Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Am J Cancer Res ; 12(1): 123-137, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35141008

RESUMO

The atezolizumab (Tecentriq), a humanized antibody against human programmed death ligand 1 (PD-L1), combined with nab-paclitaxel was granted with accelerated approval to treat unresectable locally advanced or metastatic triple-negative breast cancer (TNBC) due to the encouraging positive results of the phase 3 IMpassion130 trial using PD-L1 biomarker from immune cells to stratify patients. However, the post-market study IMpassion131 did not support the original observation, resulting in the voluntary withdrawal of atezolizumab from the indication in breast cancer by Genentech in 2021. Emerging evidence has revealed a high frequency of false negative result using the standard immunohistochemical (IHC) staining due to heavy glycosylation of PD-L1. The removal of glycosylation prevents from the false negative staining, enabling more accurate assessment of PD-L1 levels and improving prediction for response to immune checkpoint therapy. In the present study, the natural and de-glycosylated PD-L1 expression in tumor and immune cells from nine TNBC patients were analyzed by using clone 28-8 monoclonal antibody to correlate with treatment outcome. Our results demonstrate that: (1) Removal of the glycosylation indeed enhances the detection of PD-L1 by IHC staining, (2) The PD-L1 levels on tumor cell surface after removal of the glycosylation correlates well with clinical responses for atezolizumab treatment; (3) The criteria used in the IMpassion130 and IMpassion131 trials which scored the natural PD-L1 in the immune cells failed to correlate with the clinical response. Taken together, tumor cell surface staining of PD-L1 with de-glycosylation has a significant correlation with the clinical response for atezolizumab treatment, suggesting that treatment of atezolizumab may be worthy of further consideration with de-glycosylation procedure as a patient stratification strategy. A larger cohort to validate this important issue is warranted to ensure right patient population who could benefit from the existing FDA-approved drugs.

2.
Cell Rep ; 36(8): 109537, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34433039

RESUMO

Increased DNA replication and metastasis are hallmarks of cancer progression, while deregulated proliferation often triggers sustained replication stresses in cancer cells. How cancer cells overcome the growth stress and proceed to metastasis remains largely elusive. Proliferating cell nuclear antigen (PCNA) is an indispensable component of the DNA replication machinery. Here, we show that phosphorylation of PCNA on tyrosine 211 (pY211-PCNA) regulates DNA metabolism and tumor microenvironment. Abrogation of pY211-PCNA blocks fork processivity, resulting in biogenesis of single-stranded DNA (ssDNA) through a MRE11-dependent mechanism. The cytosolic ssDNA subsequently induces inflammatory cytokines through a cyclic GMP-AMP synthetase (cGAS)-dependent cascade, triggering an anti-tumor immunity by natural killer (NK) cells to suppress distant metastasis. Expression of pY211-PCNA is inversely correlated with cytosolic ssDNA and associated with poor survival in patients with cancer. Our results pave the way to biomarkers and therapies exploiting immune responsiveness to target metastatic cancer.


Assuntos
Neoplasias Experimentais/imunologia , Antígeno Nuclear de Célula em Proliferação/imunologia , Evasão Tumoral , Microambiente Tumoral/imunologia , Animais , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/mortalidade , Fosforilação , Antígeno Nuclear de Célula em Proliferação/genética , Microambiente Tumoral/genética , Tirosina/genética , Tirosina/imunologia
3.
Cell Death Dis ; 11(10): 838, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-33037177

RESUMO

An increasing number of studies have shown that long noncoding RNAs (lncRNAs) play important roles in tumor development and progression. However, their involvement in head and neck squamous cell carcinoma (HNSCC) remains largely unknown. Epigenetic regulation is one major mechanism utilized by cancer cells to control lncRNA expression. We identified that lncRNA VENTXP1 was epigenetically silenced in multiple cancer types, and its lower expression was correlated with poorer survival in HNSCC patients. Through in silico analysis and experimental validation, we identified miR-205-5p and its direct interacting partner of VENTXP1, which regulates HNSCC cell proliferation and tumorigenicity. Using RNA-seq and differential gene expression analysis, we further identified ANKRD2 as a miR-205-5p target, which plays an essential role in modulating NF-kB signaling. These findings suggest that VENTXP1 inhibits tumor growth via suppressing miR-205-5p/ANKRD2-mediated NF-kB signaling in HNSCC. Thus, pharmaceutical targeting of DNA methylation to restore VENTXP1 expression might constitute a therapeutic strategy for HNSCC.


Assuntos
Neoplasias de Cabeça e Pescoço/genética , Proteínas de Homeodomínio/metabolismo , MicroRNAs/genética , Proteínas Musculares/genética , NF-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas/genética , Movimento Celular/genética , Proliferação de Células/genética , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Homeodomínio/genética , Humanos , RNA Longo não Codificante/genética , Transdução de Sinais
4.
Am J Cancer Res ; 8(11): 2296-2310, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555745

RESUMO

ANRIL (CDKN2B antisense RNA 1, CDKN2B-AS1) is involved in the progression of various cancers. However, its role in head and neck squamous cell carcinoma (HNSCC) remains unclear. In this study, we found that ANRIL expression was upregulated in HNSCC and correlated with tumor progression. Further functional analysis showed that knockdown of ANRIL significantly inhibited proliferation in vivo and in vitro. ANRIL functioned as a ceRNA (competing endogenous RNAs) for miR-125a-3p and upregulated FGFR1 (fibroblast growth factor receptor-1), which could promote tumor growth. Moreover, we confirmed that ANRIL promoted HNSCC activity via FGFR1 with a FGFR1 inhibitor in vivo and in vitro. Thus, it could be concluded that ANRIL promoted the progression of HNSCC via miR-125a-3p/FGFR1/MAPK signaling, which might provide a new target for the diagnosis and treatment of HNSCC.

5.
Am J Cancer Res ; 7(12): 2587-2599, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29312811

RESUMO

Arginine methylation of the epidermal growth factor receptor (meEGFR) increases the binding affinity of EGFR ligands and is reported to have a role in predicting response to anti-EGFR agents. This study investigated the predictive impact of meEGFR in metastatic colorectal cancer (mCRC) patients treated with anti-EGFR agents. Two patient cohorts were evaluated. Cohort 1 consisted of mCRC patients with documented disease progression following anti-EGFR treatment. Circulating tumor cells (CTCs) were isolated and distinguished based on CD45- and Epcam+. Cohort 2 consisted of formalin fixed paraffin-embedded (FFPE) blocks from a prospective cohort. meEGFR in both cohorts was identified by positive staining for me-R198/200 EGFR signal. CTCs were identified in 30 out of 47 cases in cohort 1. Of those 30, meEGFR-CTCs were identified in 19 cases. Mean total meEGFR-CTCs counts was 2.3 (range 0-30) cells per 7.5 ml. There was no association between meEGFR-CTCs and clinic-pathological-molecular features. In RASwt/BRAFwt patients with high levels of meEGFR-CTCs ratio (≥ 0.23) had significantly inferior PFS with anti-EGFR treatment (HR = 3.4, 95% CI 1.5-7.9, P = 0.004). By contrast, high levels of meEGFR in the untreated tumor tissues had no correlation with anti-EGFR treatment duration in cohort 2. Therefore, meEGFR-CTCs may have the potential to serve as a "liquid biopsy" biomarker to predict anti-EGFR treatment efficacy.

6.
Breast Cancer Res Treat ; 98(3): 295-301, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16752078

RESUMO

The ETS protein PEA3 functions as a transcription factor to regulate gene expression. Although members of the ETS family have been reported to be involved in tumor progression, ectopic expression of PEA3 has been shown to suppress tumor formation. Despite several studies demonstrated frequent expression of PEA3 and its high association with HER-2/neu and have suggested a potential role of PEA3 in breast cancer, contradictory result has shown that the PEA3 was associated with better survival rate in breast cancer. In the current study, we address this discrepancy by examining the expression of PEA3 and HER-2/neu on 289 archived breast cancer tumor tissues and their correlation with clinicopathologic factors and prognosis. The staining of PEA3 was further validated by in situ hybridization for PEA3 mRNA. We found PEA3 was positive in 22.2% (64/289) of all cases and only 25.6% (21/82) of HER-2/neu-overexpressing cases showed co-expression of PEA3. In contrast to HER-2/neu, PEA3 expression was not correlated with prognosis or major clinicopathologic factors, except for a negative correlation with lymphovascular permeation ( p=0.007). This study demonstrates that PEA3 expression is not correlated with HER-2/neu expression in breast cancer tumor tissues, nor is it associated with adverse clinicopathologic factors or prognosis.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Receptor ErbB-2/biossíntese , Fatores de Transcrição/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença , Feminino , Humanos , Hibridização In Situ , Pessoa de Meia-Idade , Modelos Estatísticos , Prognóstico , RNA Mensageiro/metabolismo
7.
Cancer Gene Ther ; 11(9): 594-602, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15272313

RESUMO

Paclitaxel (Taxol) is a promising frontline chemotherapeutic agent for the treatment of human breast and ovarian cancers. The adenoviral type 5 E1A gene has been tested in multiple clinical trials for its anticancer activity. E1A has also been shown to sensitize paclitaxel-induced killing in E1A-expressing cells. Here, we show that E1A can sensitize paclitaxel-induced apoptosis in breast cancer cells in a gene therapy setting by an orthotopic mammary tumor model. We first showed that expression of E1A enhanced in vitro paclitaxel cytotoxicity, as compared to the control cells. We then compared the therapeutic efficacy of paclitaxel between orthotopic tumor models established with vector-transfected MDA-MB-231 (231-Vect) versus 231-E1A stable cells, using tumor weight and apoptotic index (TUNEL assay) as the parameters. We found paclitaxel was more effective in shrinking tumors and inducing apoptosis in tumor models established with stable 231-E1A cells than the control 231-Vect cells. We also tested whether E1A could directly enhance paclitaxel-induced killing in nude mice, by using a nonviral, surface-protected cationic liposome to deliver E1A gene via the mouse tail vein. We compared the therapeutic effects of E1A gene therapy with or without Taxol chemotherapy in the established orthotopic tumor model of animals inoculated with MDA-MB-231 cells, and found that a combination of systemic E1A gene therapy and paclitaxel chemotherapy significantly enhanced the therapeutic efficacy and dramatically repressed tumor growth (P < .01). In addition, survival rates were significantly higher in animals treated with combination therapy than in the therapeutic control groups (both P < .0001). Thus, the E1A gene therapy indeed enhances the sensitivity of tumor cells to chemotherapy in a gene therapy setting and, the current study provides preclinical data to support combination therapy between E1A gene and chemotherapy for future clinical trials.


Assuntos
Proteínas E1A de Adenovirus/genética , Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias da Mama/terapia , Terapia Genética/métodos , Paclitaxel/uso terapêutico , Animais , Apoptose , Neoplasias da Mama/mortalidade , Divisão Celular/efeitos dos fármacos , Terapia Combinada , Feminino , Técnicas de Transferência de Genes , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Taxa de Sobrevida , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA