Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(36): 22423-22429, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32848073

RESUMO

Metastases are the cause of the vast majority of cancer deaths. In the metastatic process, cells migrate to the vasculature, intravasate, extravasate, and establish metastatic colonies. This pattern of spread requires the cancer cells to change shape and to navigate tissue barriers. Approaches that block this mechanical program represent new therapeutic avenues. We show that 4-hydroxyacetophenone (4-HAP) inhibits colon cancer cell adhesion, invasion, and migration in vitro and reduces the metastatic burden in an in vivo model of colon cancer metastasis to the liver. Treatment with 4-HAP activates nonmuscle myosin-2C (NM2C) (MYH14) to alter actin organization, inhibiting the mechanical program of metastasis. We identify NM2C as a specific therapeutic target. Pharmacological control of myosin isoforms is a promising approach to address metastatic disease, one that may be readily combined with other therapeutic strategies.


Assuntos
Acetofenonas/farmacologia , Actomiosina/metabolismo , Citoesqueleto , Metástase Neoplásica/fisiopatologia , Actinas/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Feminino , Células HCT116 , Humanos , Camundongos , Camundongos Nus
2.
Mol Ther ; 25(7): 1588-1595, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28457664

RESUMO

Multiple therapeutic agents are typically used in concert to effectively control metastatic tumors. Recently, we described microRNAs that are associated with the oligometastatic state, in which a limited number of metastatic tumors progress to more favorable outcomes. Here, we report the effective delivery of an oligometastatic microRNA (miR-655-3p) to colorectal liver metastases using nanoscale coordination polymers (NCPs). The NCPs demonstrated a targeted and prolonged distribution of microRNAs to metastatic liver tumors. Tumor-targeted microRNA miR-655-3p suppressed tumor growth when co-delivered with oxaliplatin, suggesting additive or synergistic interactions between microRNAs and platinum drugs. This is the first known example of systemically administered nanoparticles delivering an oligometastatic microRNA to advanced metastatic liver tumors and demonstrating tumor-suppressive effects. Our results suggest a potential therapeutic strategy for metastatic liver disease by the co-delivery of microRNAs and conventional cytotoxic agents using tumor-specific NCPs.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/terapia , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/terapia , MicroRNAs/genética , Nanoestruturas/administração & dosagem , Compostos Organoplatínicos/farmacologia , Animais , Antineoplásicos/química , Colesterol/química , Colesterol/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Di-Hidroxifenilalanina/química , Di-Hidroxifenilalanina/metabolismo , Modelos Animais de Doenças , Portadores de Fármacos , Sinergismo Farmacológico , Feminino , Células HCT116 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Nus , MicroRNAs/administração & dosagem , MicroRNAs/metabolismo , Nanoestruturas/química , Compostos Organoplatínicos/química , Oxaliplatina , Polietilenoglicóis/química , Polietilenoglicóis/metabolismo , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Proc Natl Acad Sci U S A ; 111(4): E484-91, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24434553

RESUMO

An siRNA screen targeting 89 IFN stimulated genes in 14 different cancer cell lines pointed to the RIG-I (retinoic acid inducible gene I)-like receptor Laboratory of Genetics and Physiology 2 (LGP2) as playing a key role in conferring tumor cell survival following cytotoxic stress induced by ionizing radiation (IR). Studies on the role of LGP2 revealed the following: (i) Depletion of LGP2 in three cancer cell lines resulted in a significant increase in cell death following IR, (ii) ectopic expression of LGP2 in cells increased resistance to IR, and (iii) IR enhanced LGP2 expression in three cell lines tested. Studies designed to define the mechanism by which LGP2 acts point to its role in regulation of IFNß. Specifically (i) suppression of LGP2 leads to enhanced IFNß, (ii) cytotoxic effects following IR correlated with expression of IFNß inasmuch as inhibition of IFNß by neutralizing antibody conferred resistance to cell death, and (iii) mouse embryonic fibroblasts from IFN receptor 1 knockout mice are radioresistant compared with wild-type mouse embryonic fibroblasts. The role of LGP2 in cancer may be inferred from cumulative data showing elevated levels of LGP2 in cancer cells are associated with more adverse clinical outcomes. Our results indicate that cytotoxic stress exemplified by IR induces IFNß and enhances the expression of LGP2. Enhanced expression of LGP2 suppresses the IFN stimulated genes associated with cytotoxic stress by turning off the expression of IFNß.


Assuntos
Sobrevivência Celular/fisiologia , RNA Helicases DEAD-box/fisiologia , Neoplasias Experimentais/patologia , RNA Helicases/fisiologia , Radiação Ionizante , Animais , Apoptose , Neoplasias Encefálicas/patologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Glioblastoma/patologia , Humanos , Interferon Tipo I/biossíntese , Camundongos , Camundongos Knockout , Neoplasias Experimentais/metabolismo , RNA Helicases/metabolismo , Células Tumorais Cultivadas
4.
Cancer ; 122(14): 2242-50, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27206146

RESUMO

BACKGROUND: The selection of patients for oligometastasis-directed ablative therapy remains a challenge. The authors report on clinical and molecular predictors of survival from a stereotactic body radiotherapy (SBRT) dose-escalation trial for oligometastases. METHODS: Patients who had from 1 to 5 metastases, a life expectancy of >3 months, and a Karnofsky performance status of >60 received escalating SBRT doses to all known cancer sites. Time to progression, progression-free survival, and overall survival (OS) were calculated at the completion of SBRT, and clinical predictors of OS were modeled. Primary tumor microRNA expression was analyzed to identify molecular predictors of OS. RESULTS: Sixty-one evaluable patients were enrolled from 2004 to 2009. The median follow-up was 2.3 years for all patients (range, 0.2-9.3 years) and 6.8 years for survivors (range, 2.0-9.3 years). The median, 2-year, and 5-year estimated OS were 2.4 years, 57%, and 32%, respectively. The rate of progression after SBRT was associated with an increased risk of death (hazard ratio [HR], 1.44; 95% confidence interval [CI], 1.24-1.82). The time from initial cancer diagnosis to metastasis (HR, 0.98; 95% CI, 0.98-0.99), the time from metastasis to SBRT (HR, 0.98; 95% CI, 0.98-0.99), and breast cancer histology (HR, 0.12; 95% CI, 0.07-0.37) were significant predictors of OS. In an exploratory analysis, a candidate classifier using expression levels of 3 microRNAs (miR-23b, miR-449a, and miR-449b) predicted survival among 17 patients who had primary tumor microRNA expression data available. CONCLUSIONS: A subset of oligometastatic patients achieves long-term survival after metastasis-directed SBRT. Clinical features and primary tumor microRNA expression profiling, if validated in an independent dataset, may help select oligometastatic patients most likely to benefit from metastasis-directed therapy. Cancer 2016;122:2242-50. © 2016 American Cancer Society.


Assuntos
Biomarcadores Tumorais , Neoplasias/diagnóstico , Neoplasias/mortalidade , Criança , Pré-Escolar , Seguimentos , Perfilação da Expressão Gênica , Humanos , Lactente , Estimativa de Kaplan-Meier , MicroRNAs/genética , Metástase Neoplásica , Neoplasias/radioterapia , Prognóstico , Modelos de Riscos Proporcionais , Radiocirurgia/efeitos adversos , Radiocirurgia/métodos , Resultado do Tratamento
5.
Proc Natl Acad Sci U S A ; 106(14): 5837-41, 2009 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19289846

RESUMO

The mucin 1 (MUC1) oncoprotein is aberrantly overexpressed in human breast cancers. Although MUC1 modulates the activity of estrogen receptor alpha (ER), there is no information regarding the effects of MUC1 on global gene expression patterns and the potential role of MUC1-induced genes in predicting outcome for breast cancer patients. We have developed an experimental model of MUC1-induced transformation that has identified the activation of genes involved in cholesterol and fatty acid metabolism. A 38-gene set of experimentally derived MUC1-induced genes associated with lipid metabolism was applied to the analysis of ER(+) breast cancer patients treated with tamoxifen. The results obtained from 2 independent databases demonstrate that patients overexpressing MUC1 and the lipid metabolic pathways are at significantly higher risk for death and recurrence/distant metastasis. By contrast, these genes were not predictive in untreated patients. Furthermore, a positive correlation was found between expression of the 38-gene set and the ER signaling pathway. These findings indicate that (i) MUC1 regulates cholesterol and fatty acid metabolism, and (ii) activation of these pathways in ER(+) breast cancers predicts failure to tamoxifen treatment.


Assuntos
Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Redes Reguladoras de Genes , Metabolismo dos Lipídeos/genética , Mucina-1/fisiologia , Valor Preditivo dos Testes , Colesterol/metabolismo , Receptor alfa de Estrogênio , Feminino , Humanos , Metástase Neoplásica , Proteínas de Neoplasias , Prognóstico , Recidiva , Taxa de Sobrevida , Ativação Transcricional
6.
Cancer Immun ; 11: 2, 2011 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-21714479

RESUMO

Many cancers escape host immunity without losing tumor-specific rejection antigens or MHC class I expression. This study tracks the evolution of one such cancer that developed in a mouse following exposure to ultraviolet light. The primary autochthonous tumor was not highly malignant and was rejected when transplanted into naïve immunocompetent mice. Neoplastic cells isolated from the primary tumor were susceptible to the growth-inhibitory effects of IFNγ in vitro, but expressed very low levels of MHC I antigen and were resistant to tumor-specific T cells unless they were first exposed to IFNγ. Serial passage of the primary tumor cells in vivo led to a highly aggressive variant that caused fast-growing tumors in normal mice. In vitro, the variant tumor cells showed increased resistance to the growth-inhibitory effects of IFNγ but expressed high levels of immunoproteasomes and MHC I molecules and were susceptible to tumor-specific T cells even without prior exposure to IFNγ.


Assuntos
Antígenos de Neoplasias/biossíntese , Regulação Neoplásica da Expressão Gênica/imunologia , Interferon gama/farmacologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/imunologia , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes p53 , Humanos , Interferon gama/biossíntese , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Nus , Mutação , Neoplasias Experimentais/genética , Proteínas Recombinantes , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
7.
Mol Ther ; 18(5): 912-20, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20197756

RESUMO

Ad.Egr-TNF is a radioinducible adenovector currently in phase 3 trials for inoperable pancreatic cancer. The combination of Ad.Egr-TNF and ionizing radiation (IR) contributes to local tumor control through the production of tumor necrosis factor-alpha (TNFalpha) in the tumor microenvironment. Moreover, clinical and preclinical studies with Ad.Egr-TNF/IR have suggested that this local approach suppresses the growth of distant metastatic disease; however, the mechanisms responsible for this effect remain unclear. These studies have been performed in wild-type (WT) and TNFR1,2(-/-) mice to assess the role of TNFalpha-induced signaling in the suppression of draining lymph node (DLN) metastases. The results demonstrate that production of TNFalpha in the tumor microenvironment induces expression of interferon (IFNbeta). In turn, IFNbeta stimulates the production of chemokines that recruit CD8(+) T cells to the tumor. The results further demonstrate that activation of tumor antigen-specific CD8(+) CTLs contributes to local antitumor activity and suppression of DLN metastases. These findings support a model in which treatment of tumors with Ad.Egr-TNF and IR is mediated by local and distant immune-mediated antitumor effects that suppress the development of metastases.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Metástase Neoplásica/prevenção & controle , Metástase Neoplásica/terapia , Radiação Ionizante , Fator de Necrose Tumoral alfa/metabolismo , Adenoviridae/genética , Animais , Proliferação de Células , Vetores Genéticos/genética , Humanos , Interferon beta/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Metástase Neoplásica/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/genética
8.
Clin Cancer Res ; 26(24): 6437-6444, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33028595

RESUMO

PURPOSE: Multisite stereotactic body radiotherapy followed by pembrolizumab (SBRT+P) has demonstrated safety in advanced solid tumors (ASTs). However, no studies have examined the relationships between irradiated tumor response, SBRT-induced tumor gene expression, and overall survival (OS). PATIENTS AND METHODS: Patients with AST received SBRT (30-50 Gy in 3-5 fractions) to two to four metastases followed by pembrolizumab (200 mg i.v. every 3 weeks). SBRT was prescribed to a maximum tumor volume of 65 mL. Small metastases received the complete prescribed coverage (complete-Rx), while larger metastases received partial coverage (partial-Rx). Treated metastasis control (TMC) was defined as a lack of progression for an irradiated metastasis. Landmark analysis was used to assess the relationship between TMC and OS. Thirty-five biopsies were obtained from 24 patients: 19 pre-SBRT and 16 post-SBRT (11 matched) prior to pembrolizumab and were analyzed via RNA microarray. RESULTS: Sixty-eight patients (139 metastases) were enrolled with a median follow-up of 10.4 months. One-year TMC was 89.5% with no difference between complete-Rx or partial-Rx. On multivariable analysis, TMC was independently associated with a reduced risk for death (HR, 0.36; 95% confidence interval, 0.17-0.75; P = 0.006). SBRT increased expression of innate and adaptive immune genes and concomitantly decreased expression of cell cycle and DNA repair genes in the irradiated tumors. Elevated post-SBRT expression of DNASE1 correlated with increased expression of cytolytic T-cell genes and irradiated tumor response. CONCLUSIONS: In the context of SBRT+P, TMC independently correlates with OS. SBRT impacts intratumoral immune gene expression associated with TMC. Randomized trials are needed to validate these findings.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Biomarcadores Tumorais/genética , Quimiorradioterapia/mortalidade , Neoplasias/mortalidade , Radiocirurgia/mortalidade , Antineoplásicos Imunológicos/uso terapêutico , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Prognóstico , Taxa de Sobrevida
9.
Nat Commun ; 11(1): 2354, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32393794

RESUMO

Death due to sepsis remains a persistent threat to critically ill patients confined to the intensive care unit and is characterized by colonization with multi-drug-resistant healthcare-associated pathogens. Here we report that sepsis in mice caused by a defined four-member pathogen community isolated from a patient with lethal sepsis is associated with the systemic suppression of key elements of the host transcriptome required for pathogen clearance and decreased butyrate expression. More specifically, these pathogens directly suppress interferon regulatory factor 3. Fecal microbiota transplant (FMT) reverses the course of otherwise lethal sepsis by enhancing pathogen clearance via the restoration of host immunity in an interferon regulatory factor 3-dependent manner. This protective effect is linked to the expansion of butyrate-producing Bacteroidetes. Taken together these results suggest that fecal microbiota transplantation may be a treatment option in sepsis associated with immunosuppression.


Assuntos
Transplante de Microbiota Fecal , Imunidade , Sepse/imunologia , Sepse/terapia , Animais , Ácido Butírico/metabolismo , Fezes/química , Microbioma Gastrointestinal , Trato Gastrointestinal/patologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Fator Regulador 3 de Interferon/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Sepse/microbiologia , Transdução de Sinais , Transcrição Gênica
10.
BMC Med ; 7: 68, 2009 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-19891767

RESUMO

BACKGROUND: The Signal Transducer and Activator of Transcription 1 (STAT1) has traditionally been regarded as a transmitter of interferon signaling and a pro-apoptotic tumour suppressor. Recent data have identified new functions of STAT1 associated with tumourigenesis and resistance to genotoxic stress, including ionizing radiation (IR) and chemotherapy. To investigate the mechanisms contributing to the tumourigenic functions of STAT1, we performed a combined transcriptomic-proteomic expressional analysis and found that STAT1 is associated with regulation of energy metabolism with potential implication in the Warburg effect. METHODS: We generated a stable knockdown of STAT1 in the SCC61 human squamous cell carcinoma cell line, established tumour xenografts in athymic mice, and compared transcriptomic and proteomic profiles of STAT1 wild-type (WT) and knockdown (KD) untreated or irradiated (IR) tumours. Transcriptional profiling was based on Affymetrix Human GeneChip(R) Gene 1.0 ST microarrays. Proteomes were determined from the tandem mass spectrometry (MS/MS) data by searching against the human subset of the UniProt database. Data were analysed using Significance Analysis of Microarrays for ribonucleic acid and Visualize software for proteins. Functional analysis was performed with Ingenuity Pathway Analysis with statistical significance measured by Fisher's exact test. RESULTS: Knockdown of STAT1 led to significant growth suppression in untreated tumours and radio sensitization of irradiated tumours. These changes were accompanied by alterations in the expression of genes and proteins of glycolysis/gluconeogenesis (GG), the citrate cycle (CC) and oxidative phosphorylation (OP). Of these pathways, GG had the most concordant changes in gene and protein expression and demonstrated a STAT1-dependent expression of genes and proteins consistent with tumour-specific glycolysis. In addition, IR drastically suppressed the GG pathway in STAT1 KD tumours without significant change in STAT1 WT tumours. CONCLUSION: Our results identify a previously uncharacterized function of STAT1 in tumours: expressional regulation of genes encoding proteins involved in glycolysis, the citrate cycle and mitochondrial oxidative phosphorylation, with predominant regulation of glycolytic genes. STAT1-dependent expressional regulation of glycolysis suggests a potential role for STAT1 as a transcriptional modulator of genes responsible for the Warburg effect.


Assuntos
Metabolismo Energético , Regulação Neoplásica da Expressão Gênica , Redes e Vias Metabólicas/genética , Neoplasias/radioterapia , Tolerância a Radiação , Fator de Transcrição STAT1/metabolismo , Animais , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Análise de Sequência com Séries de Oligonucleotídeos , Proteoma/análise , Fator de Transcrição STAT1/genética , Espectrometria de Massas em Tandem
11.
Int J Radiat Biol ; 85(5): 421-31, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19437244

RESUMO

PURPOSE: To determine the mechanisms of Signal Transducer and Activator of Transcription 1 (Stat1)-associated radioresistance developed by nu61 tumour selected in vivo by fractionated irradiation of the parental radiosensitive tumour SCC61. MATERIALS AND METHODS: Radioresistence of nu61 and SCC61 in vitro was measured by clonogenic assay. Apoptotic response of nu61 and SCC61 cells to genotoxic stress was examined using caspase-based apoptotic assays. Co-cultivation of carboxyfluorescein diacetate, succinimidyl ester (CFDE-SE)-labeled nu61 with un-labeled SCC61 was performed at 1:1 ratio. Production of interleukin-6, interleukin-8 and soluble receptor of interleukin 6 (IL6, IL8 and sIL6R) was measured using Enzyme-Linked Immunosorbent Assay (ELISA). RESULTS: Radioresistant nu61 was also resistant to interferon-gamma (IFNgamma) and the death ligands of tumour necrosis factor alpha receptor (TNFR) family when compared to SCC61. This combined resistance is due to an impaired apoptotic response in nu61. Relative to SCC61, nu61 produced more IL6, IL8 and sIL6R. Using Stat1 knock-downs we demonstrated that IL6 and IL8 production is Stat1-dependent. Treatment with neutralising antibodies to IL6 and IL8, but not to either cytokine alone sensitised nu61 to genotoxic stress induced apoptosis. CONCLUSION: Nu61, which over-expresses Stat1 pathway, is deficient in apoptotic response to ionising radiation and cytotoxic ligands. This resistance to apoptosis is associated with Stat1-dependent production of IL6 and IL8 and suppression of caspases 8, 9 and 3.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Interleucinas/metabolismo , Neoplasias/patologia , Tolerância a Radiação , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Animais , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Técnicas de Cocultura , Citocinas/toxicidade , Citotoxinas/toxicidade , Dano ao DNA , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Interferon gama/farmacologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Neoplasias/genética , Radiação Ionizante , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação
12.
Cancer Res ; 67(19): 9214-20, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909027

RESUMO

Elsewhere, we reported that multiple serial in vivo passage of a squamous cell carcinoma cells (SCC61) concurrent with ionizing radiation (IR) treatment resulted in the selection of radioresistant tumor (nu61) that overexpresses the signal transducer and activator of transcription 1 (Stat1)/IFN-dependent pathway. Here, we report that (a) the Stat1 pathway is induced by IR, (b) constitutive overexpression of Stat1 is linked with failure to transmit a cytotoxic signal by radiation or IFNs, (c) selection of parental cell line SCC61 against IFN-alpha and IFN-gamma leads to the same IR- and IFN-resistant phenotype as was obtained by IR selection, and (d) suppression of Stat1 by short hairpin RNA renders the IR-resistant nu61 cells radiosensitive to IR. We propose a model that transient induction of Stat1 by IFN, IR, or other stress signals activates cytotoxic genes and cytotoxic response. Constitutive overexpression of Stat1 on the other hand leads to the suppression of the cytotoxic response and induces prosurvival genes that, at high levels of Stat1, render the cells resistant to IR or other inducers of cell death.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Fator de Transcrição STAT1/metabolismo , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Resistencia a Medicamentos Antineoplásicos , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/efeitos da radiação , Humanos , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , RNA Interferente Pequeno/genética , Tolerância a Radiação , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/biossíntese , Fator de Transcrição STAT1/genética , Transplante Heterólogo
13.
Int Rev Cell Mol Biol ; 344: 31-89, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30798990

RESUMO

DNA damage has been considered the primary action of ionizing radiation (IR) in normal and tumor cells resulting in cell autonomous death. However, recent findings have elucidated novel local and systemic effects of IR mediated by the induction of Type I interferons (IFN) and activation of adaptive immune responses. These responses are initiated by DNA- and RNA-dependent activation of pattern recognition receptors (PRR) which comprise an innate immunity system responsible for detection of exogenous pathogens, mounting of an anti-viral response, and activation of adaptive immunity. Activation of cytoplasmic RNA sensors by endogenous dsRNAs has emerged as an important mechanism of tumor cell-intrinsic Type I IFN induction in the response to genotoxic therapies. The fact that an effective anti-tumor effect of IR and chemotherapy response is dependent on the RNA sensing pathways indicates that tumor cells are equipped by functional innate immunity system which recognizes genotoxic stress insult as an intrusion of exogenous RNA pathogens. This indicates that DNA damage leads to the RNA stress, which is recognized as danger-associated molecular patterns (DAMPs) presented by aberrant RNA molecules interacting with cytoplasmic RNA sensors. In the current review, we will discuss the mechanistic bases for RNA-dependent innate responses in tumor cells, RNA ligands recognized as DAMPs, and the potential implications of these findings to improve cancer therapies.


Assuntos
Espaço Intracelular/metabolismo , Mamíferos/metabolismo , Neoplasias/terapia , RNA de Cadeia Dupla/metabolismo , Estresse Fisiológico , Alarminas/metabolismo , Animais , Humanos
14.
Biomaterials ; 210: 94-104, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31060867

RESUMO

Though early detection and treatment of primary tumors has significantly improved in recent years, metastatic disease remains among the most significant challenges in cancer therapy. Cancer cells can disseminate before the primary tumor is detected to form micro or gross metastases, requiring toxic systemic therapies. To prevent and suppress metastases, we have developed a nontoxic, long-circulating nanoscale coordination polymer (NCP) protecting microRNA (miRNA) in circulation and releasing it in tumors. PtIV(en)2 [en = ethylenediamine] containing NCPs (PtEN) can release a nontoxic, kinetically inert PtII(en)2 compound and carbon dioxide which aids the endosomal escape of its miRNA cargo, miR-655-3p. Without the presence of the PtEN core, the miRNA showed cellular uptake but no effect. When transfected into human colorectal HCT116 cells by NCPs, this oligometastatic miRNA limited proliferation and epithelial-to-mesenchymal transition by preventing ß-catenin nuclear translocation and tumor cell invasion. Systemic administrations of PtEN/miR-655-3p sustained effective transfection to reduce liver colonization and tumor burden in a xenogenic hepatic metastatic model of HCT116 without any observable toxicity.


Assuntos
Neoplasias Colorretais/secundário , Transição Epitelial-Mesenquimal , Técnicas de Transferência de Genes , Neoplasias Hepáticas/patologia , MicroRNAs/administração & dosagem , Nanopartículas/química , Polímeros/química , Animais , Adesão Celular , Proliferação de Células , Endossomos/metabolismo , Células HCT116 , Humanos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Invasividade Neoplásica , Metástase Neoplásica , PTEN Fosfo-Hidrolase/metabolismo , beta Catenina/metabolismo
15.
Cancer Res ; 79(3): 650-662, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30538122

RESUMO

Expression of 14q32-encoded miRNAs is a favorable prognostic factor in patients with metastatic cancer. In this study, we used genomic inhibition of DNA methylation through disruption of DNA methyltransferases DNMT1 and DNMT3B and pharmacologic inhibition with 5-Aza-2'-deoxycytidine (5-Aza-dC, decitabine) to demonstrate that DNA methylation predominantly regulates expression of metastasis-suppressive miRNAs in the 14q32 cluster. DNA demethylation facilitated CCCTC-binding factor (CTCF) recruitment to the maternally expressed gene 3 differentially methylated region (MEG3-DMR), which acts as a cis-regulatory element for 14q32 miRNA expression. 5-Aza-dC activated demethylation of the MEG3-DMR and expression of 14q32 miRNAs, which suppressed adhesion, invasion, and migration (AIM) properties of metastatic tumor cells. Cancer cells with MEG3-DMR hypomethylation exhibited constitutive expression of 14q32 miRNAs and resistance to 5-Aza-dC-induced suppression of AIM. Expression of methylation-dependent 14q32 miRNAs suppressed metastatic colonization in preclinical models of lung and liver metastasis and correlated with improved clinical outcomes in patients with metastatic cancer. These findings implicate epigenetic modification via DNA methylation in the regulation of metastatic propensity through miRNA networks and identify a previously unrecognized action of decitabine on the activation of metastasis-suppressive miRNAs. SIGNIFICANCE: This study investigates epigenetic regulation of metastasis-suppressive miRNAs and the effect on metastasis.


Assuntos
Cromossomos Humanos Par 14 , Metilação de DNA , MicroRNAs/genética , Animais , Azacitidina/farmacologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , DNA (Citosina-5-)-Metiltransferase 1/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HCT116 , Xenoenxertos , Humanos , Neoplasias Hepáticas/secundário , Células MCF-7 , Camundongos , Camundongos Nus , MicroRNAs/biossíntese , Metástase Neoplásica , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/genética , DNA Metiltransferase 3B
16.
Cancer Res ; 79(10): 2536-2548, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30940658

RESUMO

Alkylating chemotherapy is a central component of the management of glioblastoma (GBM). Among the factors that regulate the response to alkylation damage, NF-κB acts to both promote and block cytotoxicity. In this study, we used genome-wide expression analysis in U87 GBM to identify NF-κB-dependent factors altered in response to temozolomide and found the long noncoding RNA (lncRNA) MALAT1 as one of the most significantly upregulated. In addition, we demonstrated that MALAT1 expression was coregulated by p50 (p105) and p53 via novel κB- and p53-binding sites in the proximal MALAT1 coding region. Temozolomide treatment inhibited p50 recruitment to its cognate element as a function of Ser329 phosphorylation while concomitantly increasing p53 recruitment. Moreover, luciferase reporter studies demonstrated that both κB and p53 cis-elements were required for efficient transactivation in response to temozolomide. Depletion of MALAT1 sensitized patient-derived GBM cells to temozolomide cytotoxicity, and in vivo delivery of nanoparticle-encapsulated anti-MALAT1 siRNA increased the efficacy of temozolomide in mice bearing intracranial GBM xenografts. Despite these observations, in situ hybridization of GBM specimens and analysis of publicly available datasets revealed that MALAT1 expression within GBM tissue was not prognostic of overall survival. Together, these findings support MALAT1 as a target for chemosensitization of GBM and identify p50 and p52 as primary regulators of this ncRNA. SIGNIFICANCE: These findings identify NF-κB and p53 as regulators of the lncRNA MALAT1 and suggest MALAT1 as a potential target for the chemosensitization of GBM.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/metabolismo , Glioblastoma/tratamento farmacológico , NF-kappa B/metabolismo , RNA Longo não Codificante/biossíntese , Temozolomida/uso terapêutico , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Dano ao DNA/genética , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Prognóstico , RNA Longo não Codificante/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Cancer Res ; 79(7): 1465-1479, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30482772

RESUMO

Given the integral role of stimulator of interferon genes (STING, TMEM173) in the innate immune response, its loss or impairment in cancer is thought to primarily affect antitumor immunity. Here we demonstrate a role for STING in the maintenance of cellular homeostasis through regulation of the cell cycle. Depletion of STING in human and murine cancer cells and tumors resulted in increased proliferation compared with wild-type controls. Microarray analysis revealed genes involved in cell-cycle regulation are differentially expressed in STINGko compared with WT MEFs. STING-mediated regulation of the cell cycle converged on NFκB- and p53-driven activation of p21. The absence of STING led to premature activation of cyclin-dependent kinase 1 (CDK1), early onset to S-phase and mitosis, and increased chromosome instability, which was enhanced by ionizing radiation. These results suggest a pivotal role for STING in maintaining cellular homeostasis and response to genotoxic stress. SIGNIFICANCE: These findings provide clear mechanistic understanding of the role of STING in cell-cycle regulation, which may be exploited in cancer therapy because most normal cells express STING, while many tumor cells do not.See related commentary by Gius and Zhu, p. 1295.


Assuntos
Imunidade Inata , Proteínas de Membrana/genética , Animais , Proliferação de Células , Instabilidade Cromossômica , Homeostase , Humanos , Camundongos
18.
Mol Cancer Ther ; 17(4): 732-739, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29467274

RESUMO

Lung cancer is the leading cause of cancer-related deaths worldwide. Approximately 85% of all lung cancers are non-small cell histology [non-small cell lung cancer (NSCLC)]. Modern treatment strategies for NSCLC target driver oncogenes and immune checkpoints. However, less than 15% of patients survive beyond 5 years. Here, we investigated the effects of SAR302503 (SAR), a selective JAK2 inhibitor, on NSCLC cell lines and tumors. We show that SAR is cytotoxic to NSCLC cells, which exhibit resistance to genotoxic therapies, such as ionizing radiation, cisplatin, and etoposide. We demonstrate that constitutive IFN-stimulated gene expression, including an IFN-related DNA damage resistance signature, predicts for sensitivity to SAR. Importantly, tumor cell-intrinsic expression of PD-L1 is IFN-inducible and abrogated by SAR. Taken together, these findings suggest potential dual roles for JAK2 inhibitors, both as a novel monotherapy in NSCLCs resistant to genotoxic therapies, and in tandem with immune checkpoint inhibition. Mol Cancer Ther; 17(4); 732-9. ©2018 AACR.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Janus Quinase 2/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Pirrolidinas/farmacologia , Sulfonamidas/farmacologia , Animais , Apoptose , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Nat Commun ; 9(1): 4827, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30425251

RESUMO

In the originally published version of this Article, the affiliation details for Kevin P. White inadvertently omitted 'Tempus Labs, Chicago, IL, 60654, USA'. This has now been corrected in both the PDF and HTML versions of the Article.

20.
J Clin Oncol ; 36(16): 1611-1618, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29437535

RESUMO

Purpose Stereotactic body radiotherapy (SBRT) may stimulate innate and adaptive immunity to augment immunotherapy response. Multisite SBRT is an emerging paradigm for treating metastatic disease. Anti-PD-1-treatment outcomes may be improved with lower disease burden. In this context, we conducted a phase I study to evaluate the safety of pembrolizumab with multisite SBRT in patients with metastatic solid tumors. Patients and Methods Patients progressing on standard treatment received SBRT to two to four metastases. Not all metastases were targeted, and metastases > 65 mL were partially irradiated. SBRT dosing varied by site and ranged from 30 to 50 Gy in three to five fractions with predefined dose de-escalation if excess dose-limiting toxicities were observed. Pembrolizumab was initiated within 7 days after completion of SBRT. Pre- and post-SBRT biopsy specimens were analyzed in a subset of patients to quantify interferon-γ-induced gene expression. Results A total of 79 patients were enrolled; three patients did not receive any treatment and three patients only received SBRT. Patients included in the analysis were treated with SBRT and at least one cycle of pembrolizumab. Most (94.5%) of patients received SBRT to two metastases. Median follow-up for toxicity was 5.5 months (interquartile range, 3.3 to 8.1 months). Six patients experienced dose-limiting toxicities with no radiation dose reductions. In the 68 patients with imaging follow-up, the overall objective response rate was 13.2%. Median overall survival was 9.6 months (95% CI, 6.5 months to undetermined) and median progression-free survival was 3.1 months (95% CI, 2.9 to 3.4 months). Expression of interferon-γ-associated genes from post-SBRT tumor biopsy specimens significantly correlated with nonirradiated tumor response. Conclusion Multisite SBRT followed by pembrolizumab was well tolerated with acceptable toxicity. Additional studies exploring the clinical benefit and predictive biomarkers of combined multisite SBRT and PD-1-directed immunotherapy are warranted.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Radiocirurgia/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos Imunológicos/uso terapêutico , Biópsia , Terapia Combinada , Relação Dose-Resposta à Radiação , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Radiocirurgia/efeitos adversos , Dosagem Radioterapêutica , Critérios de Avaliação de Resposta em Tumores Sólidos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa