Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Vaccines (Basel) ; 12(1)2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38250876

RESUMO

Colorectal cancer (CRC) currently ranks as the third most common cancer in the United States, and its incidence is on the rise, especially among younger individuals. Despite the remarkable success of immune checkpoint inhibitors (ICIs) in various cancers, most CRC patients fail to respond due to intrinsic resistance mechanisms. While microsatellite instability-high phenotypes serve as a reliable positive predictive biomarker for ICI treatment, the majority of CRC patients with microsatellite-stable (MSS) tumors remain ineligible for this therapeutic approach. In this study, we investigated the role of centrosomal protein 55 (CEP55) in shaping the tumor immune microenvironment in CRC. CEP55 is overexpressed in multiple cancer types and was shown to promote tumorigenesis by upregulating the PI3K/AKT pathway. Our data revealed that elevated CEP55 expression in CRC was associated with reduced T cell infiltration, contributing to immune exclusion. As CRC tumors progressed, CEP55 expression increased alongside sequential mutations in crucial driver genes (APC, KRAS, TP53, and SMAD4), indicating its involvement in tumor progression. CEP55 knockout significantly impaired tumor growth in vitro and in vivo, suggesting that CEP55 plays a crucial role in tumorigenesis. Furthermore, the CEP55 knockout increased CD8+ T cell infiltration and granzyme B production, indicating improved anti-tumor immunity. Additionally, we observed reduced regulatory T cell infiltration in CEP55 knockout tumors, suggesting diminished immune suppression. Most significantly, CEP55 knockout tumors demonstrated enhanced responsiveness to immune checkpoint inhibition in a clinically relevant orthotopic CRC model. Treatment with anti-PD1 significantly reduced tumor growth in CEP55 knockout tumors compared to control tumors, suggesting that inhibiting CEP55 could improve the efficacy of ICIs. Collectively, our study underscores the crucial role of CEP55 in driving immune exclusion and resistance to ICIs in CRC. Targeting CEP55 emerges as a promising therapeutic strategy to sensitize CRC to immune checkpoint inhibition, thereby improving survival outcomes for CRC patients.

2.
IET Syst Biol ; 17(6): 352-365, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37907428

RESUMO

With increasing research on idiopathic pulmonary fibrosis (IPF) and gastroesophageal reflux disease (GERD), more and more studies have indicated that GERD is associated with IPF, but the underlying pathological mechanisms remain unclear. The aim of the present study is to identify and analyse the differentially expressed genes (DEGs) between IPF and GERD and explore the relevant molecular mechanisms via bioinformatics analysis. Four GEO datasets (GSE24206, GSE53845, GSE26886, and GSE39491) were downloaded from the GEO database, and DEGs between IPF and GERD were identified with the online tool GEO2R. Subsequently, a series of bioinformatics analyses are conducted, including Kyoto Encyclopaedia of Genes and Genomes (KEGG) and gene ontology (GO) enrichment analyses, the PPI network, biological characteristics, TF-gene interactions, TF-miRNA coregulatory networks, and the prediction of drug molecules. Totally, 71 genes were identified as DEGs in IPF and GERD. Five KEGG pathways, including Amoebiasis, Protein digestion and absorption, Relaxin signalling pathway, AGE-RAGE signalling pathway in diabetic complications, and Drug metabolism - cytochrome P450, were significantly enriched. In addition, eight hub genes, including POSTN, MMP1, COL3A1, COL1A2, CXCL12, TIMP3, VCAM1, and COL1A1 were selected from the PPI network by Cytoscape software. Then, five hub genes (MMP1, POSTN, COL3A1, COL1A2, and COL1A1) with high diagnostic values for IPF and GERD were validated by GEO datasets. Finally, TF-gene and miRNA interaction was identified with hub genes and predicted drug molecules for the IPF and GERD. And the results suggest that cetirizine, luteolin, and pempidine may have great potential therapeutic value in IPF and GERD. This study will provide novel strategies for the identification of potential biomarkers and valuable therapeutic targets for IPF and GERD.


Assuntos
Refluxo Gastroesofágico , Fibrose Pulmonar Idiopática , MicroRNAs , Humanos , Perfilação da Expressão Gênica/métodos , Metaloproteinase 1 da Matriz/genética , Biomarcadores Tumorais/genética , MicroRNAs/genética , Fibrose Pulmonar Idiopática/genética , Refluxo Gastroesofágico/diagnóstico , Refluxo Gastroesofágico/genética , Biologia Computacional/métodos
3.
Mol Ther Oncolytics ; 31: 100727, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-37822487

RESUMO

Most colorectal cancer (CRC) patients present with a microsatellite-stable phenotype, rendering them resistant to immune checkpoint inhibitors (ICIs). Among the contributors to ICI resistance, tumor-derived extracellular vesicles (TEVs) have emerged as critical players. Previously we demonstrated that autologous transfer of TEVs without miR-424 can induce tumor antigen-specific immune responses in CRC models. Therefore, we postulated that allogeneic TEVs, modified to lack miR-424 and derived from an MC38 cells, could induce CD8+ T cell responses while restraining CT26 cell-based tumor. Here, we show that prophylactic administration of MC38 TEVs, without miR-424, showed a significant augmentation in CD8+ T-cells within CT26 tumors. This allogenic TEV effect was evident in CT26 tumors but not B16-F10 melanoma. Furthermore, we demonstrated the capacity of dendritic cells (DCs) to internalize TEVs, a possible mechanism to elicit immune response. Our investigation of autologously administered DCs, which had been exposed to modified TEVs, underscores their potential to dampen tumor growth while elevating CD8+ T cell levels vis-a-vis MC38 wild-type TEVs exposed to DCs. Notably, the modified TEVs were well tolerated and did not increase peripheral blood cytokine levels. Our findings underscore the potential of modified allogeneic TEVs without immune-suppressive factors to elicit robust T cell responses and limit tumor growth.

4.
Cancers (Basel) ; 15(16)2023 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-37627225

RESUMO

In recent years, cancer immunotherapy research has made remarkable progress, completely transforming the cancer treatment landscape [...].

5.
bioRxiv ; 2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-37131597

RESUMO

Colorectal Cancer (CRC) is the second leading cause of cancer-related death in the United States. Most CRC patients present with a microsatellite stable (MSS) phenotype and are highly resistant to immunotherapies. Tumor extracellular vesicles (TEVs), secreted by tumor cells, can contribute to intrinsic resistance to immunotherapy in CRC. We previously showed that autologous TEVs without functional miR-424 induce anti-tumor immune responses. We hypothesized that allogeneic modified CRC-TEVs without miR-424 (mouse homolog miR-322) derived from an MC38 background would effectively stimulate CD8+ T cell response and limit CT26 tumor growth. Here we show that prophylactic administration of MC38 TEVs without functional miR-424 significantly increased CD8+ T cells in CT26 CRC tumors and limited tumor growth, not B16-F10 melanoma tumors. We further show that the depletion of CD4+ and CD8+ T cells abolished the protective effects of MC38 TEVs without functional miR-424. We further show that TEVs can be taken up by DCs in vitro, and subsequent prophylactic administration of autologous DCs exposed to MC38 TEVs without functional miR-424 suppressed tumor growth and increased CD8+ T cells compared to MC38 wild-type TEVs exposed to DCs, in Balb/c mice bearing CT26 tumors. Notably, the modified EVs were well tolerated and did not increase cytokine expression in peripheral blood. These findings suggest that allogeneic-modified CRC-EVs without immune suppressive miR-424 can induce antitumor CD8+ T cell responses and limit tumor growth in vivo.

6.
Pharmacol Ther ; 241: 108332, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36526013

RESUMO

Despite significant advances in the screening, diagnosis, and treatment of colorectal cancer (CRC) immune checkpoint inhibitors (ICIs) continue to have limited utility outside of microsatellite-high disease. Given the durable response to immunotherapy seen across malignancies, increasing CRC response rates to ICI therapy is an active area of clinical research. An increasing body of work has demonstrated that tumor-derived extracellular vesicles (TEVs) are key modulators in tumor signaling and the determinants of the tumor microenvironment. Pre-clinical models have shown that TEVs are directly involved in antigen presentation and are involved in radiation-induced DNA damage signaling. Both direct and indirect modifications of these TEVs can alter CRC immunogenicity and ICI treatment response, making them attractive targets for potential therapeutic development. In addition, modified TEVs can be developed using several different mechanisms, with varied cargo including micro-RNAs and small peptide molecules. Recent work has shown strong pre-clinical evidence of injected modified TEV-induced ICI activity, with knockdown of the micro-RNA miR-424 in TEVs improving CRC immunogenicity and increasing anti-PD-1 activity in mouse models. Clinical trials are ongoing in the evaluation of modified TEVs in cancer therapy, but they appear to be a promising therapeutic target in CRC.


Assuntos
Neoplasias Colorretais , Vesículas Extracelulares , MicroRNAs , Animais , Neoplasias Colorretais/tratamento farmacológico , Imunoterapia , MicroRNAs/genética , Microambiente Tumoral , Humanos
7.
Pharmacol Ther ; 231: 107981, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34480964

RESUMO

Despite significant advances over the past 2 decades in preventive screening and therapy aimed at improving patient survival, colorectal cancer (CRC) remains the second most common cause of cancer death in the United States. The average 5-year survival rate of CRC patients with positive regional lymph nodes is only 40%, while less than 5% of patients with distant metastases survive beyond 5 years. There is a critical need to develop novel therapies that can improve overall survival in patients with poor prognoses, particularly since 60% of them are diagnosed at an advanced stage. Pertinently, immune checkpoint blockade therapy has dramatically changed how we treat CRC patients with microsatellite-instable high tumors. Furthermore, accumulating evidence shows that changes in gut microbiota are associated with the regulation of host antitumor immune response and cancer progression. Appropriate animal models are essential to deciphering the complex mechanisms of host antitumor immune response and tumor-gut microbiome metabolic interactions. Here, we discuss various mouse models of colorectal cancer that are developed to address key questions on tumor immune response and tumor-microbiota interactions. These CRC models will also serve as resourceful tools for effective preclinical studies.


Assuntos
Neoplasias Colorretais , Microbioma Gastrointestinal , Animais , Neoplasias Colorretais/tratamento farmacológico , Humanos , Imunidade , Camundongos
8.
Medicine (Baltimore) ; 101(52): e32388, 2022 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-36596048

RESUMO

To investigate the effect of dexmedetomidine (DEX) on hemodynamics and recovery period after femoral shaft fracture surgery. Fifty-two patients, aged 3 to 7 years, who underwent femoral shaft fracture reduction surgery in our hospital in 2019 were randomly divided into the experimental group (n = 26) and the control group (n = 26). Both groups were given routine propofol combined with remifentanil by intravenous anesthesia. The experimental group was continuously pumped with DEX after induction of anesthesia, while the control group was continuously pumped with the same volume of normal saline. The mean arterial pressure (MAP) and heart rate (HR) were recorded before anesthesia induction (T0), when laryngeal mask was inserted (T1), when skin was cut (T2), when intramedullary needle was inserted (T3), and when laryngeal mask was removed (T4). Extubation time after anesthesia withdrawal was recorded in the 2 groups. According to the Pediatric Anesthesia Emergence Delirium score, the agitation and the incidence of agitation were recorded immediately after extubation (T5), 10 minutes after entering the recovery room (T6) and 30 minutes after entering the recovery room (T7). There was no significant difference in MAP and HR between the 2 groups at T0 and T1 time points (P > .05). The MAP and HR of the experimental group at T2 to T4 were significantly lower than those of the control group (P < .05). The extubation time of the experimental group was longer than that of the control group (P < .05), but the Pediatric Anesthesia Emergence Delirium score and the incidence of agitation in the recovery period of the experimental group were lower than those of the control group (P < .05). In femoral shaft fracture surgery, intravenous anesthesia combined with continuous pumping DEX can effectively stabilize the hemodynamics of patients, and the incidence of postoperative agitation during anesthesia recovery is low.


Assuntos
Anestésicos , Dexmedetomidina , Delírio do Despertar , Propofol , Humanos , Criança , Delírio do Despertar/prevenção & controle , Propofol/efeitos adversos , Anestesia Geral
9.
Cancers (Basel) ; 13(22)2021 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-34830808

RESUMO

The past decade has seen immunotherapy rise to the forefront of cancer treatment [...].

10.
Cancers (Basel) ; 13(19)2021 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-34638505

RESUMO

Colorectal cancer (CRC) is one of the most common malignancies in both morbidity and mortality. Immune checkpoint blockade (ICB) treatments have been successful in a portion of mismatch repair-deficient (dMMR) CRC patients but have failed in mismatch repair-proficient (pMMR) CRC patients. Atypical Chemokine Receptor 4 (ACKR4) is implicated in regulating dendritic cell (DC) migration. However, the roles of ACKR4 in CRC development and anti-tumor immunoregulation are not known. By analyzing human CRC tissues, transgenic animals, and genetically modified CRC cells lines, our study revealed an important function of ACKR4 in maintaining CRC immune response. Loss of ACKR4 in CRC is associated with poor immune infiltration in the tumor microenvironment. More importantly, loss of ACKR4 in CRC tumor cells, rather than stromal cells, restrains the DC migration and antigen presentation to the tumor-draining lymph nodes (TdLNs). Moreover, tumors with ACKR4 knockdown become less sensitive to immune checkpoint blockade. Finally, we identified that microRNA miR-552 negatively regulates ACKR4 expression in human CRC. Taken together, our studies identified a novel and crucial mechanism for the maintenance of the DC-mediated T-cell priming in the TdLNs. These new findings demonstrate a novel mechanism leading to immunosuppression and ICB treatment resistance in CRC.

11.
J Healthc Eng ; 2021: 5169803, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34336155

RESUMO

This paper aimed to study the application of local anesthetics combined with transversus abdominis plane (TAP) block in gynecological laparoscopy (GLS) surgery during perioperative period under the guidance of ultrasound image enhanced by the wavelet transform image enhancement (WTIE) algorithm. 56 patients who underwent GLS surgery in hospital were selected and classified as the infiltrating group and block group. The puncture needle was guided by ultrasound images under WTIE algorithm, and 0.375% ropivacaine was adopted to block TAP. The results showed that the dosage of propofol in the infiltrating group (313.23 ± 19.67 mg) was remarkably inferior to the infiltrating group (377.67 ± 21.56 mg) (P < 0.05). The hospitalization time of patients in the infiltrating group (2.14 ± 0.18 days) was obviously shorter than that of the infiltrating group (3.23 ± 0.27 days) (P < 0.05). 3 h, 6 h, and 12 h after the operation, the visual analogue scores (3.82 ± 1.58 points, 2.97 ± 1.53 points, and 1.38 ± 0.57 points) of the patients in the infiltration group were considerably higher than the infiltrating group (2.31 ± 1.46 points, 1.06 ± 1.28 points, and 0.95 ± 0.43 points) (P < 0.05). 3 h, 6 h, and 12 h after the operation, the number of patients in the infiltrating group who used tramadol for salvage analgesia (2 cases, 1 case, and 1 case) was notably less than that in the infiltration group (9 cases, 7 cases, and 3 cases) (P < 0.05). In short, local anesthetics combined with TAP block can reduce postoperative VAS score and postoperative nausea and vomiting (PONV) score, which also reduced the incidence of postoperative analgesia.


Assuntos
Anestésicos Locais , Laparoscopia , Músculos Abdominais , Algoritmos , Analgésicos Opioides , Humanos , Aumento da Imagem , Dor Pós-Operatória/prevenção & controle , Análise de Ondaletas
12.
Gastroenterology ; 161(2): 560-574.e11, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33895168

RESUMO

BACKGROUND & AIMS: Colorectal cancer is a major cause of cancer-related deaths worldwide. Immune checkpoint blockade therapies are effective in 30%-60% of the microsatellite instable-high subtype. Unfortunately, most patients with colorectal cancer (>85%) have microsatellite stable tumors that do not respond. In this study, we aimed to decipher the underlying tumor-intrinsic mechanisms critical for improving immunotherapy in colorectal cancer. METHODS: We used human and mouse tumor samples, cell lines, human colorectal cancer organoids, and various syngeneic orthotopic mouse models of late-stage colorectal cancer to define the effects of tumor cell-secreted extracellular vesicles (EVs) on antitumor immune response. RESULTS: Our analyses of human colorectal cancer immune profiles and tumor-immune cell interactions showed that tumor-secreted EVs containing microRNA miR-424 suppressed the CD28-CD80/86 costimulatory pathway in tumor-infiltrating T cells and dendritic cells, leading to immune checkpoint blockade resistance. Modified tumor-secreted EVs with miR-424 knocked down enhanced T-cell-mediated antitumor immune response in colorectal cancer tumor models and increased the immune checkpoint blockade response. Intravenous injections of modified tumor-secreted EVs induced tumor antigen-specific immune responses and boosted the immune checkpoint blockade efficacy in colorectal cancer models that mimic aggressively progressing, late-stage disease. CONCLUSIONS: Collectively, we show a critical role for tumor-secreted EVs in antitumor immune regulation and immunotherapy response, which could be developed as a novel treatment for immune checkpoint blockade-resistant colorectal cancer.


Assuntos
Neoplasias Colorretais/imunologia , Vesículas Extracelulares/imunologia , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Microambiente Tumoral , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígeno B7-2/genética , Antígeno B7-2/metabolismo , Antígenos CD28/genética , Antígenos CD28/metabolismo , Células CACO-2 , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Células HT29 , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Células Jurkat , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , MicroRNAs/genética , MicroRNAs/metabolismo , Fenótipo , Hipóxia Tumoral
13.
Cancers (Basel) ; 12(5)2020 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-32380703

RESUMO

Immune checkpoint blockade therapy (ICBT) has revolutionized the treatment and management of numerous cancers, yet a substantial proportion of patients who initially respond to ICBT subsequently develop resistance. Comprehensive genomic analysis of samples from recent clinical trials and pre-clinical investigation in mouse models of cancer provide insight into how tumors evade ICBT after an initial response to treatment. Here, we summarize our current knowledge on the development of acquired ICBT resistance, by examining the mechanisms related to tumor-intrinsic properties, T-cell function, and tumor-immune cell interactions. We discuss current and future management of ICBT resistance, and consider crucial questions remaining in this field of acquired resistance to immune checkpoint blockade therapies.

14.
iScience ; 23(5): 101056, 2020 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-32344378

RESUMO

Immunotherapies are used as adjuvant therapies for cancers. However, knowledge of how traditional cancer treatments affect immunotherapies is limited. Using mouse models, we demonstrate that tumor-draining lymph nodes (TdLNs) are critical for tumor antigen-specific T cell response. However, removing TdLNs concurrently with established primary tumors did not affect the immune checkpoint blockade (ICB) response on localized secondary tumor due to immunotolerance in TdLNs and distribution of antigen-specific T cells in peripheral lymphatic organs. Notably, treatment response improved with sequential administration of 5-fluorouracil (5-FU) and ICB compared with concurrent administration of ICB with 5-FU. Immune profiling revealed that using 5-FU as induction treatment increased tumor visibility to immune cells, decreased immunosuppressive cells in the tumor microenvironment, and limited chemotherapy-induced T cell depletion. We show that the effect of traditional cytotoxic treatment, not TdLNs, influences immunotherapy response in localized secondary tumors. We postulate essential considerations for successful immunotherapy strategies in clinical conditions.

15.
Clin Drug Investig ; 39(4): 341-353, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30778885

RESUMO

Lung cancer is the most prevalent and deadly cancer worldwide. Immune checkpoint therapy, which targets regulatory pathways in T cells to boost anti-tumor immune response, has revolutionized lung cancer treatment paradigms. Inhibitors of the most established immune checkpoints such as programmed death-1 (PD-1)/PD-ligand 1 (PD-L1) have been approved by the US Food and Drug Administration in the management of lung cancer. Despite the pronounced survival benefits that have been seen with immune checkpoint inhibitors, not all lung cancer patients respond to single-agent immunotherapy due to the complexity of the immune microenvironment and tumor resistance. Alternative immune checkpoints beyond PD-1/PD-L1 must be sought so that more patients can benefit from immune checkpoint therapy. Additionally, novel combination strategies of immunotherapy and conventional treatments (e.g., chemotherapy, radiotherapy, and targeted therapy) have shown promise in some clinical trials. Meanwhile, identification of predictive biomarkers is pivotal in selecting eligible patients for immunotherapy and to guide individualized clinical decision-making. The future of immune checkpoint therapy in lung cancer is not devoid of challenges, and more prospective clinical studies are awaited to translate our understanding from bench to bedside.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/tendências , Neoplasias Pulmonares/tratamento farmacológico , Animais , Pontos de Checagem do Ciclo Celular/imunologia , Previsões , Humanos , Imunoterapia/métodos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Estudos Prospectivos
16.
Hum Pathol ; 77: 54-62, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29634976

RESUMO

Synthesis of cytochrome c oxidase 2 (SCO2) and TP53-induced glycolysis and apoptosis regulator (TIGAR) are 2 p53-mediated proteins that can play a regulatory role in cancer energy metabolism. However, no study has examined the association of SCO2 and TIGAR with the prognosis of patients with lung adenocarcinoma (AC). In our study, the expression of SCO2 and TIGAR proteins in lung AC was detected, and the potential relation to prognosis was evaluated, aiming to take a further view of lung AC progression. Quantum dots-based immunofluorescence histochemistry staining was performed to observe the expression of p53, SCO2, and TIGAR in 75 specimens of lung AC. Of these, 51 (68.0%) showed high expression of SCO2, and 59 (78.7%) showed high expression of TIGAR. High TIGAR expression was significantly associated with a history of smoking (P = .017) and being male (P = .006). The correlation between high SCO2 expression and age also was significant (P = .042). Moreover, high TIGAR expression was positively correlated with high SCO2 expression (P = .019; rs = 0.271). High expression of the SCO2 and TIGAR proteins predicted poorer survival and a higher mortality rate (P = .024 and .030, respectively). High expression of SCO2 and TIGAR proteins is significantly associated with lung AC progression, suggesting their potential use as prognostic markers and therapeutic targets.


Assuntos
Adenocarcinoma de Pulmão/metabolismo , Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Mitocondriais/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Metabolismo Energético/fisiologia , Feminino , Glicólise , Humanos , Masculino , Pessoa de Meia-Idade , Chaperonas Moleculares
17.
Transl Res ; 196: 62-70, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29518351

RESUMO

Immune checkpoint blockade therapy (ICBT) has resulted in extended overall survival for some patients with certain types of cancer, most prominently including colorectal cancer (CRC) associated with microsatellite instability (MSI). However, most patients with CRC whose phenotypes have microsatellite stability (MSS) are unresponsive to ICBT. In efforts to understand the responsiveness of CRC tumors to ICBT, genotypic and phenotypic signatures of CRC tumors are now being investigated. The MSI and MSS classification has been clinically validated as helpful in predicting response vs nonresponse to ICBT in patients with CRC. Other potential predictive markers include mutational and neoantigen loads, T-cell receptor diversity, and the immune score system, all of which have mechanistic connections to ICBT response. These novel predictive signatures could provide unprecedented insights into patients with CRC associated with MSS. Clinical trials or prospective cohort studies using standardized methodologies for biomarker quantification should be illuminating. Further validation of these novel predictive signatures will be essential to tailoring treatment of patients whose CRC is most likely to respond to ICBT.


Assuntos
Antígeno CTLA-4/antagonistas & inibidores , Neoplasias Colorretais/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Neoplasias Encefálicas/complicações , Neoplasias Colorretais/complicações , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Dano ao DNA , Genótipo , Humanos , Instabilidade de Microssatélites , Síndromes Neoplásicas Hereditárias/complicações , Fenótipo
18.
Asian Pac J Cancer Prev ; 19(2): 401-406, 2018 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-29479989

RESUMO

Background: Lung cancer is the leading causes of cancer-related deaths around the world. Abnormal activation of the hedgehog (Hh) signaling pathway has been found to be involved in the occurrence, invasion, and metastasis of cancers. Autophagy also plays a significant role in the growth and metastasis of cancers. However, the correlation between the Hh signaling pathway and autophagy in small cell lung cancer (SCLC) is still poorly understood. This study aimed to investigate the significance of Hh signaling pathway and autophagy in SCLC. Materials and Methods: The expression of the Hh-induced transcriptional factor, glioma associated oncogene-1 (Gli-1) and the autophagy-related molecule caveolin-1 (Cav-1) and their clinical significance was performed to detect and assay by immunohistochemistry in tissue microarray including 70 patients with SCLC. Results: In our study, 47 (67.1%) patients had positive Gli-1 expression, 49 (70.0%) patients had positive Cav-1 expression, and 44 (62.9%) patients had negative fibroblastic Cav-1 expression. In SCLC, Gli-1 expression increased markedly, and was closely associated with decreased fibroblastic Cav-1 expression. Furthermore, we also found that Gli-1 expression was closely associated with increased Cav-1 expression. Conclusions: Our findings suggested that abnormal activation of the Hh signaling pathway is closely related to autophagy in SCLC. We envision that novel targets may come with the further investigation of Gli-1 and Cav-1 in carcinogenesis of SCLC.


Assuntos
Biomarcadores Tumorais/metabolismo , Caveolina 1/metabolismo , Neoplasias Pulmonares/patologia , Recidiva Local de Neoplasia/patologia , Carcinoma de Pequenas Células do Pulmão/secundário , Proteína GLI1 em Dedos de Zinco/metabolismo , Adulto , Idoso , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Recidiva Local de Neoplasia/metabolismo , Prognóstico , Transdução de Sinais , Carcinoma de Pequenas Células do Pulmão/metabolismo , Análise Serial de Tecidos , Adulto Jovem
19.
Cancer Res ; 78(5): 1321-1333, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29259015

RESUMO

Resident fibroblasts that contact tumor epithelial cells (TEC) can become irreversibly activated as cancer-associated-fibroblasts (CAF) that stimulate oncogenic signaling in TEC. In this study, we evaluated the cross-talk between CAF and TEC isolated from tumors generated in a mouse model of KRAS/mut p53-induced pancreatic cancer (KPC mice). Transcriptomic profiling conducted after treatment with the anticancer compound Minnelide revealed deregulation of the TGFß signaling pathway in CAF, resulting in an apparent reversal of their activated state to a quiescent, nonproliferative state. TEC exposed to media conditioned by drug-treated CAFs exhibited a decrease in oncogenic signaling, as manifested by downregulation of the transcription factor Sp1. This inhibition was rescued by treating TEC with TGFß. Given promising early clinical studies with Minnelide, our findings suggest that approaches to inactivate CAF and prevent tumor-stroma cross-talk may offer a viable strategy to treat pancreatic cancer.Significance: In an established mouse model of pancreatic cancer, administration of the promising experimental drug Minnelide was found to actively deplete reactive stromal fibroblasts and to trigger tumor regression, with implications for stromal-based strategies to attack this disease. Cancer Res; 78(5); 1321-33. ©2018 AACR.


Assuntos
Fibroblastos Associados a Câncer/patologia , Carcinogênese , Carcinoma Ductal Pancreático/patologia , Modelos Animais de Doenças , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pancreáticas/patologia , Animais , Apoptose , Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Proliferação de Células , Diterpenos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Compostos de Epóxi , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Organofosfatos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Fenantrenos/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
20.
Curr Cancer Drug Targets ; 18(6): 558-566, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29189160

RESUMO

Glioma-associated oncogenes (GLIs) are zinc finger protein family members and downstream regulatory factors of the classic Hedgehog (Hh) signaling pathway. GLI proteins influence the growth and development of organisms and aid in tissue repair. However, aberrant expression of the GLI family member GLI1 promotes carcinogenesis by inducing epithelial-mesenchymal transition (EMT), angiogenesis, and other signaling pathways. Overexpression of GLI1 is thought to be an indicator of poor prognosis as well as a potential therapeutic target for cancers. GLI inhibitors such as zerumbone, GANT61, resveratrol, and cyclopamine depress the Hh pathway in vitro and in vivo cancer research, and other non-canonical pathways may also activate expression of GLI1. Here, we summarize GLI function in carcinogenesis and cancer-targeted therapy.


Assuntos
Antineoplásicos/uso terapêutico , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Glioma/tratamento farmacológico , Glioma/patologia , Terapia de Alvo Molecular , Proteína GLI1 em Dedos de Zinco/antagonistas & inibidores , Carcinogênese/metabolismo , Glioma/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA