Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Res Sq ; 2023 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-37790365

RESUMO

TCF1high progenitor CD8+ T cells mediate the efficacy of PD-1 blockade, however the mechanisms that govern their generation and maintenance are poorly understood. Here, we show that targeting glycolysis through deletion of pyruvate kinase muscle 2 (PKM2) results in elevated pentose phosphate pathway (PPP) activity, leading to enrichment of a TCF1high central memory-like phenotype and increased responsiveness to PD-1 blockade in vivo. PKM2KO CD8+ T cells showed reduced glycolytic flux, accumulation of glycolytic intermediates and PPP metabolites, and increased PPP cycling as determined by 1,2 13C glucose carbon tracing. Small molecule agonism of the PPP without acute glycolytic impairment skewed CD8+ T cells towards a TCF1high population, generated a unique transcriptional landscape, enhanced tumor control in mice in combination with PD-1 blockade, and promoted tumor killing in patient-derived tumor organoids. Our study demonstrates a new metabolic reprogramming that contributes to a progenitor-like T cell state amenable to checkpoint blockade.

2.
Nat Commun ; 14(1): 120, 2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36624093

RESUMO

IRE1α-XBP1 signaling is emerging as a central orchestrator of malignant progression and immunosuppression in various cancer types. Employing a computational XBP1s detection method applied to TCGA datasets, we demonstrate that expression of the XBP1s mRNA isoform predicts poor survival in non-small cell lung cancer (NSCLC) patients. Ablation of IRE1α in malignant cells delays tumor progression and extends survival in mouse models of NSCLC. This protective effect is accompanied by alterations in intratumoral immune cell subsets eliciting durable adaptive anti-cancer immunity. Mechanistically, cancer cell-intrinsic IRE1α activation sustains mPGES-1 expression, enabling production of the immunosuppressive lipid mediator prostaglandin E2. Accordingly, restoring mPGES-1 expression in IRE1αKO cancer cells rescues normal tumor progression. We have developed an IRE1α gene signature that predicts immune cell infiltration and overall survival in human NSCLC. Our study unveils an immunoregulatory role for cancer cell-intrinsic IRE1α activation and suggests that targeting this pathway may help enhance anti-tumor immunity in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Endorribonucleases , Neoplasias Pulmonares , Proteínas Serina-Treonina Quinases , Animais , Humanos , Camundongos , Carcinoma Pulmonar de Células não Pequenas/genética , Endorribonucleases/genética , Endorribonucleases/metabolismo , Neoplasias Pulmonares/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo
3.
Prehosp Emerg Care ; 27(2): 213-220, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35020551

RESUMO

BACKGROUND: Several studies have demonstrated the high frequency of medication errors in pediatric patients by prehospital providers during both patient care and simulation. In 2015, our hospital-based urban EMS system introduced the HandtevyTM Field Guide that provides precalculated pediatric doses in milliliters (mL) by patient age. We hypothesized that implementation of the Field Guide would increase the percentage of correct pediatric medication doses to greater than 85%. METHODS: We performed a single center retrospective cohort study of medications administered to patients < 13 years of age from August 2017 to July 2019 compared to 2014 baseline data through electronic medical record review. We excluded nebulized medications and online medication direction cases. Our primary outcome was the percentage of correct doses defined as a dose within 80-120% of the Field Guide dose recommendation. Each dosing error was reviewed by two investigators. RESULTS: We analyzed 483 drug administrations in 375 patients for the Field Guide study period. Doses were correct in 89.4% of medication administrations with 68.5% reportedly administered exactly as dictated by the Field Guide compared to 51.1% in the baseline period (p < 0.001). During the Field Guide study period, the following medications had 100% appropriate dosing: adenosine, dextrose 10%, diphenhydramine, epinephrine 1:10,000, glucagon, naloxone and oral ondansetron. Overdoses accounted for 4.4% of medication errors and underdoses accounted for 6.2% of medications errors. The most overdosed medications were intranasal (IN) midazolam (11.8%) and intravenous fentanyl (9.4%). The most underdosed medications were IN midazolam (23.5%) and intramuscular epinephrine 1:1000 (12.5%). The highest percentage of errors (20%) were seen in the zero to one-year-old age group. CONCLUSION: After implementation of a precalculated mL dose system by patient age for EMS providers, most pediatric medications were reportedly administered within the appropriate dose range. A field guide with precalculated doses (in mL) may be an effective tool for reducing pediatric medication dosing errors by EMS providers.


Assuntos
Overdose de Drogas , Serviços Médicos de Emergência , Criança , Humanos , Recém-Nascido , Lactente , Midazolam , Estudos Retrospectivos , Erros de Medicação , Epinefrina
4.
Sci Transl Med ; 14(636): eabe8195, 2022 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-35294260

RESUMO

Most patients with non-small cell lung cancer (NSCLC) do not achieve durable clinical responses from immune checkpoint inhibitors, suggesting the existence of additional resistance mechanisms. Nicotinamide adenine dinucleotide (NAD)-induced cell death (NICD) of P2X7 receptor (P2X7R)-expressing T cells regulates immune homeostasis in inflamed tissues. This process is mediated by mono-adenosine 5'-diphosphate (ADP)-ribosyltransferases (ARTs). We found an association between membranous expression of ART1 on tumor cells and reduced CD8 T cell infiltration. Specifically, we observed a reduction in the P2X7R+ CD8 T cell subset in human lung adenocarcinomas. In vitro, P2X7R+ CD8 T cells were susceptible to ART1-mediated ADP-ribosylation and NICD, which was exacerbated upon blockade of the NAD+-degrading ADP-ribosyl cyclase CD38. Last, in murine NSCLC and melanoma models, we demonstrate that genetic and antibody-mediated ART1 inhibition slowed tumor growth in a CD8 T cell-dependent manner. This was associated with increased infiltration of activated P2X7R+CD8 T cells into tumors. In conclusion, we describe ART1-mediated NICD as a mechanism of immune resistance in NSCLC and provide preclinical evidence that antibody-mediated targeting of ART1 can improve tumor control, supporting pursuit of this approach in clinical studies.


Assuntos
ADP Ribose Transferases , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Subpopulações de Linfócitos T , ADP Ribose Transferases/genética , ADP Ribose Transferases/metabolismo , Difosfato de Adenosina , Animais , Carcinoma Pulmonar de Células não Pequenas/imunologia , Proteínas Ligadas por GPI/genética , Humanos , Neoplasias Pulmonares/imunologia , Camundongos
6.
J Asthma ; 59(5): 937-945, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-33504232

RESUMO

OBJECTIVE: To evaluate the frequency of EMS protocol non-adherence during pediatric asthma encounters and its association with emergency department (ED) length of stay (LOS) and hospital admission. METHODS: This is a retrospective review of asthma encounters aged 2-17 years transported by EMS to a pediatric ED from 2012 to 2017. Our primary outcome was hospital admission based on prehospital protocol adherence defined as: (1) bronchodilator administration, (2) treatment of hypoxia with oxygen, or (3) administration of intramuscular (IM) epinephrine in encounters with high severity of distress. Multivariable logistic regression estimated the association between protocol non-adherence and hospital admission. RESULTS: During the study period, 290 EMS encounters met inclusion criteria. Median age was 9 years (IQR 5-12), 63% were male, 40% had moderate to severe exacerbations, and 24% were admitted. Protocol non-adherence occurred in 32% of encounters with failure to administer bronchodilators in 27% and failure to administer IM epinephrine when indicated in 83%. Prehospital steroids were administered in 8% of encounters. After adjusting for covariates, protocol non-adherence was not statistically associated with likelihood of inpatient admission (OR 1.3; 95% CI: 0.6-2.6). CONCLUSIONS: Among prehospital pediatric asthma encounters, EMS protocol non-adherence is common but not associated with a higher frequency of hospital admission. Hospital admission was associated with acute exacerbation severity suggesting further research is needed to develop a valid prehospital asthma severity assessment scoring tool.Supplemental data for this article can be accessed at publisher's website.


Assuntos
Asma , Serviços Médicos de Emergência , Asma/tratamento farmacológico , Broncodilatadores/uso terapêutico , Criança , Pré-Escolar , Serviços Médicos de Emergência/métodos , Serviço Hospitalar de Emergência , Epinefrina/uso terapêutico , Feminino , Humanos , Masculino , Estudos Retrospectivos
7.
J Hepatol ; 76(1): 148-159, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34689996

RESUMO

BACKGROUND & AIMS: Regulatory T cell (Treg) depletion increases antitumor immunity. However, severe autoimmunity can occur following systemic loss of Tregs, which could be avoided by selectively depleting intratumoral Tregs. Herein, we aimed to investigate the role of tumor-infiltrating CCR4+ Tregs in hepatocellular carcinoma (HCC) and to provide a potential target strategy for immunotherapy. METHODS: CCR4+ Tregs were analyzed by flow cytometry in murine models and clinical samples. The function of tumor-infiltrating and induced CCR4+ Tregs was interrogated by genetic and epigenetic approaches. To block CCR4+ Treg chemotaxis, we developed an N-terminus recombinant protein of CCR4 (N-CCR4-Fc) as a neutralizing pseudo-receptor that effectively bound to its ligand CCL22. The efficacy of CCR4 antagonism as an immunotherapeutic agent was evaluated by tumor weights, growth kinetics and survival curves. RESULTS: CCR4+ Tregs were the predominant type of Tregs recruited to hepatitis B-associated HCC (HBV+ HCC), correlating with sorafenib resistance and HBV load titers. Compared with CCR4- Tregs, CCR4+ Tregs exhibited increased IL-10 and IL-35 expression, and enhanced functionality in suppressing CD8+ T cells. CCR4+ Tregs also displayed PD-1+TCF1+ stem-like properties. ATAC-seq data revealed substantial chromatin remodeling between tumor-infiltrating Tregs (TIL-Tregs) and induced Tregs, suggesting that long-term chromatin reprogramming accounted for the acquisition of enhanced immunosuppressive stem-like specificity by CCR4+ TIL-Tregs. Treatment with a CCR4 antagonist or N-CCR4-Fc blocked intratumoral Treg accumulation, overcame sorafenib resistance, and sensitized tumors to PD-1 checkpoint blockade. CONCLUSIONS: Intratumoral stem-like CCR4+ Tregs orchestrated immunosuppressive resource cells in the tumor microenvironment. CCR4 could be targeted to enhance antitumor immunity by specifically blocking infiltration of Tregs into the tumor microenvironment and inhibiting maintenance of the TIL-Treg pool. LAY SUMMARY: Targeting regulatory T cells is a promising approach in cancer immunotherapy; however, severe autoimmunity can occur following systemic regulatory T cell loss. This could be avoided by selectively depleting intratumoral regulatory T cells. Herein, targeting intratumoral stem-like CCR4+ regulatory T cells helped to overcome sorafenib resistance and sensitize tumors to immune checkpoint blockade in mouse models of liver cancer. This approach could have wide clinical applicability.


Assuntos
Carcinoma Hepatocelular/etiologia , Hepatite B/complicações , Hospedeiro Imunocomprometido/efeitos dos fármacos , Receptores CCR4/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/imunologia , China , Modelos Animais de Doenças , Hepatite B/imunologia , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/patogenicidade , Hospedeiro Imunocomprometido/genética , Hospedeiro Imunocomprometido/imunologia , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/imunologia , Camundongos , Receptores CCR4/imunologia , Linfócitos T Reguladores/imunologia
8.
Nat Cancer ; 2(9): 919-931, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34917944

RESUMO

Radiation therapy (RT) in combination with immune checkpoint inhibitor (ICI) represents a promising regimen for non-small cell lung cancer (NSCLC), however, the underlying mechanisms are poorly characterized. We identified a specific dose of RT that conferred tumor regression and improved survival in NSCLC models when combined with ICI. The immune-modulating functions of RT was ascribed to activated lung-resident Scgb1a1+ club cells. Importantly, mice with club cell-specific knockout of synaptosome-associated protein 23 failed to benefit from the combination treatment, indicating a pivotal role of club cell secretome. We identified 8 club cells secretory proteins, which inhibited immunosuppressive myeloid cells, reduced pro-tumor inflammation, and enhanced anti-tumor immunity. Notably, CC10, a member of club cell secretome was increased in plasma of NSCLC patients responding to the combination therapy. By revealing an immune-regulatory role of club cells, our studies have the potential to guide future clinical trials of ICI in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Uteroglobina/uso terapêutico
9.
Cancer Cell ; 39(7): 973-988.e9, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34115989

RESUMO

Immune checkpoint blockade (ICB) has been a remarkable clinical advance for cancer; however, the majority of patients do not respond to ICB therapy. We show that metastatic disease in the pleural and peritoneal cavities is associated with poor clinical outcomes after ICB therapy. Cavity-resident macrophages express high levels of Tim-4, a receptor for phosphatidylserine (PS), and this is associated with reduced numbers of CD8+ T cells with tumor-reactive features in pleural effusions and peritoneal ascites from patients with cancer. We mechanistically demonstrate that viable and cytotoxic anti-tumor CD8+ T cells upregulate PS and this renders them susceptible to sequestration away from tumor targets and proliferation suppression by Tim-4+ macrophages. Tim-4 blockade abrogates this sequestration and proliferation suppression and enhances anti-tumor efficacy in models of anti-PD-1 therapy and adoptive T cell therapy in mice. Thus, Tim-4+ cavity-resident macrophages limit the efficacy of immunotherapies in these microenvironments.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/imunologia , Regulação Neoplásica da Expressão Gênica , Macrófagos/imunologia , Proteínas de Membrana/metabolismo , Microambiente Tumoral , Animais , Apoptose , Proliferação de Células , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Prognóstico , Estudos Retrospectivos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Res ; 81(9): 2386-2398, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33653774

RESUMO

Hepatitis B-associated hepatocellular carcinoma (HCC) is often accompanied by severe vascular invasion and portal vein tumor thrombus, leading to a poor prognosis. However, the underlying mechanism of this disease remains obscure. In this study, we demonstrate that the hepatitis B virus (HBV)-encoded gene HBx induces high IL8 production through MEK-ERK signal activation, leading to enhanced endothelial permeability to facilitate tumor vascular invasion. In a vascular metastatic model using a tail vein injection in a transgenic mouse with selective expression of human CXCR1 in the endothelium, activation of the IL8-CXCR1 cascade by overexpression of IL8 in tumor cells dramatically enhanced liver metastasis. Mechanistically, IL8 selectively induced GARP-latent-TGFß in liver sinusoidal endothelial cells and subsequently provoked preferential regulatory T-cell polarization to suppress antitumor immunity. Collectively, these findings reveal a hepatitis B-associated IL8-CXCR1 signaling axis that mediates vascular invasion and local microenvironmental immune escape of HCC to induce intrahepatic metastasis, which may serve as potential therapeutic targets for HBV-associated HCC. SIGNIFICANCE: This study identifies a hepatitis B-induced IL8/CXCR1/TGFß signaling cascade that suppresses antitumor immunity and enhances metastasis in hepatocellular carcinoma, providing new potential targets for therapeutic intervention.


Assuntos
Carcinoma Hepatocelular/secundário , Carcinoma Hepatocelular/virologia , Vírus da Hepatite B/metabolismo , Hepatite B/complicações , Interleucina-8/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/virologia , Fígado/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Carcinoma Hepatocelular/metabolismo , Modelos Animais de Doenças , Células HEK293 , Células Hep G2 , Hepatite B/virologia , Vírus da Hepatite B/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Interleucina-8/genética , Interleucina-8/farmacologia , Neoplasias Hepáticas/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Proteínas Recombinantes/farmacologia , Transativadores/genética , Transativadores/metabolismo , Transfecção , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo
11.
Pharmacol Res ; 157: 104800, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32278046

RESUMO

The HBV-initiated hepatocellular carcinoma (HCC) frequently develops from or accompanies long-term chronic hepatitis, inflammation, and cirrhosis, and has a poor prognosis. Sorafenib, an orally active multi-kinase inhibitor, currently the most common approved drug for first-line systemic treatment of advanced HCC, only improves overall survival of three months, suggesting the need for new therapeutic strategies. In this study, we identified that sorafenib selectively resisted in immune competent C57BL/6 mice but not nude mice. The chemokines CCL22 and CCL17 were upregulated by sorafenib, which elevated dramatically higher in HBV-associated HCC. Mechanically, sorafenib accelerates CCL22 expression via TNF-α-RIP1-NF-κB signaling pathway. Blocking CCL22 signaling with antagonist C-021 and sorafenib treated in combination can inhibit tumor growth and enhance the antitumor response, whereas no significant differences in tumor burden were observed in nude mice upon addition of C-021. These findings strongly suggest that CCL22 signaling pathway strongly contributes to sorafenib resistance in HBV-associated HCC, indicating a potential therapeutic strategy for immunological chemotherapy complementing first-line agents against HBV-associated HCC.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Quimiocina CCL22/metabolismo , Resistencia a Medicamentos Antineoplásicos , Neoplasias Hepáticas/tratamento farmacológico , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Sorafenibe/farmacologia , Linfócitos T/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Quimiocina CCL22/antagonistas & inibidores , Quimiocina CCL22/genética , Células Hep G2 , Hepatite B/complicações , Hepatite B/virologia , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Quinazolinas/farmacologia , Receptores CCR4/antagonistas & inibidores , Receptores CCR4/genética , Receptores CCR4/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Carga Tumoral/efeitos dos fármacos
12.
Cell Rep ; 30(3): 755-770.e6, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31968251

RESUMO

Epigenetic changes are increasingly being appreciated as key events in breast cancer progression. However, breast cancer subtype-specific epigenetic regulation remains poorly investigated. Here we report that EZH2 is a leading candidate of epigenetic modulators associated with the TNBC subtype and that it predicts poor overall survival in TNBC patients. We demonstrate that specific pharmacological or genetic inhibition of EZH2 catalytic activity impairs distant metastasis. We further define a specific EZH2high population with enhanced invasion, mammosphere formation, and metastatic potential that exhibits marked sensitivity to EZH2 inhibition. Mechanistically, EZH2 inhibition differentiates EZH2high basal cells to a luminal-like phenotype by derepressing GATA3 and renders them sensitive to endocrine therapy. Furthermore, dissection of human TNBC heterogeneity shows that EZH2high basal-like 1 and mesenchymal subtypes have exquisite sensitivity to EZH2 inhibition compared with the EZH2low luminal androgen receptor subtype. These preclinical findings provide a rationale for clinical development of EZH2 as a targeted therapy against TNBC metastasis.


Assuntos
Biocatálise , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Animais , Sequência de Bases , Compartimento Celular , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Epigênese Genética , Feminino , Fator de Transcrição GATA3/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Camundongos SCID , Proteínas Mutantes/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Fator 3 de Transcrição de Octâmero/metabolismo , Fenótipo , Fatores de Transcrição SOXB1/metabolismo , Neoplasias de Mama Triplo Negativas/genética
13.
Nat Rev Cancer ; 19(1): 9-31, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30532012

RESUMO

Lung cancer is a major global health problem, as it is the leading cause of cancer-related deaths worldwide. Major advances in the identification of key mutational alterations have led to the development of molecularly targeted therapies, whose efficacy has been limited by emergence of resistance mechanisms. US Food and Drug Administration (FDA)-approved therapies targeting angiogenesis and more recently immune checkpoints have reinvigorated enthusiasm in elucidating the prognostic and pathophysiological roles of the tumour microenvironment in lung cancer. In this Review, we highlight recent advances and emerging concepts for how the tumour-reprogrammed lung microenvironment promotes both primary lung tumours and lung metastasis from extrapulmonary neoplasms by contributing to inflammation, angiogenesis, immune modulation and response to therapies. We also discuss the potential of understanding tumour microenvironmental processes to identify biomarkers of clinical utility and to develop novel targeted therapies against lung cancer.


Assuntos
Proliferação de Células/fisiologia , Neoplasias Pulmonares/patologia , Metástase Neoplásica/patologia , Microambiente Tumoral/fisiologia , Animais , Biomarcadores Tumorais/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Neoplasias Pulmonares/metabolismo , Processos Neoplásicos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Prognóstico
14.
JCI Insight ; 3(13)2018 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-29997286

RESUMO

Success of immune checkpoint inhibitors in advanced non-small-cell lung cancer (NSCLC) has invigorated their use in the neoadjuvant setting for early-stage disease. However, the cellular and molecular mechanisms of the early immune responses to therapy remain poorly understood. Through an integrated analysis of early-stage NSCLC patients and a Kras mutant mouse model, we show a prevalent programmed cell death 1/programmed cell death 1 ligand 1 (PD-1/PD-L1) axis exemplified by increased intratumoral PD-1+ T cells and PD-L1 expression. Notably, tumor progression was associated with spatiotemporal modulation of the immune microenvironment with dominant immunosuppressive phenotypes at later phases of tumor growth. Importantly, PD-1 inhibition controlled tumor growth, improved overall survival, and reprogrammed tumor-associated lymphoid and myeloid cells. Depletion of T lymphocyte subsets demonstrated synergistic effects of those populations on PD-1 inhibition of tumor growth. Transcriptome analyses revealed T cell subset-specific alterations corresponding to degree of response to the treatment. These results provide insights into temporal evolution of the phenotypic effects of PD-1/PD-L1 activation and inhibition and motivate targeting of this axis early in lung cancer progression.


Assuntos
Antígeno B7-H1/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Neoplasias Pulmonares/imunologia , Receptor de Morte Celular Programada 1/efeitos dos fármacos , Receptor de Morte Celular Programada 1/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Imunoterapia , Pulmão/patologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas p21(ras) , Linfócitos T , Microambiente Tumoral/imunologia
15.
J Biol Chem ; 292(2): 748-759, 2017 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-27903634

RESUMO

Targeted inhibitors of the human epidermal growth factor receptor 2 (HER2), such as trastuzumab and lapatinib, are among the first examples of molecularly targeted cancer therapy and have proven largely effective for the treatment of HER2-positive breast cancers. However, approximately half of those patients either do not respond to these therapies or develop secondary resistance. Although a few signaling pathways have been implicated, a comprehensive understanding of mechanisms underlying HER2 inhibitor drug resistance is still lacking. To address this critical question, we undertook a concerted approach using patient expression data sets, HER2-positive cell lines, and tumor samples biopsied both before and after trastuzumab treatment. Together, these methods revealed that high expression and activation of a specific subset of receptor tyrosine kinases (RTKs) was strongly associated with poor clinical prognosis and the development of resistance. Mechanistically, these RTKs are capable of maintaining downstream signal transduction to promote tumor growth via the suppression of cellular senescence. Consequently, these findings provide the rationale for the design of therapeutic strategies for overcoming drug resistance in breast cancer via combinational inhibition of the limited number of targets from this specific subset of RTKs.


Assuntos
Neoplasias da Mama , Resistencia a Medicamentos Antineoplásicos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Quinazolinas/farmacologia , Transdução de Sinais , Trastuzumab/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Lapatinib , Receptor ErbB-2 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
16.
Adv Exp Med Biol ; 927: 297-315, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27376740

RESUMO

The cellular signaling network plays a fundamental role during development and disease, especially cancer progression. By deregulating signaling pathways, cancer cells acquire hallmarks of the disease including uncontrolled proliferation, evasion from cell death, activation of angiogenesis, invasion, and metastasis. Noncoding RNAs make substantial contributions to regulating signal transduction in cancer, thereby promoting or suppressing different biological processes during tumorigenesis. This chapter provides an overview on the regulatory functions of noncoding RNAs in the signaling network in cancer cells. It summarizes examples of noncoding RNAs that act as oncogenes or tumor-suppressing genes involved in key signal pathways as well as signal crosstalk in cancer cells.


Assuntos
Carcinogênese/genética , Neoplasias/genética , Neovascularização Patológica/genética , RNA Longo não Codificante/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , Transdução de Sinais/genética
17.
Adv Exp Med Biol ; 899: 197-209, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27325268

RESUMO

Glioblastoma multiforme (GBM, WHO grade IV astrocytoma) is the most common and lethal primary brain tumor in adults, with an average survival of slightly more than 1 year after initial diagnosis. GBMs display significant heterogeneity within the tumor mass, among which a subpopulation of cells called glioma-initiating cells (GICs) is responsible for tumorigenesis and resistance to conventional therapies. Therefore, understanding the mechanism underlying the biological properties of GICs would help develop better therapies to target this population for GBM treatment. This protocol provides detailed procedures to isolate GICs and non-GICs from patient's specimen and glioma xenografts, which serves as the first step for the biological studies of GICs. Upon separation of GICs and non-GICs, a series of studies, such as expression profiling and functional screen, etc., can be performed to identify signal pathways responsible for the malignant nature of GICs. Besides, translational studies can also be conducted to examine drug responses of GICs. In sum, isolation of GICs with reliable methods will provide the basis for the further biological studies.


Assuntos
Separação Celular/métodos , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Antígenos CD/metabolismo , Biomarcadores Tumorais/metabolismo , Citometria de Fluxo , Humanos
18.
Oncotarget ; 7(15): 20381-94, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26967387

RESUMO

Triple-negative breast cancer (TNBC) presents a major challenge in the clinic due to its lack of reliable prognostic markers and targeted therapies. Accumulating evidence strongly supports the notion that microRNAs (miRNAs) are involved in tumorigenesis and could serve as biomarkers for diagnostic purposes. To identify miRNAs that functionally suppress metastasis of TNBC, we employed a concerted approach with selecting miRNAs that display differential expression profiles from bioinformatic analyses of breast cancer patient databases and validating top candidates with functional assays using breast cancer cell lines and mouse models. We have found that miR-148a exhibits properties as a tumor suppressor as its expression is inversely correlated with the ability of both human and mouse breast cancer cells to colonize the lung in mouse xenograft tumor models. Mechanistically, miR-148a appears to suppress the extravasation process of cancer cells, likely by targeting two genes WNT1 and NRP1 in a cell non-autonomous manner. Importantly, lower expression of miR-148a is detected in higher-grade tumor samples and correlated with increased likelihood to develop metastases and poor prognosis in subsets of breast cancer patients, particularly those with TNBC. Thus, miR-148a is functionally defined as a suppressor of breast cancer metastasis and may serve as a prognostic biomarker for this disease.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Pulmonares/secundário , MicroRNAs/genética , Neuropilina-1/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Proteína Wnt1/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Neuropilina-1/genética , Prognóstico , Taxa de Sobrevida , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Células Tumorais Cultivadas , Proteína Wnt1/genética , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Cancer Res ; 76(8): 2394-405, 2016 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-26893476

RESUMO

Chronic inflammation in liver tissue is an underlying cause of hepatocellular carcinoma. High levels of inflammatory cytokine IL18 in the circulation of patients with hepatocellular carcinoma correlates with poor prognosis. However, conflicting results have been reported for IL18 in hepatocellular carcinoma development and progression. In this study, we used tissue specimens from hepatocellular carcinoma patients and clinically relevant mouse models of hepatocellular carcinoma to evaluate IL18 expression and function. In a mouse model of liver fibrosis that recapitulates a tumor-promoting microenvironment, global deletion of the IL18 receptor IL18R1 enhanced tumor growth and burden. Similarly, in a carcinogen-induced model of liver tumorigenesis, IL18R1 deletion increased tumor burden. Mechanistically, we found that IL18 exerted inflammation-dependent tumor-suppressive effects largely by promoting the differentiation, activity, and survival of tumor-infiltrating T cells. Finally, differences in the expression of IL18 in tumor tissue versus nontumor tissue were more predictive of patient outcome than overall tissue expression. Taken together, our findings resolve a long-standing contradiction regarding a tumor-suppressive role for IL18 in established hepatocellular carcinoma and provide a mechanistic explanation for the complex relationship between its expression pattern and hepatocellular carcinoma prognosis. Cancer Res; 76(8); 2394-405. ©2016 AACR.


Assuntos
Carcinoma Hepatocelular/patologia , Interleucina-18/metabolismo , Neoplasias Hepáticas/patologia , Linfócitos do Interstício Tumoral/imunologia , Transdução de Sinais , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Knockout , Prognóstico , Receptores de Interleucina-18/genética
20.
Methods Mol Biol ; 1344: 325-39, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26520135

RESUMO

The transforming growth factor-ß (TGF-ß) is known to regulate a large number of biological processes and is involved in various aspects of tumor development. Recent studies have shown that the biogenesis of miRNAs can be regulated by TGF-ß signaling directly via Smad-dependent mechanisms and/or other unknown mechanisms, which may induce autoregulatory feedback loops in response to the activation of TGF-ß signaling, influencing the fate of tumor cells. In this chapter, we summarize the currently described mechanisms underlying TGF-ß's regulation of miRNA biogenesis, and the functional role of TGF-ß-regulated miRNAs in tumor initiation, epithelial-mesenchymal transition, and tumor microenvironment modulation. Finally, we introduce methods to study TGF-ß-regulated miRNAs and their functions in tumor progression and metastasis using an example of publication from our lab demonstrating the presence of a TGF-ß-miR-34a-CCL22 signaling axis, which serves as a potent etiological pathway for the development of hepatocellular carcinoma venous metastases.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Neoplasias/genética , Neoplasias/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Metástase Neoplásica , Neoplasias/patologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Microambiente Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...