAR1A gene, coding for the type 1A regulatory subunit of protein kinase A (PKA), cause primary pigmented nodular adrenocortical disease and the multiple endocrine neoplasia syndrome Carney complex. PRKAR1A mutations and 17q22-24 chromosomal losses have been found in sporadic adrenal tumors and are associated with aberrant PKA signaling. OBJECTIVE: The objective of the study was to examine whether somatic 17q22-24 changes, PRKAR1A mutations, and/or PKA abnormalities are present in AIMAH. PATIENTS: We studied fourteen patients with Cushing syndrome due to AIMAH. METHODS: Fluorescent in situ hybridization with a PRKAR1A-specific probe was used for investigating chromosome 17 allelic losses. The PRKAR1A gene was sequenced in all samples, and tissue was studied for PKA activity, cAMP responsiveness, and PKA subunit expression. RESULTS: We found 17q22-24 allelic losses in 73% of the samples. There were no PRKAR1A-coding sequence mutations. The RIIbeta PKA subunit was overexpressed by mRNA, whereas the RIalpha, RIbeta, RIIalpha, and Calpha PKA subunits were underexpressed. These findings were confirmed by immunohistochemistry. Total PKA activity and free PKA activity were higher in AIMAH than normal adrenal glands, consistent with the up-regulation of the RIIbeta PKA subunit. CONCLUSIONS: PRKAR1A mutations are not found in AIMAH. Somatic losses of the 17q22-24 region and PKA subunit and enzymatic activity changes show that PKA signaling is altered in AIMAH in a way that is similar to that of other adrenal tumors with 17q losses or PRKAR1A mutations.


Asunto(s)
Cromosomas Humanos Par 17/genética , Síndrome de Cushing/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/patología , Alelos , Síndrome de Cushing/enzimología , AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Hiperplasia , Inmunohistoquímica , Hibridación Fluorescente in Situ , Mutación , ARN/química , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Clin Endocrinol Metab ; 91(6): 2380-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16569736

RESUMEN

CONTEXT: Primary pigmented nodular adrenocortical disease, associated with Carney complex, is caused by mutations in PRKAR1A (mt-PRKAR1A), a gene that codes for the regulatory subunit type 1alpha (RIalpha) of cAMP-dependent protein kinase (PKA). PRKAR1A inactivation is associated with dysregulated PKA activity that is thought to result in tumorigenesis. mt-PRKAR1A-bearing lymphocytes from Carney complex patients exhibit enhanced cell proliferation associated with increased expression of the MAPK ERK1/2 pathway. OBJECTIVE: The objective of the study was to determine how PKA and its subunits and ERK1/2 and their molecular partners change in the presence of PRKAR1A mutations in adrenocortical tissue. DESIGN: PKA activity and subunit expression, ERK1/2, other immunoassays, and immunohistochemistry on adrenocortical samples from patients with germline normal or mt-PRKAR1A were analyzed. RESULTS: Increased cAMP-stimulated total kinase activity was associated with mt-PRKAR1A. PKA subunit expression analysis in mt-PRKAR1A tissues, by quantitative mRNA assay and immunoblotting, showed a 2.4-fold (P = 0.02) and 1.8-fold (P = 0.09) decrease in RIalpha's message and protein, respectively, and increases in other PKA subunits. Immunoassays showed 2-fold (P = 0.03) and 6-fold (P = 0.03) decreases in baseline ERK1/2, with corresponding increases in phosphorylated (p) ERK1/2 in mt-PRKAR1A samples. B-raf kinase, p-MEK1/2, and p-c-Myc, but not p-Akt/protein kinase B, were significantly increased. Immunohistochemistry studies supported these data. CONCLUSIONS: mt-PRKAR1A causes increased total cAMP-stimulated kinase activity, likely the result of up-regulation of other PKA subunits caused by down-regulation of RIalpha, as seen in human lymphocytes and mouse animal models. These changes, associated with enhanced MAPK activity, may be, in part, responsible for the proliferative signals that result in primary pigmented nodular adrenocortical disease.


Asunto(s)
Corteza Suprarrenal/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Mutación , Transducción de Señal/fisiología , Adulto , Niño , Preescolar , AMP Cíclico/fisiología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/análisis , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Inmunohistoquímica , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Fosforilación , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Mensajero/análisis
11.
Am J Surg Pathol ; 30(1): 42-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16330941

RESUMEN

Functioning paraganglioma and gastrointestinal stromal tumor (GIST) are uncommon tumors that occur mostly in a sporadic and isolated form, occasionally as components of multiple neoplasia syndromes, either separately or together. Separately, they occur in several inherited syndromes including multiple endocrine neoplasia 2, and the GIST, lentigines, and mast cell tumor syndrome. Together, they are variably prominent components of three syndromes: the familial paraganglioma and gastric GIST syndrome, neurofibromatosis type 1, and the Carney triad. The two former conditions are inherited as autosomal dominant traits; the latter does not appear to be inherited and affects young women predominantly. This article reports the nonfamilial occurrence of functioning paraganglioma and GIST of the jejunum in 3 women, 1 young (22 years) at initial presentation. The occurrences were unexpected because of the infrequency of the tumors. The neoplasms, respectively, did not show germline SDHA, SDHB, SDHC, and SDHD, and KIT mutations associated with familial paraganglioma and familial GIST. The paraganglioma-jejunal GIST combination may be the harbinger of a rare genetic syndrome, a variant of the Carney triad or the paraganglioma-gastric stromal sarcoma syndrome, or be coincidental.


Asunto(s)
Tumores del Estroma Gastrointestinal/patología , Neoplasias del Yeyuno/patología , Neoplasias del Yeyuno/fisiopatología , Neoplasias Primarias Múltiples/patología , Paraganglioma/patología , Adulto , Anciano , Femenino , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/fisiopatología , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Neoplasias del Yeyuno/genética , Persona de Mediana Edad , Neoplasias Primarias Múltiples/genética , Neoplasias Primarias Múltiples/fisiopatología , Paraganglioma/genética , Paraganglioma/fisiopatología , Reacción en Cadena de la Polimerasa , Embarazo
12.
Genes Chromosomes Cancer ; 44(2): 204-11, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16001434

RESUMEN

Odontogenic myxomas are rare benign neoplasms affecting the jaw. Myxomas of bones and other sites occur as part of Carney complex (CNC), a multiple neoplasia syndrome caused by mutations in the PRKAR1A gene, which codes for the regulatory subunit of protein kinase A (PKA). In the present study, 17 odontogenic myxomas from patients without CNC were screened for PRKAR1A mutations and PRKAR1A protein expression by immunohistochemistry (IHC). Mutations of the coding region of the PRKAR1A gene were identified in 2 tumors; both these lesions showed no or significantly decreased immunostaining of PRKAR1A in the tumor compared to that in the surrounding normal tissue. One mutation (c.725C>A) led to a nonconservative amino acid substitution in a highly conserved area of the gene (A213D); the other was a single base-pair deletion that led to a frameshift (del774C) and a stop codon 11 amino acids downstream of the mutation site; both tumors were heterozygous for the respective mutations. Of the remaining tumors, 7 of the 15 without mutations showed almost no PRKAR1A in the tumor cells, whereas IHC showed that the protein was abundant in nontumorous cells. We concluded that PRKAR1A may be involved by its down-regulation in the pathogenesis of odontogenic myxomas caused by mutations and/or other genetic mechanisms. Of the sporadic, nonfamilial tumors associated with PRKAR1A mutations, the odontogenic type was the first myxomatous lesion found to harbor somatic PRKAR1A sequence changes.


Asunto(s)
Mixoma/genética , Tumores Odontogénicos/genética , Proteínas/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico , ADN de Neoplasias/genética , Humanos , Inmunohistoquímica , Mutación , Mixoma/diagnóstico , Mixoma/metabolismo , Tumores Odontogénicos/diagnóstico , Tumores Odontogénicos/metabolismo , Proteínas/metabolismo
13.
J Clin Endocrinol Metab ; 90(6): 3773-9, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15741255

RESUMEN

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is an autosomal dominant disorder caused by mutations in the fumarate hydratase (FH) gene on chromosome 1q42.3-43. Massive macronodular adrenocortical disease (MMAD) is a heterogeneous condition associated with Cushing syndrome (CS) and bilateral hyperplasia of the adrenal glands. In MMAD, cortisol secretion is often mediated by ectopic, adrenocortical expression of receptors for a variety of substances; however, to date, no consistent genetic defects have been identified. In a patient with HLRCC caused by a germline-inactivating FH mutation, we diagnosed atypical (subclinical) CS due to bilateral, ACTH-independent adrenocortical hyperplasia. A clinical protocol for the detection of ectopic expression of various hormone receptors was employed. Histology was consistent with MMAD. The tumor tissue harbored the germline FH mutation and demonstrated allelic losses of the 1q42.3-43 FH locus. We then searched the National Institutes of Health (NIH) databases of patients with MMAD or HLRCC and found at least three other cases with MMAD that had a history of tumors that could be part of HLRCC; among patients with HLRCC, there were several with some adrenal nodularity noted on computed tomography but none with imaging findings consistent with MMAD. From two of the three MMAD patients, adrenocortical tumor DNA was available and sequenced for coding FH mutations; there were none. We conclude that in a patient with HLRCC, adrenal hyperplasia and CS were due to MMAD. The latter was likely due to the FH germline mutation because in tumor cells, only the mutant allele was retained. However, other patients with MMAD and HLRCC, or HLRCC patients with adrenal imaging findings consistent with MMAD, or MMAD patients with somatic FH mutations were not found among the NIH series. Although a fortuitous association cannot be excluded, HLRCC may be added to the short list of monogenic disorders that have been reported to be associated with the development of adrenal tumors; FH may be considered a candidate gene for MMAD.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Síndrome de Cushing/genética , Leiomiomatosis/genética , Neoplasias de la Corteza Suprarrenal/complicaciones , Neoplasias de la Corteza Suprarrenal/patología , Glándulas Suprarrenales/patología , Anciano , Mapeo Cromosómico , Cromosomas Humanos Par 1 , Síndrome de Cushing/complicaciones , ADN/sangre , ADN/genética , ADN/aislamiento & purificación , Femenino , Lateralidad Funcional , Humanos , Hibridación Fluorescente in Situ , Leiomiomatosis/cirugía , Piel/patología , Resultado del Tratamiento
14.
Cancer Res ; 64(24): 8811-5, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15604237

RESUMEN

Mutations of the human type Ialpha regulatory subunit (RIalpha) of cyclic AMP-dependent protein kinase (PKA; PRKAR1A) lead to altered kinase activity, primary pigmented nodular adrenocortical disease, and tumors of the thyroid and other tissues. To bypass the early embryonic lethality of Prkar1a(-/-) mice, we established transgenic mice carrying an antisense transgene for Prkar1a exon 2 (X2AS) under the control of a tetracycline-responsive promoter. Down-regulation of Prkar1a by up to 70% was achieved in transgenic mouse tissues and embryonic fibroblasts, with concomitant changes in kinase activity and increased cell proliferation, respectively. Mice developed thyroid follicular hyperplasia and adenomas, adrenocortical hyperplasia, and other features reminiscent of primary pigmented nodular adrenocortical disease, histiocytic and epithelial hyperplasias, lymphomas, and other mesenchymal tumors. These were associated with allelic losses of the mouse chromosome 11 Prkar1a locus, an increase in total type II PKA activity, and higher RIIbeta protein levels. This mouse provides a novel, useful tool for the investigation of cyclic AMP, RIalpha, and PKA functions and confirms the critical role of Prkar1a in tumorigenesis in endocrine and other tissues.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/enzimología , Trastornos Linfoproliferativos/enzimología , Proteínas/fisiología , Neoplasias de la Tiroides/enzimología , Adenoma/enzimología , Adenoma/genética , Neoplasias de la Corteza Suprarrenal/genética , Animales , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , ADN sin Sentido/genética , Regulación hacia Abajo , Exones , Femenino , Trastornos Linfoproliferativos/genética , Mesodermo/enzimología , Mesodermo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Regiones Promotoras Genéticas , Proteínas/genética , Tetraciclina/farmacología , Neoplasias de la Tiroides/genética , Transactivadores/genética
15.
Front Horm Res ; 32: 253-64, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15281351

RESUMEN

Carney complex (CNC) is a familial multiple neoplasia and lentiginosis syndrome with features overlapping those of McCune-Albright syndrome (MAS) and other multiple endocrine neoplasia (MEN) syndromes like MEN type 1 (MEN 1). Pituitary tumors have been described in a number of patients with CNC; all have been growth hormone (GH) and prolactin (PRL)-producing. In at least some patients, pituitary gland involvement is manifested by hyperplastic areas; hyperplasia appears to involve somatomammotrophs only and to precede GH-producing tumor formation, in a pathway similar to that seen in MAS-related pituitary tumors (and in oncogenesis in other CNC tissues). One patient with CNC and advanced acromegaly had a GH-producing macroadenoma that showed extensive genetic changes at the chromosomal level. These changes appeared to represent secondary or tertiary genetic 'hits' involved in pituitary oncogenesis and were confirmed at the molecular level. So far, almost half of the patients with CNC have germline-inactivating mutations in the PRKAR1A gene; in their pituitary tumors, the normal allele of the PRKAR1A gene is lost. Loss of heterozygosity suggests that PRKAR1A, which codes for the regulatory subunit type 1alpha of the cAMP-dependent protein kinase A (PKA), may act as a tumor-suppressor gene in pituitary tissue. These data provide evidence for a PKA-induced somatomammotroph hyperplasia in the pituitary tissue of CNC patients; hyperplasia leads to additional genetic changes at the somatic level, which in turn cause the formation of adenomas in some, but not all, patients.


Asunto(s)
Enfermedades del Sistema Endocrino/patología , Mutación de Línea Germinal , Mixoma/patología , Neurilemoma/patología , Trastornos de la Pigmentación/patología , Proteínas/genética , Pigmentación de la Piel , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico , Enfermedades del Sistema Endocrino/genética , Humanos , Lentigo/genética , Lentigo/patología , Neoplasia Endocrina Múltiple/genética , Neoplasia Endocrina Múltiple/patología , Mixoma/genética , Neurilemoma/genética , Trastornos de la Pigmentación/genética , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Síndrome
16.
Endocr Res ; 30(4): 903-11, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15666843

RESUMEN

Mice with complete inactivation of the type Ialpha regulatory subunit (RIalpha) of cyclic (c) AMP-dependent protein kinase (PKA) (coded by the Prkar1a gene) die early in embryonic life. To bypass the early embryonic lethality of Prkar1a-/- mice, we established transgenic mice carrying an antisense transgene for Prkar1a exon 2 (X2AS) under the control of a tetracycline-responsive promoter. Mice developed thyroid follicular hyperplasia and adenomas, adrenocortical hyperplasia, and other features reminiscent of PPNAD, and histiocytic and epithelial hyperplasias, lymphomas, and other mesenchymal tumors. This mouse provides a useful tool for the investigation of cAMP, RIalpha, and PKA functions and confirms Prkar1a's critical role in tumorigenesis in endocrine and other tissues.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Transgénicos , Neoplasia Endocrina Múltiple/genética , Proteínas/genética , Animales , AMP Cíclico/farmacología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Ratones , Neoplasia Endocrina Múltiple/metabolismo , Neoplasia Endocrina Múltiple/patología , ARN Mensajero/metabolismo , Distribución Tisular
17.
Pituitary ; 7(2): 73-82, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15761655

RESUMEN

Carney complex (CNC) is a familial multiple neoplasia syndrome with features overlapping those of McCune-Albright syndrome (MAS) and multiple endocrine neoplasia (MEN) type 1 (MEN 1). Like MAS and MEN 1 patients, patients with CNC develop growth hormone (GH)-producing pituitary tumors. Occasionally, these tumors are also prolactin-producing, but there are no isolated prolactinomas or other types of pituitary tumors. In at least some patients with CNC, the pituitary gland is characterized by hyperplastic areas; hyperplasia appears to involve somatomammotrophs only. Hyperplasia most likely precedes the formation of GH-producing adenomas in CNC, as has been suggested in MAS-related somatotropinomas, but has never been seen in MEN 1 patients. In at least one case of a patient with CNC and advanced acromegaly, a GH-producing macroadenoma showed extensive genetic changes at the chromosomal level. So far, half of the patients with CNC have germline inactivating mutations in the PRKAR1A gene; in their pituitary tumors, the normal allele of the PRKAR1A gene is lost. Loss-of-hererozygosity suggests that PRKAR1A, which codes for the regulatory subunit type 1alpha of the cAMP-dependent protein kinase A (PKA) may act as a tumor-suppressor gene in CNC somatomammotrophs. These data provide evidence for a PRKAR1A-induced somatomammotroph hyperpasia in the pituitary tissue of CNC patients; hyperplasia, in turn may lead to additional genetic changes at the somatic level, which then cause the formation of adenomas in some, but not all, patients.


Asunto(s)
Displasia Fibrosa Poliostótica/patología , Neoplasia Endocrina Múltiple Tipo 1/patología , Hipófisis/patología , Neoplasias Hipofisarias/patología , Acromegalia/patología , Adulto , Femenino , Hormona de Crecimiento Humana/metabolismo , Humanos , Hiperplasia , Masculino , Neoplasia Endocrina Múltiple Tipo 1/ultraestructura , Hipófisis/ultraestructura , Neoplasias Hipofisarias/ultraestructura
18.
Cancer Res ; 63(17): 5308-19, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500362

RESUMEN

Germ-line protein kinase A (PKA) regulatory-subunit type-Ialpha (RIalpha; PRKAR1A)-inactivating mutations and loss-of-heterozygosity (LOH) of its 17q22-24 locus have been found in Cushing syndrome (CS) caused by primary pigmented nodular adrenocortical disease (PPNAD). We examined whether somatic 17q22-24, PRKAR1A, or PKA changes are present in 44 sporadic adrenocortical tumors (29 adenomas and 15 cancers); 26 of these tumors were responsible for CS. A probe containing the PRKAR1A gene-mapped by fluorescent in situ hybridization to 17q22-24-and corresponding microsatellite markers were used to study allelic losses; PRKAR1A was sequenced in all samples. 17q22-24 losses were seen in 23 and 53% of adenomas and cancers, respectively. In three tumors, somatic, PRKAR1A-inactivating mutations were identified: (a) a nonsense mutation in exon 6 (A751G); (b) a splicing mutation (9IVS-1G/A); and (c) a transition (1050T>C) followed by a 22-bp deletion, also in exon 9; all predicted premature RIalpha protein terminations. Quantitative message and protein studies showed RIalpha down-regulation in tumors with genetic changes; their cortisol secretion pattern was similar to that of PPNAD, and they had higher PKA activity by enzymatic studies. We conclude that somatic allelic losses of the 17q22-24 region, PRKAR1A-inactivating mutations or down-regulation, and corresponding PKA activity changes are present in at least some sporadic adrenocortical tumors, especially those with a PPNAD-like clinical presentation of CS.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Adenoma Corticosuprarrenal/genética , Cromosomas Humanos Par 17/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Neoplasias de la Corteza Suprarrenal/enzimología , Adenoma Corticosuprarrenal/enzimología , Adulto , Anciano , Alelos , Western Blotting , Mapeo Cromosómico , Síndrome de Cushing/complicaciones , Síndrome de Cushing/enzimología , Síndrome de Cushing/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Humanos , Inmunohistoquímica , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Mutación , ARN Mensajero/biosíntesis , ARN Mensajero/genética
19.
FEBS Lett ; 546(1): 59-64, 2003 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-12829237

RESUMEN

Carney complex (CNC) is a multiple neoplasia syndrome that consists of endocrine (thyroid, pituitary, adrenocortical and gonadal), non-endocrine (myxomas, nevi and other cutaneous pigmented lesions), and neural (schwannomas) tumors. Primary pigmented nodular adrenocortical disease (PPNAD) is the most common endocrine manifestation of CNC and the only inherited form of Cushing syndrome known to date. In the search of genes responsible for CNC, two chromosomal loci were identified; one (17q22-24) harbored the gene encoding the type I-alpha regulatory subunit (RIalpha) of protein kinase A (PKA), PRKAR1A, a critical component of the cAMP signaling pathway. Here we review CNC and the implications of this discovery for the cAMP and/or PKA's involvement in human tumorigenesis.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/genética , Neoplasia Endocrina Múltiple/enzimología , Neoplasia Endocrina Múltiple/genética , Mutación/genética , Cromosomas Humanos Par 17 , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Genes Supresores de Tumor , Heterogeneidad Genética , Humanos , Modelos Biológicos , Neoplasia Endocrina Múltiple/diagnóstico , Síndrome
20.
IEEE Trans Neural Syst Rehabil Eng ; 11(4): 416-21, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14960118

RESUMEN

We present a device that allows three-dimensional (3-D) space perception by sonification of range information obtained via a point laser range sensor. The laser range sensor is worn by a blindfolded user, who scans space by pointing the laser beam in different directions. The resulting stream of range measurements is then converted to an auditory signal whose frequency or amplitude varies with the range. Our device differs from existing navigation aids for the visually impaired. Such devices use sonar ranging whose primary purpose is to detect obstacles for navigation, a task to which sonar is well suited due to its wide beam width. In contrast, the purpose of our device is to allow users to perceive the details of 3-D space that surrounds them, a task to which sonar is ill suited, due to artifacts generated by multiple reflections and due to its limited range. Preliminary trials demonstrate that the user is able to easily and accurately detect corners and depth discontinuities and to perceive the size of the surrounding space.


Asunto(s)
Acústica/instrumentación , Análisis de Falla de Equipo/métodos , Auxiliares Sensoriales , Percepción Espacial , Interfaz Usuario-Computador , Trastornos de la Visión/rehabilitación , Adolescente , Percepción de Distancia , Diseño de Equipo , Femenino , Humanos , Terapia por Láser , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad , Procesamiento de Señales Asistido por Computador , Localización de Sonidos , Análisis y Desempeño de Tareas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...