Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Clin Chem ; 66(1): 169-177, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31672856

RESUMO

BACKGROUND: Blood molecular profiling of circulating tumor cells (CTCs) can enable monitoring of patients with metastatic melanoma during checkpoint inhibitor immunotherapy (CII) and in combination with targeted therapies. We developed a microfluidics-based CTC platform to explore CTC profiling utility in CII-treated patients with melanoma using a melanoma messenger RNA (mRNA)/DNA biomarker panel. METHODS: Blood samples (n = 213) were collected prospectively from 75 American Joint Committee on Cancer-staged III/IV melanoma patients during CII treatment and those enriched for CTCs. CTC profiling was performed using 5 known melanoma mRNA biomarkers and BRAF V600E DNA mutation. CTC biomarker status associations with clinical outcomes were assessed. RESULTS: CTCs were detected in 88% of blood samples from patients with melanoma. CTC-derived biomarkers and clinical variables analyzed using classification and regression tree analysis revealed that a combination of lactate dehydrogenase, CTC-mRNA biomarkers, and tumor BRAF-mutation status was indicative of clinical outcomes for patients with stage IV melanoma (n = 52). The panel stratified low-risk and high-risk patients, whereby the latter had poor disease-free (P = 0.03) and overall survival (P = 0.02). Incorporation of a DNA biomarker with mRNA profiling increased overall CTC-detection capability by 57% compared to mRNA profiling only. RNA sequencing of isolated CTCs identified significant catenin beta 1 (CTNNB1) overexpression (P <0.01) compared to nondisease donor blood. CTC-CTNNB1 was associated with progressive disease/stable disease compared to complete-responder patient status (P = 0.02). Serial CTC profiling identified subclinical disease in patients who developed progressive disease during treatment/follow-up. CONCLUSIONS: CTC-derived mRNA/DNA biomarkers have utility for monitoring CII, targeted, and combinatorial therapies in metastatic melanoma patients.


Assuntos
Melanoma/terapia , Células Neoplásicas Circulantes/metabolismo , Idoso , Anticorpos Monoclonais Humanizados/uso terapêutico , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Intervalo Livre de Doença , Feminino , Humanos , Imunoterapia , Estimativa de Kaplan-Meier , Masculino , Melanoma/mortalidade , Melanoma/patologia , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Modelos de Riscos Proporcionais , Estudos Prospectivos , Proteínas Proto-Oncogênicas B-raf/genética , RNA Mensageiro/metabolismo , Fatores de Risco , Regulação para Cima , beta Catenina/genética , beta Catenina/metabolismo
2.
Ann Surg ; 247(6): 1003-10, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18520228

RESUMO

OBJECTIVE: To investigate a cohort of melanoma patients with false negative (FN) sentinel node (SN) biopsies (SNBs) to identify the reasons for the FN result. SUMMARY OF BACKGROUND DATA: SNB is a highly efficient staging method in melanoma patients. However, with long-term follow-up FN SNB results of up to 25% have been reported. METHODS: Seventy-four SNs from 33 patients found to have had an FN SNB were analyzed by reviewing the lymphoscintigraphy, surgical data, and histopathology, and by assessing nodal tissue using multimarker real-time quantitative reverse transcription (qRT) polymerase chain reaction, and antimony concentration measurements (as a marker of "true" SN status) using inductively coupled plasma mass spectroscopy. RESULTS: Nine SNs (12%) from 9 patients (27%) had evidence of melanoma on histopathologic review. Twelve SNs (16%) from 10 patients (30%) were qRT(+). Four of these 12 SNs were positive on histopathology review and 8 were negative. Four patients (12%) were upstaged by qRT. Sixteen patients had their SNB histology, lymphoscintigraphy, and surgical data reviewed. Identifiable causes of the FN SNBs were not found after review of all modalities in 4 patients. SNs from all 4 patients had antimony levels indicative of an SN. Of the SNs evaluable by qRT, 1 was qRT(+) and 7 SNs from 2 patients were qRT(-). CONCLUSIONS: An FN SN can occur because of deficiencies in nuclear medicine, surgery, or pathology. qRT can detect "occult" metastatic melanoma in SNs that have been identified as negative by histopathology.


Assuntos
Melanoma/patologia , Biópsia de Linfonodo Sentinela , Neoplasias Cutâneas/patologia , Antimônio , Biomarcadores Tumorais/análise , Erros de Diagnóstico , Reações Falso-Negativas , Humanos , Imuno-Histoquímica , Metástase Linfática/diagnóstico por imagem , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Cintilografia , Compostos Radiofarmacêuticos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Compostos de Tecnécio
3.
Clin Cancer Res ; 23(16): 4831-4842, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28389511

RESUMO

Purpose: Abnormal activation of the NF-κB pathway induces a more aggressive phenotype of cutaneous melanoma. Understanding the mechanisms involved in melanoma NF-κB activation may identify novel targets for this pathway. KPC1, an E3 ubiquitin ligase, is a regulator of the NF-κB pathway. The objective of this study was to investigate the mechanisms regulating KPC1 expression and its clinical impact in melanoma.Experimental Design: The clinical impact of KPC1 expression and its epigenetic regulation were assessed in large cohorts of clinically well-annotated melanoma tissues (tissue microarrays; n = 137, JWCI cohort; n = 40) and The Cancer Genome Atlas database (TCGA cohort, n = 370). Using melanoma cell lines, we investigated the functional interactions between KPC1 and NF-κB, and the epigenetic regulations of KPC1, including DNA methylation and miRNA expression.Results: We verified that KPC1 suppresses melanoma proliferation by processing NF-κB1 p105 into p50, thereby modulating NF-κB target gene expression. Concordantly, KPC1 expression was downregulated in American Joint Committee on Cancer stage IV melanoma compared with early stages (stage I/II P = 0.013, stage III P = 0.004), and low KPC1 expression was significantly associated with poor overall survival in stage IV melanoma (n = 137; HR 1.810; P = 0.006). Furthermore, our data showed that high miR-155-5p expression, which is controlled by DNA methylation at its promoter region (TCGA; Pearson's r -0.455; P < 0.001), is significantly associated with KPC1 downregulation (JWCI; P = 0.028, TCGA; P = 0.003).Conclusions: This study revealed novel epigenetic regulation of KPC1 associated with NF-κB pathway activation, promoting metastatic melanoma progression. These findings suggest the potential utility of KPC1 and its epigenetic regulation as theranostic targets. Clin Cancer Res; 23(16); 4831-42. ©2017 AACR.


Assuntos
Epigênese Genética , Melanoma/genética , NF-kappa B/genética , Ubiquitina-Proteína Ligases/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Estudos de Coortes , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Melanoma/metabolismo , Melanoma/patologia , MicroRNAs/genética , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/genética , Interferência de RNA , Transdução de Sinais/genética , Ubiquitina-Proteína Ligases/metabolismo
4.
J Clin Oncol ; 23(12): 2744-53, 2005 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-15837989

RESUMO

PURPOSE: Liver metastasis is the predominant cause of colorectal cancer (CRC) related mortality. Chemokines, soluble factors that orchestrate hematopoetic cell movement, have been implicated in directing cancer metastasis, although their clinical relevance in CRC has not been defined. Our hypothesis was that the chemokine receptor CXCR4 expressed by CRC is a prognostic factor for poor disease outcome. METHODS: CRC cell lines (n = 6) and tumor specimens (n = 139) from patients with different American Joint Committee on Cancer (AJCC) stages of CRC were assessed. Microarray screening of select specimens and cell lines identified CXCR4 as a prominent chemokine receptor. CXCR4 expression in tumor and benign specimens was assessed by quantitative real-time reverse transcription polymerase chain reaction and correlated with disease recurrence and overall survival. RESULTS: High CXCR4 expression in tumor specimens (n = 57) from AJCC stage I/II patients was associated with increased risk for local recurrence and/or distant metastasis (risk ratio, 1.35; 95% CI, 1.09 to 1.68; P = .0065). High CXCR4 expression in primary tumor specimens (n = 35) from AJCC stage IV patients correlated with worse overall median survival (9 months v 23 months; RR, 2.53; 95% CI, 1.19 to 5.40; P = .016). CXCR4 expression was significantly higher in liver metastases (n = 39) compared with primary CRC tumors (n = 100; P < .0001). CONCLUSION: CXCR4, a well-characterized chemokine receptor for T-cells, is differentially expressed in CRC. CXCR4 gene expression in primary CRC demonstrated significant associations with recurrence, survival, and liver metastasis. The CXCR4-CXCL12 signaling mechanism may be clinically relevant for patients with CRC and represents a potential novel target for disease-directed therapy.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Perfilação da Expressão Gênica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundário , Recidiva Local de Neoplasia/genética , Receptores CXCR4/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica/fisiopatologia , Prognóstico , Fatores de Risco , Transdução de Sinais , Análise de Sobrevida , Células Tumorais Cultivadas
6.
J Clin Med ; 4(10): 1890-907, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26512704

RESUMO

An increasing number of studies have focused on circulating microRNAs (cmiRNA) in cancer patients' blood for their potential as minimally-invasive biomarkers. Studies have reported the utility of assessing specific miRNAs in blood as diagnostic/prognostic biomarkers; however, the methodologies are not validated or standardized across laboratories. Unfortunately, there is often minimum limited overlap in techniques between results reported even in similar type studies on the same cancer. This hampers interpretation and reliability of cmiRNA as potential cancer biomarkers. Blood collection and processing, cmiRNA extractions, quality and quantity control of assays, defined patient population assessment, reproducibility, and reference standards all affect the cmiRNA assay results. To date, there is no reported definitive method to assess cmiRNAs. Therefore, appropriate and reliable methodologies are highly necessary in order for cmiRNAs to be used in regulated clinical diagnostic laboratories. In this review, we summarize the developments made over the past decade towards cmiRNA detection and discuss the pros and cons of the assays.

7.
Oncotarget ; 6(9): 7053-64, 2015 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-25749524

RESUMO

Circulating cell-free(cf) microRNAs (miRNAs) have been reported to exist in plasma. MicroRNA-210(miR-210) is known to play important roles in the tumor hypoxic state. We hypothesized that the expression levels of cf-miR-210 in plasma would predict early clinical recurrence in melanoma patients. A direct miRNA assay on plasma (RT-qPCR-DP) was developed to improve cf-miRNA assay logistics, eliminate RNA extraction, and reduce specimen amount required. RNA was extracted from formalin-fixed paraffin-embedded (FFPE) melanoma tissues (n = 108) and assessed by RT-qPCR. Plasma (10 µl; n = 264) was procured from AJCC Stage III/IV patients in phase III clinical trials. A RT-qPCR-DP was performed to detect cf-miR-210. MiR-210 was significantly higher in metastatic tumors compared to primary tumors. Cf-miR-210 was significantly higher in melanoma patients versus healthy donor controls. In serial bloods within individual patients, cf-miR-210 < 3 months prior to disease recurrence significantly increased compared to baseline levels (p = 0.012). ROC curve analysis demonstrated that patients with elevated cf-miR-210 were more likely to have disease recurrence. Moreover, cf-miR-210 increase significantly correlated with poorer prognosis (p < 0.001). Lactate dehydrogenase (LDH) level was also assessed within patients, and the AIC values for proportional hazards regression models of cf-miR-210(120.01) and LDH (122.91) demonstrated that cf-miR-210 is a better recurrence indicator. We concluded enhanced cf-miR-210 provides identification of early systemic melanoma recurrence.


Assuntos
Hipóxia , Melanoma/genética , Melanoma/patologia , MicroRNAs/sangue , Recidiva Local de Neoplasia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Adulto , Idoso , Estudos de Coortes , Reações Falso-Positivas , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , L-Lactato Desidrogenase/metabolismo , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Metástase Neoplásica , Projetos Piloto , Prognóstico , Modelos de Riscos Proporcionais , Curva ROC , Reprodutibilidade dos Testes
8.
Methods Mol Biol ; 1102: 513-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24258996

RESUMO

Detection of circulating tumor cells (CTC) in peripheral blood has been investigated for its prognostic ability, and its potential to measure the effectiveness of treatment(s) in patients with melanoma. However, a highly sensitive and specific assay is required to detect CTC in patients' blood. We have developed a multimarker quantitative real-time reverse transcriptase polymerase chain reaction (RT-qPCR) assay for detecting CTC directly from peripheral blood specimens without the need of separating CTC from leukocytes (PBL). We selected and optimized four mRNA biomarkers (MART-1/Melan-A, MAGE-A3, PAX3, and GalNAc-T) for detection and prediction of clinical outcome in melanoma patients. Our protocol has both high sensitivity and specificity for CTC in blood specimens-detecting approximately one to five melanoma cells in 10(7) PBL. We have demonstrated the significance of this assay for serial bleed assessment of CTC in clinical trials and for daily clinical usage.


Assuntos
Biomarcadores Tumorais/sangue , Melanoma/sangue , Células Neoplásicas Circulantes/patologia , Neoplasias Cutâneas/sangue , Separação Celular , Humanos , Estimativa de Kaplan-Meier , Linfócitos/metabolismo , Melanoma/genética , Melanoma/patologia , Células Neoplásicas Circulantes/metabolismo , Prognóstico , RNA Neoplásico/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Melanoma Maligno Cutâneo
9.
J Clin Invest ; 124(12): 5291-304, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25384218

RESUMO

Patients with triple-negative breast cancer (TNBC) have a high incidence of early relapse and metastasis; however, the molecular basis for recurrence in these individuals remains poorly understood. Here, we demonstrate that RASAL2, which encodes a RAS-GTPase-activating protein (RAS-GAP), is a functional target of anti-invasive microRNA-203 and is overexpressed in a subset of triple-negative or estrogen receptor-negative (ER-negative) breast tumors. As opposed to luminal B ER-positive breast cancers, in which RASAL2 has been shown to act as a RAS-GAP tumor suppressor, we found that RASAL2 is oncogenic in TNBC and drives mesenchymal invasion and metastasis. Moreover, high RASAL2 expression was predictive of poor disease outcomes in patients with TNBC. RASAL2 acted independently of its RAS-GAP catalytic activity in TNBC; however, RASAL2 promoted small GTPase RAC1 signaling, which promotes mesenchymal invasion, through binding and antagonizing the RAC1-GAP protein ARHGAP24. Together, these results indicate that activation of a RASAL2/ARHGAP24/RAC1 module contributes to TNBC tumorigenesis and identify a context-dependent role of RASAL2 in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Ativação Enzimática/genética , Feminino , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Neoplasias/genética , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Proteínas rac1 de Ligação ao GTP/genética
10.
J Nucl Med ; 54(12): 2039-45, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24101687

RESUMO

UNLABELLED: (18)F-FDG PET/CT is used in the follow-up of patients with head and neck squamous cell cancer (HNSCC). However, its impact on clinical decision making and patient outcome is not fully established. The objective of this study was to determine the prognostic value of (18)F-FDG PET/CT for overall survival (OS) of HNSCC patients when performed in addition to clinical assessment between 4 and 24 mo after treatment. METHODS: This was a retrospective study at a single tertiary center. The institutional review board approved this study, and the requirement to obtain informed consent was waived. The study included 134 biopsy-proven HNSCC patients with 227 follow-up PET/CT scans. The primary outcome measure was OS. Median follow-up was 40 mo (range, 7-145 mo). Survival is presented as Kaplan-Meier plots with Mantel-Cox log-rank test. The multivariate Cox model included clinical covariates. RESULTS: Of the 227 PET/CT scans, 41 (18%) were positive for tumor and 186 (82%) were negative for tumor. PET/CT identified recurrence in 5% (9/194) of scans performed without prior clinical concern and ruled out tumor in 51.5% (17/33) of scans performed to evaluate clinical suspicion or uncertainty of recurrence. The median survival of PET-positive and -negative groups from the date of the scan was 20 and 30.5 mo, respectively (P < 0.0001). There was a significant difference in OS from the scan date between patients who had a positive PET/CT result for tumor and those who had a negative result (log-rank, P < 0.0001), with a hazard ratio of 29.74. Human papillomavirus status (P = 0.001) and PET/CT result (P = 0.04) were the only factors significantly associated with OS, adjusted for all other covariates. CONCLUSION: (18)F-FDG PET/CT performed between 4 and 24 mo after treatment adds value to clinical assessment at the time of the study, especially when there is clinical suspicion or uncertainty, and can serve as a prognostic marker of OS in HNSCC.


Assuntos
Fluordesoxiglucose F18 , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Neoplasias de Células Escamosas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada por Raios X , Adulto , Feminino , Seguimentos , Humanos , Interpretação de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Imagem Multimodal , Prognóstico , Estudos Retrospectivos , Análise de Sobrevida , Fatores de Tempo
11.
Imaging Med ; 4(6): 633-647, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23482696

RESUMO

This article discusses the value of 18F-fluoro-2-deoxyglucose PET/CT imaging biomarkers in head and neck squamous cell carcinoma. 18F-fluoro-2-deoxyglucose PET/CT is valuable at baseline staging, radiotherapy planning, therapy response assessment and in the follow-up of patients with head and neck squamous cell carcinoma. Maximum and peak standardized uptake value (SUVmax and SUVpeak), metabolic tumor volume and total lesion glycolysis are the common 18F-fluoro-2-deoxyglucose quantitative parameters that have been studied, along with qualitative assessments. These parameters will be evaluated with respect to their established or potential role as noninvasive biomarkers for patient risk stratification, treatment response and survival outcome.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA