Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 12(534)2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32161105

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer that does not respond to endocrine therapy or human epidermal growth factor receptor 2 (HER2)-targeted therapies. Individuals with TNBC experience higher rates of relapse and shorter overall survival compared to patients with receptor-positive breast cancer subtypes. Preclinical discoveries are needed to identify, develop, and advance new drug targets to improve outcomes for patients with TNBC. Here, we report that MYCN, an oncogene typically overexpressed in tumors of the nervous system or with neuroendocrine features, is heterogeneously expressed within a substantial fraction of primary and recurrent TNBC and is expressed in an even higher fraction of TNBCs that do not display a pathological complete response after neoadjuvant chemotherapy. We performed high-throughput chemical screens on TNBC cell lines with varying amounts of MYCN expression and determined that cells with higher expression of MYCN were more sensitive to bromodomain and extraterminal motif (BET) inhibitors. Combined BET and MEK inhibition resulted in a synergistic decrease in viability, both in vitro and in vivo, using cell lines and patient-derived xenograft (PDX) models. Our preclinical data provide a rationale to advance a combination of BET and MEK inhibitors to clinical investigation for patients with advanced MYCN-expressing TNBC.


Assuntos
Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Proteínas/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas , Animais , Linhagem Celular Tumoral , Humanos , Proteína Proto-Oncogênica N-Myc/genética , Recidiva Local de Neoplasia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Clin Cancer Res ; 26(9): 2111-2123, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31822498

RESUMO

PURPOSE: Preclinical data demonstrating androgen receptor (AR)-positive (AR+) triple-negative breast cancer (TNBC) cells are sensitive to AR antagonists, and PI3K inhibition catalyzed an investigator-initiated, multi-institutional phase Ib/II study TBCRC032. The trial investigated the safety and efficacy of the AR-antagonist enzalutamide alone or in combination with the PI3K inhibitor taselisib in patients with metastatic AR+ (≥10%) breast cancer. PATIENTS AND METHODS: Phase Ib patients [estrogen receptor positive (ER+) or TNBC] with AR+ breast cancer received 160 mg enzalutamide in combination with taselisib to determine dose-limiting toxicities and the maximum tolerated dose (MTD). Phase II TNBC patients were randomized to receive either enzalutamide alone or in combination with 4 mg taselisib until disease progression. Primary endpoint was clinical benefit rate (CBR) at 16 weeks. RESULTS: The combination was tolerated, and the MTD was not reached. The adverse events were hyperglycemia and skin rash. Overall, CBR for evaluable patients receiving the combination was 35.7%, and median progression-free survival (PFS) was 3.4 months. Luminal AR (LAR) TNBC subtype patients trended toward better response compared with non-LAR (75.0% vs. 12.5%, P = 0.06), and increased PFS (4.6 vs. 2.0 months, P = 0.082). Genomic analyses revealed subtype-specific treatment response, and novel FGFR2 fusions and AR splice variants. CONCLUSIONS: The combination of enzalutamide and taselisib increased CBR in TNBC patients with AR+ tumors. Correlative analyses suggest AR protein expression alone is insufficient for identifying patients with AR-dependent tumors and knowledge of tumor LAR subtype and AR splice variants may identify patients more or less likely to benefit from AR antagonists.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Receptores Androgênicos/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Antagonistas de Receptores de Andrógenos/administração & dosagem , Antagonistas de Receptores de Andrógenos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Benzamidas , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Feminino , Humanos , Imidazóis/administração & dosagem , Imidazóis/efeitos adversos , Pessoa de Meia-Idade , Metástase Neoplásica , Nitrilas , Oxazepinas/administração & dosagem , Oxazepinas/efeitos adversos , Feniltioidantoína/administração & dosagem , Feniltioidantoína/efeitos adversos , Feniltioidantoína/análogos & derivados , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Taxa de Sobrevida , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
3.
PLoS One ; 14(6): e0218458, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31216312

RESUMO

p63 is a transcriptional regulator of ectodermal development that is required for basal cell proliferation and stem cell maintenance. p73 is a closely related p53 family member that is expressed in select p63-positive basal cells and can heterodimerize with p63. p73-/- mice lack multiciliated cells and have reduced numbers of basal epithelial cells in select tissues; however, the role of p73 in basal epithelial cells is unknown. Herein, we show that p73-deficient mice exhibit delayed wound healing despite morphologically normal-appearing skin. The delay in wound healing is accompanied by decreased proliferation and increased levels of biomarkers of the DNA damage response in basal keratinocytes at the epidermal wound edge. In wild-type mice, this same cell population exhibited increased p73 expression after wounding. Analyzing single-cell transcriptomic data, we found that p73 was expressed by epidermal and hair follicle stem cells, cell types required for wound healing. Moreover, we discovered that p73 isoforms expressed in the skin (ΔNp73) enhance p63-mediated expression of keratinocyte genes during cellular reprogramming from a mesenchymal to basal keratinocyte-like cell. We identified a set of 44 genes directly or indirectly regulated by ΔNp73 that are involved in skin development, cell junctions, cornification, proliferation, and wound healing. Our results establish a role for p73 in cutaneous wound healing through regulation of basal keratinocyte function.


Assuntos
Ectoderma/metabolismo , Pele/metabolismo , Proteína Tumoral p73/genética , Cicatrização/genética , Animais , Proliferação de Células/genética , Dano ao DNA/genética , Ectoderma/crescimento & desenvolvimento , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Humanos , Queratinócitos/metabolismo , Camundongos , Camundongos Knockout , Análise de Célula Única , Pele/crescimento & desenvolvimento , Pele/lesões , Nicho de Células-Tronco/genética , Transativadores/genética
4.
iScience ; 8: 236-249, 2018 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-30340069

RESUMO

We report that p73 is expressed in ovarian granulosa cells and that loss of p73 leads to attenuated follicle development, ovulation, and corpus luteum formation, resulting in decreased levels of circulating progesterone and defects in mammary gland branching. Ectopic progesterone in p73-deficient mice completely rescued the mammary branching and partially rescued the ovarian follicle development defects. Performing RNA sequencing (RNA-seq) on transcripts from murine wild-type and p73-deficient antral follicles, we discovered differentially expressed genes that regulate biological adhesion programs. Through modulation of p73 expression in murine granulosa cells and transformed cell lines, followed by RNA-seq and chromatin immunoprecipitation sequencing, we discovered p73-dependent regulation of a gene set necessary for cell adhesion and migration and components of the focimatrix (focal intra-epithelial matrix), a basal lamina between granulosa cells that promotes follicle maturation. In summary, p73 is essential for ovarian folliculogenesis and functions as a key regulator of a gene network involved in cell-to-cell adhesion and migration.

5.
Ozone Sci Eng ; 39(1): 61-66, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29170573

RESUMO

Whole blood is a complex mixture of biological and chemical species. Its pretreatment, which is often conducted by dry ashing, is needed before the analyses of trace metals in whole blood. Recently photo-Fenton Advanced Oxidation Process (AOP) process has been used in the pretreatment of whole blood. Two new AOP processes using simple heating and microwave irradiation have been developed in the current work to pretreat blood samples. The treatments are based on a Fenton-like AOP with acid deactivation of the enzyme catalase. The first treatment is performed with a lab oven over 5 h, while the second uses microwave irradiation for 6 min. These methods allow for either cost-effective pretreatment through the use of the lab oven, or time savings through the use of the microwave oven. The degradations of blood and pure hemoglobin samples are compared through UV/visible spectroscopy, and the copper concentration in the treated samples were analyzed via anodic stripping voltammetry as a demonstration of analyzing trace metals in the pretreated whole blood.

6.
PLoS One ; 11(6): e0157368, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27310713

RESUMO

Triple-negative breast cancer (TNBC) is a heterogeneous disease that can be classified into distinct molecular subtypes by gene expression profiling. Considered a difficult-to-treat cancer, a fraction of TNBC patients benefit significantly from neoadjuvant chemotherapy and have far better overall survival. Outside of BRCA1/2 mutation status, biomarkers do not exist to identify patients most likely to respond to current chemotherapy; and, to date, no FDA-approved targeted therapies are available for TNBC patients. Previously, we developed an approach to identify six molecular subtypes TNBC (TNBCtype), with each subtype displaying unique ontologies and differential response to standard-of-care chemotherapy. Given the complexity of the varying histological landscape of tumor specimens, we used histopathological quantification and laser-capture microdissection to determine that transcripts in the previously described immunomodulatory (IM) and mesenchymal stem-like (MSL) subtypes were contributed from infiltrating lymphocytes and tumor-associated stromal cells, respectively. Therefore, we refined TNBC molecular subtypes from six (TNBCtype) into four (TNBCtype-4) tumor-specific subtypes (BL1, BL2, M and LAR) and demonstrate differences in diagnosis age, grade, local and distant disease progression and histopathology. Using five publicly available, neoadjuvant chemotherapy breast cancer gene expression datasets, we retrospectively evaluated chemotherapy response of over 300 TNBC patients from pretreatment biopsies subtyped using either the intrinsic (PAM50) or TNBCtype approaches. Combined analysis of TNBC patients demonstrated that TNBC subtypes significantly differ in response to similar neoadjuvant chemotherapy with 41% of BL1 patients achieving a pathological complete response compared to 18% for BL2 and 29% for LAR with 95% confidence intervals (CIs; [33, 51], [9, 28], [17, 41], respectively). Collectively, we provide pre-clinical data that could inform clinical trials designed to test the hypothesis that improved outcomes can be achieved for TNBC patients, if selection and combination of existing chemotherapies is directed by knowledge of molecular TNBC subtypes.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Terapia Neoadjuvante/métodos , Proteínas de Neoplasias/genética , Neoplasias de Mama Triplo Negativas/classificação , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Antineoplásicos/uso terapêutico , Biologia Computacional , Conjuntos de Dados como Assunto , Progressão da Doença , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Microdissecção e Captura a Laser , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/patologia , Análise em Microsséries , Gradação de Tumores , Proteínas de Neoplasias/metabolismo , Estudos Retrospectivos , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Análise de Sobrevida , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/mortalidade
7.
Mol Cancer ; 9: 95, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20429933

RESUMO

BACKGROUND: Autophagy is characterized by the sequestration of cytoplasm and organelles into multimembrane vesicles and subsequent degradation by the cell's lysosomal system. It is linked to many physiological functions in human cells including stress response, protein degradation, organelle turnover, caspase-independent cell death and tumor suppression. Malignant transformation is frequently associated with deregulation of autophagy and several tumor suppressors can modulate autophagic processes. The tumor suppressor p53 can induce autophagy after metabolic or genotoxic stress through transcriptionally-dependent and -independent mechanisms. In this study we expand on the former mechanism by functionally characterizing a p53 family target gene, ISG20L1 under conditions of genotoxic stress. RESULTS: We identified a p53 target gene, ISG20L1, and show that transcription of the gene can be regulated by all three p53 family members (p53, p63, and p73). We generated an antibody to ISG20L1 and found that it localizes to the nucleolar and perinucleolar regions of the nucleus and its protein levels increase in a p53- and p73-dependent manner after various forms of genotoxic stress. When ectopically expressed in epithelial cancer-derived cell lines, ISG20L1 expression decreased clonogenic survival without a concomitant elevation in apoptosis and this effect was partially rescued in cells that were ATG5 deficient. Knockdown of ISG20L1 did not alter 5-FU induced apoptosis as assessed by PARP and caspase-3 cleavage, sub-G1 content, and DNA laddering. Thus, we investigated the role of ISG20L1 in autophagy, a process commonly associated with type II cell death, and found that ISG20L1 knockdown decreased levels of autophagic vacuoles and LC3-II after genotoxic stress as assessed by electron microscopy, biochemical, and immunohistochemical measurements of LC3-II. CONCLUSIONS: Our identification of ISG20L1 as a p53 family target and discovery that modulation of this target can regulate autophagic processes further strengthens the connection between p53 signaling and autophagy. Given the keen interest in targeting autophagy as an anticancer therapeutic approach in tumor cells that are defective in apoptosis, investigation of genes and signaling pathways involved in cell death associated with autophagy is critical.


Assuntos
Autofagia/fisiologia , Dano ao DNA/fisiologia , Exodesoxirribonucleases/metabolismo , Regulação da Expressão Gênica , Proteína Supressora de Tumor p53/metabolismo , Western Blotting , Linhagem Celular , Separação Celular , Imunoprecipitação da Cromatina , Exodesoxirribonucleases/genética , Citometria de Fluxo , Imunofluorescência , Humanos , Imuno-Histoquímica , Microscopia Eletrônica de Transmissão , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Proteína Supressora de Tumor p53/genética
8.
Clin Cancer Res ; 16(2): 681-90, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20068102

RESUMO

PURPOSE: To identify molecular markers of pathologic response to neoadjuvant paclitaxel/radiation treatment, protein and gene expression profiling were done on pretreatment biopsies. EXPERIMENTAL DESIGN: Patients with high-risk, operable breast cancer were treated with three cycles of paclitaxel followed by concurrent paclitaxel/radiation. Tumor tissue from pretreatment biopsies was obtained from 19 of the 38 patients enrolled in the study. Protein and gene expression profiling were done on serial sections of the biopsies from patients that achieved a pathologic complete response (pCR) and compared to those with residual disease, non-pCR (NR). RESULTS: Proteomic and validation immunohistochemical analyses revealed that alpha-defensins (DEFA) were overexpressed in tumors from patients with a pCR. Gene expression analysis revealed that MAP2, a microtubule-associated protein, had significantly higher levels of expression in patients achieving a pCR. Elevation of MAP2 in breast cancer cell lines led to increased paclitaxel sensitivity. Furthermore, expression of genes that are associated with the basal-like, triple-negative phenotype were enriched in tumors from patients with a pCR. Analysis of a larger panel of tumors from patients receiving presurgical taxane-based treatment showed that DEFA and MAP2 expression as well as histologic features of inflammation were all statistically associated with response to therapy at the time of surgery. CONCLUSION: We show the utility of molecular profiling of pretreatment biopsies to discover markers of response. Our results suggest the potential use of immune signaling molecules such as DEFA as well as MAP2, a microtubule-associated protein, as tumor markers that associate with response to neoadjuvant taxane-based therapy.


Assuntos
Biomarcadores Farmacológicos/análise , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Paclitaxel/uso terapêutico , Taxoides/uso terapêutico , Biomarcadores Farmacológicos/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Carcinoma/tratamento farmacológico , Carcinoma/radioterapia , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genômica , Humanos , Metaboloma/efeitos dos fármacos , Terapia Neoadjuvante , Análise de Sequência com Séries de Oligonucleotídeos , Proteômica , Taxoides/farmacologia , Células Tumorais Cultivadas
9.
Dev Biol ; 329(1): 130-9, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19272371

RESUMO

p63, a homolog of the tumor suppressor p53, is critical for the development and maintenance of complex epithelia. The developmentally regulated p63 isoform, DeltaNp63, can act as a transcriptional repressor, but the link between the transcriptional functions of p63 and its biological roles is unclear. Based on our initial finding that the mesoderm-inducing factor activin A is suppressed by DeltaNp63 in human keratinocytes, we investigated the role of DeltaNp63 in regulating mesoderm induction during early Xenopus laevis development. We find that down-regulation of DeltaNp63 by morpholino injection in the early Xenopus embryo potentiates mesoderm formation whereas ectopic expression of DeltaNp63 inhibits mesoderm formation. Furthermore, we show that mesodermal induction after down-regulation of DeltaNp63 is dependent on p53. We propose that a key function for p63 in defining a squamous epithelial phenotype is to actively suppress mesodermal cell fates during early development. Collectively, we show that there is a distinct requirement for different p53 family members during the development of both mesodermal and ectodermal tissues. These findings have implications for the role of p63 and p53 in both development and tumorigenesis of human epithelia.


Assuntos
Mesoderma/fisiologia , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Transativadores/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/metabolismo , Ativinas/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Embrião não Mamífero , Epitélio/metabolismo , Humanos , Imuno-Histoquímica , Queratinócitos/citologia , Queratinócitos/metabolismo , Mesoderma/metabolismo , Modelos Biológicos , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Cultura de Órgãos , Fosfoproteínas/genética , RNA Interferente Pequeno/metabolismo , Transativadores/genética , Fatores de Transcrição , Transfecção , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/farmacologia , Proteínas de Xenopus/genética , Xenopus laevis/genética , Xenopus laevis/fisiologia
10.
Cancer Res ; 65(6): 2314-20, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15781645

RESUMO

DeltaNp63alpha is a nuclear transcription factor that maintains epithelial progenitor cell populations, is overexpressed in several epithelial cancers, and can negatively regulate apoptosis. However, the mechanisms by which DeltaNp63alpha promotes cell survival are unclear. DeltaNp63alpha has been reported to act as a transcriptional repressor, but specific target genes directly repressed by DeltaNp63alpha remain unidentified. Here, we present evidence that DeltaNp63alpha functions to negatively regulate the proapoptotic protein IGFBP-3. Disruption of p63 expression in squamous epithelial cells increases IGFBP-3 expression, whereas ectopic expression of DeltaNp63alpha down-regulates IGFBP-3. DeltaNp63alpha binds to sites in the IGFBP-3 gene in vivo and can modulate transcription through these sites. Furthermore, DeltaNp63alpha and IGFBP-3 expression patterns are inversely correlated in normal squamous epithelium and squamous cell carcinomas. These data suggest that IGFBP-3 is a target of transcriptional repression by DeltaNp63alpha and that this repression represents a mechanism by which tumors that overexpress p63 may be protected from apoptosis.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Fosfoproteínas/genética , Transativadores/genética , Apoptose/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Genes Supressores de Tumor , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Queratinócitos/citologia , Queratinócitos/metabolismo , Queratinócitos/fisiologia , Fosfoproteínas/biossíntese , Transativadores/biossíntese , Fatores de Transcrição , Transcrição Gênica/fisiologia , Transfecção , Proteínas Supressoras de Tumor , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...