Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
NPJ Precis Oncol ; 7(1): 50, 2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37258779

RESUMEN

Adjuvant chemotherapy (ACT) is usually used to reduce the risk of disease relapse and improve survival for stage II/III colorectal cancer (CRC). However, only a subset of patients could benefit from ACT. Thus, there is an urgent need to identify improved biomarkers to predict survival and stratify patients to refine the selection of ACT. We used high-throughput proteomics to analyze tumor and adjacent normal tissues of stage II/III CRC patients with /without relapse to identify potential markers for predicting prognosis and benefit from ACT. The machine learning approach was applied to identify relapse-specific markers. Then the artificial intelligence (AI)-assisted multiplex IHC was performed to validate the prognostic value of the relapse-specific markers and construct a proteomic-derived classifier for stage II/III CRC using 3 markers, including FHL3, GGA1, TGFBI. The proteomics profiling-derived signature for stage II/III CRC (PS) not only shows good accuracy to classify patients into high and low risk of relapse and mortality in all three cohorts, but also works independently of clinicopathologic features. ACT was associated with improved disease-free survival (DFS) and overall survival (OS) in stage II (pN0) patients with high PS and pN2 patients with high PS. This study demonstrated the clinical significance of proteomic features, which serve as a valuable source for potential biomarkers. The PS classifier provides prognostic value for identifying patients at high risk of relapse and mortality and optimizes individualized treatment strategy by detecting patients who may benefit from ACT for survival.

2.
Clin Transl Gastroenterol ; 13(5): e00475, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35297794

RESUMEN

INTRODUCTION: Antiprogrammed cell death protein-1 (PD-1) immunotherapy has substantially broadened in scope for the treatment of colorectal cancer (CRC). However, comparative safety, efficacy and survival outcome of anti-PD-1 therapy in CRC patients with and without hepatitis B virus (HBV) infection remain unclear. METHODS: This multicenter, retrospective cohort study included 180 advanced-stage CRC patients with available serological markers for HBV infection treated with anti-PD-1 therapy at the Sixth Affiliated Hospital, Sun Yat-sen University and Sun Yat-sen University Cancer Center between December 2016 and December 2019. A propensity score-matched analysis was performed to compare the safety, efficacy, and survival outcome between HBV and non-HBV groups. RESULTS: The incidences of deficient mismatch repair and metastatic disease were significantly different between HBV and non-HBV groups (both P < 0.05). After propensity score-matched analysis, any grade immune-related adverse events and grade ≥ 3 immune-related adverse events were 47% vs 38% (P = 0.25) and 5% vs 6% (P = 1.0) between HBV and non-HBV groups, respectively. The overall response rate was 39% with 17 complete responses and 13 partial responses for the HBV infection cohort and 39% with 11 complete responses and 19 partial responses for the non-HBV infection cohort (P = 1.0). Two-year progression-free survival rates were 38% vs 40% (P = 0.596) and 2-year overall survival rates were 55% vs 63% (P = 0.401) for HBV vs non-HBV infection cohorts. DISCUSSION: The incidences of toxicity, efficacy and survival outcome were similar between patients with HBV infection and non-HBV patients receiving anti-PD-1 therapy, which supports to include CRC patients with HBV in clinical trials of anti-PD-1 therapy.


Asunto(s)
Neoplasias Colorrectales , Hepatitis B , Estudios de Cohortes , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Hepatitis B/complicaciones , Hepatitis B/tratamiento farmacológico , Virus de la Hepatitis B/genética , Humanos , Inmunoterapia/efectos adversos , Estudios Retrospectivos
3.
Front Immunol ; 13: 809971, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35185898

RESUMEN

Purpose: Deficient mismatch repair (dMMR) is an established biomarker for the response to the programmed cell death (PD)-1 inhibitors in metastatic colorectal cancer (mCRC). Although patients with dMMR mCRC could achieve a high incidence of disease control and favorable progression-free survival (PFS), reported response rates to PD-1 inhibitors are variable from 28% to 52%. We aimed to explore the additional predictive biomarkers associated with response to anti-PD-1 immunotherapy in patients with dMMR mCRC. Methods: This multicenter cohort study enrolled patients with dMMR mCRC receiving anti-PD-1 immunotherapy at the Sixth Affiliated Hospital of Sun Yat-sen University and Sun Yat-sen University Cancer Center between December 2016 and December 2019. The total information of 20 peripheral blood biomarkers, including T cells (frequency of CD4+ T cell, frequency of CD8+ T cell, and ratio of CD4+/CD8+), carcinoembryonic antigen (CEA), inflammatory markers, and lipid metabolism markers, was collected. The association between response or survival and peripheral blood parameters was analyzed. Results: Among the tested parameters, the ratio of CD4+/CD8+ and frequency of CD4+ T cell were significantly associated with PFS (p = 0.023, p = 0.012) and overall survival (OS; p = 0.027, p = 0.019) in a univariate analysis. A lower level of CD4+/CD8+ ratio or frequency of CD4+ T cell showed a significant association with better overall response rates (ORRs; p = 0.03, p = 0.01). The ratio of CD4+/CD8+ and frequency of CD4+ T cell maintained significance in multivariate Cox model for PFS (HR = 9.23, p = 0.004; HR = 4.83, p = 0.02) and OS (HR = 15.22, p = 0.009; HR = 16.21, p = 0.025). Conclusion: This study indicated that the ratio of CD4+/CD8+ and the frequency of CD4+ T cell might be crucial independent biomarkers within dMMR mCRC to better identify patients for anti-PD-1 immunotherapy. If validated in prospective clinical trials, the ratio of CD4+/CD8+ and the frequency of CD4+ T cell might aid in guiding the treatment of PD-1 inhibitors among patients with dMMR mCRC.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Reparación de la Incompatibilidad de ADN , Adulto , Anciano , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Estudios de Cohortes , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/inmunología , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Masculino , Persona de Mediana Edad , Supervivencia sin Progresión , Adulto Joven
4.
NPJ Precis Oncol ; 4: 26, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32964128

RESUMEN

Prior reports have indicated that defective mismatch repair (MMR) has a favorable impact on outcome in colorectal cancer patients treated with surgery, immunotherapy, or adjuvant chemotherapy. However, the impact of MMR status on response to neoadjuvant radiotherapy in rectal cancer is not well understood. Here we report that dMMR was associated with improved disease-free survival (DFS) (P = 0.034) in patients receiving neoadjuvant chemotherapy (NCT). Patients with dMMR tumors who received neoadjuvant chemoradiotherapy (NCRT) achieved significantly worse DFS (P = 0.026) than those treated with NCT. Conversely, NCRT improved DFS (P = 0.043) in patients with pMMR tumors, especially for stage III disease with improved DFS (P = 0.02). The presence of dMMR was associated with better prognosis in rectal cancer patients treated with NCT. NCT benefited patients with dMMR tumors; while NCRT benefited patients with stage III disease and pMMR tumors. Patients stratified by MMR status may provide a more tailored approach to rectal cancer neoadjuvant therapy.

5.
Front Oncol ; 10: 214, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32266127

RESUMEN

Purpose: To investigate the predictive biomarker value of estrogen receptor 1 (ESR1) expression in tumor tissue on adjuvant chemotherapy in curatively resected colorectal cancer (CRC). Methods: A total of 467 CRC patients in 2007-2010 were retrospectively evaluated. Clinical information and follow-up data were retrieved from hospital registries and patient files. What's more, we used an external independent cohort (n = 511) from GSE39582 for further validation. Overall survival was estimated by the Kaplan-Meier method, and the survival curves were compared by log-rank tests. Cox proportional hazards models were used for multivariate analyses to calculate the hazard ratios (HRs) and test independent significance. Immunohistochemistry and Western blot were applied to detect protein expression of ESR1 in CRC patients and cell lines. The stable knockdown and overexpressed cells were transduced with the lentivirus. Cell viability was measured by an MTS reagent. Results: The predictive value of ESR1 was investigated in locally advanced CRC patients. Kaplan-Meier analysis indicated that ESR1 expression was significantly correlated with OS in patients receiving adjuvant chemotherapy from these cohorts, with p = 0.015 and p < 0.001, respectively. ESR1 expression was significantly correlated with 5-flurouracil (5-FU)-based adjuvant chemotherapy in training with an HR of 1.792 (95%CI: 1.100-2.921, p = 0.019). Downregulation of ESR1 was related with enhanced chemosensitivity to 5-FU in CRC cell lines, while upregulation of ESR1 was correlated with decreased chemosensitivity. Conclusions: The present study manifest clinical validity of ESR1 expression as a predictive biomarker on 5-FU-based adjuvant chemotherapy in stage II-III CRC.

6.
Ann Transl Med ; 8(4): 96, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32175389

RESUMEN

BACKGROUND: Metastatic colorectal cancer (mCRC) is a heterogeneous disease. Predictive biomarkers are in great demand to optimize patient selection at high risk for death and to provide a novel insight into potential targeted therapy. METHODS: The present study retrospectively analyzed the gene expression profiles of tumor tissue samples from 4 public CRC cohorts, including 1 RNA-Seq data set from The Cancer Genome Atlas (TCGA) CRC cohort and 3 microarray data sets from GEO. Prognostic analysis was performed to test the predictive value of prognostic gene signature. RESULTS: Of 192 patients, 108 patients (56.3%) were men and median age was 65 years. A prognostic gene signature that consisted of 15 unique genes was generated in the discovery cohort. In the meta-validation cohorts, the signature significantly classified patients into high-risk and low-risk groups with regard to overall survival (OS) in mCRC patients with advanced stage disease and remained as an independent prognostic marker in multivariable analysis (1.57; 95% CI: 1.16-2.11; P=0.003) after adjusting for clinical parameters and molecular types. Gene Set Enrichment Analysis showed that several biological processes, including angiogenesis (P<0.001), epithelial mesenchymal transit (P<0.001) and inflammatory response (P=0.001), were enriched among this prognostic gene signature. CONCLUSIONS: The proposed prognostic gene signature is a promising prognostic tool to estimate OS in mCRC. Prospective larger studies to examine the clinical utility of the biomarkers to guide individualized treatment of mCRC are warranted.

7.
BMC Cancer ; 19(1): 933, 2019 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-31590647

RESUMEN

BACKGROUND: Prognostic assessment is crucial for optimal treatment. The aim of our study was to investigate the potential impact of estrogen receptor-α (ER-α) and progesterone receptor (PR) on the prognosis of colorectal cancer (CRC) patients who received curative resection. METHODS: Retrospective evaluation of two independent cohorts of CRC patients maintained prospectively in 2009-2010 (training set) (n = 148) and 2007-2009 (internal validation set) (n = 485). Furthermore, we used an external independent CRC cohort from The Cancer Genome Atlas (TCGA) (n = 511) for further validation. ER-α and PR expression as well as other potential prognostic factors were retrospectively evaluated in training set with respect to overall survival (OS), local relapse free survival (LRFS) and distant metastasis free survival (DMFS). The prognostic factors found in training set will be validated in two validation cohorts. RESULTS: On univariate analysis for the training set, OS, LRFS and DMFS were not associated with PR expression. While patients with ER-αexpression were found to have poor prognosis. In addition, multivariate analysis showed that ER-αexpression maintained significance with respect to OS (HR, 5.06; p = 0.002), LRFS (HR, 8.81; p = 0.002) and DMFS (HR, 8.07; p = 0.004). Similarly, ER-α expression showed prognostic significance with respect to OS with hazard ratios (HRs) of 1.572 (95% CI: 1.001-2.467, p = 0.049) and 1.624 (95% CI: 1.047-2.520, p = 0.031) for the internal and external validation cohort, respectively. CONCLUSION: ER-α expression was a biomarker of poor prognosis and it might inform treatment decision for high risk CRC patients. However, PR expression was not associated with survival outcomes.


Asunto(s)
Neoplasias Colorrectales/mortalidad , Receptor alfa de Estrógeno/metabolismo , Receptores de Progesterona/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/cirugía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
8.
Cell Death Differ ; 26(11): 2314-2328, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30816302

RESUMEN

Stimulator of interferon genes (STING), a major adaptor protein in antiviral innate immune signaling, is considered as one of the most important regulators of antiviral and antitumor immunity. Although STING agonists are now intensively studied in clinical trials as a new class of adjuvants to boost cancer immunotherapy, the tumor-intrinsic role of the STING pathway in shaping the tumor microenvironment remains controversial. Here, we discovered that STING plays a vital role in regulation of myeloid-derived suppressor cell (MDSC) differentiation and antitumor immunity in Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC). Mechanistic analyses reveal that STING represses NPC-derived MDSC induction by enhancing SOCS1 expression in both tumor cells and MDSCs. SOCS1 physically interacts with STAT3 through its SH2 domain to prevent STAT3 phosphorylation and dimerization, resulting in reduced MDSC induction via inhibition of GM-CSF and IL-6 production. Notably, reduced tumoral STING expression was found to be significantly associated with a poor prognosis for NPC patients. Our findings reveal a novel mechanism linking STING to tumor microenvironmental cytokine production and MDSC induction.


Asunto(s)
Proteínas de la Membrana/metabolismo , Células Supresoras de Origen Mieloide/citología , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/patología , Microambiente Tumoral/fisiología , Sistemas CRISPR-Cas , Diferenciación Celular/inmunología , Línea Celular Tumoral , Dimerización , Infecciones por Virus de Epstein-Barr/patología , Técnicas de Inactivación de Genes , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Células HEK293 , Herpesvirus Humano 4/inmunología , Humanos , Interleucina-6/antagonistas & inhibidores , Proteínas de la Membrana/genética , Células Supresoras de Origen Mieloide/inmunología , Carcinoma Nasofaríngeo/inmunología , Carcinoma Nasofaríngeo/virología , Neoplasias Nasofaríngeas/inmunología , Neoplasias Nasofaríngeas/virología , Fosforilación/fisiología , Pronóstico , Factor de Transcripción STAT3/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Microambiente Tumoral/efectos de los fármacos
9.
PLoS Pathog ; 13(7): e1006503, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28732079

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are expanded in tumor microenvironments, including that of Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC). The link between MDSC expansion and EBV infection in NPC is unclear. Here, we show that EBV latent membrane protein 1 (LMP1) promotes MDSC expansion in the tumor microenvironment by promoting extra-mitochondrial glycolysis in malignant cells, which is a scenario for immune escape initially suggested by the frequent, concomitant detection of abundant LMP1, glucose transporter 1 (GLUT1) and CD33+ MDSCs in tumor sections. The full process has been reconstituted in vitro. LMP1 promotes the expression of multiple glycolytic genes, including GLUT1. This metabolic reprogramming results in increased expression of the Nod-like receptor family protein 3 (NLRP3) inflammasome, COX-2 and P-p65 and, consequently, increased production of IL-1ß, IL-6 and GM-CSF. Finally, these changes in the environment of malignant cells result in enhanced NPC-derived MDSC induction. One key step is the physical interaction of LMP1 with GLUT1 to stabilize the GLUT1 protein by blocking its K48-ubiquitination and p62-dependent autolysosomal degradation. This work indicates that LMP1-mediated glycolysis regulates IL-1ß, IL-6 and GM-CSF production through the NLRP3 inflammasome, COX-2 and P-p65 signaling pathways to enhance tumor-associated MDSC expansion, which leads to tumor immunosuppression in NPC.


Asunto(s)
Carcinoma/fisiopatología , Infecciones por Virus de Epstein-Barr/fisiopatología , Herpesvirus Humano 4/metabolismo , Células Supresoras de Origen Mieloide/citología , Neoplasias Nasofaríngeas/fisiopatología , Proteínas de la Matriz Viral/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/virología , Línea Celular Tumoral , Proliferación Celular , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Regulación Neoplásica de la Expresión Génica , Glucólisis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/virología , Transducción de Señal , Proteínas de la Matriz Viral/genética
10.
J Pathol ; 240(3): 329-340, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27538493

RESUMEN

Recent studies have shown that extracellular microRNAs are not only potential biomarkers but are also involved in cell interactions to regulate the intercommunication between cancer cells and their microenvironments in various types of malignancies. In this study, we isolated exosomes from nasopharyngeal carcinoma (NPC) cell lines and patient sera (T-EXOs), or control NP69 cells and healthy donor sera (HD-EXOs). We found that miR-24-3p was markedly enriched in T-EXOs as compared with HD-EXOs; the serum exosomal miR-24-3p level was correlated with worse disease-free survival of patients (p < 0.05). Knockdown of exosomal miR-24-3p (miR-24-3p-sponge-T-EXOs) by a sponge RNA targeting miR-24-3p restored the T-EXO-mediated (control-sponge-T-EXO) inhibition of T-cell proliferation and Th1 and Th17 differentiation, and the induction of regulatory T cells (Tregs). Mechanistic analyses revealed that administration of exosomal miR-24-3p increased P-ERK, P-STAT1 and P-STAT3 expression while decreasing P-STAT5 expression during T-cell proliferation and differentiation. Moreover, by in vivo and in vitro assessments, we found FGF11 to be a direct target of miR-24-3p. However, both miR-24-3p-sponge-T-EXOs and T-EXOs (control-sponge-T-EXOs) impeded proliferation and Th1 and Th17 differentiation, but induced Treg differentiation, of lenti-shFGF11-transfected T cells. The levels of phosphorylated ERK and STAT proteins were different in lenti-ScshRNA-transfected T cells and lenti-shFGF11-transfected T cells following administration of miR-24-3p-sponge-T-EXO. Interestingly, tumour FGF11 expression was positively correlated with the number of CD4+ and CD8+ T cells in vivo, and predicted favourable patient DFS (p < 0.05). Additionally, hypoxia increased cellular and exosomal miR-24-3p levels and enhanced the inhibitory effect of T-EXO on T-cell proliferation and differentiation. Collectively, our findings suggest that exosomal miR-24-3p is involved in tumour pathogenesis by mediating T-cell suppression via repression of FGF11, and may serve as a potential prognostic biomarker in NPC. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Carcinoma/genética , Exosomas/genética , Factores de Crecimiento de Fibroblastos/genética , MicroARNs/genética , Neoplasias Nasofaríngeas/genética , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Carcinoma/diagnóstico , Carcinoma/inmunología , Carcinoma/patología , Comunicación Celular , Diferenciación Celular , Proliferación Celular , Supervivencia sin Enfermedad , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/diagnóstico , Neoplasias Nasofaríngeas/inmunología , Neoplasias Nasofaríngeas/patología , Pronóstico , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
11.
Br J Radiol ; 89(1065): 20140160, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27376481

RESUMEN

OBJECTIVE: To report a novel approach for craniospinal irradiation (CSI) using a supine isocentric technique. METHODS: Patients were treated in the supine position using CT simulation. Half-beam-blocked lateral cranial fields and superior spinal fields have the same isocentre, and their beam divergences match. Tangential irradiation provides a non-divergent junction for the other two full-beam spinal fields. Shielding for cranial fields was generated, and dose distribution was calculated using a three-dimensional planning system. When sacral spinal fields were required, two lateral opposite fields were designed to protect the urogenital organs. All treatment portals were filmed once per week. RESULTS: At a median follow-up of 49.8 months, 5 relapses and no cases of radiation myelitis developed in 26 consecutive patients. In the junctions of the brain-spine or spine-spine field, no failure occurred. Three failures occurred in the primary site alone, two in the spinal axis alone. CONCLUSION: The results of our study have shown that our novel approach for CSI was not associated with increased failures at the field junction and deaths. In addition, no radiation myelitis, pneumonia, severe damage to the heart and gastrointestinal tract, and second cancers occurred in our study. ADVANCES IN KNOWLEDGE: This new approach is an optimal alternative in cancer centre without tomotherapy because of its convenience for immobilization, repeatability, optimal dose distribution and satisfactory clinical outcome.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Irradiación Craneoespinal/métodos , Adolescente , Adulto , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/cirugía , Niño , Preescolar , Terapia Combinada , Ependimoma/tratamiento farmacológico , Ependimoma/radioterapia , Ependimoma/cirugía , Femenino , Humanos , Masculino , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/radioterapia , Meduloblastoma/cirugía , Recurrencia Local de Neoplasia/etiología , Neoplasias de Células Germinales y Embrionarias/tratamiento farmacológico , Neoplasias de Células Germinales y Embrionarias/radioterapia , Neoplasias de Células Germinales y Embrionarias/cirugía , Dosificación Radioterapéutica , Estudios Retrospectivos , Posición Supina , Teratoma/tratamiento farmacológico , Teratoma/radioterapia , Teratoma/cirugía , Resultado del Tratamiento , Adulto Joven
12.
Oncoimmunology ; 4(11): e1044712, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26451317

RESUMEN

The expansion of myeloid-derived suppressor cells (MDSCs) is a common feature of cancer, but its biological roles and molecular mechanism remain unclear. Here, we investigated a molecular link between MDSC expansion and tumor cell metastasis in nasopharyngeal carcinoma (NPC). We demonstrated that MDSCs expanded and were positively correlated with the elevated tumor COX-2 expression and serum IL-6 levels in NPC patients. Importantly, COX-2 and MDSCs were poor predictors of patient disease-free survival (DFS). Knocking down tumor COX-2 expression hampered functional TW03-mediated-MDSC cell (T-MDSC) induction with IL-6 blocking. We identified that T-MDSCs promoted NPC cell migration and invasion by triggering the epithelial-mesenchymal transition (EMT) on cell-to-cell contact, and T-MDSCs enhanced tumor experimental lung metastasis in vivo. Interestingly, the contact between T-MDSCs and NPC cells enhanced tumor COX-2 expression, which subsequently activated the ß-catenin/TCF4 pathway, resulting in EMT of the cancer cells. Blocking transforming growth factor ß (TGFß) or inducible nitric oxide synthase (iNOS) significantly abolished the T-MDSC-induced upregulation of COX-2 and EMT scores in NPC cells, whereas the administration of TGFß or L-arginine supplements upregulated COX-2 expression and EMT scores in NPC cells. These findings reveal that COX-2 is a key factor mediating the interaction between MDSCs and tumor cells, suggesting that the inhibition of COX-2 or MDSCs has the potential to suppress NPC metastasis.

13.
Cancer Immunol Immunother ; 64(12): 1587-99, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26497849

RESUMEN

The expansion of myeloid-derived suppressor cells (MDSCs) and its correlation with advanced disease stage have been shown in solid cancers. Here, we investigated the functional features and clinical significance of MDSCs in extranodal NK/T cell lymphoma (ENKL). A higher percentage of circulating HLA-DR(-)CD33(+)CD11b(+) MDSCs was observed in ENKL patients than in healthy controls (P < 0.05, n = 32) by flow cytometry analysis. These MDSCs from ENKL patients (ENKL-MDSCs) consisted of CD14(+) monocytic (Mo-MDSCs, >60 %) and CD15(+) granulocytic (PMN-MDSCs, <20 %) MDSCs. Furthermore, these ENKL-MDSCs expressed higher levels of Arg-1, iNOS and IL-17 compared to the levels of MDSCs from healthy donors, and they expressed moderate levels of TGFß and IL-10 but lower levels of CD66b. The ENKL-MDSCs strongly suppressed the anti-CD3-induced allogeneic and autologous CD4 T cell proliferation (P < 0.05), but they only slightly suppressed CD8 T cell proliferation (P > 0.05). Interestingly, ENKL-MDSCs inhibited the secretion of IFNγ but promoted IL-10, IL-17 and TGFß secretion as well as Foxp3 expression in T cells. The administration of inhibitors of iNOS, Arg-1 and ROS significantly reversed the suppression of anti-CD3-induced T cell proliferation by MDSCs (P < 0.05). Importantly, based on multivariate Cox regression analysis, the HLA-DR(-)CD33(+)CD11b(+) cells and CD14(+) Mo-MDSCs were independent predictors for disease-free survival (DFS, P = 0.013 and 0.016) and overall survival (OS, P = 0.017 and 0.027). Overall, our results identified for the first time that ENKL-MDSCs (mainly Mo-MDSCs) have a prognostic value for patients and a suppressive function on T cell proliferation.


Asunto(s)
Linfoma Extranodal de Células NK-T/fisiopatología , Células Mieloides/inmunología , Acetilcisteína/farmacología , Adolescente , Adulto , Anciano , Arginina/análogos & derivados , Arginina/farmacología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Proliferación Celular/efectos de los fármacos , Citocinas/genética , Inhibidores Enzimáticos/farmacología , Femenino , Depuradores de Radicales Libres/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Activación de Linfocitos/inmunología , Linfoma Extranodal de Células NK-T/diagnóstico , Linfoma Extranodal de Células NK-T/inmunología , Masculino , Persona de Mediana Edad , Óxido Nítrico/antagonistas & inhibidores , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Adulto Joven , omega-N-Metilarginina/farmacología
14.
J Transl Med ; 13: 47, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25638150

RESUMEN

BACKGROUND: Expansions of myeloid-derived suppressor cells (MDSCs) have been identified in human solid tumors, including colorectal cancer (CRC). However, the nature of these tumor-associated MDSCs and their interactions with tumor cells in CRC are still poorly understood. METHODS: The percentages and phenotype of MDSCs in peripheral blood and tumorous and paraneoplastic tissues from CRC patients, as well as the clinical relevance of these MDSCs, were assessed. Age-matched healthy donors were included as controls. The interaction between MDSCs and T cells or tumor cells was investigated in a coculture system in vitro, and the molecular mechanism of the effect of MDSCs on T cells or tumor cells was evaluated. RESULTS: We discovered that CRC patients had elevated levels of CD33(+)CD11b(+)HLA-DR(-) MDSCs in primary tumor tissues and in peripheral blood, and the elevated circulating MDSCs were correlated with advanced TNM stages and lymph node metastases. Radical resection significantly decreases the proportions of circulating MDSCs and CD4(+)CD25(high)FOXP3(+) regulatory T cells. In vitro, CRC cells mediate the promotion of MDSC induction. Moreover, these tumor-induced MDSCs could suppress T cell proliferation and promote CRC cell growth via cell-to-cell contact. Such effects could be abolished by the inhibition of oxidative metabolism, including the production of nitric oxide (NO), and reactive oxygen species (ROS). CONCLUSIONS: Our results reveal the functional interdependence between MDSCs, T cells and cancer cells in CRC pathogenesis. Understanding the impact of MDSCs on T cells and tumor cells will be helpful to establish an immunotherapeutic strategy in CRC patients.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Células Mieloides/metabolismo , Antígenos CD/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Células Mieloides/inmunología , Estadificación de Neoplasias , Oxidación-Reducción , Fenotipo , Linfocitos T Reguladores/inmunología
15.
Int J Clin Exp Pathol ; 7(7): 3887-97, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25120765

RESUMEN

The expression of hypoxia-induced factor (HIF)-1α is up-regulated in tumor microenvironments under hypoxia condition. However, the prognostic significance of HIF-1α in esophageal squamous cell carcinoma (ESCC) is still elusive. We measured the HIF-1α expression by immunochemistry in tumor specimens from 136 resected ESCC; in the current study, the HIF-1α expression in tumor cells was significantly associated with tumor stage (P = 0.003) and lymph node metastasis (P = 0.006); whereas the HIF-1α expression in tumor-infiltrating lymphocytes (TILs) had no relationship with patients' clinicopathological parameters. Patients with high HIF-1α expression in tumor cells or in TILs showed worse survival related to those with low HIF-1α expression. Multivariate analysis demonstrated that expression of HIF-1α in TILs was an independent factor for DFS (P = 0.007) and OS (P = 0.013). Additionally, the expression of HIF-1α in tumor cells was an independent factor for DFS (P = 0.037) and OS (P = 0.033) in locoregional ESCC patients, whereas the expression of HIF-1α in TILs was an independent factor for DFS (P = 0.048) and OS (P = 0.039) in metastatic ESCC patients. Correlation analysis revealed that expressions of HIF-1α in tumor cells and in TILs were positively correlated, and patients with combined high HIF-1α in both tumor cells and TILs had the worst survivals (P < 0.05). These findings suggest that the HIF-1α expressions in different cell populations of ESCC microenvironments have different clinical relevance and prognostic impact on patients.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Linfocitos Infiltrantes de Tumor/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/mortalidad , Supervivencia sin Enfermedad , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/mortalidad , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Inmunohistoquímica , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Microambiente Tumoral/inmunología
16.
Oncotarget ; 5(14): 5439-52, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-24978137

RESUMEN

Tumor-derived exosomes contain biologically active proteins and messenger and microRNAs (miRNAs). These particles serve as vehicles of intercellular communication and are emerging mediators of tumorigenesis and immune escape. Here, we isolated 30-100 nm exosomes from the serum of patients with nasopharyngeal carcinoma (NPC) or the supernatant of TW03 cells. Increased circulating exosome concentrations were correlated with advanced lymphoid node stage and poor prognosis in NPC patients (P< 0.05). TW03-derived exosomes impaired T-cell function by inhibiting T-cell proliferation and Th1 and Th17 differentiation and promoting Treg induction by NPC cells in vitro. These results are associated with decreases in ERK, STAT1, and STAT3 phosphorylation and increases in STAT5 phosphorylation in exosome-stimulated T-cells. TW03-derived exosomes increased the proinflammatory cytokines IL-1ß, IL-6, and IL-10 but decreased IFNγ, IL-2, and IL-17 release from CD4+ or CD8+ T-cells. Furthermore, five commonly over-expressed miRNAs were identified in the exosomes from patient sera or NPC cells: hsa-miR-24-3p, hsa-miR-891a, hsa-miR-106a-5p, hsa-miR-20a-5p, and hsa-miR-1908. These over-expressed miRNA clusters down-regulated the MARK1 signaling pathway to alter cell proliferation and differentiation. Overall, these observations reveal the clinical relevance and prognostic value of tumor-derived exosomes and identify a unique intercellular mechanism mediated by tumor-derived exosomes to modulate T-cell function in NPC.


Asunto(s)
Exosomas/genética , MicroARNs/genética , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/inmunología , Linfocitos T/inmunología , Carcinoma , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Exosomas/inmunología , Humanos , MicroARNs/inmunología , MicroARNs/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Fosforilación , Transducción de Señal
17.
Int J Clin Exp Pathol ; 7(5): 2402-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24966950

RESUMEN

Kimura's disease (KD) is a rare chronic disease with unknown origin. It remains controversial in KD's diagnosis, treatment, transformation and need further research. The aim of this study is to investigate the clinicopathologic features of KD and the relationship between the expression of Notch-1, Ki-67 receptor and the recurrence of KD. The hematoxylin and eosin sections and clinical data of 40 patients diagnosed with KD were examined retrospectively. Specimens were available in these 40 cases. Notch-1 and Ki-67 expression were examined using IHC (immunohistochemistry staining) analysis. Of 40 cases of KD (average age, 38.4 years; median age, 36.0 years), 34 cases (85.0%) were clinically seen to involve swelling of the head and neck region. Notch-1 and Ki-67 have a high expression in recurrent patients. High expression of Notch-1 receptor and Ki-67 tended to be found in patients who relapsed. This is the first study to discuss the correlation among Notch-1, Ki-67 and recurrent KD. These results suggest both of the markers may act as promising predictors for the recurrence and prognosis of KD. However, Notch-1 immunoexpression had no statistically significant association with the Ki-67 proliferation index.


Asunto(s)
Hiperplasia Angiolinfoide con Eosinofilia/metabolismo , Antígeno Ki-67/análisis , Receptor Notch1/análisis , Tejido Subcutáneo/química , Adolescente , Adulto , Anciano , Hiperplasia Angiolinfoide con Eosinofilia/mortalidad , Hiperplasia Angiolinfoide con Eosinofilia/patología , Hiperplasia Angiolinfoide con Eosinofilia/terapia , Biomarcadores/análisis , Biopsia , Proliferación Celular , Niño , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Recurrencia , Estudios Retrospectivos , Factores de Riesgo , Tejido Subcutáneo/patología , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
18.
J Transl Med ; 11: 60, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23497377

RESUMEN

BACKGROUND: Tumor-derived cytokines and their receptors usually take important roles in the disease progression and prognosis of cancer patients. In this survey, we aimed to detect the expression levels of MIF and CXCR4 in different cell populations of tumor microenvironments and their association with survivals of patients with esophageal squamous cell carcinoma (ESCC). METHODS: MIF and CXCR4 levels were measured by immunochemistry in tumor specimens from 136 resected ESCC. Correlation analyses and independent prognostic outcomes were determined using Pearson's chi-square test and Cox regression analysis. RESULTS: The expression of CXCR4 in tumor cells was positively associated with tumor status (P = 0.045) and clinical stage (P = 0.044); whereas the expression of CXCR4 in tumor-infiltrating lymphocytes (TILs) and the expression of MIF in tumor cells and in TILs were not associated with clinical parameters of ESCC patients. High MIF expression in tumor cells or in TILs or high CXCR4 expression in tumor cells was significantly related to poor survival of ESCC patients (P < 0.05). Multivariate analysis showed that the expression of MIF or CXCR4 in tumor cells and the expression of MIF in TILs were adverse independent factors for disease-free survival (DFS) and overall survival (OS) in the whole cohort of patients (P < 0.05). Furthermore, the expression of MIF and CXCR4 in tumor cells were independent factors for reduced DFS and OS in metastatic/recurrent ESCC patients (P < 0.05). Interestingly, the expressions of MIF and CXCR4 in tumor cells and in TILs were significantly positively correlated (P < 0.05), and the combined MIF and CXCR4 expression in tumor cells was an independent adverse predictive factor for DFS and OS (P < 0.05). CONCLUSION: The expressions of MIF and CXCR4 proteins in tumor cells and TILs have different clinically predictive values in ESCC.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Neoplasias Esofágicas/diagnóstico , Regulación Neoplásica de la Expresión Génica , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Receptores CXCR4/metabolismo , Microambiente Tumoral , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Estudios de Cohortes , Supervivencia sin Enfermedad , Neoplasias Esofágicas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunoquímica , Masculino , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Análisis de Regresión , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...