Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Sci Rep ; 10(1): 6600, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313120

ABSTRACT

Understanding cancer-prone environments is important to efficiently detect and prevent cancers. The associations between miRNA and cancer-prone environments are still largely unknown in gastric cancer (GC). Six miRNAs that are differentially expressed during gastric carcinogenesis were selected, and quantitative real-time PCR was performed in an independent training set (fresh non-tumor and tumor samples from 18 GC patients) and validation sets (set 1 with formalin-fixed paraffin-embedded non-tumor and tumor samples from 19 solitary GC and set 2 with 37 multiple GC patients). The results were compared with those of 37 gastric mucosa from 20 healthy volunteers. The expression levels of miR-26a, miR-375, and miR-1260 in gastric mucosa from healthy volunteers were statistically higher than that of non-tumorous gastric mucosa located 3 cm apart from the GC in the training set (miR-26a, P < 0.0001; miR-375, P = 0.0049; miR-1260, P = 0.0172), validation set 1 (miR-26a and miR-375, P < 0.0001; miR-1260, P = 0.0008), and validation set 2 (miR-26a, miR-375, and miR-1260, P < 0.0001). And a combination of miR-26a and miR-1260 showed the highest area under the curve value of 0.89. miRNAs are differentially expressed in non-neoplastic gastric mucosa and can be used as a biomarker to predict cancer-prone environments.


Subject(s)
Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Stomach Neoplasms/genetics , Aged , Case-Control Studies , Female , Humans , Male , MicroRNAs/metabolism , ROC Curve
2.
Pathol Res Pract ; 216(5): 152941, 2020 May.
Article in English | MEDLINE | ID: mdl-32241597

ABSTRACT

CDH1 mutation is the most frequent genetic alteration in hereditary diffuse gastric cancer (GC) and early onset diffuse GC patients. However, the incidence of CDH1 mutations in sporadic GC with or without family history has not been studied. This retrospective study includes a total of 993 Korean patients with primary advanced GC who underwent surgery and received palliative chemotherapy. Targeted deep sequencing was performed in all cases and family history of GC was searched with survival analysis. We found CDH1 alterations in 146 of 993 patients (14.7 %) and 8 were germline (0.8 %). Out of 146 patients with CDH1 mutations, 25 (17.1 %) had a family history of GC in one of their first relatives, and 12 patients (8.2 %) were diagnosed with familial GC (FGC). All cases with FGC were diffuse type by Lauren classification, and only one harbored a previously reported germline mutation of CDH1 (c.2638 G > A) and the remaining 11 harbored known somatic CDH1 mutations. Among all patients with CDH1 mutation, there was no significant survival difference between patients with family history or FGC. In the 847 patients without CDH1 mutation, 189 (22.3 %) had a family history of GC and 92 patients (10.9 %) were FGC. CDH1 mutations were more frequent in patients with early onset (<45 years) GC (45.5 %) compared with patients with late onset GC (10.9 %) (p = 0.001), but were not significantly associated with the family history of GC (p > 0.05). CDH1 mutations are mostly somatic and typically are not associated with family history.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Genetic Predisposition to Disease/genetics , Stomach Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Mutation , Retrospective Studies , Young Adult
3.
Virchows Arch ; 476(2): 243-250, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31463730

ABSTRACT

Programmed death ligand-1 (PD-L1) immunohistochemistry is used to predict response to anti-PD-L1 therapy. However, intra- and inter-observer variability influence the prediction of PD-L1 expression. Unlike evaluations of non-small cell lung cancer according to PD-L1 expression determined by tumor proportion score, gastric cancer is evaluated using combined positive scores. We aimed to compare the results of digital image analysis and pathologists' interpretations for predicting response to pembrolizumab in gastric cancer patients. Gastric cancer tissues from patients enrolled in a clinical trial of pembrolizumab were reviewed, and 39 cases were analyzed. PD-L1 22C3 PharmDx (Dako) immunohistochemistry slides were interpreted by digital image analysis and by the consensus of two pathologists, and the results of interpretations were compared with the eventual clinical responses to pembrolizumab. In direct comparisons of digital image analyses and pathologists' interpretations, 33 (84.6%) out of 39 cases showed concordant results. In 6 (15.4%) cases, weak staining of PD-L1, anthracotic pigment deposits, or decalcification artifacts caused discordant results, and all the cases were challenging. In statistical analyses, PD-L1 expression as interpreted by pathologists and digital image analysis did not differ significantly for predicting responses to pembrolizumab (P = 0.1856). Digital image analyses and pathologists' interpretations showed concordant results for PD-L1 combined positive scores in most cases, and the prediction performances of each were not significantly different.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , B7-H1 Antigen/drug effects , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Adult , Aged , B7-H1 Antigen/analysis , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/metabolism , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Middle Aged , Observer Variation , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
4.
Mol Biol Rep ; 46(6): 5859-5866, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31452046

ABSTRACT

The statins are a group of therapeutic drugs widely used for lowering plasma cholesterol level, while it has also been reported to induce cell death in human acute myeloid leukemia (AML) cells. To determine antitumor activity triggered by simvastatin, four AML cell lines-U937, KG1, THP1 (NRASG12D mutant) and HL60 (NRASQ61L mutant)-were cultured with simvastatin and cell viability was assessed using the CellTiter-Glo reagent. For understanding mechanism of antitumor activity, immunoblot analysis for pAkt (Ser473), Akt, pMEK, MEK, pERK (Thr202/Tyr204) and ERK (Thr202/Tyr204) was performed. Apoptotic cell population was calculated using the Annexin V-FITC assay, and cell cycle state was assessed by flow cytometry. Simvastatin showed different cytotoxic effect among AML cells, of which NRASG12D mutant THP1 was the most statin sensitive cell line (IC50 values: 1.96 uM in HL60, 7.87 uM in KG1, 0.83 uM in THP1 and 1.37 uM in U937). Western blot analysis revealed that Ras downstream signaling molecules including Akt, MEK, and ERK1/2 were markedly inhibited in THP1 cells compared to other AML cells when exposed to simvastatin. In addition, only in THP1 cells, increased apoptosis and cell cycle arrest by simvastatin was observed. The combination of simvastatin and MEK inhibitor AZD6244 synergistically reduced THP1 cell proliferation compared to simvastatin alone and AZD6244 alone (IC50 values: 0.88 uM in simvastatin, 0.32 uM in AZD6244, and 0.23 uM in combination of simvastatin and AZD6244). Simvastatin exhibited anti-leukemic effect in human AML cells in vitro, especially at NRASG12D mutant AML cell line.


Subject(s)
Antineoplastic Agents/pharmacology , GTP Phosphohydrolases/genetics , Leukemia, Myeloid, Acute , Membrane Proteins/genetics , Simvastatin/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , GTP Phosphohydrolases/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Membrane Proteins/metabolism , Mutation/genetics
5.
Sci Rep ; 8(1): 14393, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30258124

ABSTRACT

Intestinal-type gastric carcinoma exhibits a multistep carcinogenic sequence from adenoma to carcinoma with a gradual increase in genomic alterations. But the roles of microRNAs (miRNA) in this multistage cascade are not fully explored. To identify differentially expressed miRNA (DEM) during early gastric carcinogenesis, we performed miRNA microarray profiling with 24 gastric cancers and precursor lesions (7 early gastric cancer [EGC], 3 adenomas with high-grade dysplasia, 4 adenomas with low-grade dysplasia, and 10 adjacent normal tissues). Alterations in the expression of 132 miRNA were detected; these were categorized into three groups based on their expression patterns. Of these, 42 miRNAs were aberrantly expressed in EGC. Five miRNA (miR-26a, miR-375, miR-574-3p, miR-145, and miR-15b) showed decreased expression since adenoma. Expression of two miRNA, miR-200C and miR-29a, was down-regulated in EGCs compared to normal mucosa or adenomas. Six miRNA (miR-601, miR-107, miR-18a, miR-370, miR-300, and miR-96) showed increased expression in gastric cancer compared to normal or adenoma samples. Five representative miRNAs were further validated with RT-qPCR in independent 77 samples. Taken together, these results suggest that the dysregulated miRNA show alterations at the early stages of gastric tumorigenesis and may be used as a candidate biomarker.


Subject(s)
Adenoma/genetics , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Stomach Neoplasms/genetics , Transcriptome , Adenoma/pathology , Aged , Carcinogenesis/pathology , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Stomach/pathology , Stomach Neoplasms/pathology
6.
Oncotarget ; 8(57): 96656-96667, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29228560

ABSTRACT

INTRODUCTION: The role of MerTK has not been assessed in gastric cancer (GC). The aim of this study was to identify a subgroup of GC patients with MerTK tumor overexpression, and to evaluate MerTK as a potential therapeutic target in this disease. METHODS: Protein and mRNA expression of MerTK were evaluated, and other various in vitro analyses including shRNA transfection, cell cycle anslysis, MTS assay and colony forming assay were carried out with GC cell lines and GC patient-derived cells (PDCs). RESULTS: shRNA-mediated knockdown of MerTK resulted in inhibition of cell growth, as well as increased cellular apoptosis in MerTK positive GC cells. Out of 192 GC patients, 16 (8.3%) patients showed strong protein expression and they had a significantly shorter overall survival compared to those with no MerTK expression. In 54 cases of GC PDCs, 4 cases (7.4%) showed mRNA overexpression, which was comparable to the protein expression rate. When we administered UNC1062, a novel MerTK-selective small molecular tyrosine kinase inhibitor, proliferation of MerTK overexpressing GC cells and PDCs were considerably inhibited. CONCLUSION: MerTK may be involved in GC carcinogenesis, and it could be a potential novel therapeutic target in GC patients.

7.
Transl Oncol ; 10(4): 469-475, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28501555

ABSTRACT

BACKGROUND: FGFR2 amplification is associated with aggressive gastric cancer (GC), and targeted drugs have been developed for treatment of GC. We evaluated the antitumor activity of an FGFR inhibitor in FGFR2-amplified GC patients with peritoneal carcinomatosis. METHODS: Two GC patients with FGFR2 amplification confirmed by fluorescence in situ hybridization showed peritoneal seeding and malignant ascites. We used the patient-derived xenograft model; patient-derived cells (PDCs) from malignant ascites were used to assess FGFR2 expression and its downstream pathway using immunofluorescence analysis and immunoblot assay in vitro. Apoptosis and cell cycle arrest after treatment of FGFR inhibitor were analyzed by Annexin V-FITC assay and cell cycle analysis. RESULTS: FGFR2 amplification was verified in both PDC lines. AZD4547 as an FGFR inhibitor decreased proliferation of PDCs, and the IC50 value was estimated to be 250 nM in PDC#1 and 210 nM in PDC#2. FGFR inhibitor also significantly decreased levels of phosphorylated FGFR2 and downstream signaling molecules in FGFR2-amplified PDC lines. In cell cycle analysis, apoptosis was significantly increased in AZD4547-treated cells compared with nontreated cells. The proportion of cells in the sub-G1 stage was significantly higher in AZD4547-treated PDCs than in control cells. CONCLUSION: Our findings suggest that FGFR2 amplification is a relevant therapeutic target in GC with peritoneal carcinomatosis.

8.
Transl Oncol ; 10(3): 304-310, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28325666

ABSTRACT

PURPOSE: Although targeting angiogenesis with tyrosine kinase inhibitors (TKIs) has become standard of care in the treatment of clear cell renal cell carcinoma (RCC), resistance mechanism are not fully understood, and there is a need to develop new therapeutic options overcoming them. METHODS AND MATERIALS: To develop a preclinical model that predicts clinical activity of novel agents in 19 RCC patients, we established patient-derived cell (PDC) and xenograft (PDX) models derived from malignant effusions or surgical specimen. RESULTS: Successful PDCs, defined as cells that maintained growth following two passages, were established in 5 of 15 malignant effusions and 1 of 4 surgical specimens. One PDC, clinically refractory to TKIs, was implanted and engrafted in mice, resulting in a comparable histology to the primary tumor. The PDC-PDX model also showed similar genomic features when tested using targeted sequencing of cancer-related genes. When we examined the drug effects of the PDX model, the tumor cells showed resistance to TKIs and everolimus in vitro. CONCLUSION: The results suggest that the PDC-PDX preclinical model we developed using malignant effusions can be a useful preclinical model to interrogate sensitivity to targeted agents based on genomic alterations.

9.
Oncotarget ; 8(9): 15014-15022, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28122360

ABSTRACT

BACKGROUND: Fibroblast growth factor 2 (FGFR2) amplification, occurring in ~2-9% of gastric cancers (GC), is associated with poor overall survival. RESULTS: RNA sequencing identified a novel FGFR2-ACSL5 fusion in the resistant tumor that was absent from the matched pre-treatment tumor. The FGFR2-amplified PDC line was sensitive to FGFR inhibitors whereas the PDC line with concomitant FGFR2 amplification and FGFR2-ACSL5 fusion exhibited resistance. Additionally, the FGFR2-amplified GC PDC line, which was initially sensitive to FGFR2 inhibitors, subsequently also developed resistance. MATERIALS AND METHODS: We identified an FGFR2-amplified patient with GC, who demonstrated a dramatic and long-term response to LY2874455, a pan-FGFR inhibitor, but eventually developed an acquired LY2874455 resistance. Following resistance development, an endoscopic biopsy was performed for transcriptome sequencing and patient-derived tumor cell line (PDC) establishment to elucidate the underlying molecular alterations. CONCLUSIONS: FGFR inhibitors may function against FGFR2-amplified GC, and a novel FGFR2-ACSL5 fusion identified by transcriptomic characterization may underlie clinically acquired resistance. IMPLICATIONS FOR PRACTICE: Poor treatment response represents a substantial concern in patients with gastric cancer carrying multiple FGFR2 gene copies. Here, we show the utility of a general FGFR inhibitor for initial response prior to treatment resistance and report the first characterization of a potential resistance mechanism involving an FGFR2-ACSL5 fusion protein.


Subject(s)
Coenzyme A Ligases/genetics , Drug Resistance, Neoplasm/genetics , Gene Amplification , Indazoles/pharmacology , Oncogene Proteins, Fusion/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stomach Neoplasms/genetics , Adult , Female , Humans , Prognosis , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology
10.
Transl Oncol ; 9(3): 173-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27267833

ABSTRACT

BACKGROUND: Biliary tract cancers (BTCs) are rare and heterogeneous group of tumors classified anatomically into intrahepatic and extrahepatic bile ducts and gallbladder adenocarcinomas. Patient-derived tumor cell (PDC) models with genome analysis can be a valuable platform to develop a method to overcome the clinical barrier on BTCs. MATERIAL AND METHODS: Between January 2012 and June 2015, 40 BTC patients' samples were collected. PDCs were isolated and cultured from surgical specimens, biopsy tissues, or malignant effusions including ascites and pleural fluid. Genome analysis using targeted panel sequencing as well as digital multiplexed gene analysis was applied to PDCs as well as primary tumors. RESULTS: Extrahepatic cholangiocarcinoma (N=15, 37.5%), intrahepatic cholangiocarcinoma (N=10, 25.0%), gallbladder cancer (N=14, 35.0%), and ampulla of Vater cancer (N=1, 2.5%) were included. We identified 15 mutations with diverse genetic alterations in 19 cases of BTC from primary tumor specimens. The most common molecular alterations were in TP53 (8/19, 42.1%), including missense mutations such as C242Y, E285K, G112S, P19T, R148T, R248Q, and R273L. We also detected two NRAS mutations (G12C and Q61L), two KRAS mutations (G12A and G12S), two ERBB2 mutations (V777L and pM774delinsMA) and amplification, and three PIK3CA mutations (N345K, E545K, and E521K). PDC models were successfully established in 27 of 40 samples (67.5%), including 22/24 from body fluids (91.7%) and 5/16 from tissue specimens (31.3%). CONCLUSIONS: PDC models are promising tools for uncovering driver mutations and identifying rational therapeutic strategies in BTC. Application of this model is expected to inform clinical trials of drugs for molecular-based targeted therapy.

11.
Transl Oncol ; 9(3): 197-202, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27267837

ABSTRACT

BACKGROUND: Although pazopanib treatment has become the standard chemotherapy in salvage setting for metastatic sarcoma patients, most patients progress after pazopanib treatment in 4 to 6 months. After failure to pazopanib, patients have limited options for treatment. Therefore, subsequent therapy in patients who failed to pazopanib is urgently needed and the use of patient derived cells or patient derived tumors for accompanying testing with various pharmacological inhibitors could offer additional treatment options for these patients. METHODS: Patient derived tumor cells were collected from ascites at the time of progression to pazopanib and a 13-drug panel was tested for drug sensitivity. We confirmed the results using in vitro cell viability assay and immunoblot assay. We also performed the genomic profiling of PDX model. RESULTS: The growth of patient derived tumor cells was significantly reduced by exposure to 1.0 µM AZD2014 compared with control (control versus AZD2014, mean growth = 100.0% vs 16.04%, difference = 83.96%, 95% CI = 70.01% to 97.92%, P = .0435). Similarly, 1.0 µM BEZ235 profoundly inhibited tumor cell growth in vitro when compared to control (control versus BEZ235, mean growth = 100.0% vs 7.308%, difference = 92.69%, 95% CI = 78.87% to 106.5%, P < .0001). Despite the presence of CDK4 amplification in the patient-derived tumor cells, LEE011 did not considerably inhibit cell proliferation when compared with control (control vs LEE011, mean growth = 100.0% vs 80.23%, difference = 19.77%, 95% CI = 1.828% to 37.72%, P = .0377). The immunoblot analysis showed that BEZ235 treatment decreased pAKT, pmTOR and pERK whereas AZD2014 decreased only pmTOR. CONCLUSION: Taken together, upregulation of mTOR/AKT pathway in sarcoma patient derived cells was considerably inhibited by the treatment of AZD2014 and BEZ235 with downregulation of AKT pathway (greater extent for BEZ235). These molecules may be considered as treatment option in STS patient who have failed to pazopanib in the context of clinical trials.

12.
Oncotarget ; 7(15): 19610-9, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26909603

ABSTRACT

BACKGROUND: We aimed to establish a prospectively enrolled colorectal cancer (CRC) cohort for targeted sequencing of primary tumors from CRC patients. In parallel, we established collateral PDC models from the matched primary tumor tissues, which may be later used as preclinical models for genome-directed targeted therapy experiments. RESULTS: In all, we identified 27 SNVs in the 6 genes such as PIK3CA (N = 16), BRAF (N = 6), NRAS (N = 2), and CTNNB1 (N = 1), PTEN (N = 1), and ERBB2 (N = 1). RET-NCOA4 translocation was observed in one out of 105 patients (0.9%). PDC models were successfully established from 62 (55.4%) of the 112 samples. To confirm the genomic features of various tumor cells, we compared variant allele frequency results of the primary tumor and progeny PDCs. The Pearson correlation coefficient between the variants from primary tumor cells and PDCs was 0.881. METHODS: Between April 2014 and June 2015, 112 patients with CRC who underwent resection of the primary tumor were enrolled in the SMC Oncology Biomarker study. The PDC culture protocol was performed for all eligible patients. All of the primary tumors from the 112 patients who provided written informed consent were genomically sequenced with targeted sequencing. In parallel, PDC establishment was attempted for all sequenced tumors. CONCLUSIONS: We have prospectively sequenced a CRC cohort of 105 patients and successfully established 62 PDC in parallel. Each genomically characterized PDCs can be used as a preclinical model especially in rare genomic alteration event.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Mutation , Adult , Aged , Aged, 80 and over , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Female , GTP Phosphohydrolases/genetics , Gene Frequency , Humans , Kaplan-Meier Estimate , Male , Membrane Proteins/genetics , Middle Aged , PTEN Phosphohydrolase/genetics , Polymorphism, Single Nucleotide , Prospective Studies , Proto-Oncogene Proteins B-raf/genetics , Receptor, ErbB-2/genetics , Sequence Analysis, DNA/methods , Tumor Cells, Cultured , beta Catenin/genetics
13.
Oncotarget ; 6(36): 39028-35, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26472021

ABSTRACT

BACKGROUND: We have investigated the incidence of NTRK1 rearrangements in metastatic gastrointestinal cancer patients and demonstrated the potential for clinical response of these patients to targeted therapy. METHODS: We prospectively collected tumor tissue specimens for one year and simultaneously generated patient-derived tumor cells (PDCs). Specimens were initially screened for TrkA protein expression using TrkA immunohistochemistry (IHC). In the case of TrkA IHC positive results, samples were further examined by fluorescence in situ hybridization (FISH) and next generation sequencing (NGS) to confirm the presence of NTRK1 rearrangements. RESULTS: From January 2014 to December 2014, a total of 74 metastatic colorectal cancer (CRC) patients and 66 gastric cancer (GC) patients were initially screened by TrkA IHC. Two of the 74 CRC patients (2.7%) and one of the 66 GC patients (1.5%) were positive for TrkA expression by IHC. All three IHC positive cases had evidence of NTRK1 rearrangements by FISH. NGS was performed on the 3 IHC positive cases and confirmed TPM3-NTRK1 rearrangements in the two CRC cases. One GC patient with TrkA expression by IHC did not harbor an NTRK1 rearrangement. PDCs established from the NTRK1 positive CRC patients were positive for the NTRK1 rearrangement. Entrectinib, a pan-TRK inhibitor, profoundly inhibited cell proliferation of NTRK1-rearranged PDCs with such inhibition associated with inactivation of TrkA, and down-regulation of downstream signaling pathways. CONCLUSIONS: TrkA IHC is an effective, initial screening method for NTRK1 rearrangement detection in the clinic. Inhibition of the TrkA kinase is a promising targeted therapy for cancer patients whose tumors harbor a NTRK1 rearrangement.


Subject(s)
Colorectal Neoplasms/genetics , Receptor, trkA/genetics , Adult , Aged , Aged, 80 and over , Benzamides/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Female , Gene Rearrangement , Humans , Immunohistochemistry , Indazoles/pharmacology , Male , Middle Aged , Molecular Targeted Therapy , Prospective Studies , Receptor, trkA/antagonists & inhibitors , Tumor Cells, Cultured
14.
Oncotarget ; 6(29): 28211-22, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26375439

ABSTRACT

Recently, MET exon 14 deletion (METex14del) has been postulated to be one potential mechanism for MET protein overexpression. We screened for the presence of METex14del transcript by multiplexed fusion transcript analysis using nCounter assay followed by confirmation with quantitative reverse transcription PCR with correlation to MET protein expression by immunohistochemistry (IHC) and MET amplification by fluorescence in situ hybridization (FISH). We extracted RNAs from 230 patients enrolled onto the prospective molecular profiling clinical trial (NEXT-1) (NCT02141152) between November 2013 and August 2014. Thirteen METex14del cases were identified including 3 gastric cancer, 4 colon cancer, 5 non-small cell lung cancer, and one adenocarcinoma of unknown primary. Of these 13 METex14del cases, 11 were MET IHC 3+ and 2 were 2+. Only one out of the 13 METex14del cases was MET amplified (MET/CEP ratio > 2.0). Growths of two (gastric, colon) METex14del+ patient tumor derived cell lines were profoundly inhibited by both MET tyrosine kinase inhibitors and a monoclonal antibody targeting MET. In conclusion, METex14del is a unique molecular aberration present in gastrointestinal (GI) malignancies corresponding with overexpression of MET protein but rarely with MET amplification. Substantial growth inhibition of METex14del+ patient tumor derived cell lines by several MET targeting drugs strongly suggests METex14del is a potential actionable driver mutation in GI malignancies.


Subject(s)
Exons/genetics , Gastrointestinal Neoplasms/genetics , Proto-Oncogene Proteins c-met/genetics , Sequence Deletion , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Anilides/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Mutational Analysis/methods , Female , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Molecular Sequence Data , Prospective Studies , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/immunology , Proto-Oncogene Proteins c-met/metabolism , Pyridines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Oncotarget ; 6(28): 25619-30, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26296973

ABSTRACT

BACKGROUND: In this study, we established patient-derived tumor cell (PDC) models using tissues collected from patients with metastatic cancer and assessed whether these models could be used as a tool for genome-based cancer treatment. METHODS: PDCs were isolated and cultured from malignant effusions including ascites and pleural fluid. Pathological examination, immunohistochemical analysis, and genomic profiling were performed to compare the histological and genomic features of primary tumors, PDCs. An exploratory gene expression profiling assay was performed to further characterize PDCs. RESULTS: From January 2012 to May 2013, 176 samples from patients with metastatic cancer were collected. PDC models were successfully established in 130 (73.6%) samples. The median time from specimen collection to passage 1 (P1) was 3 weeks (range, 0.5-4 weeks), while that from P1 to P2 was 2.5 weeks (range, 0.5-5 weeks). Sixteen paired samples of genomic alterations were highly concordant between each primary tumor and progeny PDCs, with an average variant allele frequency (VAF) correlation of 0.878. We compared genomic profiles of the primary tumor (P0), P1 cells, P2 cells, and patient-derived xenografts (PDXs) derived from P2 cells and found that three samples (P0, P1, and P2 cells) were highly correlated (0.99-1.00). Moreover, PDXs showed more than 100 variants, with correlations of only 0.6-0.8 for the other samples. Drug responses of PDCs were reflective of the clinical response to targeted agents in selected patient PDC lines. CONCLUSION(S): Our results provided evidence that our PDC model was a promising model for preclinical experiments and closely resembled the patient tumor genome and clinical response.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Gene Expression Profiling , Genome, Human , Neoplasms/drug therapy , Neoplasms/genetics , Precision Medicine , Adult , Aged , Aged, 80 and over , Animals , Ascitic Fluid/pathology , Female , Gene Expression Profiling/methods , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Mice, Nude , Middle Aged , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasms/pathology , Patient Selection , Phenotype , Pleural Effusion, Malignant/pathology , Predictive Value of Tests , Primary Cell Culture , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult
16.
Oncotarget ; 6(27): 24320-32, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26172300

ABSTRACT

PURPOSE: Anaplastic lymphoma kinase (ALK) rearrangement has been detected in colorectal carcinoma (CRC) using advanced molecular diagnostics tests including exon scanning, fluorescence in situ hybridization (FISH), and next generation sequencing (NGS). We investigated if immunohistochemistry (IHC) can be used to detect ALK rearrangement in gastrointestinal malignancies. EXPERIMENTAL DESIGNS: Tissue microarrays (TMAs) from consecutive gastric carcinoma (GC) and CRC patients who underwent surgical resection at Samsung Medical Center, Seoul, Korea were screened by IHC using ALK monoclonal antibody 5A4. IHC positive cases were confirmed by FISH, nCounter assays, and NGS-based comprehensive genomic profiling (CGP). ALK IHC was further applied to CRC patients enrolled in a pathway-directed therapeutic trial. RESULTS: Four hundred thirty-two GC and 172 CRC cases were screened by IHC. No GC sample was ALK IHC positive. One CRC (0.6%) was ALK IHC positive (3+) that was confirmed by ALK FISH and a novel CAD-ALK (C35; A20) fusion variant that resulted from a paracentric inversion event inv(2)(p22-21p23) was identified by CGP. One out of 50 CRC patients enrolled in a pathway-directed therapeutic trial was ALK IHC positive (3+) confirmed by ALK FISH and found to harbor the EML4-ALK (E21, A20) fusion variant by CGP. Growth of a tumor cell line derived from this EML4-ALK CRC patient was inhibited by ALK inhibitors crizotinib and entrectinib. CONCLUSIONS: ALK IHC is a viable screening strategy for identifying ALK rearrangement in CRC. ALK rearrangement is a potential actionable driver mutation in CRC based on survival inhibition of patient tumor-derived cell line by potent ALK inhibitors.


Subject(s)
Colorectal Neoplasms/genetics , Gastrointestinal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Receptor Protein-Tyrosine Kinases/genetics , Adult , Aged , Aged, 80 and over , Anaplastic Lymphoma Kinase , Antibodies, Monoclonal/chemistry , Benzamides/chemistry , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Crizotinib , Female , Gastrointestinal Neoplasms/metabolism , Gene Expression Profiling , Genomics , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Indazoles/chemistry , Male , Middle Aged , Mutation , Phosphorylation , Pyrazoles/chemistry , Pyridines/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Reproducibility of Results , Republic of Korea , Sensitivity and Specificity , Tissue Array Analysis , Young Adult
17.
Int J Cancer ; 136(7): 1629-35, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25157953

ABSTRACT

Growth factor receptors, often carrying tyrosine kinase activities in their cytoplasmic domains, are overexpressed in many cancers. Coactivation of receptor tyrosine kinases (RTKs) plays a critical role in tumor response to targeted therapeutics. We examined concomitant overexpression of EGFR and MET in patients with HER2(+) and HER2(-) gastric cancers (GCs). Tissue microarray samples obtained from 1,589 GC patients who received R0 gastrectomy with extensive node dissection and adjuvant chemoradiationtherapy were analyzed by immunohistochemistry and fluorescence in situ hybridization. HER2(+) was observed in 169 patients (11%). Out of 169 HER2(+) patients, 15 (9%) were EGFR(+) and MET(+) , 29 (17%) were EGFR(+) , 37 (22%) were MET(+) and the remaining 88 patients (52%) were HER2(+) only, without concomitant EGFR or MET overexpression. Greater number of overexpressed RTKs correlated with younger age (p < 0.001), larger tumor size (p = 0.027), intestinal histology (p < 0.001) and shorter overall survival (p = 0.002). The mean overall survival was 113 months for HER2(-) /EGFR(-) /MET(-) and 63 months for HER2(+) /EGFR(+) /MET(+) subgroups. Patients with HER2(+) /EGFR(+) /MET(+) GCs had a substantial risk of death with a hazard ratio of 3.01 (95% CI: 1.54-5.90), compared with HER2(-) /EGFR(-) /MET(-) GC patients. Using patient-derived tumor cell models isolated from pericardial effusion of HER2(+) and MET(+) GC cases, we demonstrated that the combination of HER2-inhibitor (lapatinib) and MET-inhibitor offered a more profound inhibition in the ERK/AKT pathway and cell proliferation than lapatinib alone. Co-overexpression of RTKs was demonstrated in small subsets of GC associated with aggressive behavior and in these cases, combination therapy may be considered as potential treatment options.


Subject(s)
ErbB Receptors/genetics , Gene Expression , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/genetics , Stomach Neoplasms/genetics , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , ErbB Receptors/metabolism , Female , Gene Amplification , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Stomach Neoplasms/therapy , Tumor Burden
18.
J Gastrointest Oncol ; 5(6): 447-56, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25436124

ABSTRACT

BACKGROUND: The aims of this study were to demonstrate the tumorigenicity of CD133+ colon cancer cells in vitro, analyze the correlations between spheroid formation and clinicopathologic variables, and screen for overexpressed genes in CD133+ colon cancer stem cells. Moreover, the aim of this study was to establish a living tumor tissue bank using surgically resected specimens. METHODS: Using LoVo cell line, we isolated CD133+ cells and performed clonogenic assay and animal experiment to test tumorigenicity of CD133+ cells. Twenty-nine surgical samples were freshly collected from 27 patients who received curative or palliative surgery, and the samples were mechanically and enzymatically dissociated into single cells. RESULTS: We confirmed the enhanced tumorigenicity of CD133+ cells isolated from LoVo cell line both in vitro and in vivo. Of these 29 samples, 8 (28%) contained >3% CD133+ cells. Sphere formation was significantly higher in samples from patients with lymphatic invasion than in those without lymphatic invasion [54.5% (6/11) vs. 12.5% (2/16); P=0.033] and in samples containing >3% of CD133+ cells than in those containing ≤3% of CD133+ cells [36.4% (4/11) vs. 0% (0/16); P=0.019]. CONCLUSIONS: These findings indicate that CD133 is a valid marker for identifying cancer stem cells from fresh surgically resected colorectal cancer tissues. Furthermore, we successfully established a living tumor tissue bank using surgically resected colorectal tissues with a viability of >70%.

19.
Sci Rep ; 4: 7592, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25534230

ABSTRACT

To investigate the prognostic role of the estrogen receptor (ER) in gastric cancer (GC) patients, tumor tissues from 932 patients with advanced GC were assessed for ER expression using immunohistochemistry, and their clinicopathologic features were evaluated. Forty patients (4.3%) had ER expression and they were more frequently associated with diffuse type gastric cancer and shorter disease free survival. Furthermore, we carried out in vitro analysis to evaluate the effect of ER modulation on the proliferation of GC cell lines. Estradiol enhanced proliferation of ER positive GC cells while it did not show any effect on ER negative GC cells. When ER was inhibited by fulvestrant and ER siRNA, estradiol-induced proliferation of ER positive GC cell was suppressed. Paclitaxel showed synergistic anti-proliferative impacts with fulvestrant. Suppressing ER by fulvestrant, paclitaxel and ER siRNA showed increased expression of E-cadherin, which is a crucial factor in diffuse-type carcinogenesis.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Estradiol/analogs & derivatives , Gene Expression Regulation, Neoplastic/drug effects , Receptors, Estrogen/biosynthesis , Stomach Neoplasms , Adult , Aged , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Disease-Free Survival , Estradiol/pharmacology , Female , Fulvestrant , Humans , Immunohistochemistry , Male , Middle Aged , Paclitaxel/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate
20.
J Clin Invest ; 124(12): 5145-58, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25401468

ABSTRACT

Patients with gastric and esophageal (GE) adenocarcinoma tumors in which the oncogene ERBB2 has been amplified are routinely treated with a combination of cytotoxic chemotherapy and the ERBB2-directed antibody trastuzumab; however, the addition of trastuzumab, even when tested in a selected biomarker-positive patient population, provides only modest survival gains. To investigate the potential reasons for the modest impact of ERBB2-directed therapies, we explored the hypothesis that secondary molecular features of ERBB2-amplified GE adenocarcinomas attenuate the impact of ERBB2 blockade. We analyzed genomic profiles of ERBB2-amplified GE adenocarcinomas and determined that the majority of ERBB2-amplified tumors harbor secondary oncogenic alterations that have the potential to be therapeutically targeted. These secondary events spanned genes involved in cell-cycle regulation as well as phosphatidylinositol-3 kinase and receptor tyrosine kinase signaling. Using ERBB2-amplified cell lines, we demonstrated that secondary oncogenic events could confer resistance to ERBB2-directed therapies. Moreover, this resistance could be overcome by targeting the secondary oncogene in conjunction with ERBB2-directed therapy. EGFR is commonly coamplified with ERBB2, and in the setting of ERBB2 amplification, higher EGFR expression appears to mark tumors with greater sensitivity to dual EGFR/ERBB2 kinase inhibitors. These data suggest that combination inhibitor strategies, guided by secondary events in ERBB2-amplified GE adenocarcinomas, should be evaluated in clinical trials.


Subject(s)
Adenocarcinoma , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor , Esophageal Neoplasms , Gene Amplification , Receptor, ErbB-2 , Stomach Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , ErbB Receptors/genetics , ErbB Receptors/metabolism , Esophageal Neoplasms/diet therapy , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Gene Amplification/drug effects , Gene Amplification/genetics , Humans , Male , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Trastuzumab
SELECTION OF CITATIONS
SEARCH DETAIL
...