Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Cancer Discov ; 11(5): 1100-1117, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33419761

RESUMO

The clinical benefit of PD-1 blockade can be improved by combination with CTLA4 inhibition but is commensurate with significant immune-related adverse events suboptimally limiting the doses of anti-CTLA4 mAb that can be used. MEDI5752 is a monovalent bispecific antibody designed to suppress the PD-1 pathway and provide modulated CTLA4 inhibition favoring enhanced blockade on PD-1+ activated T cells. We show that MEDI5752 preferentially saturates CTLA4 on PD-1+ T cells versus PD-1- T cells, reducing the dose required to elicit IL2 secretion. Unlike conventional PD-1/CTLA4 mAbs, MEDI5752 leads to the rapid internalization and degradation of PD-1. Moreover, we show that MEDI5752 preferentially localizes and accumulates in tumors providing enhanced activity when compared with a combination of mAbs targeting PD-1 and CTLA4 in vivo. Following treatment with MEDI5752, robust partial responses were observed in two patients with advanced solid tumors. MEDI5752 represents a novel immunotherapy engineered to preferentially inhibit CTLA4 on PD-1+ T cells. SIGNIFICANCE: The unique characteristics of MEDI5752 represent a novel immunotherapy engineered to direct CTLA4 inhibition to PD-1+ T cells with the potential for differentiated activity when compared with current conventional mAb combination strategies targeting PD-1 and CTLA4. This molecule therefore represents a step forward in the rational design of cancer immunotherapy.See related commentary by Burton and Tawbi, p. 1008.This article is highlighted in the In This Issue feature, p. 995.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma de Células Claras/tratamento farmacológico , Antígeno CTLA-4/metabolismo , Humanos , Imunoterapia , Neoplasias Renais/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Linfócitos T/imunologia
2.
Dalton Trans ; 49(28): 9811-9819, 2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32633296

RESUMO

1,4-Di(1-ethyl-1,2,3-triazol-5-yl)butane (bbtre) was prepared by lithiation of 1-ethyl-1,2,3-triazole, followed by alkylation with 1,4-dibromobutane. The ligand bbtre forms a three-dimensional network with Fe(ii), [Fe(bbtre)3](ClO4)2·2CH3CN, that exhibits thermally induced spin crossover (SCO). A change of temperature or change of spin state results in various types of structural transformation, leading to different structures that are stable in strictly defined temperature ranges. As a result, there are three spin crossover transitions arranged via two different paths. Thus, cooling below 280 K involves a HT(HS) → LT(HS) (HT, high temperature structure; LT, low temperature structure; HS, high spin) phase transition (PT), which is associated with conformational changes of the bbtre molecules and with deformation of the polymeric skeleton. In the LT phase incomplete and reversible LT(HS) ⇄ LT(HS/LS) spin crossover occurs (LS, low spin). In contrast, rapid cooling (of a sample not previously thermally treated) allows the HT(HS) → LT(HS) phase transition to be avoided, and so complete HT(HS) → HT1(LS) SCO occurs. This means that the PT plays the role of a switch, which allows a choice of one of two ways in which the SCO will proceed. After rapid cooling, further heating to 150 K and subsequent cooling results in a reversible HT1(HS) ⇄ HT1(LS) spin crossover (T↓1/2 = 130 K, T↑1/2 = 131 K). However, raising the temperature to 170-200 K leads to formation of a modulated structure HT2(HS) exhibiting the next reversible HT2(HS) ⇄ HT2(LS) SCO (T↓1/2 = 121 K, T↑1/2 = 123 K). Finally, heating above 200 K involves the HT2(HS) → LT(HS) PT and results in a LT(HS) structure exhibiting incomplete LT(HS) ⇄ LT(HS/LS) spin crossover.

3.
Mayo Clin Proc ; 93(3): 307-320, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29502561

RESUMO

OBJECTIVE: To evaluate myeloid differentiation primary response gene 88 (MyD88) and Toll-like receptor 4 (TLR4) expression in relation to clinical features of epithelial ovarian cancer, histologic subtypes, and overall survival. PATIENTS AND METHODS: We conducted centralized immunohistochemical staining, semi-quantitative scoring, and survival analysis in 5263 patients participating in the Ovarian Tumor Tissue Analysis consortium. Patients were diagnosed between January 1, 1978, and December 31, 2014, including 2865 high-grade serous ovarian carcinomas (HGSOCs), with more than 12,000 person-years of follow-up time. Tissue microarrays were stained for MyD88 and TLR4, and staining intensity was classified using a 2-tiered system for each marker (weak vs strong). RESULTS: Expression of MyD88 and TLR4 was similar in all histotypes except clear cell ovarian cancer, which showed reduced expression compared with other histotypes (P<.001 for both). In HGSOC, strong MyD88 expression was modestly associated with shortened overall survival (hazard ratio [HR], 1.13; 95% CI, 1.01-1.26; P=.04) but was also associated with advanced stage (P<.001). The expression of TLR4 was not associated with survival. In low-grade serous ovarian cancer (LGSOC), strong expression of both MyD88 and TLR4 was associated with favorable survival (HR [95% CI], 0.49 [0.29-0.84] and 0.44 [0.21-0.89], respectively; P=.009 and P=.02, respectively). CONCLUSION: Results are consistent with an association between strong MyD88 staining and advanced stage and poorer survival in HGSOC and demonstrate correlation between strong MyD88 and TLR4 staining and improved survival in LGSOC, highlighting the biological differences between the 2 serous histotypes.


Assuntos
Carcinoma Epitelial do Ovário/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Neoplasias Ovarianas/metabolismo , Receptor 4 Toll-Like/metabolismo , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Carcinoma Epitelial do Ovário/mortalidade , Feminino , Humanos , Imuno-Histoquímica/métodos , Pessoa de Meia-Idade , Neoplasias Ovarianas/mortalidade , Análise de Sobrevida , Análise Serial de Tecidos/métodos
4.
JAMA Oncol ; 3(12): e173290, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29049607

RESUMO

Importance: Cytotoxic CD8+ tumor-infiltrating lymphocytes (TILs) participate in immune control of epithelial ovarian cancer; however, little is known about prognostic patterns of CD8+ TILs by histotype and in relation to other clinical factors. Objective: To define the prognostic role of CD8+ TILs in epithelial ovarian cancer. Design, Setting, and Participants: This was a multicenter observational, prospective survival cohort study of the Ovarian Tumor Tissue Analysis Consortium. More than 5500 patients, including 3196 with high-grade serous ovarian carcinomas (HGSOCs), were followed prospectively for over 24 650 person-years. Exposures: Following immunohistochemical analysis, CD8+ TILs were identified within the epithelial components of tumor islets. Patients were grouped based on the estimated number of CD8+ TILs per high-powered field: negative (none), low (1-2), moderate (3-19), and high (≥20). CD8+ TILs in a subset of patients were also assessed in a quantitative, uncategorized manner, and the functional form of associations with survival was assessed using penalized B-splines. Main Outcomes and Measures: Overall survival time. Results: The final sample included 5577 women; mean age at diagnosis was 58.4 years (median, 58.2 years). Among the 5 major invasive histotypes, HGSOCs showed the most infiltration. CD8+ TILs in HGSOCs were significantly associated with longer overall survival; median survival was 2.8 years for patients with no CD8+ TILs and 3.0 years, 3.8 years, and 5.1 years for patients with low, moderate, or high levels of CD8+ TILs, respectively (P value for trend = 4.2 × 10−16). A survival benefit was also observed among women with endometrioid and mucinous carcinomas, but not for those with the other histotypes. Among HGSOCs, CD8+ TILs were favorable regardless of extent of residual disease following cytoreduction, known standard treatment, and germline BRCA1 pathogenic mutation, but were not prognostic for BRCA2 mutation carriers. Evaluation of uncategorized CD8+ TIL counts showed a near-log-linear functional form. Conclusions and Relevance: This study demonstrates the histotype-specific nature of immune infiltration and provides definitive evidence for a dose-response relationship between CD8+ TILs and HGSOC survival. That the extent of infiltration is prognostic, not merely its presence or absence, suggests that understanding factors that drive infiltration will be the key to unraveling outcome heterogeneity in this cancer.


Assuntos
Antígenos CD8/metabolismo , Carcinoma Epitelial do Ovário/tratamento farmacológico , Cistadenocarcinoma Seroso/imunologia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Ovarianas/imunologia , Proteína BRCA2/genética , Carcinoma Epitelial do Ovário/imunologia , Carcinoma Epitelial do Ovário/patologia , Estudos de Coortes , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Mutação , Gradação de Tumores , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Estudos Prospectivos , Análise de Sobrevida , Resultado do Tratamento
5.
Cancer Res ; 77(18): 4921-4933, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28720576

RESUMO

The core LATS kinases of the Hippo tumor suppressor pathway phosphorylate and inhibit the downstream transcriptional co-activators YAP and TAZ, which are implicated in various cancers. Recent studies have identified various E3 ubiquitin ligases that negatively regulate the Hippo pathway via ubiquitination, yet few deubiquitinating enzymes (DUB) have been implicated. In this study, we report the DUB USP9X is an important regulator of the core kinases of this pathway. USP9X interacted strongly with LATS kinase and to a lesser extent with WW45, KIBRA, and Angiomotin, and LATS co-migrated exclusively with USP9X during gel filtration chromatography analysis. Knockdown of USP9X significantly downregulated and destabilized LATS and resulted in enhanced nuclear translocation of YAP and TAZ, accompanied with activation of their target genes. In the absence of USP9X, cells exhibited an epithelial-to-mesenchymal transition phenotype, acquired anchorage-independent growth in soft agar, and led to enlarged, disorganized, three-dimensional acini. YAP/TAZ target gene activation in response to USP9X knockdown was suppressed by knockdown of YAP, TAZ, and TEAD2. Deletion of USP9X in mouse embryonic fibroblasts resulted in significant downregulation of LATS. Furthermore, USP9X protein expression correlated positively with LATS but negatively with YAP/TAZ in pancreatic cancer tissues as well as pancreatic and breast cancer cell lines. Overall, these results strongly indicate that USP9X potentiates LATS kinase to suppress tumor growth. Cancer Res; 77(18); 4921-33. ©2017 AACR.


Assuntos
Neoplasias da Mama/patologia , Endopeptidases/fisiologia , Neoplasias Pancreáticas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina Tiolesterase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Angiomotinas , Animais , Apoptose , Biomarcadores Tumorais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Seguimentos , Via de Sinalização Hippo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos , Estadiamento de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Prognóstico , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Taxa de Sobrevida , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitinação , Proteínas de Sinalização YAP
6.
Oncotarget ; 7(12): 13706-16, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26885614

RESUMO

Dysregulation of Hippo pathway results in activation of transcriptional co-activators YAP/TAZ in breast cancer. Previously, we showed that overexpression of TAZ in breast cancer promotes cell migration, invasion and tumorigenesis. Here, we show that upregulation of TAZ in breast cancers could also be due to dysregulation of TAZ transcription. Heregulin ß1 (HRG1) increases TAZ mRNA level in breast cancer cells. TAZ is a direct target of MRTF/SRF transcriptional factors which are activated by HRG1. Both MRTF/SRF and TAZ are the important downstream effectors enhancing cell migration induced by HRG1. TAZ mRNA level is correlated with nuclear localization of MRTF in breast cancer cells and the mRNA level of MRTF/SRF direct target genes in breast cancers, indicating the correlation between MRTF/SRF activity and TAZ expression. Our results provide new insights into the transcriptional regulation of TAZ and dysregulation mechanism of TAZ in breast cancer, which could be a new therapeutic strategy for breast cancer.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Hemeproteínas/metabolismo , Fator de Resposta Sérica/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/genética , Aciltransferases , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Proliferação de Células , Feminino , Hemeproteínas/genética , Humanos , Fator de Resposta Sérica/genética , Transdução de Sinais , Transativadores/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA