Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Histopathology ; 81(6): 818-825, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36102613

RESUMEN

Diagnosis of spindle cell/sarcomatoid melanoma may be challenging due to frequent loss of expression of melanocytic marker(s) and histomorphologic resemblance to various mesenchymal tumours, particularly malignant peripheral nerve sheath tumour (MPNST). Overexpression of PReferentially expressed Antigen in MElanoma (PRAME) supports a diagnosis of melanoma when evaluating challenging melanocytic tumours. PRAME expression in MPNST and other cutaneous sarcomatoid neoplasms, however, has not been well characterised. We aimed to determine the utility of PRAME immunostain in distinguishing spindle cell melanoma from MPNST and other sarcomatoid mimics. PRAME expression was scored by extent (0 to 4+) and intensity (0 to 3) of staining. A strong positive correlation was observed between the extent and intensity scores (r = 0.84). An extent score of 4+, defined by staining in 76-100% of tumour cells, was seen in 56% (23/41) of spindle cell melanomas, 18% (7/38) of MPNSTs, 15% (4/27) of cutaneous sarcomatoid squamous cell carcinomas (SCCs), 33% (5/15) of poorly differentiated cutaneous angiosarcomas, 12% (4/33) of atypical fibroxanthomas (AFXs), 4% (1/25) of pleomorphic dermal sarcomas (PDSs), and none (0/16) of the high-grade cutaneous leiomyosarcomas. A significant difference was found between spindle cell melanoma and all other examined sarcomatoid neoplasms except angiosarcoma. While diffuse (and often strong) PRAME expression is more frequently observed in spindle cell melanoma than MPNST, sarcomatoid SCC, AFX, PDS, and high-grade leiomyosarcoma, its limited sensitivity and specificity caution against its use as a standalone diagnostic marker. PRAME may complement other epigenetic or lineage-specific markers and should only be used as part of an immunohistochemical panel when evaluating these sarcomatoid neoplasms.


Asunto(s)
Leiomiosarcoma , Melanoma , Neurofibrosarcoma , Sarcoma , Neoplasias Cutáneas , Humanos , Antígenos de Neoplasias , Biomarcadores de Tumor/análisis , Diagnóstico Diferencial , Inmunohistoquímica , Leiomiosarcoma/diagnóstico , Melanoma/patología , Neurofibrosarcoma/diagnóstico , Sarcoma/diagnóstico , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/patología
2.
Immunity ; 39(3): 611-21, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-24012420

RESUMEN

Myeloid-derived suppressor cells (MDSCs) and cancer stem cells (CSCs) are important cellular components in the cancer microenvironment and may affect cancer phenotype and patient outcome. The nature of MDSCs and their interaction with CSCs in ovarian carcinoma are unclear. We examined the interaction between MDSCs and CSCs in patients with ovarian carcinoma and showed that MDSCs inhibited T cell activation and enhanced CSC gene expression, sphere formation, and cancer metastasis. MDSCs triggered miRNA101 expression in cancer cells. miRNA101 subsequently repressesed the corepressor gene C-terminal binding protein-2 (CtBP2), and CtBP2 directly targeted stem cell core genes resulting in increased cancer cell stemness and increasing metastatic and tumorigenic potential. Increased MDSC density and tumor microRNA101 expression predict poor survival, as does decreased tumor CtBP2 expression, independent of each other. Collectively, our work identifies an immune-associated cellular, molecular, and clinical network involving MDSCs-microRNA101-CtBP2-stem cell core genes, which extrinsically controls cancer stemness and impacts patient outcome.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , MicroARNs/metabolismo , Células Mieloides/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Ováricas/inmunología , Oxidorreductasas de Alcohol/antagonistas & inhibidores , Oxidorreductasas de Alcohol/genética , Comunicación Celular , Proteínas Co-Represoras , Femenino , Humanos , Activación de Linfocitos , MicroARNs/genética , Células Mieloides/citología , Células Mieloides/inmunología , Metástasis de la Neoplasia , Células Madre Neoplásicas/inmunología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Neoplasias Ováricas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Linfocitos T/inmunología
3.
Proc Natl Acad Sci U S A ; 114(30): 8065-8070, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28698375

RESUMEN

Osteosarcoma occurs predominantly in children and young adults. High-grade tumors require multidisciplinary treatment consisting of chemotherapy in the neoadjuvant and adjuvant settings, along with surgical intervention. Despite this approach, death from respiratory failure secondary to the development and progression of pulmonary metastases remains a significant problem. Here, we identify the IL-11 receptor α subunit (IL-11Rα) as a cell surface marker of tumor progression that correlates with poor prognosis in patients with osteosarcoma. We also show that both IL-11Rα and its ligand, IL-11, are specifically up-regulated in human metastatic osteosarcoma cell lines; engagement of this autocrine loop leads to tumor cell proliferation, invasion, and anchorage-independent growth in vitro. Consistently, IL-11Rα promotes lung colonization by human metastatic osteosarcoma cells in vivo in an orthotopic mouse model. Finally, we evaluate the IL-11Rα-targeted proapoptotic agent bone metastasis-targeting peptidomimetic (BMTP-11) in preclinical models of primary intratibial osteosarcomas, observing marked inhibition of both tumor growth and lung metastases. This effect was enhanced when BMTP-11 was combined with the chemotherapeutic drug gemcitabine. Our combined data support the development of approaches targeting IL-11Rα, and establish BMTP-11 as a leading drug candidate for clinical translation in patients with high-risk osteosarcoma.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Subunidad alfa del Receptor de Interleucina-11/antagonistas & inhibidores , Osteosarcoma/tratamiento farmacológico , Péptidos/uso terapéutico , Animales , Comunicación Autocrina , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Subunidad alfa del Receptor de Interleucina-11/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Masculino , Ratones Desnudos , Metástasis de la Neoplasia , Osteosarcoma/metabolismo , Péptidos/farmacología , Investigación Biomédica Traslacional
4.
Proc Natl Acad Sci U S A ; 114(25): 6581-6586, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28607076

RESUMEN

Identification of biomarkers and therapeutic targets is a critical goal of precision medicine. N-glycoproteins are a particularly attractive class of proteins that constitute potential cancer biomarkers and therapeutic targets for small molecules, antibodies, and cellular therapies. Using mass spectrometry (MS), we generated a compendium of 1,091 N-glycoproteins (from 40 human primary lymphomas and cell lines). Hierarchical clustering revealed distinct subtype signatures that included several subtype-specific biomarkers. Orthogonal immunological studies in 671 primary lymphoma tissue biopsies and 32 lymphoma-derived cell lines corroborated MS data. In anaplastic lymphoma kinase-positive (ALK+) anaplastic large cell lymphoma (ALCL), integration of N-glycoproteomics and transcriptome sequencing revealed an ALK-regulated cytokine/receptor signaling network, including vulnerabilities corroborated by a genome-wide clustered regularly interspaced short palindromic screen. Functional targeting of IL-31 receptor ß, an ALCL-enriched and ALK-regulated N-glycoprotein in this network, abrogated ALK+ALCL growth in vitro and in vivo. Our results highlight the utility of functional proteogenomic approaches for discovery of cancer biomarkers and therapeutic targets.


Asunto(s)
Biomarcadores de Tumor/genética , Linfoma/genética , Transcriptoma/genética , Línea Celular Tumoral , Humanos , Proteogenómica/métodos , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/genética
5.
Proc Natl Acad Sci U S A ; 112(50): E6882-8, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621735

RESUMEN

Whether human cancer follows a hierarchical or stochastic model of differentiation is controversial. Furthermore, the factors that regulate cancer stem-like cell (CSC) differentiation potential are largely unknown. We used a novel microfluidic single-cell culture method to directly observe the differentiation capacity of four heterogeneous ovarian cancer cell populations defined by the expression of the CSC markers aldehyde dehydrogenase (ALDH) and CD133. We evaluated 3,692 progeny from 2,833 cells. We found that only ALDH(+)CD133(+) cells could generate all four ALDH(+/-)CD133(+/-) cell populations and identified a clear branched differentiation hierarchy. We also observed a single putative stochastic event. Within the hierarchy of cells, bone morphologenetic protein 2 (BMP2) is preferentially expressed in ALDH(-)CD133(-) cells. BMP2 promotes ALDH(+)CD133(+) cell expansion while suppressing the proliferation of ALDH(-)CD133(-) cells. As such, BMP2 suppressed bulk cancer cell growth in vitro but increased tumor initiation rates, tumor growth, and chemotherapy resistance in vivo whereas BMP2 knockdown reduced CSC numbers, in vivo growth, and chemoresistance. These data suggest a hierarchical differentiation pattern in which BMP2 acts as a feedback mechanism promoting ovarian CSC expansion and suppressing progenitor proliferation. These results explain why BMP2 suppresses growth in vitro and promotes growth in vivo. Together, our results support BMP2 as a therapeutic target in ovarian cancer.


Asunto(s)
Proteína Morfogenética Ósea 2/fisiología , Neoplasias Ováricas/patología , Antígeno AC133 , Aldehído Deshidrogenasa/metabolismo , Antígenos CD/metabolismo , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Proteína Morfogenética Ósea 2/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Glicoproteínas/metabolismo , Humanos , Microfluídica , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Péptidos/metabolismo
6.
Breast Cancer Res Treat ; 161(2): 375-384, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27900490

RESUMEN

PURPOSE: To investigate the discordance between original and central laboratories in estrogen receptor (ER) status, in tumors originally deemed to be ER-negative, and in HER2 status in a diverse population-based sample. METHODS: In a follow-up study of 1785 women with Stage I-III breast cancer diagnosed between 2005 and 2007 in the Detroit and Los Angeles County SEER registry catchment areas, participants were asked to consent to reassessment of ER (in tumors originally deemed to be ER-negative) and HER2 status on archival tumor samples approximately four years after diagnosis. Blocks were centrally prepared and analyzed for ER and HER2 using standardized methods and the guidelines of the American Society of Clinical Oncology and the College of American Pathologists. Analyses determined the discordance between original and central laboratories. RESULTS: 132 (31%) of those eligible for ER reassessment and 367 (21%) eligible for HER2 reassessment had archival blocks reassessed centrally. ER discordance was only 6%. HER2 discordance by immunohistochemistry (IHC) was 26%, but final HER2 results-employing FISH in tumors that were IHC 2+ at the central laboratory-were discordant in only 6%. Half of the original laboratories did not perform their own assays. CONCLUSIONS: Discordance between original and central laboratories in two large metropolitan areas was low in this population-based sample compared to previously reported patient samples. Centralization of testing for key pathology variables appears to be occurring in many hospitals. In addition, quality improvement efforts may have preceded the publication and dissemination of specialty society guidelines.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/metabolismo , Servicios de Laboratorio Clínico/normas , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/diagnóstico , Femenino , Humanos , Inmunohistoquímica/métodos , Inmunohistoquímica/normas , Hibridación Fluorescente in Situ/métodos , Hibridación Fluorescente in Situ/normas , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Vigilancia de la Población , Receptor ErbB-2/genética , Receptores de Estrógenos/genética , Reproducibilidad de los Resultados , Programa de VERF , Adulto Joven
7.
Gynecol Oncol ; 147(2): 351-357, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28844540

RESUMEN

OBJECTIVE: Melanoma originating from gynecologic sites (MOGS), including the vulva, vagina, and cervix, is a rare and aggressive form of melanoma with poor long-term clinical outcome. The clinicopathologic features of vulvar and non-vulvar tumors remain relatively understudied, and in contrast to cutaneous melanomas at non-sun-exposed sites, MOGS typically do not harbor BRAF mutations. Thus, we sought to analyze the clinicopathologic and molecular features of MOGS. METHODS: A large retrospective cohort of patients with MOGS (n=59) at a single large academic institution over a 28-year period was identified. Associations among clinicopathologic characteristics were assessed via standard statistical approaches, and clinical outcome was examined using Cox regression analysis. Sanger sequencing was utilized to identify mutations in hotspot regions of BRAF, KIT, NRAS, and CTNNB1. RESULTS: Tumors involving the vagina and/or cervix (non-vulvar) are significantly associated with high-risk clinicopathologic features, including increased tumor thickness, ulceration, positive resection margins, lymph node metastasis, and poor long-term clinical outcome (with increased risk of death due to disease). The aggressive clinical behavior of non-vulvar tumors is independent of advanced clinical stage and lymph node metastasis in multivariate analysis. Targeted molecular analysis confirms an overall low rate of oncogenic mutations in our MOGS cohort, although KIT mutations (particularly in exon 11) are relatively enriched. CONCLUSIONS: Overall, our results show that non-vulvar MOGS are aggressive tumors with poor long-term clinical outcome and indicate that few targeted therapeutic options are currently available to patients with MOGS.


Asunto(s)
Neoplasias de los Genitales Femeninos/genética , Neoplasias de los Genitales Femeninos/patología , Melanoma/genética , Melanoma/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Estudios de Cohortes , Femenino , GTP Fosfohidrolasas/genética , Humanos , Proteínas de la Membrana/genética , Persona de Mediana Edad , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-kit/genética , Estudios Retrospectivos , Adulto Joven , beta Catenina/genética
8.
Cancer ; 122(6): 868-74, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26710211

RESUMEN

BACKGROUND: Dasatinib exhibited activity in preclinical models of sarcoma. The Sarcoma Alliance for Research through Collaboration (SARC) conducted a multicenter, phase 2 trial of dasatinib in patients with advanced sarcoma. METHODS: Patients received dasatinib twice daily. The primary objective was to estimate the clinical benefit rate (CBR) (complete response or partial response within 6 months or stable disease duration of ≥6 months) with a target of ≥25%. Patients were enrolled into 1 of 7 different cohorts and assessed by imaging every 8 weeks using Choi criteria tumor response and a Bayesian hierarchical design. For each subtype, enrollment was stopped after a minimum of 9 patients were treated if there was a <1% chance the CBR was ≥25%. RESULTS: A total of 200 patients were enrolled. Accrual was stopped early in 5 cohorts because of low CBR. The leiomyosarcoma (LMS) and undifferentiated pleomorphic sarcoma (UPS) cohorts fully accrued and 6 of 47 and 8 of 42 evaluable patients, respectively, exhibited clinical benefit. The probability that the CBR was ≥25% in the LMS and UPS cohorts was 0.008 and 0.10, respectively. The median progression-free survival ranged from 0.9 months in patients with rhabdomyosarcoma to 2.2 months in patients with LMS. The median overall survival was 8.6 months. The most frequent adverse events were constitutional, gastrointestinal, and respiratory, and 36% of patients required dose reduction for toxicity. Serious adverse events attributed to therapy occurred in 11% of patients. CONCLUSIONS: Dasatinib may have activity in patients with UPS but is inactive as a single agent in the other sarcoma subtypes included herein. The Bayesian design allowed for the early termination of accrual in 5 subtypes because of lack of drug activity.


Asunto(s)
Antineoplásicos/uso terapéutico , Dasatinib/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Sarcoma/tratamiento farmacológico , Sarcoma/patología , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Teorema de Bayes , Dasatinib/administración & dosificación , Dasatinib/efectos adversos , Supervivencia sin Enfermedad , Esquema de Medicación , Femenino , Humanos , Estimación de Kaplan-Meier , Leiomiosarcoma/tratamiento farmacológico , Leiomiosarcoma/patología , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Resultado del Tratamiento
9.
Breast J ; 22(3): 282-6, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26843318

RESUMEN

Malignant phyllodes tumor (PT) infrequently displays heterologous differentiation, and when present is most often liposarcomatous. We identified five cases of malignant PT with regions identical to well-differentiated liposarcoma (WDLS) of soft tissue and evaluated them for MDM2 and CDK4 gene expression and amplification using immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH), respectively. Despite indistinguishable morphology all cases of malignant PT with WDLS-like liposarcomatous differentiation were negative for MDM2 and CDK4 IHC and FISH, supporting different underlying pathogenesis.


Asunto(s)
Neoplasias de la Mama/patología , Liposarcoma/patología , Tumor Filoide/patología , Adulto , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Liposarcoma/genética , Liposarcoma/metabolismo , Persona de Mediana Edad , Tumor Filoide/genética , Tumor Filoide/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología
10.
Lab Invest ; 95(11): 1278-90, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26280220

RESUMEN

Retinoblastoma (RB) is the most common primary intraocular cancer in children, and the third most common cancer overall in infants. No molecular-targeted therapy for this lethal tumor exists. Since the tumor suppressor RB1, whose genetic inactivation underlies RB, is upstream of the epigenetic regulator EZH2, a pharmacologic target for many solid tumors, we reasoned that EZH2 might regulate human RB tumorigenesis. Histologic and immunohistochemical analyses were performed using an EZH2 antibody in sections from 43 samples of primary, formalin-fixed, paraffin-embedded human RB tissue, cryopreserved mouse retina, and in whole cell lysates from human RB cell lines (Y79 and WERI-Rb1), primary human fetal retinal pigment epithelium (RPE) and fetal and adult retina, mouse retina and embryonic stem (ES) cells. Although enriched during fetal human retinal development, EZH2 protein was not present in the normal postnatal retina. However, EZH2 was detected in all 43 analyzed human RB specimens, indicating that EZH2 is a fetal protein expressed in postnatal human RB. EZH2 expression marked single RB cell invasion into the optic nerve, a site of invasion whose involvement may influence the decision for systemic chemotherapy. To assess the role of EZH2 in RB cell survival, human RB and primary RPE cells were treated with two EZH2 inhibitors (EZH2i), GSK126 and SAH-EZH2 (SAH). EZH2i impaired intracellular adenosine triphosphate (ATP) production, an indicator of cell viability, in a time and dose-dependent manner, but did not affect primary human fetal RPE. Thus, aberrant expression of a histone methyltransferase protein is a feature of human RB. This is the first time this mechanism has been implicated for an eye, adnexal, or orbital tumor. The specificity of EZH2i toward human RB cells, but not RPE, warrants further in vivo testing in animal models of RB, especially those EZH2i currently in clinical trials for solid tumors and lymphoma.


Asunto(s)
Epigénesis Genética/fisiología , Complejo Represivo Polycomb 2/efectos de los fármacos , Neoplasias de la Retina/metabolismo , Retinoblastoma/metabolismo , Animales , Línea Celular Tumoral , Preescolar , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Humanos , Lactante , Masculino , Ratones , Complejo Represivo Polycomb 2/metabolismo , Complejo Represivo Polycomb 2/fisiología , Neoplasias de la Retina/patología , Retinoblastoma/patología
11.
Mod Pathol ; 28(1): 57-68, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24947144

RESUMEN

Recent molecular advances have identified a novel, clinically aggressive subgroup of undifferentiated round cell sarcomas defined molecularly by oncogenic fusion of the gene, CIC, and either DUX4 or its paralog, DUX4L, herein termed CIC-DUX sarcomas. Morphologically, CIC-DUX sarcomas are round cell sarcomas with high-grade nuclear features, including vesicular chromatin and nucleoli, patchy clear cell foci, myxoid change, and necrosis. Here, we studied a cohort of 10 cases, including 6 newly identified cases, 2 with paired metastases. Given our prior observation of trisomy 8 in these tumors, we assayed for amplification and expression of MYC (c-Myc) and representative downstream targets. Trisomy 8 was detected in 5/7 testable cases, with further amplification of MYC locus in 6/7 testable cases and immunohistochemical expression of MYC in 10/10. The canonical MYC transcriptional target, p21, but not MTDH, was differentially expressed compared with Ewing sarcomas. Given prior observation of induction of ETS-family transcription factors by the fusion oncoprotein, we assayed and identified highly prevalent positivity for ERG (9/10) and FLI1 (8/8). These findings are cautionary regarding use of these immunostains in prospective case workup, whereas the prevalent MYC amplification may represent a therapeutically targetable oncogenic pathway in CIC-DUX sarcomas.


Asunto(s)
Genes myc/genética , Proteínas Proto-Oncogénicas c-ets/biosíntesis , Sarcoma de Células Pequeñas/genética , Neoplasias de los Tejidos Blandos/genética , Adulto , Femenino , Amplificación de Genes , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Proteínas de Fusión Oncogénica , Reacción en Cadena de la Polimerasa , Proteínas Represoras/genética , Estudios Retrospectivos , Sarcoma de Células Pequeñas/patología , Neoplasias de los Tejidos Blandos/patología , Adulto Joven
12.
Proc Natl Acad Sci U S A ; 109(11): 4251-6, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22375031

RESUMEN

Gene amplification is a tumor-specific event during malignant transformation. Recent studies have proposed a lineage-dependency (addiction) model of human cancer whereby amplification of certain lineage transcription factors predisposes a survival mechanism in tumor cells. These tumor cells are derived from tissues where the lineage factors play essential developmental and maintenance roles. Here, we show that recurrent amplification at 18q11.2 occurs in 21% of esophageal adenocarcinomas (EAC). Utilization of an integrative genomic strategy reveals a single gene, the embryonic endoderm transcription factor GATA6, as the selected target of the amplification. Overexpression of GATA6 is found in EACs that contain gene amplification. We find that EAC patients whose tumors carry GATA6 amplification have a poorer survival. We show that ectopic expression of GATA6, together with FGFR2 isoform IIIb, increases anchorage-independent growth in immortalized Barrett's esophageal cells. Conversely, siRNA-mediated silencing of GATA6 significantly reduces both cell proliferation and anchorage-independent growth in EAC cells. We further demonstrate that induction of apoptotic/anoikis pathways is triggered upon silencing of GATA6 in EAC cells but not in esophageal squamous cells. We show that activation of p38α signaling and up-regulation of TNF-related apoptosis-inducing ligand are detected in apoptotic EAC cells upon GATA6 deprivation. We conclude that selective gene amplification of GATA6 during EAC development sustains oncogenic lineage-survival of esophageal adenocarcinoma.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Linaje de la Célula/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Factor de Transcripción GATA6/genética , Apoptosis/genética , Esófago de Barrett/genética , Esófago de Barrett/patología , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Cromosomas Humanos Par 18/genética , Hibridación Genómica Comparativa , Fragmentación del ADN , Factor de Transcripción GATA6/metabolismo , Amplificación de Genes/genética , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Genoma Humano/genética , Humanos , ARN Interferente Pequeño/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Surg Res ; 187(1): 6-13, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24418519

RESUMEN

BACKGROUND: When presenting with advanced stage disease, lung cancer patients have <5% 5-y survival. The overexpression of checkpoint kinase 1 (CHK1) is associated with poorer outcomes and may contribute to therapy resistance. Targeting CHK1 with small-molecule inhibitors in p53 mutant tumors might improve the effectiveness of chemotherapy and radiotherapy in non-small cell lung cancer (NSCLC). METHODS: We evaluated CHK1 messenger RNA and protein levels in multiple NSCLC cell lines. We assessed cell line sensitization to gemcitabine, pemetrexed, and radiotherapy by CHK1 inhibition with the small molecule AZD7762 using proliferation and clonogenic cell survival assays. We analyzed CHK1 signaling by Western blotting to confirm that AZD7762 inhibits CHK1. RESULTS: We selected two p53 mutant NSCLC cell lines with either high (H1299) or low (H1993) CHK1 levels for further analysis. We found that AZD7762 sensitized both cell lines to gemcitabine, pemetrexed, and radiotherapy. Chemosensitization levels were greater, however, for the higher CHK1 protein expressing cell line, H1299, when compared with H1993. Furthermore, analysis of the CHK1 signaling pathway showed that H1299 cells have an increased dependence on the CHK1 pathway in response to chemotherapy. There was no increased sensitization to radiation in H1299 versus H1993. CONCLUSIONS: CHK1 inhibition by AZD7762 preferentially sensitizes high CHK1 expressing cells, H1299, to anti-metabolite chemotherapy as compared with low CHK1 expressing H1993 cells. Thus, CHK1 inhibitors may improve the efficacy of standard lung cancer therapies, especially for those subgroups of tumors harboring higher expression levels of CHK1 protein.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Proteínas Quinasas/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma de Células Grandes/tratamiento farmacológico , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Resistencia a Antineoplásicos/genética , Glutamatos/uso terapéutico , Guanina/análogos & derivados , Guanina/uso terapéutico , Humanos , Neoplasias Pulmonares/metabolismo , Pemetrexed , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/metabolismo , Tiofenos/uso terapéutico , Urea/análogos & derivados , Urea/uso terapéutico , Gemcitabina
14.
J Am Acad Dermatol ; 71(1): 100-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24656728

RESUMEN

BACKGROUND: Distinction of rosacea and cutaneous lupus erythematosus (LE) can be challenging because of significant clinical and histologic overlap. A controlled study comparing these conditions is lacking. OBJECTIVE: We compared the histologic features, T-cell subsets, and plasmacytoid dendritic cells in rosacea and LE. METHODS: Biopsy specimens of rosacea (n = 27) and facial LE (n = 30) were retrospectively reviewed and reacted with Alcian blue and periodic acid-Schiff stains, and CD4, CD8, CD25, and CD123 immunostains. RESULTS: LE demonstrates a lower CD4:CD8 ratio (1.74 vs 2.80, P = .0064), fewer CD4(+)CD25(+) regulatory T cells (13% vs 31%, P < .0001), and more CD123(+) plasmacytoid dendritic cells (18% vs 6%, P = .0137) than rosacea. The plasmacytoid dendritic cells in LE are more likely to form clusters (P = .0137) and comprise at least 20% of the infiltrate (P = .0340). Also associated with LE are follicular plugging (P = .0039), perineural lymphocytic infiltrate (P = .0211), abundant mucin deposition (P = .0031), and conspicuous basement membrane thickening (P = .0073), whereas Demodex infestation (P = .0064) and sebaceous hyperplasia (P = .0029) are significantly associated with rosacea. LIMITATIONS: Although statistically significant, the immunophenotypic differences are rather small and limited for routine use. CONCLUSION: The infiltrates in rosacea and LE differ immunophenotypically, and may aid in their distinction in addition to conventional histologic examination.


Asunto(s)
Células Dendríticas/patología , Lupus Eritematoso Cutáneo/patología , Rosácea/patología , Subgrupos de Linfocitos T/patología , Adulto , Células Dendríticas/inmunología , Diagnóstico Diferencial , Femenino , Humanos , Inmunohistoquímica , Inmunofenotipificación , Lupus Eritematoso Cutáneo/inmunología , Lupus Eritematoso Cutáneo/metabolismo , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Rosácea/inmunología , Rosácea/metabolismo , Subgrupos de Linfocitos T/inmunología
15.
Am J Pathol ; 181(3): 1017-33, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22800756

RESUMEN

Dysregulation of the WNT and insulin-like growth factor 2 (IGF2) signaling pathways has been implicated in sporadic and syndromic forms of adrenocortical carcinoma (ACC). Abnormal ß-catenin staining and CTNNB1 mutations are reported to be common in both adrenocortical adenoma and ACC, whereas elevated IGF2 expression is associated primarily with ACC. To better understand the contribution of these pathways in the tumorigenesis of ACC, we examined clinicopathological and molecular data and used mouse models. Evaluation of adrenal tumors from 118 adult patients demonstrated an increase in CTNNB1 mutations and abnormal ß-catenin accumulation in both adrenocortical adenoma and ACC. In ACC, these features were adversely associated with survival. Mice with stabilized ß-catenin exhibited a temporal progression of increased adrenocortical hyperplasia, with subsequent microscopic and macroscopic adenoma formation. Elevated Igf2 expression alone did not cause hyperplasia. With the combination of stabilized ß-catenin and elevated Igf2 expression, adrenal glands were larger, displayed earlier onset of hyperplasia, and developed more frequent macroscopic adenomas (as well as one carcinoma). Our results are consistent with a model in which dysregulation of one pathway may result in adrenal hyperplasia, but accumulation of a second or multiple alterations is necessary for tumorigenesis.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/patología , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Factor II del Crecimiento Similar a la Insulina/metabolismo , beta Catenina/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Corticoesteroides/metabolismo , Neoplasias de la Corteza Suprarrenal/genética , Animales , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/genética , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica , Impresión Genómica , Humanos , Hiperplasia , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Ratones Noqueados , Análisis Multivariante , Mutación/genética , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , Estabilidad Proteica , Transporte de Proteínas , Regulación hacia Arriba/genética
16.
J Immunother Cancer ; 11(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36653070

RESUMEN

BACKGROUND: Cell therapies for solid tumors are thwarted by the hostile tumor microenvironment (TME) and by heterogeneous expression of tumor target antigens. We address both limitations with a novel class of chimeric antigen receptors based on plant lectins, which recognize the aberrant sugar residues that are a 'hallmark' of both malignant and associated stromal cells. We have expressed in T cells a modified lectin from banana, H84T BanLec, attached to a chimeric antigen receptor (H84T-CAR) that recognizes high-mannose (asparagine residue with five to nine mannoses). Here, we tested the efficacy of our novel H84T CAR in models of pancreatic ductal adenocarcinoma (PDAC), intractable tumors with aberrant glycosylation and characterized by desmoplastic stroma largely contributed by pancreatic stellate cells (PSCs). METHODS: We transduced human T cells with a second-generation retroviral construct expressing the H84T BanLec chimeric receptor, measured T-cell expansion, characterized T-cell phenotype, and tested their efficacy against PDAC tumor cells lines by flow cytometry quantification. In three-dimensional (3D) spheroid models, we measured H84T CAR T-cell disruption of PSC architecture, and T-cell infiltration by live imaging. We tested the activity of H84T CAR T cells against tumor xenografts derived from three PDAC cell lines. Antitumor activity was quantified by caliper measurement and bioluminescence signal and used anti-human vimentin to measure residual PSCs. RESULTS: H84T BanLec CAR was successfully transduced and expressed by T cells which had robust expansion and retained central memory phenotype in both CD4 and CD8 compartments. H84T CAR T cells targeted and eliminated PDAC tumor cell lines. They also disrupted PSC architecture in 3D models in vitro and reduced total tumor and stroma cells in mixed co-cultures. H84T CAR T cells exhibited improved T-cell infiltration in multicellular spheroids and had potent antitumor effects in the xenograft models. We observed no adverse effects against normal tissues. CONCLUSIONS: T cells expressing H84T CAR target malignant cells and their stroma in PDAC tumor models. The incorporation of glycan-targeting lectins within CARs thus extends their activity to include both malignant cells and their supporting stromal cells, disrupting the TME that otherwise diminishes the activity of cellular therapies against solid tumors.


Asunto(s)
Carcinoma Ductal Pancreático , Musa , Neoplasias Pancreáticas , Receptores Quiméricos de Antígenos , Humanos , Musa/metabolismo , Lectinas/metabolismo , Linfocitos T , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
17.
Histopathology ; 60(4): 597-608, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22260386

RESUMEN

AIMS: High-grade, poorly differentiated, infiltrative carcinomas involving the renal sinus region often pose challenging differential diagnostic considerations, specifically differentiation of urothelial carcinoma (UC) from renal cell carcinoma (RCC) subtypes. Accurate classification, especially the distinction of UC from RCC, is critical, as therapeutic approaches differ. METHODS AND RESULTS: Cluster analysis was performed on immunohistochemical data from 18 invasive UCs, six CDCs, two RMCs, 18 type 2 papillary renal cell carcinomas (PRCCs) and 20 high-grade clear cell renal cell carcinomas (CRCCs) using a broad panel of traditional and novel immunohistochemical markers. The initial analysis with all antibodies segregates almost all the RCCs (45 of 46, 98%) from all the UCs based on the lack of expression of p63 in all (100%) RCCs, along with predominant strong expression of paired box gene 8 (PAX8) and vimentin, predominant lack of expression of high molecular weight cytokeratin (HMCK) and CK7 and variable expression of RCC, CD10, CA1X and PAX2. All the UCs cluster together with strong, diffuse reactivity for p63, predominant reactivity for CK7 and high molecular weight kininogen (HMWK), and absent to minimal staining with PAX8, RCC antigen, PAX2, alpha-methylacyl-CoA racemase (AMACR), carbonic anhydrase IX (CAIX) and vimentin. After removing antibodies with significant overlap and/or minimal impact, a second analysis with a limited panel including p63, CK7, vimentin, integrase interactor 1 (INI-1) and PAX8 was performed. Again, the majority of UCs cluster into one group and p63 positivity separates all UCs from RCCs. CONCLUSIONS: Lack of INI-1 expression, noted exclusively in RMCs, segregates RMCs into a separate cluster. PAX8 is rarely positive (17%) in UC, is commonly expressed in CDC, RMC, PRCC and CRCC and is superior to PAX2.


Asunto(s)
Carcinoma de Células Renales/diagnóstico , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Queratina-7/metabolismo , Neoplasias Renales/diagnóstico , Factores de Transcripción Paired Box/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Urotelio/patología , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/metabolismo , Diagnóstico Diferencial , Humanos , Neoplasias Renales/metabolismo , Factor de Transcripción PAX8 , Proteína SMARCB1 , Análisis de Matrices Tisulares , Urotelio/metabolismo
18.
Oral Oncol ; 135: 106226, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36323071

RESUMEN

OBJECTIVES: In an evolving era of immunotherapeutic options for persistent or recurrent laryngeal squamous cell carcinoma (LSCC), there is a need for improved biomarkers of treatment response and survival to inform optimal treatment selection and prognostication. Herein, our primary objective was to explore correlations between tumor infiltrating lymphocytes (TILs) and PD-L1 Combined Positive Score (CPS). Secondarily, we sought to explore their combined association with survival outcomes in patients with persistent or recurrent LSCC treated with salvage surgery. MATERIALS AND METHODS: This was a retrospective cohort study at a single academic medical center. Immunohistochemistry staining for TILs and PD-L1 was performed on a tissue microarray of persistent or recurrent LSCC pathologic specimens. Correlations between TIL subsets and PD-L1 CPS were examined using Pearson's correlation coefficient and survival outcomes were analyzed with the Kaplan-Meier method and log-rank tests. RESULTS: Only CD103+ TILs showed a statistically significant, weakly-positive correlation with PD-L1 CPS (r2 = 0.264, p < 0.015). No other TIL subsets correlated with PD-L1 CPS in our cohort. The most favorable survival outcomes were seen in patients with pathologic N0 tumors showing high CD103+ TILs and/or high PD-L1 CPS staining. CONCLUSION: Among patients with persistent or recurrent LSCC, CD103+ TILs only modestly correlated with PD-L1 CPS. A combined biomarker score incorporating CD103+ TILs and PD-L1 CPS greatly enhanced survival discrimination. This model may have additional utility in predicting the clinical benefit of immunotherapies in persistent or recurrent LSCC in the future.


Asunto(s)
Neoplasias de Cabeza y Cuello , Linfocitos Infiltrantes de Tumor , Humanos , Linfocitos Infiltrantes de Tumor/patología , Antígeno B7-H1 , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Estudios Retrospectivos , Neoplasias de Cabeza y Cuello/patología , Biomarcadores de Tumor
19.
Histopathology ; 58(2): 169-79, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21323945

RESUMEN

AIMS: To develop an immunohistochemical strategy for distinguishing renal oncocytoma (RO) from the eosinophilic variant of chromophobe (ChRCC), and papillary (PRCC) and clear cell (CRCC) renal cell carcinoma containing eosinophilic cytoplasm in core biopsy specimens. METHODS AND RESULTS: Cluster analysis was performed on immunohistochemical data from 21 RO, 16 ChRCC, 16 CRCC and 20 PRCC patients. A panel of CK7, C-kit, S100A1 and vimentin clustered into four groups. Cluster A (94% ChRCC) expressed C-kit and CK7 and lacked S100A1 and vimentin. Cluster B (95% RO) expressed C-kit, S100A1, focal CK7 (single or small clusters of cells) and lacked vimentin. Cluster C comprised a mixture of PRCC and CRCC with no expression of C-kit or CK7 and variable S100A1 and vimentin. PRCC with strong expression of CK7 clustered into group D. A panel of S100A1 (positive) and focal CK7 expression distinguished RO from ChRCC with 91% sensitivity and 93% specificity. A panel of vimentin (negative) and C-kit (positive) distinguished RO from CRCC with 83% sensitivity and 86% specificity and RO from PRCC with 79% sensitivity and 88% specificity. CONCLUSIONS: Hierarchical cluster analysis is an effective approach to analyse high-volume immunohistochemical data to generate an optimal panel in the differential diagnosis of oncocytoma from its mimics.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Renales/diagnóstico , Adenoma Oxifílico/química , Adenoma Oxifílico/diagnóstico , Carcinoma de Células Renales/química , Carcinoma de Células Renales/diagnóstico , Análisis por Conglomerados , Diagnóstico Diferencial , Humanos , Inmunohistoquímica , Queratina-7/análisis , Neoplasias Renales/química , Proteínas Proto-Oncogénicas c-kit/análisis , Proteínas S100/análisis , Sensibilidad y Especificidad , Análisis de Matrices Tisulares , Vimentina/análisis
20.
Nat Med ; 8(8): 816-24, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12118244

RESUMEN

Histopathology is insufficient to predict disease progression and clinical outcome in lung adenocarcinoma. Here we show that gene-expression profiles based on microarray analysis can be used to predict patient survival in early-stage lung adenocarcinomas. Genes most related to survival were identified with univariate Cox analysis. Using either two equivalent but independent training and testing sets, or 'leave-one-out' cross-validation analysis with all tumors, a risk index based on the top 50 genes identified low-risk and high-risk stage I lung adenocarcinomas, which differed significantly with respect to survival. This risk index was then validated using an independent sample of lung adenocarcinomas that predicted high- and low-risk groups. This index included genes not previously associated with survival. The identification of a set of genes that predict survival in early-stage lung adenocarcinoma allows delineation of a high-risk group that may benefit from adjuvant therapy.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Perfilación de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Adenocarcinoma/clasificación , Adenocarcinoma/patología , Estudios de Cohortes , Humanos , Neoplasias Pulmonares/clasificación , Neoplasias Pulmonares/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Valor Predictivo de las Pruebas , Pronóstico , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA