Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Anticancer Res ; 41(11): 5431-5444, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34732412

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/metabolism , Cytotoxicity, Immunologic , Microsatellite Instability , Signaling Lymphocytic Activation Molecule Family/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Cell Movement , Cell Proliferation , Coculture Techniques , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HCT116 Cells , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , HT29 Cells , Humans , Immune Checkpoint Inhibitors/pharmacology , Neoplasm Invasiveness , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Signaling Lymphocytic Activation Molecule Family/genetics , THP-1 Cells , Triggering Receptor Expressed on Myeloid Cells-1/antagonists & inhibitors , Triggering Receptor Expressed on Myeloid Cells-1/genetics
2.
Anticancer Res ; 41(9): 4651-4658, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34475094

ABSTRACT

BACKGROUND/AIM: We evaluated the predictive value of candidate serum biomarkers for recurrence in stage II and III colorectal cancer (CRC) after curative surgery. PATIENTS AND METHODS: A total of 33 and 120 patients with CRC with or without recurrence at 5 years after curative surgery were included in the training set and the validation set, respectively. Possible serum biomarkers were examined for associations with CRC recurrence using receiver operating characteristics (ROC) curve analysis. RESULTS: In the training set, the expression levels of the 14 biomarkers were compared according to recurrence. Among them, five biomarkers that had significantly different expression levels were validated in 60 patients with recurrence at 5 years after curative surgery and 60 patients without. Multivariate analysis showed that natural log-transformed values of carcinoembryonic antigen (CEA), cyclin-dependent kinase regulatory subunit 2 (CKS2), 2'-5'-oligoadenylate synthetase 2 (OAS2), and autophagy-related gene 5 (ATG5) in preoperative serum were significantly related to recurrence. ROC analysis showed that these biomarkers were able to discriminate patients with recurrence from those without (area under the curve=0.828, 95% confidence interval=0.755-0.990). CONCLUSION: Preoperative serum levels of CEA, CKS2, OAS2 and ATG5 were independent risk factors for recurrence. A combination of serum CEA, CKS2, OAS2 and ATG5 predicted tumor recurrence well in patients with stage II and III CRC.


Subject(s)
Biomarkers, Tumor/blood , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Neoplasm Recurrence, Local/diagnosis , 2',5'-Oligoadenylate Synthetase/blood , Aged , Autophagy-Related Protein 5/blood , CDC2-CDC28 Kinases/blood , Carcinoembryonic Antigen/blood , Cell Cycle Proteins/blood , Colorectal Neoplasms/blood , Female , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/blood , Neoplasm Staging , ROC Curve
3.
J Cancer Res Clin Oncol ; 147(1): 117-128, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32960359

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genetic Testing/methods , Microsatellite Instability , Mutation , High-Throughput Nucleotide Sequencing/methods , Humans , Prognosis
4.
Exp Mol Med ; 51(10): 1-12, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578316

ABSTRACT

Approximately half of colorectal cancer (CRC) patients experience disease recurrence and metastasis, and these individuals frequently fail to respond to treatment due to their clinical and biological diversity. Here, we aimed to identify a prognostic signature consisting of a small gene group for precisely predicting CRC heterogeneity. We performed transcriptomic profiling using RNA-seq data generated from the primary tissue samples of 130 CRC patients. A prognostic index (PI) based on recurrence-associated genes was developed and validated in two larger independent CRC patient cohorts (n = 795). The association between the PI and prognosis of CRC patients was evaluated using Kaplan-Meier plots, log-rank tests, a Cox regression analysis and a RT-PCR analysis. Transcriptomic profiling in 130 CRC patients identified two distinct subtypes associated with systemic recurrence. Pathway enrichment and RT-PCR analyses revealed an eleven gene signature incorporated into the PI system, which was a significant prognostic indicator of CRC. Multivariate and subset analyses showed that PI was an independent risk factor (HR = 1.812, 95% CI = 1.342-2.448, P < 0.001) with predictive value to identify low-risk stage II patients who responded the worst to adjuvant chemotherapy. Finally, a comparative analysis with previously reported Consensus Molecular Subgroup (CMS), high-risk patients classified by the PI revealed a distinct molecular property similar to CMS4, associated with a poor prognosis. This novel PI predictor based on an eleven gene signature likely represents a surrogate diagnostic tool for identifying high-risk CRC patients and for predicting the worst responding patients for adjuvant chemotherapy.


Subject(s)
Colorectal Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Prognosis , Transcriptome/genetics , Aged , Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local/pathology , Risk Factors
5.
Anticancer Res ; 39(9): 4853-4864, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31519588

ABSTRACT

BACKGROUND/AIM: Colorectal cancer (CRC) is the leading cause of cancer mortality worldwide. Its poor prognosis can be ascribed primarily to high recurrence rates. Accordingly, the aim of this study was to identify novel prognostic biomarkers and therapeutic targets for management of CRC. MATERIALS AND METHODS: To develop prognostic biomarkers, we performed RNA-seq analysis and real-time RT-PCR in primary cancer tissues with or without systemic recurrence. To characterize the molecular functions of the encoded proteins, CRC cells underexpressing or overexpressing the candidate genes were established and appropriate cell-based assays were applied. RESULTS: ITGB1 and RHOC mRNA levels were up-regulated in the recurrence group of CRC patients. Overexpression of ITGB1 or RHOC stimulated CRC cell proliferation, invasion and migration, whereas the opposite effects were observed in cells underexpressing either protein. Five-year recurrence-free survival rates were significantly higher in the ITGB1- and RHOC-underexpression groups than those in the overexpression. CONCLUSION: ITGB1 and RHOC are potential predictors of recurrence and therapeutic targets for CRC, possibly predicting a high-risk group of stage II patients.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Integrin beta1/metabolism , rhoC GTP-Binding Protein/metabolism , Aged , Biomarkers, Tumor , Cell Proliferation , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , RNA, Small Interfering/genetics , Recurrence , Survival Analysis
6.
World J Gastroenterol ; 25(11): 1341-1354, 2019 Mar 21.
Article in English | MEDLINE | ID: mdl-30918427

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/mortality , Signaling Lymphocytic Activation Molecule Family/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cell Line, Tumor , Coculture Techniques , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Down-Regulation , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/immunology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Molecular Targeted Therapy/methods , Neoplasm Staging , Patient Selection , Prognosis , Progression-Free Survival , Real-Time Polymerase Chain Reaction , Retrospective Studies , Risk Assessment/methods , Risk Factors , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology , Triggering Receptor Expressed on Myeloid Cells-1/genetics , Triggering Receptor Expressed on Myeloid Cells-1/immunology , Up-Regulation
7.
PLoS One ; 13(8): e0202856, 2018.
Article in English | MEDLINE | ID: mdl-30148861

ABSTRACT

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.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Gelsolin/metabolism , Neoplasm Recurrence, Local/pathology , 2',5'-Oligoadenylate Synthetase/genetics , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gelsolin/genetics , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Prognosis , Survival Rate , Tumor Cells, Cultured
8.
Anticancer Res ; 38(1): 271-277, 2018 01.
Article in English | MEDLINE | ID: mdl-29277783

ABSTRACT

The ultraviolent irradiation resistance-associated gene (UVRAG), a component of the Beclin 1/autophagy-related 6 complex, regulates the autophagy initiation step and functions in the DNA-damage response. UVRAG is frequently mutated in various cancer types, and mutations of UVRAG increase sensitivity to chemotherapy by impairing DNA-damage repair. In this study, we addressed the epigenetic regulation of UVRAG in colorectal cancer cells. UVRAG expression was increased in cells treated with histone deacetylase (HDAC) inhibitors, such as valproic acid and suberoylanilide hydroxamic acid. Down-regulation of HDAC1 enhanced UVRAG expression in colorectal cancer cells. In addition, both chemical and genetic inhibition of HDAC1 reduced the activation of caspase-3 and cytotoxicity in 5-fluorouracil (5FU)-treated cancer cells. In contrast, UVRAG overexpression inhibited caspase activation and cell death in 5FU-treated cells. Taken together, our findings suggest that up-regulation of UVRAG by HDAC1 inhibition potentiates DNA-damage-mediated cell death in colorectal cancer cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colorectal Neoplasms/metabolism , Fluorouracil/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , Cell Death/drug effects , Colorectal Neoplasms/drug therapy , DNA Damage , Epigenesis, Genetic , HCT116 Cells , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Up-Regulation
9.
Int J Radiat Oncol Biol Phys ; 98(5): 1164-1173, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28721901

ABSTRACT

PURPOSE: The ability to predict individual responsiveness to cancer therapy is urgently needed. This is particularly true for patients with locally advanced rectal cancer (LARC) because a large proportion are resistant to preoperative chemoradiation therapy (CRT). In this study, we sought to identify markers that could predict response by comparing the gene expression profiles of the tumors of patients who received preoperative CRT. METHODS AND MATERIALS: The basal gene expression profiles of tumors from 22 LARC patients who were responders (n=9) and nonresponders (n=13) to preoperative CRT were analyzed using RNA sequencing (RNA-Seq). To validate the RNA-Seq findings, real-time reverse transcriptase polymerase chain reaction (RT-PCR) was performed on tumor samples from an additional 40 LARC patients (n=20 responders; n=20 nonresponders). Candidate genes were stably overexpressed or knocked down in colorectal cancer (CRC) cell lines, and the effect on response to radiation was tested in vitro and also in vivo in a mouse xenograft model. RESULTS: Eight differentially expressed (>16-fold) genes (B3GALT4, HSPA1B, KRBOX1, PPBP, PPP1R18, PSMB8, SLC39A7, and TAP2) associated with the preoperative CRT response were identified (P<.0005). Among these genes, real-time RT-PCR showed that PSMB8 and SLC39A7 were upregulated in the responsive group of the additional 40 LARC patients. In CRC cell lines, PSMB8 overexpression significantly reduced colony formation and increased the apoptosis-inducing molecules cleaved caspase-3 and cleaved PARP after 6-Gy irradiation. PSMB8 knockdown increased colony formation and decreased caspase-3 activation and cleaved PARP levels after irradiation. SLC39A7 overexpression had no significant effects on irradiated CSC cells. After irradiation of the xenografted mice, tumors that arose from CRC cell line HCT116 overexpressing PSMB8 grew more slowly than did those from HCT116 with vector alone. CONCLUSION: These results suggest that PSMB8 is a predictive marker of preoperative radiosensitivity in LARC patients. Clinical validation in a larger cohort is now required.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling/methods , Proteasome Endopeptidase Complex/genetics , Radiation Tolerance/genetics , Rectal Neoplasms/genetics , Rectal Neoplasms/radiotherapy , Sequence Analysis, RNA/methods , Animals , Biomarkers, Tumor/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Death , Cell Line, Tumor , Gene Knockdown Techniques , Gene Silencing , Genetic Markers , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Preoperative Care/methods , Proteasome Endopeptidase Complex/metabolism , Real-Time Polymerase Chain Reaction , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery , Tumor Stem Cell Assay , Up-Regulation
10.
Anticancer Res ; 37(3): 1297-1303, 2017 03.
Article in English | MEDLINE | ID: mdl-28314295

ABSTRACT

AIM: The present study investigated how well the results of integrative tumor-response assay (ITRA) compared to those of clinical response to chemotherapy in patients with metastatic colorectal cancer (CRC). PATIENTS AND METHODS: A total of 129 patients with metastatic CRC were prospectively enrolled. ITRA consisted of two sequential histoculture drug-response assays (HDRAs). First-stage HDRAs were performed using 5-fluorouracil with leucovorin and oxaliplatin (FX), or with irinotecan (FR). Second-stage HDRAs (ITRA) were performed for cells surviving after the first-stage HDRA, using FX, FR, and their combinations with bevacizumab and cetuximab. RESULTS: Among 129 patients, 42 (32.6%) completed second-line chemotherapy, results that correlated with those of ITRA. The accuracy of ITRA for predicting response to second-line chemotherapy was 61.9% (26/42), with a sensitivity of 44.4% (8/18) and a specificity of 75% (18/24). CONCLUSION: Despite its relatively low accuracy, ITRA might be a useful technique for predicting therapeutic efficacy and selecting for appropriate first-line and second-line anticancer regimens for patients.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Screening Assays, Antitumor/methods , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Female , Fluorouracil/administration & dosage , Humans , Irinotecan , Leucovorin/administration & dosage , Male , Middle Aged , Molecular Targeted Therapy , Neoplasm Metastasis , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Prospective Studies , Reproducibility of Results , Sensitivity and Specificity
11.
Cancer Lett ; 385: 21-27, 2017 01 28.
Article in English | MEDLINE | ID: mdl-27836735

ABSTRACT

Autophagy plays complex roles in tumor initiation and development, and the expression of autophagy-related genes (ATGs) is differentially regulated in various cancer cells, depending on their environment. In this study, we analyzed the expressional relationship between polypyrimidine tract-binding protein 1 (PTBP1) and ATG10 in metastatic colorectal cancer. PTBP1 is associated with tumor metastasis in primary colorectal tumors and colorectal cancer liver metastasis (CLM) tissues. In addition, PTPB1 directly interacts with mRNA of ATG10, and regulates ATG10 expression level in colorectal cancer cells. Ectopic expression of PTBP1 decreased ATG10 expression, whereas down-regulation of PTBP1 increased ATG10 level. In contrast to PTBP1, expression of ATG10 was decreased in CLM tissues. Knock down of ATG10 promoted cell migration and invasion of colorectal cancer cells. Moreover, depletion of ATG10 modulated epithelial-mesenchymal transition-associated proteins in colorectal cancer cells: N-cadherin, TCF-8/ZEB1, and CD44 were up-regulated, whereas E-cadherin was down-regulated. Taken together, our findings suggest that expression of ATG10 negatively regulated by PTBP1 is associated with metastasis of colorectal cancer cells.


Subject(s)
Autophagy-Related Proteins/metabolism , Cell Movement , Colorectal Neoplasms/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Vesicular Transport Proteins/metabolism , Antigens, CD/metabolism , Autophagy-Related Proteins/genetics , Cadherins/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Down-Regulation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , HCT116 Cells , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Hyaluronan Receptors/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Polypyrimidine Tract-Binding Protein/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transfection , Vesicular Transport Proteins/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
12.
Ann Surg Treat Res ; 91(4): 165-171, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27757393

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the prognostic significance of serum CEA (s-CEA) changes in colorectal cancer (CRC) patients with sustained elevated postoperative s-CEA levels. METHODS: Between January 1999 and December 2008, 9,380 CRC patients underwent surgery. Curative resection was performed in 1,242 CRC patients with high preoperative s-CEA levels (>6 ng/mL). High s-CEA levels were normalized in 924 patients (74.4%) within 2 weeks from surgery, whereas high s-CEA levels were persistent in 318 patients (25.6%). Patients were divided into 2 groups according to their postoperative s-CEA levels: group 1 (37 patients with a 1-year postoperative s-CEA>6 ng/mL) and group 2 (281 patients with a 1-year postoperative s-CEA≤6 ng/mL). RESULTS: A postoperative recurrence was identified in 24 patients (64.9%) in group 1 and 65 patients (23.1%) in group 2 (P < 0.001). A curative resection after recurrence was performed in 22 patients (33.8%) from group 2, but no patients from group 1 (P = 0.001). The 5-year overall survival and time to recurrence were significantly lower in patients with recurrent cancer in group 1 (P < 0.001). CONCLUSION: Patients with persistent elevated postoperative s-CEA levels are at high risk for recurrence and a low survival rate. More intensive surveillance of patients with high postoperative s-CEA levels should be mandatory.

13.
J Cancer Res Clin Oncol ; 142(8): 1705-14, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27177629

ABSTRACT

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.


Subject(s)
Annexins/genetics , Bevacizumab/therapeutic use , Colorectal Neoplasms/drug therapy , Neoplasm Metastasis , Polymorphism, Single Nucleotide , Protein Phosphatase 1/genetics , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Feasibility Studies , Humans , Male , Mice , Mice, Inbred BALB C
14.
PLoS One ; 11(5): e0155160, 2016.
Article in English | MEDLINE | ID: mdl-27152521

ABSTRACT

Using our data set (GSE50760) previously established by RNA sequencing, the present study aimed to identify upregulated genes associated with colorectal cancer (CRC) liver metastasis (CLM) and verify their biological behavior. The potential roles of candidate genes in tumors were assessed using cell proliferation and invasion assays. Tissue samples were collected from 18 CRC patients with synchronous CLM and two CRC cell lines (SW480 and SW620) were used for transfection and cloning. The roles of the genes identified in CLM were verified using immunohistochemistry in 48 nude mice after intrasplenic transplantation of CRC cells. mRNA and protein expression was determined by quantitative real-time reverse transcription polymerase chain reaction and western blot, respectively. Nine genes were initially selected according to the relevance of their molecular function and biological process and, finally, ALDH1A1 and IGFBP1 were chosen based on differential mRNA expression and a positive correlation with protein expression. The overexpression of ALDH1A1 and IGFBP1 significantly and time-dependently decreased cell proliferation (p ≤ 0.001-0.003) and suppressed invasiveness by ≥3-fold over control cells (p < 0.001) in the SW480 cell line, whereas they had a slight effect on reducing SW620 cell proliferation. The protein expression levels of E-cadherin, N-cadherin, claudin-1, and vimentin were significantly higher in CLM than in primary tumor tissues (p < 0.05). However, the cadherin switch, namely, N-cadherin overexpression with reduced E-cadherin expression, was not observed in CLM tissues and transfected CRC cells. Irrespective of reduced proliferation and invasion found on in vitro cell assays, persistent overexpression of ß-catenin, vimentin, and ZO-1 in IGFBP1-overexpressing SW480 cells possibly contributed to CLM development in mice implanted with IGFBP1-overexpressing SW480 cells (CLM occurrences: SW480/IGFBP1-transfected mice vs. SW480/vector- and SW480/ALDH1A1-transfected mice, 4/8 vs. 0/10, p = 0.023). In conclusion, ALDH1A1 and IGFBP1 are differentially overexpressed in CLM and may play a dual role, functioning as both tumor suppressors and metastasis promoters in CRC.


Subject(s)
Aldehyde Dehydrogenase/genetics , Colorectal Neoplasms/pathology , Insulin-Like Growth Factor Binding Protein 1/genetics , Liver Neoplasms/secondary , Aldehyde Dehydrogenase 1 Family , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Retinal Dehydrogenase
15.
Anticancer Res ; 36(5): 2397-406, 2016 May.
Article in English | MEDLINE | ID: mdl-27127149

ABSTRACT

AIM: Zinc finger with KRAB and SCAN domain 3 (ZKSCAN3) is overexpressed in invasive colorectal cancer (CRC) cells and regulates the expression of several genes favoring tumor progression, including vascular endothelial growth factor (VEGF) and integrin ß4. We evaluated the association of ZKSCAN3 and colorectal cancer liver metastasis (CLM) to determine whether it is related to invasive signaling pathways. MATERIALS AND METHODS: The ratios of expression by primary tumor to normal tissue and metastatic tumor to normal tissue were compared between ZKSCAN3-overexpressing and underexpressing primary tumor groups. RESULTS: In terms of CLM, the ZKSCAN3 overexpression was positively correlated with carcinoembryonic antigen (CEA), VEGF, and AKT expression. The protein-expression analysis showed that ZKSCAN-specific siRNA knockdown reduced CEA expression in LoVo and LS174T CRC cells. Matrigel invasion by ZKSCAN3-overexpressing HCT116 cells was increased when examined on CEA-coated filters compared with phosphate-buffered saline-treated controls. Additionally, matrix metalloproteinase 9 (MMP9) expression was greater in cells with reference allele (GG) than substitution allele (CC) for ZKSCAN3 rs733743 (p=0.032). ZKSCAN3 protein expression of the high serum CEA group was increased in hepatic metastatic tissue compared with the primary tumor tissue, while in the group with normal serum CEA it decreased or was similar. Reference ZKSCAN3 alleles were correlated with male dominance, a family history of malignancy, high serum CEA concentration and stage IV CRC in 450 patients with sporadic CRC. In conclusion, ZKSCAN3 appears to promote colorectal tumor progression and invasion. ZKSCAN3 may facilitate hepatic metastasis of CRC associated with CEA particularly in cases with CEA-producing tumor.


Subject(s)
Carcinoembryonic Antigen/blood , Colorectal Neoplasms/pathology , Liver Neoplasms/secondary , Transcription Factors/physiology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Humans , Liver Neoplasms/immunology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism
16.
Oncotarget ; 6(26): 22179-90, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26109429

ABSTRACT

To characterize the mutation profiles of colorectal cancer (CRC) primary tumors (PTs) and liver metastases (CLMs), we performed both whole-exome and RNA sequencing. Ten significantly mutated genes, including BMI1, CARD11, and NRG1, were found in 34 CRCs with CLMs. We defined three mutation classes (Class 1 to 3) based on the absence or presence of mutations during liver metastasis. Most mutations were classified into Class 1 (shared between PTs and CLMs), suggesting the common clonal origin of PTs and CLMs. Class 1 was more strongly associated with the clinical characteristics of advanced cancer and was more frequently superimposed with chromosomal deletions in CLMs than Class 2 (PT-specific). The integration of exome and RNA sequencing revealed that variant-allele frequencies (VAFs) of mutations in the transcriptome tended to have stronger functional implications than those in the exome. For instance, VAFs of the TP53 and APC mutations in the transcriptome significantly correlated with the expression level of their target genes. Additionally, mutations with high functional impact were enriched with high VAFs in the CLM transcriptomes. We identified 11 mutation-associated splicing events in the CRC transcriptomes. Thus, the integration of the exome and the transcriptome may elucidate the underlying molecular events responsible for CLMs.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Exome , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Genome-Wide Association Study/methods , Humans , Mutation , Neoplasm Metastasis , Transcriptome
17.
Mol Oncol ; 8(8): 1653-66, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25049118

ABSTRACT

Colorectal cancer (CRC) patients frequently experience disease recurrence and distant metastasis. This study aimed to identify prognostic indicators, including individual responses to chemotherapy, in CRC patients. RNA-seq data was generated using 54 samples (normal colon, primary CRC, and liver metastases) from 18 CRC patients and genes associated with CRC aggressiveness were identified. A risk score based on these genes was developed and validated in four independent CRC patient cohorts (n = 1063). Diverse statistical methods were applied to validate the risk scoring system, including a generalized linear model likelihood ratio test, Kaplan-Meier curves, a log-rank test, and the Cox model. TREM1 and CTGF were identified as two activated regulators associated with CRC aggressiveness. A risk score based on 19 genes regulated by TREM1 or CTGF activation (TCA19) was a significant prognostic indicator. In multivariate and subset analyses based on pathological staging, TCA19 was an independent risk factor (HR = 1.894, 95% CI = 1.227-2.809, P = 0.002). Subset stratification in stage III patients revealed that TCA19 had prognostic potential and identified patients who would benefit from adjuvant chemotherapy, regardless of age. The TCA19 predictor represents a novel diagnostic tool for identifying high-risk CRC patients and possibly predicting the response to adjuvant chemotherapy.


Subject(s)
Colorectal Neoplasms/pathology , Aged , Colorectal Neoplasms/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Triggering Receptor Expressed on Myeloid Cells-1
18.
Am J Clin Pathol ; 140(3): 341-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23955452

ABSTRACT

OBJECTIVES: To evaluate the association of microsatellite instability (MSI) with clinicopathologic features and oncologic outcomes in patients with poorly differentiated colorectal cancer (PD). METHODS: Study patients were divided into well-differentiated colorectal cancer (WD) and PD, which were compared according to histologic differentiation and MSI status. RESULTS: Among 1,941 patients, PD was more frequent among microsatellite-unstable tumors (23.6%) than among microsatellite-stable (MSS) tumors (4.2%, P < .001). Patients with PD had worse 4-year overall survival rates than patients with WD (78.6% vs 88.2%, P = 0.010). Compared with MSS-PD tumors, MSI-PD tumors were characterized by right-colon predilection, larger size, and infrequent lymph node metastasis (P < .001 to P = .007). CONCLUSIONS: The clinicopathologic characteristics of PD were closely associated with those of MSI. The outcomes of MSI-PD tumors were better than those of MSS-PD tumors, but this finding did not reach statistical significance.


Subject(s)
Cell Differentiation/genetics , Colorectal Neoplasms/pathology , Microsatellite Instability , Adult , Aged , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Female , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Survival Rate
19.
J Korean Surg Soc ; 83(1): 21-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22792530

ABSTRACT

PURPOSE: Identification of subgroups of patients who differ in their response to treatment could help to establish which of the best available chemotherapeutic options are best, based on biological activity. In metastatic colorectal cancer (CRC), novel molecular-targeted agents that act on pathways that regulate cell growth, the cell cycle, apoptosis, angiogenesis, and invasion are being developed. Here, we employed an in vitro chemosensitivity assay to evaluate the biological efficacy of conventional monotherapies and combination chemotherapy with targeted drugs. METHODS: The chemosensitivities of 12 CRC cell lines to the established regimens FOLFOX (5-fluorouracil [5-FU] + leucovorin + oxaliplatin) and FOLFIRI (5-FU + leucovorin + irinotecan) and to therapy with these regimens in combination with the biologically targeted drugs bevacizumab or cetuximab were comparatively evaluated for their effects on apoptotic and autophagic cell death processes, angiogenesis, and invasion. RESULTS: Each of the chemotherapeutic regimens promoted apoptotic cell death and invasion. All drug regimens caused significantly greater apoptotic cell death with activation of caspase-3 in SW480 cells compared to other cells, effects that were associated with a remarkable reduction in matrix metalloproteinase-9 activity. The FOLFOX regimen more effectively promoted apoptotic cell death, angiogenesis, and invasion than the FOLFIRI regimen. Combination therapy with FOLFOX/FOLFIRI regimen and bevacizumab produced a moderate angiogenesis-blocking effect in most cell lines. CONCLUSION: The results validate our in vitro chemosensitivity assay, and suggest that it may be applied to help determine adequate regimens in individual CRC patients based on the biological characteristics of their tumors.

20.
Anticancer Res ; 32(8): 3581-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22843949

ABSTRACT

AIM: The present study, using the histoculture drug response assay (HDRA) compared chemosensitivity with the clinical response of a treatment regime in patients with advanced colorectal cancer (CRC). PATIENTS AND METHODS: A total of 324 patients with primary CRC were prospectively enrolled. HDRAs were performed using seven combinations of anticancer drugs, including 5-fluorouracil with leucovorin (FL), FL with oxaliplatin (FOLFOX), irinotecan (FOLFIRI), and their combinations with bevacizumab and cetuximab. RESULTS: Among 324 HDRA results, tumor inhibition rates of regimes using FOLFOX (34.2-39.2%) were higher than those using FOLFIRI (24.2-32.7%, p<0.001). Out of 86 evaluated chemotherapeutic regimes, the correlation rate of HDRA to the clinical effect of chemotherapy was calculated to be 66.3% (57/86), with a 72.7% (40/55) sensitivity and a 54.7% (17/31) specificity. CONCLUSION: HDRA might be a feasible and useful technique for predicting therapy efficacy and selecting the appropriate anticancer regime for individual patients, notwithstanding its low accuracy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Aged , Female , Humans , Male , Middle Aged , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...