Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Anticancer Res ; 41(11): 5431-5444, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34732412

RESUMEN

BACKGROUND/AIM: The aim of this study was to identify the association between SLAMF7 and TREM1 and anti-PD-1 drugs, and to determine whether they are molecular targets or predictors of responses to immunotherapy through induction of immunogenic cell death. MATERIALS AND METHODS: CRC cell lines over-expressing SLAMF7 and TREM1 were used to examine immunogenic and biological traits (e.g., proliferation and invasiveness) associated with factors related to anti-cancer immunity. In addition, multiplex immunofluorescence was used to examine immune cells in microsatellite instability-high (MSI-H) CRC and microsatellite stable (MSS) CRC. RESULTS: Proliferation rate and invasiveness of TREM1-over-expressing CRC cells were significantly greater than those of control cells (p<0.001 and 0.031, respectively), whereas SLAMF7-over-expressing CRC cells showed the opposite traits (p=0.005 and 0.002, respectively). SLAMF7-over-expressing DLD-1 cells harboring MSI-H showed increased apoptosis when treated with anti-PD-1 drugs, unlike SLAMF7-over-expressing SW480 cells harboring MSS. SLAMF7-over-expressing DLD1 and SW480 cells showed a marked increase in expression of the major cytokine mediator HMGB1 when exposed to anti-PD-1 drugs. Co-administration of anti-PD-1 drugs and TREM1 inhibitors induced apoptosis only in MSI-H HCT116 cells; HMGB1 was over-expressed regardless of microsatellite status. CONCLUSION: Expression of TREM1 and SLAMF7 is closely associated with immunogenic cell death, and TREM1 inhibitors may be an effective adjuvant that enhances anti-PD-1-mediated immunogenic cell death in MSS CRC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Citotoxicidad Inmunológica , Inestabilidad de Microsatélites , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis , Movimiento Celular , Proliferación Celular , Técnicas de Cocultivo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Células HT29 , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Invasividad Neoplásica , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Células THP-1 , Receptor Activador Expresado en Células Mieloides 1/antagonistas & inhibidores , Receptor Activador Expresado en Células Mieloides 1/genética
2.
J Cancer Res Clin Oncol ; 147(1): 117-128, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32960359

RESUMEN

PURPOSE: As few genotype-phenotype correlations are available for nonsyndromic hereditary colorectal cancer (CRC), we implemented genomic analysis on the basis of the revised Bethesda guideline (RBG) and extended (12 items) to verify possible subtypes. METHODS: Patients with sporadic CRC (n = 249) were enrolled, stratified according to the revised Bethesda guidelines (RBG+ and RBG- groups) plus additional criteria. Exome/transcriptome analyses (n = 98) and cell-based functional assays were conducted. RESULTS: We detected 469 somatic and 830 germline gene mutations differing significantly between the positive and negative groups, associated with 12 RBG items/additional criteria. Twenty-one genes had significantly higher mutation rates in left, relative to right, colon cancer, while USP40, HCFC1, and HSPG2 mutation rates were higher in rectal than colon cancer. FAT4 mutation rates were lower in early-onset CRC, in contrast to increased rates in microsatellite instability (MSI)-positive tumors, potentially defining an early-onset microsatellite-stable subtype. The mutation rates of COL6A5 and MGAM2 were significantly and SETD5 was assumably, associated CRC pedigree with concurrent gastric cancer (GC). The predicted deleterious/damaging germline variants, SH2D4A rs35647122, was associated with synchronous/metachronous CRC with related tumors, while NUP160 rs381660 and KRTAP27-1 rs2244485 were potentially associated with a GC pedigree and less strictly defined hereditary CRC, respectively. SH2D4A and NUP160 acted as oncogenic facilitators. CONCLUSION: Our limited genomic analysis for RBG and additional items suggested that specific somatic alterations in the respective items may enlighten relevant pathogenesis along with the knowledge of germline mutations. Further validation is needed to indicate appropriate surveillance in suspected individuals.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Pruebas Genéticas/métodos , Inestabilidad de Microsatélites , Mutación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Pronóstico
3.
World J Gastroenterol ; 25(11): 1341-1354, 2019 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-30918427

RESUMEN

BACKGROUND: Genomic profiling of tumors has contributed to the understanding of colorectal cancer (CRC), facilitating diagnosis, prognosis and selection of treatments, including targeted regimens. A report suggested that a 19-gene-based risk classifier (TCA19) was a prognostic tool for patients with stage III CRC. The survival outcomes in patients with stage IV CRC are still poor and appropriate selection of targeted therapies and immunotherapies is challenging. AIM: To assess clinical implication of TCA19 in patients with stage IV CRC, and to identify TCA19 with involvement in immune-oncology. METHODS: A retrospective review of the medical records of 60 patients with stage IV CRC was conducted, assessing clinicopathological variables and progression-free survival (PFS). TCA19 gene expression was determined by quantitative polymerase chain reaction (qPCR) in matched normal and tumor tissues taken from the study cohort. Expression of potential immune-oncology regulatory proteins and targets was examined by immunohistochemistry (IHC), western blot, immunofluorescence staining in tissues from a validation cohort of 10 patients, and in CRC cell lines co-cultured with monocyte in vitro. RESULTS: In the patients with TCA19 score higher than the median, the PFS rates of eight patients who received the targeted regimens were significantly higher than the PFS rates of four patients who received 5-fluorouracil-based regimen (P = 0.041). In multivariate analysis, expression of signaling lymphocytic activation molecule family, member 7 (SLAMF7) and triggering receptor expressed on myeloid cells 1 (TREM1) was associated with PFS in the 60-patient cohort. After checking another 10 validate set, the expression of the IHC, the level of real-time qPCR, and the level of western blot were lower for SLAMF7 and higher for TREM7 in primary and metastatic tumors than in normal tissues. In CRC cells expressing SLAMF7 that were co-cultured with a monocytic cell line, levels of CD 68 and CD 73 were significantly lower at day 5 of co-culture than at day 0. CONCLUSION: The TCA19 score might be prognostic for target-regimen-specific PFS in stage IV CRC. Down-regulation of SLAMF7 and up-regulation of TREM1 occur in primary and metastatic tumor tissues.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/mortalidad , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Estadificación de Neoplasias , Selección de Paciente , Pronóstico , Supervivencia sin Progresión , Reacción en Cadena en Tiempo Real de la Polimerasa , Estudios Retrospectivos , Medición de Riesgo/métodos , Factores de Riesgo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Receptor Activador Expresado en Células Mieloides 1/genética , Receptor Activador Expresado en Células Mieloides 1/inmunología , Regulación hacia Arriba
4.
PLoS One ; 13(8): e0202856, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30148861

RESUMEN

The present study aimed to identify molecules associated with lymphovascular invasion (LVI) and perineural invasion (PNI) and to examine their biological behavior in colorectal cancer (CRC). LVI- and PNI-associated molecules were identified and verified using sequential processes including (1) identification of 117 recurrence-associated genes differentially expressed on RNA-seq analysis using primary cancer tissues from 130 CRC patients with and without systemic recurrence; (2) analysis of molecules associated with LVI and PNI; (3) assessment of biological properties by measuring proliferation, anoikis, invasion/migration, epithelial-mesenchymal transition and autophagy flux; and (4) verification of disease-free survival using public datasets. Gelsolin (GSN) and 2'-5'-oligoadenylate synthetase 2 (OAS2) were associated with PNI and LVI, respectively. Invasion potential was >2-fold greater in GSN-overexpressing LoVo cells than in control cells (p<0.001-0.005), whereas OAS2-overexpressing RKO cells showed reduced invasion (p<0.001-0.005). GSN downregulated E-cadherin, ß-catenin, claudin-1 and snail, and upregulated N-cadherin and ZEB1, whereas OAS2 overexpression had the opposite effects. Several autophagy-related proteins including ATG5-12, ATG6/BECN1, ATG7 and ATG101 were downregulated in GSN-overexpressing LoVo cells, whereas the opposite pattern was observed in OAS2-overexpressing RKO cells. Patients with low GSN expression had significantly higher 5-year recurrence-free survival (RFS) rates than those with GSN overexpression (73.6% vs. 64.7%, p = 0.038), whereas RFS was longer in patients with OAS2 overexpression than in those with underexpression (73.4% vs. 63.7%, p = 0.01). In conclusion, GSN and OAS2 were positively and negatively associated with recurrence, respectively, suggesting their potential value as predictors of recurrence or therapeutic targets in CRC patients.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Gelsolina/metabolismo , Recurrencia Local de Neoplasia/patología , 2',5'-Oligoadenilato Sintetasa/genética , Apoptosis , Biomarcadores de Tumor/genética , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Gelsolina/genética , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
5.
J Cancer Res Clin Oncol ; 142(8): 1705-14, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27177629

RESUMEN

PURPOSE: Bevacizumab improves survival in patients with metastatic colorectal cancer (mCRC) under chemotherapy, but few predictive markers have been identified. METHODS: To investigate chemosensitive single nucleotide polymorphisms (SNPs) of mCRC, we performed exome sequencing and RNA sequencing in 19 patients. A clinical association analysis was performed with the other 116 patients who had received chemotherapy to bevacizumab regimens. In vivo biodistribution studies and [(18)F]FDG-PET imaging were performed on mice bearing human colorectal cancer (HCT116 and SW480) xenografts after injection of bevacizumab with 5-FU, leucovorin, and irinotecan (FOLFIRI). RESULTS: PPP1R15A rs557806 showed the most significant association with FRB-driven tumor IR in exome sequencing and the highest correlation (r = 0.74) with drug responses in RNA sequencing. Patients homozygous for the reference alleles (GG) of PPP1R15A rs557806 exhibited greater disease control rate and a tendency toward greater objective response rate (ORR) than those with homozygous or heterozygous substitution alleles (GC and CC; P = 0.027 and 0.073, respectively). In xenografted mice, HCT116 clones transfected with the G allele at PPP1R15A rs557806 were more sensitive to bevacizumab regimens than those with the C allele. Tumor volume of xenografts with the G allele was significantly lower than that of xenografts with the C allele (P = 0.004, day 13). [(18)F]FDG uptake decreased to 75 % in HCT116 xenograft-bearing mice with the G allele, whereas [(18)F]FDG uptake was 42 % in mice xenografts with the C allele (P = 0.032). ANXA11 rs1049550, a predictive biomarker of SNP described in our previous study, was validated using the xenograft model. Tumor volume and [(18)F]FDG uptake analyses showed that tumors in the SW480 xenografts expressing the substitution allele (T) at ANXA11 rs1049550 were more susceptible to FOLFIRI plus bevacizumab-induced suppression than those expressing the reference allele (C) (P = 0.001 and 0.026, respectively). CONCLUSION: ANXA11 rs1049550 and PPP1R15A rs557806 may improve the identification of mCRC patients sensitive to bevacizumab regimens, and further validation is required in large cohorts.


Asunto(s)
Anexinas/genética , Bevacizumab/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Metástasis de la Neoplasia , Polimorfismo de Nucleótido Simple , Proteína Fosfatasa 1/genética , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Estudios de Factibilidad , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C
6.
PLoS One ; 7(4): e33618, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496756

RESUMEN

BACKGROUND: There is increasing evidence that phloroglucinol, a compound from Ecklonia cava, induces the apoptosis of cancer cells, eventually suppressing tumor angiogenesis. METHODOLOGY/PRINCIPAL FINDINGS: This is the first report on phloroglucinol's ability to potentially inhibit the functional bioactivities of endothelial progenitor cells (EPCs) and thereby attenuate tumor growth and angiogenesis in the Lewis lung carcinoma (LLC)-tumor-bearing mouse model. Although Phloroglucinol did not affect their cell toxicity, it specifically inhibited vascular endothelial growth factor (VEGF) dependent migration and capillary-like tube formation of EPCs. Our matrigel plug assay clearly indicated that orally injected phloroglucinol effectively disrupts VEGF-induced neovessel formation. Moreover, we demonstrated that when phloroglucinol is orally administered, it significantly inhibits tumor growth and angiogenesis as well as CD45(-)/CD34(+) progenitor mobilization into peripheral blood in vivo in the LLC-tumor-bearing mouse model. CONCLUSIONS/SIGNIFICANCE: These results suggest a novel role for Phloroglucinol: Phloroglucinol might be a modulator of circulating EPC bioactivities, eventually suppressing tumorigenesis. Therefore, phloroglucinol might be a candidate compound for biosafe drugs that target tumor angiogenesis.


Asunto(s)
Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/prevención & control , Endotelio Vascular/efectos de los fármacos , Indicadores y Reactivos/administración & dosificación , Neovascularización Patológica/prevención & control , Floroglucinol/administración & dosificación , Células Madre/efectos de los fármacos , Administración Oral , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Carcinoma Pulmonar de Lewis/patología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Combinación de Medicamentos , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Laminina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteoglicanos/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Cancer Biol Ther ; 13(7): 504-15, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22406998

RESUMEN

Accumulating evidence suggests that Ginsenoside Rg3 appears to inhibit tumor growth including Lewis lung carcinoma, intestinal adenocarcinomas or B16 melanoma by inhibiting cell proliferation, tumor cell invasion and metastasis. Endothelial progenitor cells (EPCs) appear to play a key role in the growth of early tumors by intervening with the angiogenic switch promoting tumor neovessel formation by producing angiogenic cytokines during tumor progression. This paper reports a novel mechanism of Ginsenoside Rg3, a candidate anticancer bio-molecule, on tumor angiogenesis by inhibiting the multiple bioactivities of EPCs. When Ginsenoside Rg3 was applied to the ex vivo cultured outgrowth ECs, a type of EPCs, it inhibited the cell proliferation, cell migration and tubular formation of EPCs. Importantly, Ginsenoside Rg3 attenuated the phosphorylation cascade of the VEGF dependent p38/ERK signaling in vitro. The xenograft tumor model clearly showed that Ginsenoside Rg3 suppresses tumor growth and tumor angiogenesis by inhibiting the mobilization of EPCs from the bone marrow microenvironment to the peripheral circulation and modulates VEGF-dependent tumor angiogenesis. In conclusion, this study provides a potential therapeutic molecule, Ginsenoside Rg3, as an anticancer drug by inhibiting the EPC bioactivities.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Células Endoteliales/efectos de los fármacos , Ginsenósidos/farmacología , Neovascularización Patológica , Células Madre/efectos de los fármacos , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/metabolismo , Expresión Génica/efectos de los fármacos , Ginsenósidos/administración & dosificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Células Madre/metabolismo , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Phytother Res ; 26(9): 1286-93, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22253055

RESUMEN

Endothelial progenitor cells (EPCs) play a critical role both in vascular repair after cell transplantation for ischemic diseases and in the growth of early tumors by intervening with the angiogenic switch during tumor progression. This paper reports on the effect of ginsenoside Rg3 in EPCs as a candidate angiogenesis inhibitor for in vitro functional assays. CD34⁺ cells were isolated from human cord blood and the study investigated whether or not ginsenoside Rg3 regulated EPC bioactivities including cell proliferation, differentiation, migration and tube formation. Although ginsenoside Rg3 did not affect the ex vivo expansion of CD34 and/or KDR (VEGFR2) stem/progenitor cells, treatment with ginsenoside Rg3 led to a significant decrease in CD34-expressing cells, specifically the absolute number of expanded CD34⁺ cells. Importantly, a significantly decreased number of EPC colony-forming units among human cord blood-derived CD34⁺ cells was observed, implying that ginsenoside Rg3 inhibited EPC differentiation, in particular, the commitment to primitive EPC colonies (the early stage of EPC differentiation). Moreover, treatment of CD34-derived EPCs with ginsenoside Rg3 resulted in the attenuation of VEGF-dependent Akt/eNOS signaling as well as the inhibition of migration and tube formation. In conclusion, this study provides in vitro evidence for ginsenoside Rg3 as a potential therapeutic molecule, specifically as an angiogenesis inhibitor that functions by attenuating EPC bioactivities.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Ginsenósidos/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Madre/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Antígenos CD34 , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Sangre Fetal/citología , Humanos , Transducción de Señal , Células Madre/efectos de los fármacos
9.
Biomol Ther (Seoul) ; 20(2): 158-64, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24116289

RESUMEN

Inhibiting the bioactivities of circulating endothelial progenitor cells (EPCs) results in significant inhibition of neovessel formation during tumor angiogenesis. To investigate the potential effect of phloroglucinol as an EPC inhibitor, we performed several in vitro functional assays using CD34(+) cells isolated from human umbilical cord blood (HUCB). Although a high treatment dose of phloroglucinol did not show any cell toxicity, it specifically induced the cell death of EPCs under serum free conditions through apoptosis. In the EPC colony-forming assay (EPC-CFA), we observed a significant decreased in the small EPC-CFUs for the phloroglucinol group, implying that phloroglucinol inhibited the early stage of EPC commitment. In addition, in the in vitro expansion assay using CD34(+) cells, treatment with phloroglucinol was shown to inhibit endothelial lineage commitment, as demonstrated by the decrease in endothelial surface markers of EPCs including CD34(+), CD34(+)/CD133(+), CD34(+)/CD31(+) and CD34(+)/CXCR4(+). This is the first report to demonstrate that phloroglucinol can inhibit the functional bioactivities of EPCs, indicating that phloroglucinol may be used as an EPC inhibitor in the development of biosafe anti-tumor drugs that target tumor angiogenesis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA