Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Adv Drug Deliv Rev ; 168: 158-180, 2021 01.
Article in English | MEDLINE | ID: mdl-32360576

ABSTRACT

The clustered, regularly interspaced, short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas) systems are efficient and versatile gene editing tools, which offer enormous potential to treat cancer by editing genome, transcriptome or epigenome of tumor cells and/or immune cells. A large body of works have been done with CRISPR/Cas systems for genetic modification, and 16 clinical trials were conducted to treat cancer by ex vivo or in vivo gene editing approaches. Now, promising preclinical works have begun using CRISPR/Cas systems in vivo. However, efficient and safe delivery of CRISPR/Cas systems in vivo is still a critical challenge for their clinical applications. This article summarizes delivery of CRISPR/Cas systems by physical methods, viral vectors and non-viral vectors for cancer gene therapy and immunotherapy. The prospects for the development of physical methods, viral vectors and non-viral vectors for delivery of CRISPR/Cas systems are reviewed, and promising advances in cancer treatment using CRISPR/Cas systems are discussed.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Genetic Therapy/methods , Neoplasms/genetics , Neoplasms/therapy , Animals , Apoptosis/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Delivery Systems , Epigenesis, Genetic/physiology , Gene Editing , Genes, Neoplasm/physiology , Genetic Vectors/administration & dosage , Humans , Tumor Microenvironment
2.
JAMA Netw Open ; 3(12): e2028644, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33295976

ABSTRACT

Importance: The incidence of appendiceal cancer (AC) is rising, particularly among individuals younger than 50 years (early-onset AC), with unexplained etiologies. The unique spectrum of somatic cancer gene variations among patients with early-onset AC is largely undetermined. Objective: To characterize the frequency of somatic variations and genomic patterns among patients with early-onset (age <50 years) vs late-onset (age ≥50 years) AC. Design, Setting, and Participants: This cohort study included individuals aged 18 years and older diagnosed with pathologically verified AC. Cases with clinical-grade targeted sequencing data from January 1, 2011, to December 31, 2019, were identified from the international clinicogenomic data-sharing consortium American Association for Cancer Research Project Genomics Evidence Neoplasia Information Exchange (GENIE). Data analysis was conducted from May to September 2020. Exposures: Age at disease onset. Main Outcomes and Measures: Somatic variation prevalence and spectrum in AC patients was determined. Variation comparisons between early-onset and late-onset AC were evaluated using multivariable logistic regression with adjustment for sex, race/ethnicity, histological subtype, sequencing center, and sample type. Results: In total 385 individuals (mean [SD] age at diagnosis, 56.0 [12.4] years; 187 [48.6%] men; 306 [79.5%] non-Hispanic White individuals) with AC were included in this study, and 109 patients (28.3%) were diagnosed with early-onset AC. Race/ethnicity differed by age at disease onset; non-Hispanic Black individuals accounted for a larger proportion of early-onset vs late-onset cases (9 of 109 [8.3%] vs 11 of 276 [4.0%]; P = 0.04). Compared with patients aged 50 years or older at diagnosis, patients with early-onset AC had significantly higher odds of presenting with nonsilent variations in PIK3CA, SMAD3, and TSC2 (PIK3CA: odds ratio [OR], 4.58; 95% CI, 1.72-12.21; P = .002; SMAD3: OR, 7.37; 95% CI, 1.24-43.87; P = .03; TSC2: OR, 12.43; 95% CI, 1.03-149.59; P = .047). In contrast, patients with early-onset AC had a 60% decreased odds of presenting with GNAS nonsilent variations compared with patients with late-onset AC (OR, 0.40; 95% CI, 0.21-0.76, P = .006). By histological subtype, young patients with mucinous adenocarcinomas of the appendix had 65% decreased odds of variations in GNAS compared with late-onset cases in adjusted models (OR, 0.35; 95% CI, 0.15-0.79; P = .01). Similarly, patients with early-onset nonmucinous appendiceal adenocarcinomas had 72% decreased odds of presenting with GNAS variations vs late-onset cases, although these findings did not reach significance (OR, 0.28; 95% CI, 0.07-1.14; P = .08). GNAS and TP53 variations were mutually exclusive in ACs among early-onset and late-onset cases (P < .05). Conclusions and Relevance: In the study, AC diagnosed among younger individuals harbored a distinct genomic landscape compared with AC diagnosed among older individuals. Development of therapeutic modalities that target these unique molecular features may yield clinical implications specifically for younger patients.


Subject(s)
Adenocarcinoma, Mucinous , Appendiceal Neoplasms , Genes, Neoplasm/physiology , Adenocarcinoma, Mucinous/epidemiology , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Age of Onset , Appendiceal Neoplasms/epidemiology , Appendiceal Neoplasms/genetics , Appendiceal Neoplasms/pathology , Appendix/pathology , Biopsy/methods , Chromogranins/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Drug Discovery , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Male , Middle Aged , Pharmacogenomic Variants , Tuberous Sclerosis Complex 2 Protein/genetics , United States/epidemiology
3.
Medicine (Baltimore) ; 99(37): e22194, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32925794

ABSTRACT

DNA methylation is an important epigenetic regulatory mechanism in esophageal carcinoma (EC) and is associated with genomic instability and carcinogenesis. In the present study, we aimed to identify tumor biomarkers for predicting prognosis of EC patients.We downloaded mRNA expression profiles and DNA methylation profiles associated with EC from the Gene Expression Omnibus database. Differentially expressed and differentially methylated genes between tumor tissues and adjacent normal tissue samples were identified. Functional enrichment analyses were performed, followed by the construction of protein-protein interaction networks. Data were validated based on methylation profiles from The Cancer Genome Atlas. Candidate genes were further verified according to survival analysis and Cox regression analysis.We uncovered multiple genes with differential expression or methylation in tumor samples compared with normal samples. After taking the intersection of 3 differential gene sets, we obtained a total of 232 overlapping genes. Functional enrichment analysis revealed that these genes are related to pathways such as "glutathione metabolism," "p53 signaling pathway," and "focal adhesion." Furthermore, 8 hub genes with inversed expression and methylation correlation were identified as candidate genes. The abnormal expression levels of MSN, PELI1, and MTHFD2 were correlated with overall survival times in EC patients (P < .05). Only MTHFD2 was significantly associated with a pathologic stage according to univariate analysis (P = .037) and multivariate analysis (P = .043).Our study identified several novel EC biomarkers with prognostic value by integrated analysis of transcriptomic data and methylation profiles. MTHFD2 could serve as an independent biomarker for predicting prognosis and pathological stages of EC.


Subject(s)
Aminohydrolases/biosynthesis , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Methylenetetrahydrofolate Dehydrogenase (NADP)/biosynthesis , Multifunctional Enzymes/biosynthesis , Biomarkers, Tumor , DNA Methylation , Databases, Genetic , Gene Expression Profiling , Genes, Neoplasm/physiology , Humans , Prognosis , Protein Interaction Maps , RNA, Messenger , Regression Analysis , Survival Analysis
4.
Cell ; 177(6): 1375-1383, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150618

ABSTRACT

Recent studies of the tumor genome seek to identify cancer pathways as groups of genes in which mutations are epistatic with one another or, specifically, "mutually exclusive." Here, we show that most mutations are mutually exclusive not due to pathway structure but to interactions with disease subtype and tumor mutation load. In particular, many cancer driver genes are mutated preferentially in tumors with few mutations overall, causing mutations in these cancer genes to appear mutually exclusive with numerous others. Researchers should view current epistasis maps with caution until we better understand the multiple cause-and-effect relationships among factors such as tumor subtype, positive selection for mutations, and gross tumor characteristics including mutational signatures and load.


Subject(s)
Epistasis, Genetic/genetics , Genes, Neoplasm/genetics , Neoplasms/genetics , Algorithms , Computational Biology/methods , Epistasis, Genetic/physiology , Genes, Neoplasm/physiology , Humans , Models, Genetic , Mutation/genetics , Oncogenes/genetics
5.
BJU Int ; 122(1): 152-159, 2018 07.
Article in English | MEDLINE | ID: mdl-29542849

ABSTRACT

OBJECTIVE: To evaluate the presence of circulating tumour cells (CTCs) at different stages of prostate cancer using the AdnaTest® ProstateCancerDetect kit (Qiagen). Moreover, we aimed to assess the expression of transcripts that are specific for cancer stem cells (AdnaTest StemCell) and epithelial-mesenchymal transition (EMT) in CTCs (AdnaTest EMT), as well as additional genes that are known to promote prostate cancer progression. PATIENTS AND METHODS: In this prospective study, we included 81 patients who underwent treatment for prostate cancer between 07/2014 and 02/2015, including: Group A, 18 patients (22.2%) with low-risk clinically localised prostate cancer; Group B, 25 patients (30.9%) with high-risk clinically localised prostate cancer; Group C, 11 patients (13.6%) with metastatic castration-sensitive prostate cancer (mCSPC); and Group D, 27 patients (33.3%) with metastatic castration-resistant prostate cancer (mCRPC). AdnaTest ProstateCancer and AdnaTest StemCell/EMT were performed in all cases. In addition, expression of the androgen receptor (AR), c-met, c-kit and thymidylate synthase (TYMS) in CTCs was assessed using specific polymerase chain reaction assays. RESULTS: A positive AdnaTest ProstateCancer was present in three (16.7%), two (8.0%), six (54.5%) and 19 (70.5%) patients in groups A, B, C and D, respectively (P < 0.01, chi-squared test). The AdnaTest EMT and AdnaTest StemCell were positive in zero (0.0%), zero (0.0%), one (9.1%), and two (7.4%); and in five (27.8%), four (16.0%), three (27.3%), and 11 (40.7%) patients in groups A, B, C and D, respectively, with no significant differences noted between groups. CTCs expressing TYMS (44.4% and 50.0% vs 13.9%) or AR (18.2% and 25.9% vs 0.0%) were seen more commonly in patients in groups C and D vs patients with non-metastatic disease (all P < 0.05). Expression of c-kit and c-met were rare events, with only two patients positive for either marker. CONCLUSIONS: AdnaTest ProstateCancerDetect exhibits positive results mainly in patients with metastatic disease. Expression of AR and TYMS are frequent events in CTCs of patients with advanced disease, whereas c-met and c-kit gene expression is seen in only a small proportion of patients. The implications of these results for the use of CTC analysis as a decision factor for personalised treatment strategies in advanced prostate cancer remain to be determined.


Subject(s)
Genes, Neoplasm/physiology , Neoplastic Cells, Circulating , Prostatic Neoplasms, Castration-Resistant/genetics , Aged , Aged, 80 and over , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Neoplasm Metastasis , Prospective Studies , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-met/metabolism
6.
Pril (Makedon Akad Nauk Umet Odd Med Nauki) ; 37(2-3): 89-97, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27883322

ABSTRACT

Malignant skin melanoma is a tumor deriving from transformed skin melanocytes as a result of complex interactions between genetic and environmental factors. This melanoma has a potential to metastasize early and very often it is resistant to the existing modalities of the systemic therapy. As in any other neoplasms, certain types of melanoma may skip certain stages of progression. The progression from one stage to another is accompanied by specific biological changes. Several key changes in the melanoma tumorogenesis influence the regulation of the cell proliferation and vitality, including the RAS-RAF-ERK, PI3K-AKT, and p16INK4/CDK4/RB pathways. A key role in the dissreguarity of the RAS-RAF-ERK (MAPK) pathway in the malignant melanoma development have been demonstrated by many studies. To date, the molecular genetic alterations during melanoma development have been partially known. In the pathogenesis of the malignant melanoma, there are mutations of various genes such as NRAS, BRAF, and PTEN and mutations and deletions of CDKN2A. In the past years, great advance has been made in the insights of the molecular aspects of the melanoma pathogenesis. However, this field yet poses a challenge to discover new details about the melanoma molecular characteristics. The research results are focused towards the improvement of the melanoma patients prognosis by introducing personalized targeted therapy.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Genes, Neoplasm/physiology , Humans , Signal Transduction/physiology
7.
PLoS One ; 11(11): e0165830, 2016.
Article in English | MEDLINE | ID: mdl-27824899

ABSTRACT

The effects of sarcosine on the processes driving prostate cancer (PCa) development remain still unclear. Herein, we show that a supplementation of metastatic PCa cells (androgen independent PC-3 and androgen dependent LNCaP) with sarcosine stimulates cells proliferation in vitro. Similar stimulatory effects were observed also in PCa murine xenografts, in which sarcosine treatment induced a tumor growth and significantly reduced weight of treated mice (p < 0.05). Determination of sarcosine metabolism-related amino acids and enzymes within tumor mass revealed significantly increased glycine, serine and sarcosine concentrations after treatment accompanied with the increased amount of sarcosine dehydrogenase. In both tumor types, dimethylglycine and glycine-N-methyltransferase were affected slightly, only. To identify the effects of sarcosine treatment on the expression of genes involved in any aspect of cancer development, we further investigated expression profiles of excised tumors using cDNA electrochemical microarray followed by validation using the semi-quantitative PCR. We found 25 differentially expressed genes in PC-3, 32 in LNCaP tumors and 18 overlapping genes. Bioinformatical processing revealed strong sarcosine-related induction of genes involved particularly in a cell cycle progression. Our exploratory study demonstrates that sarcosine stimulates PCa metastatic cells irrespectively of androgen dependence. Overall, the obtained data provides valuable information towards understanding the role of sarcosine in PCa progression and adds another piece of puzzle into a picture of sarcosine oncometabolic potential.


Subject(s)
Cell Cycle/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm/physiology , Prostatic Neoplasms/metabolism , Sarcosine/pharmacology , Animals , Cell Cycle/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Glycine N-Methyltransferase/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Polymerase Chain Reaction , Prostatic Neoplasms/physiopathology , Sarcosine/metabolism , Sarcosine Dehydrogenase/metabolism , Transcriptome , Up-Regulation
8.
Neoplasma ; 63(6): 856-864, 2016.
Article in English | MEDLINE | ID: mdl-27565323

ABSTRACT

This study aimed to investigate the role and potential mechanism of miR-22 in clear cell ovarian cancer (CCOC) progression. The gene expression profile of GSE16568, including 3 CCOC samples with miR-22 overexpression and 3 negative controls, was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened using the limma package in R. Gene Ontology (GO) and pathway enrichment analysis of DEGs were performed by using The Database for Annotation, Visualization and Integrated Discovery (DAVID). Furthermore, protein-protein interaction (PPI) network of the DEGs was constructed using the Search Tool for the Retrieval of Interacting Genes (STRING) database. Besides, the miR-22-mRNA interaction pairs were predicted to explore the critical genes involved in the cancer. Totally, 95 up-regulated DEGs and 51 down-regulated DEGs were identified. The DEGs were enriched in different GO terms and pathways. The up-regulated genes cyclin-dependent kinases (CDK6), MDM2 oncogene, E3 ubiquitin protein ligase (MDM2), and thrombospondin 1 (THBS1) were involved in the p53 signaling pathway. The up-regulated gene FBJ murine osteosarcoma viral oncogene homolog (FOS) was a hub protein in the PPI network of the DEGs. The down-regulated DEGs including lymphoid enhancer-binding factor 1 (LEF1) and v-myb avian myeloblastosis viral oncogene homolog (MYB) were mainly associated with immunity. Nine DEGs as target genes were identified to be recognized by miR-22. Our study suggested that several key genes such as CDK6, MDM2, LEF1, MYB, and FOS that involved in different pathways including p53 signaling pathway were associated with CCOC progression. miR-22 may play an essential role in cell migration and invasion in CCOC through targeting responsive genes.


Subject(s)
Genes, Neoplasm/physiology , MicroRNAs/physiology , Ovarian Neoplasms/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Cell Movement , Disease Progression , Down-Regulation , Female , Gene Expression Profiling , Humans , Ovarian Neoplasms/pathology , Protein Interaction Maps
9.
Zhonghua Zhong Liu Za Zhi ; 37(2): 107-12, 2015 Feb.
Article in Chinese | MEDLINE | ID: mdl-25975686

ABSTRACT

OBJECTIVE: To investigate the polymorphism in the promoter region of PCA3 gene and its relationship with risk of prostate cancer (PCa). METHODS: The promoter region of PCA3 gene of the DNA of peripheral blood mononuclear cells was detected by sequence analysis in the 186 PCa and 141 BPH patients and 135 healthy control individuals. If the samples were detected with polymorphism of insection/deletion, clone sequence analysis was used with pBS-T carrier to verify it. RESULTS: There were 5 polymorphisms. TAAA repeat times: 4, 5, 6, 7, 8, and 8 genotypes (TAAA 4/5, TAAA 4/6, TAAA 5/5, TAAA 5/6, TAAA 5/7, TAAA 5/8, TAAA 6/6, and TAAA 6/7) were detected in the promoter region of PCA3 gene. The eight genotypes were divided into three groups: ≤10TAAA, 11TAAA, ≥12TAAA. Unconditional logistic regression analysis models were used to analyze the relationship between different genotypes and cancer risks adjusted by sex and age. The type 11TAAA and ≥12TAAA was associated with higher relative risk for prostate cancer than the group ≤10TAAA [OR=1.74, 95% CI=1.06-2.87 (for type 11TAAA); OR=5.63, 95% CI=1.85-17.19 (for type ≥12TAAA)]. In the 186 PCa patients, there was 62.4% allele of PCA3 gene with AG/CA mutation found in the promoter 18-19 bp region of PCA3 gene and it had a close relation with the development of prostate cancer. CONCLUSIONS: Short tandem repeats are found in the promoter region of the PCA3 gene in PCa patients, and the increase of TAAA repeat sequences highly enhance the relative risk of prostate cancer development. The occurrence of such STR might be related to the mutations in their upstream loci.


Subject(s)
Antigens, Neoplasm/genetics , Genes, Neoplasm/physiology , Prostatic Neoplasms/genetics , Antigens, Neoplasm/metabolism , Base Sequence , Genotype , Humans , Leukocytes, Mononuclear , Male , Microsatellite Repeats , Mutation , Polymorphism, Genetic , Promoter Regions, Genetic , Prostatic Neoplasms/epidemiology , Risk
10.
Clin Exp Ophthalmol ; 43(3): 259-67, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25132102

ABSTRACT

BACKGROUND: Regulation of apoptosis is a complex process that involves a number of genes, including Bcl-2, Bcl-x, Bax and other Bcl-2 family members. The aim of the present study is to assess the expression of Bcl- 2 and Bax in retinoblastoma, and correlate them with clinical and histopathological parameters. METHODS: The expression of Bcl-2 and Bax proteins were examined using immunohistochemistry, Western blotting and reverse transcriptase-polymerase chain reaction in a series of 60 prospective cases of primary retinoblastoma tissues. RESULTS: Immunohistochemistry showed expression of Bcl-2 in 40/60 (66.6%), whereas Bax expression was found only in 18/60 (30%) cases, and these correlated with mRNA expression. The Western blotting results also correlated well with the immunohistochemical expression of Bcl-2 (25 kDa) and Bax (21 kDa) proteins. Bcl-2 was expressed in 96% (24/25) of invasive tumours and in 45.7% (16/35) of non-invasive tumours. Expression of Bcl-2 significantly correlated with tumour invasiveness (P = 0.0274) and poor differentiation (P = 0.0163), whereas loss of Bax correlated with massive choroidal invasion and Pathological Tumor-Node-Metastasis (pTNM) (P = 0.0341). However, no correlation was found between Bax and Bcl-2 expression. CONCLUSIONS: Our findings suggest that these apoptotic regulatory proteins may serve as poor prognostic markers and can be used as a therapeutic target for the treatment of invasive retinoblastoma. Further functional studies are required to explore the role of Bax and Bcl-2 in retinoblastoma.


Subject(s)
Apoptosis , Biomarkers, Tumor/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , bcl-2-Associated X Protein/metabolism , Biomarkers, Tumor/genetics , Blotting, Western , Child, Preschool , Female , Gene Expression/physiology , Genes, Neoplasm/physiology , Humans , Immunohistochemistry , Male , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , Retinal Neoplasms/genetics , Retinal Neoplasms/pathology , Retinoblastoma/genetics , Retinoblastoma/pathology , Reverse Transcriptase Polymerase Chain Reaction , bcl-2-Associated X Protein/genetics
12.
Vet Pathol ; 51(4): 868-81, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24091811

ABSTRACT

Metastasis is the primary cause of death in breast cancer patients, yet there are challenges to modeling this process in vivo. The goal of this study was to analyze the effects of injection site on tumor growth and metastasis and gene expression of breast cancer cells in vivo using the MMTV-PymT breast cancer model (Met-1 cells). Met-1 cells were injected into 5 sites (subcutaneous, mammary fat pad, tail vein, intracardiac, and intratibial), and tumors and metastases were monitored using bioluminescent imaging and confirmed with gross necropsy and histopathology. Met-1 tumors were analyzed based on morphology and changes in gene expression in each tissue microenvironment. There were 6 permissible sites of Met-1 tumor growth (mammary gland, subcutis, lung, adrenal gland, ovary, bone). Met-1 cells grew faster in the subcutis compared to mammary fat pad tumors (highest Ki-67 index). Morphologic differences were evident in each tumor microenvironment. Finally, 7 genes were differentially expressed in the Met-1 tumors in the 6 sites of growth or metastasis. This investigation demonstrates that breast cancer progression and metastasis are regulated by not only the tumor cells but also the experimental model and unique molecular signals from the tumor microenvironment.


Subject(s)
Disease Models, Animal , Gene Expression Regulation, Neoplastic/physiology , Genes, Neoplasm/physiology , Mammary Neoplasms, Animal/pathology , Neoplasm Metastasis/physiopathology , Tumor Microenvironment/physiology , Analysis of Variance , Animals , Cell Line, Tumor , Female , Gene Expression Profiling/veterinary , Gene Expression Regulation, Neoplastic/genetics , Genes, Neoplasm/genetics , Histological Techniques/veterinary , Immunohistochemistry/veterinary , Mice , Neoplasm Metastasis/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary
13.
Nat Genet ; 45(7): 756-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685747

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are sarcomas of Schwann cell lineage origin that occur sporadically or in association with the inherited syndrome neurofibromatosis type 1. To identify genetic drivers of MPNST development, we used the Sleeping Beauty (SB) transposon-based somatic mutagenesis system in mice with somatic loss of transformation-related protein p53 (Trp53) function and/or overexpression of human epidermal growth factor receptor (EGFR). Common insertion site (CIS) analysis of 269 neurofibromas and 106 MPNSTs identified 695 and 87 sites with a statistically significant number of recurrent transposon insertions, respectively. Comparison to human data sets identified new and known driver genes for MPNST formation at these sites. Pairwise co-occurrence analysis of CIS-associated genes identified many cooperating mutations that are enriched in Wnt/ß-catenin, PI3K-AKT-mTOR and growth factor receptor signaling pathways. Lastly, we identified several new proto-oncogenes, including Foxr2 (encoding forkhead box R2), which we functionally validated as a proto-oncogene involved in MPNST maintenance.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, Neoplasm , Genetic Testing/methods , Nerve Sheath Neoplasms/genetics , Animals , Cell Line, Tumor , DNA Mutational Analysis , DNA Transposable Elements/genetics , Genes, Neoplasm/physiology , Genetic Association Studies , Humans , Mice , Mice, Transgenic , Mutation/physiology , Neurofibroma/genetics , Proto-Oncogene Mas , Signal Transduction/genetics
15.
Blood ; 120(6): 1274-81, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22740451

ABSTRACT

Anaplastic large-cell lymphomas (ALCLs) are a group of clinically and biologically heterogeneous diseases including the ALK(+) and ALK(-) systemic forms. Whereas ALK(+) ALCLs are molecularly characterized and can be readily diagnosed, specific immunophenotypic or genetic features to define ALK(-) ALCL are missing, and their distinction from other T-cell non-Hodgkin lymphomas (T-NHLs) remains controversial. In the present study, we undertook a transcriptional profiling meta-analysis of 309 cases, including ALCL and other primary T-NHL samples. Pathway discovery and prediction analyses defined a minimum set of genes capable of recognizing ALK(-) ALCL. Application of quantitative RT-PCR in independent datasets from cryopreserved and formalin-fixed paraffin-embedded samples validated a 3-gene model (TNFRSF8, BATF3, and TMOD1) able to successfully separate ALK(-) ALCL from peripheral T-cell lymphoma not otherwise specified, with overall accuracy near 97%. In conclusion, our data justify the possibility of translating quantitative RT-PCR protocols to routine clinical settings as a new approach to objectively dissect T-NHL and to select more appropriate therapeutic protocols.


Subject(s)
Biomarkers, Tumor/genetics , Genes, Neoplasm , Lymphoma, Large-Cell, Anaplastic/diagnosis , Lymphoma, Large-Cell, Anaplastic/genetics , Molecular Diagnostic Techniques/methods , Receptor Protein-Tyrosine Kinases/genetics , Adult , Anaplastic Lymphoma Kinase , Biomarkers, Tumor/isolation & purification , Biomarkers, Tumor/physiology , Case-Control Studies , Diagnosis, Differential , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/physiology , Humans , Microarray Analysis , Models, Statistical , Predictive Value of Tests , Prognosis , Receptor Protein-Tyrosine Kinases/metabolism
16.
Cancer Res ; 72(15): 3720-4, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22628426

ABSTRACT

The Global Cancer Genomics Consortium (GCGC) is an international collaborative platform that amalgamates cancer biologists, cutting-edge genomics, and high-throughput expertise with medical oncologists and surgical oncologists; they address the most important translational questions that are central to cancer research and treatment. The annual GCGC symposium was held at the Advanced Centre for Treatment Research and Education in Cancer, Mumbai, India, from November 9 to 11, 2011. The symposium showcased international next-generation sequencing efforts that explore cancer-specific transcriptomic changes, single-nucleotide polymorphism, and copy number variations in various types of cancers, as well as the structural genomics approach to develop new therapeutic targets and chemical probes. From the spectrum of studies presented at the symposium, it is evident that the translation of emerging cancer genomics knowledge into clinical applications can only be achieved through the integration of multidisciplinary expertise. In summary, the GCGC symposium provided practical knowledge on structural and cancer genomics approaches, as well as an exclusive platform for focused cancer genomics endeavors.


Subject(s)
Genomics/trends , Medical Oncology/methods , Medical Oncology/trends , Neoplasms/genetics , Congresses as Topic , Genes, Neoplasm/physiology , Genome, Human/physiology , Genomics/methods , Genomics/organization & administration , Humans , India , International Cooperation , Medical Oncology/organization & administration , Neoplasms/therapy , Public-Private Sector Partnerships/organization & administration , Public-Private Sector Partnerships/trends , Societies, Medical , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends
17.
J Pathol ; 226(3): 471-81, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22009689

ABSTRACT

Nasopharyngeal carcinoma (NPC) is an EBV-associated epithelial malignancy which is prevalent in south-east Asia and southern China. Despite the multiple genetic and epigenetic changes reported, the contribution of dysregulated signalling pathways to this distinct type of head and neck cancer is not well understood. Here we demonstrate the up-regulation of NOTCH ligands (JAG1 or DLL4) and effector (HEY1) in the majority of EBV-positive tumour lines and primary tumours. Among the NOTCH receptors, NOTCH3 was over-expressed in all EBV-positive tumour lines and 92.5% of primary tumours. Aberrant activation of NOTCH3 signalling was consistently detected in all these samples. These findings imply that NOTCH3 may play an crucial role in the development of NPC. By NOTCH3 specific siRNA, NOTCH3 signalling was suppressed and thereby significant growth inhibition and apoptosis induction occurred in NPC cells. Down-regulation of a number of targets involved in cell proliferation, eg CCND1, C-MYC,NFKB1, and survival, eg BCL2, BCL-XL, SURVIVIN, was confirmed in the NOTCH3 knockdown NPC cells. Importantly, NOTCH3 knockdown highly enhanced the sensitivity of NPC cells to cisplatin treatment. Furthermore, we revealed that the ability of NPC cells to form spheroids in vitro and tumours in nude mice was also significantly decreased after knockdown of NICD3 expression. These findings indicate that activation of NOTCH3 pathway is a critical oncogenic event in NPC tumourigenesis. Targeting NOTCH3 signalling may serve as a potential therapeutic approach for treating patients suffering from EBV-associated NPC.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Epstein-Barr Virus Infections/complications , Nasopharyngeal Neoplasms/drug therapy , Receptors, Notch/antagonists & inhibitors , Signal Transduction/physiology , Animals , Apoptosis/physiology , Carcinoma , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Gene Knockdown Techniques , Genes, Neoplasm/physiology , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/virology , Neoplasm Transplantation , RNA, Small Interfering/metabolism , Receptor, Notch3 , Receptors, Notch/metabolism , Spheroids, Cellular/physiology , Transfection , Transplantation, Heterologous
18.
Neoplasia ; 13(11): 1081-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22131883

ABSTRACT

The human and Old World primate genomes possess conserved endogenous retrovirus sequences that have been implicated in evolution, reproduction, and carcinogenesis. Human endogenous retrovirus (HERV)-K with 5'LTR-gag-pro-pol-env-rec/np9-3'LTR sequences represents the newest retrovirus family that integrated into the human genome 1 to 5 million years ago. Although a high-level expression of HERV-K in melanomas, breast cancers, and teratocarcinomas has been demonstrated, the mechanism of the lineage-specific activation of the long terminal repeat (LTR) remains obscure. We studied chromosomal HERV-K expression in MeWo melanoma cells in comparison with the basal expression in human embryonic kidney 293 (HEK293) cells. Cloned LTR of HERV-K (HML-2.HOM) was also characterized by mutation and transactivation experiments. We detected multiple transcriptional initiator (Inr) sites in the LTR by rapid amplification of complementary DNA ends (5' RACE). HEK293 and MeWo showed different Inr usage. The most potent Inr was associated with a TATA box and three binding motifs of microphthalmia-associated transcription factor (MITF). Both chromosomal HERV-K expression and the cloned LTR function were strongly activated in HEK293 by transfection with MITF-M, a melanocyte/melanoma-specific isoform of MITF. Coexpression of MITF and the HERV-K core antigen was detected in retinal pigmented epithelium by an immunofluorescence analysis. Although malignant melanoma lines MeWo, G361, and SK-MEL-28 showed enhanced HERV-K transcription compared with normal melanocytes, the level of MITF-M messenger RNA persisted from normal to transformed melanocytes. Thus, MITF-M may be a prerequisite for the pigmented cell lineage-specific function of HERV-K LTR, leading to the high-level expression in malignant melanomas.


Subject(s)
Endogenous Retroviruses/genetics , Microphthalmia-Associated Transcription Factor/physiology , Terminal Repeat Sequences/genetics , Transcriptional Activation , Adult , Animals , Base Sequence , Cells, Cultured , Female , Fetus/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Genes, Neoplasm/physiology , HEK293 Cells , Humans , Macaca mulatta , Male , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Molecular Sequence Data , Skin Neoplasms/genetics , Transcriptional Activation/genetics
19.
Neuroendocrinology ; 94(3): 177-90, 2011.
Article in English | MEDLINE | ID: mdl-21893937

ABSTRACT

Pancreatic neuroendocrine tumors (pancreatic NET) are relatively rare, slowly growing tumors, although their incidence is increasing, and patients may survive for several years with metastatic disease. Apart from symptomatic relief, there have been few treatment options for these tumors in the past. More recently, investigators have explored the potential of molecularly targeted agents in treating pancreatic NET, with some success. In this review, we consider the data supporting exploitation of different targets in pancreatic NET, including peptide receptors, receptor tyrosine kinases (involved in tumor angiogenesis and more directly supporting tumor growth), and intracellular targets, such as the mammalian target of rapamycin (mTOR), which has a central role in regulating cell growth, metabolism, and apoptosis. Probably due to the paucity of pancreatic NET, many clinical trials to date have included heterogeneous NET populations, and there are few randomized studies of this specific patient population. Very recently, promising results have been achieved in placebo-controlled, phase III trials with the multitargeted tyrosine kinase inhibitor, sunitinib, and the mTOR inhibitor, everolimus. These agents have been approved or are currently being reviewed by authorities for use in patients with pancreatic NET. Here we review potential molecular targets in pancreatic NET and summarize the available data for targeted agents from phase II and III trials open to patients with this tumor.


Subject(s)
Medical Oncology/trends , Molecular Targeted Therapy/methods , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/therapeutic use , Genes, Neoplasm/physiology , Humans , Medical Oncology/methods , Models, Biological , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics
20.
Horm Cancer ; 2(5): 261-71, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21826535

ABSTRACT

Primary breast carcinomas expressing both estrogen and progesterone receptors are most likely to respond to tamoxifen therapy, especially in patients with early-stage lesions. However, certain patients exhibit clinicopathologic features suggesting good prognosis relapse within 10 years, justifying a search for biomarkers identifying patients at risk for recurrence. Nine candidate genes associated with estrogen signaling were selected from microarray studies and combined with those for conventional biomarkers (ESR1, PGR, ERBB2). Expression of this 12-gene subset was analyzed by RT-qPCR in frozen tissue specimens from 60 early-stage, estrogen receptor (ER)+/progestin receptor (PR)+ breast cancers from patients treated with adjuvant tamoxifen. A multivariate model was created by Cox regression using a training data set and applied to an independent validation set. A five-gene model was developed from the training set (n = 36) that exhibited significant correlations with both relapse-free and overall survival. Applying this model to Kaplan-Meier regression, patients were separated into low-risk (100% relapse-free at 150 months) and high-risk (60% relapse-free at 150 months) groups (P = 0.03). When this model was applied to the validation set (n = 24), similar risk stratification was achieved for both relapse-free and overall survival (P = 0.01 and 0.04, respectively). We developed a five-gene model composed of PgR, BCL2, ERBB4 JM-a, RERG, and CD34 that identified early-stage, ER+/PR+ breast cancers in patients treated with tamoxifen that relapsed, although they exhibited clinicopathologic features suggesting good prognosis. Within this multivariate model, increased expression of PgR, ERBB4 JM-a, RERG, and CD34 was associated with increased survival, while increased expression of BCL2 was associated with decreased survival.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Carcinoma/diagnosis , Carcinoma/drug therapy , Genes, Neoplasm , Tamoxifen/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Chemotherapy, Adjuvant , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/physiology , Humans , Middle Aged , Neoplasm Staging , Prognosis , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...