Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Eur J Endocrinol ; 2023 May 22.
Article in English | MEDLINE | ID: mdl-37216402

ABSTRACT

OBJECTIVE: 20% of sporadic MTC has no RET/RAS somatic alterations or other known gene alterations. Aim of this study was to investigate RET/RAS negative MTC for the presence of NF1 alterations. METHODS: we studied 18 sporadic RET/RAS negative MTC cases: Next generation sequencing of tumoral and blood DNA was performed using a custom panel including the entire coding region of the NF1 gene. The effect of NF1 alterations on the transcripts were characterized by RT-PCR and the loss of heterozygosity of the other NF1 allele was investigated with Multiplex Ligation-dependent Probe Amplification. RESULTS: Two cases showed bi-allelic inactivation of NF1 with a prevalence of about 11% of RET/RAS negative cases. In a patient affected by neurofibromatosis there was a somatic intronic point mutation determining the transcript alteration in one allele and a germline loss of heterozygosity (LOH) in the other. In the other case described both the point mutation and the LOH were somatic events; this latter finding shows, for the first time, a driver role of NF1 inactivation in MTC independent of RET/RAS alterations and the presence of neurofibromatosis. CONCLUSIONS: About 11% of our series of sporadic RET/RAS negative MTC harbor biallelic inactivation of NF1 suppressor gene also regardless neurofibromatosis status. According to our results, NF1 alterations should be searched in all RET/RAS negative MTC as possible driver. Moreover, this finding reduces the number of negative sporadic MTCs and may have important clinical implications in the management of these tumors.

2.
J Mol Cell Biol ; 14(11)2023 04 06.
Article in English | MEDLINE | ID: mdl-36460033

ABSTRACT

Peritoneal metastases (PM) from colorectal cancer (CRC) are associated with poor survival. The extracellular matrix (ECM) plays a fundamental role in modulating the homing of CRC metastases to the peritoneum. The mechanisms underlying the interactions between metastatic cells and the ECM, however, remain poorly understood, and the number of in vitro models available for the study of the peritoneal metastatic process is limited. Here, we show that decellularized ECM of the peritoneal cavity allows the growth of organoids obtained from PM, favoring the development of three-dimensional (3D) nodules that maintain the characteristics of in vivo PM. Organoids preferentially grow on scaffolds obtained from neoplastic peritoneum, which are characterized by greater stiffness than normal scaffolds. A gene expression analysis of organoids grown on different substrates reflected faithfully the clinical and biological characteristics of the organoids. An impact of the ECM on the response to standard chemotherapy treatment for PM was also observed. The ex vivo 3D model, obtained by combining patient-derived decellularized ECM with organoids to mimic the metastatic niche, could be an innovative tool to develop new therapeutic strategies in a biologically relevant context to personalize treatments.


Subject(s)
Colorectal Neoplasms , Peritoneal Neoplasms , Humans , Decellularized Extracellular Matrix , Peritoneum , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Organoids , Colorectal Neoplasms/metabolism
4.
Biomedicines ; 10(5)2022 May 23.
Article in English | MEDLINE | ID: mdl-35625944

ABSTRACT

Pancreatic cancer has a high morbidity and mortality with the majority being PC ductal adenocarcinomas (PDAC). Whole genome sequencing provides a wide description of genomic events involved in pancreatic carcinogenesis and identifies putative biomarkers for new therapeutic approaches. However, currently, there are no approved treatments targeting driver mutations in PDAC that could produce clinical benefit for PDAC patients. A proportion of 5-10% of PDAC have a hereditary origin involving germline variants of homologous recombination genes, such as Mismatch Repair (MMR), STK11 and CDKN2A genes. Very recently, BRCA genes have been demonstrated as a useful biomarker for PARP-inhibitor (PARPi) treatments. In this study, a series of 21 FFPE PDACs were analyzed using OncoPan®, a strategic next-generation sequencing (NGS) panel of 37 genes, useful for identification of therapeutic targets and inherited cancer syndromes. Interestingly, this approach, successful also on minute pancreatic specimens, identified biomarkers for personalized therapy in five PDAC patients, including two cases with HER2 amplification and three cases with mutations in HR genes (BRCA1, BRCA2 and FANCM) and potentially eligible to PARPi therapy. Molecular analysis on normal tissue identified one PDAC patient as a carrier of a germline BRCA1 pathogenetic variant and, noteworthy, this patient was a member of a family affected by inherited breast and ovarian cancer conditions. This study demonstrates that the OncoPan® NGS-based panel constitutes an efficient methodology for the molecular profiling of PDAC, suitable for identifying molecular markers both for therapy and risk assessment. Our data demonstrate the feasibility and utility of these NGS analysis in the routine setting of PDAC molecular characterization.

5.
Int J Biol Markers ; 37(1): 102-109, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34931559

ABSTRACT

INTRODUCTION: Disease recurrence after surgery is a crucial predictor of poor prognosis in colorectal cancer, where disseminated disease at the time of intervention can also be observed in localized early-stage cases. We evaluated the ability to predict disease recurrence of miRNAs from two signatures that we have found linked to the presence of colorectal cancer (CL signature) or adenoma (HgA signature) in higher-risk subjects. METHODS: miRNAs from the signatures were studied longitudinally by quantitative real-time polymerase chain reaction in plasma from 24 patients with resectable colorectal cancer collected at the time of surgery and during scheduled follow-up across 36 months. Patients either showed relapse within 36 months (alive with disease (AWD)), or remained disease-free (no evidence of disease (NED)) for the same period. RESULTS: Although the signatures did not predict recurrence, expression of the miRNAs from the CL signature decreased 1 year after surgery, and one miRNA of the signature, miR-378a-3p, almost reached significance in the NED subgroup (Wilcoxon signed-rank test: p-value = 0.078). Also, miR-335-5p from the HgA signature was higher in AWD patients before surgery (Kruskal-Wallis test: p-value = 0.019). CONCLUSIONS: These data, although from a small cohort of patients, support the possible use of miRNAs as non-invasive biomarkers in liquid biopsy-based tests to identify patients at risk of relapse and to monitor them during follow-up.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/surgery , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Recurrence, Local/genetics , Prognosis
6.
Breast ; 58: 121-129, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34022715

ABSTRACT

Genetic testing for hereditary breast and ovarian cancer following genetic counseling is based on guidelines that take into account particular features of the personal and family history, and clinical criteria conferring a probability of having a BRCA mutation greater than 10% as a threshold for accessing the test. However, besides reducing mortality and social impact, the extension of screening programs also for healthy family members would allow a huge saving of the rising costs associated with these pathologies, supporting the choice of the "Test" strategy versus a "No Test" one. Analyses of different health care systems show that by applying the "Test" strategy on patients and their families, a decrease in breast and ovarian cancer cases is achieved, as well as a substantial decrease in costs of economic resources, including the costs of the clinical management of early detected tumors. In this review, we analyzed the most recent papers published on this topic and we summarized the findings on the economic evaluations related to breast and ovarian cancer population screenings. These results proved and validated that the population-wide testing approach is a more accurate screening and preventive intervention than traditional guidelines based on personal/family history and clinical criteria to reduce breast and ovarian cancer risk.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Cost-Benefit Analysis , Early Detection of Cancer , Female , Genetic Predisposition to Disease , Genetic Testing , Humans , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics
7.
Int J Cancer ; 146(4): 1164-1173, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31304978

ABSTRACT

Colorectal cancer (CRC) screening programs help diagnose cancer precursors and early cancers and help reduce CRC mortality. However, currently recommended tests, the fecal immunochemical test (FIT) and colonoscopy, have low uptake. There is therefore a pressing need for screening strategies that are minimally invasive and consequently more acceptable to patients, most likely blood based, to increase early CRC identification. MicroRNAs (miRNAs) released from cancer cells are detectable in plasma in a remarkably stable form, making them ideal cancer biomarkers. Using plasma samples from FIT-positive (FIT+) subjects in an Italian CRC screening program, we aimed to identify plasma circulating miRNAs that detect early CRC. miRNAs were initially investigated by quantitative real-time PCR in plasma from 60 FIT+ subjects undergoing colonoscopy at Fondazione IRCCS Istituto Nazionale dei Tumori, then tested on an internal validation cohort (IVC, 201 cases) and finally in a large multicenter prospective series (external validation cohort [EVC], 1121 cases). For each endoscopic lesion (low-grade adenoma [LgA], high-grade adenoma [HgA], cancer lesion [CL]), specific signatures were identified in the IVC and confirmed on the EVC. A two-miRNA-based signature for CL and six-miRNA signatures for LgA and HgA were selected. In a multivariate analysis including sex and age at blood collection, the areas under the receiver operating characteristic curve (95% confidence interval) of the signatures were 0.644 (0.607-0.682), 0.670 (0.626-0.714) and 0.682 (0.580-0.785) for LgA, HgA and CL, respectively. A miRNA-based test could be introduced into the FIT+ workflow of CRC screening programs so as to schedule colonoscopies only for subjects likely to benefit most.


Subject(s)
Colorectal Neoplasms/genetics , MicroRNAs/blood , Aged , Colorectal Neoplasms/blood , Early Detection of Cancer , Female , Humans , Male , MicroRNAs/genetics , Middle Aged
8.
Sci Rep ; 7(1): 15992, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29167573

ABSTRACT

Metformin is a widely used and well-tolerated anti-diabetic drug that can reduce cancer risk and improve the prognosis of certain malignancies. However, the mechanism underlying its anti-cancer effect is still unclear. We studied the anti-cancer activity of metformin on colorectal cancer (CRC) by using the drug to treat HT29, HCT116 and HCT116 p53-/- CRC cells. Metformin reduced cell proliferation and migration by inducing cell cycle arrest in the G0/G1 phase. This was accompanied by a sharp decrease in the expression of c-Myc and down-regulation of IGF1R. The anti-proliferative action of metformin was mediated by two different mechanisms: AMPK activation and increase in the production of reactive oxygen species, which suppressed the mTOR pathway and its downstream targets S6 and 4EBP1. A reduction in CD44 and LGR5 expression suggested that the drug had an effect on tumour cells with stem characteristics. However, a colony formation assay showed that metformin slowed the cells' ability to form colonies without arresting cell growth, as confirmed by absence of apoptosis, autophagy or senescence. Our finding that metformin only transiently arrests CRC cell growth suggests that efforts should be made to identify compounds that combined with the biguanide can act synergistically to induce cell death.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Colorectal Neoplasms/metabolism , Metformin/pharmacology , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Female , HCT116 Cells , Humans , In Situ Hybridization , Membrane Potential, Mitochondrial/drug effects , Signal Transduction/drug effects
9.
J Exp Clin Cancer Res ; 36(1): 128, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28903768

ABSTRACT

The increasing use of genomics to define the pattern of actionable mutations and to test and validate new therapies for individual cancer patients, and the growing application of liquid biopsy to dynamically track tumor evolution and to adapt molecularly targeted therapy according to the emergence of tumor clonal variants is shaping modern medical oncology., In order to better describe this new therapeutic paradigm we propose the term "Liquid dynamic medicine" in the place of "Personalized or Precision medicine". Clinical validation of the "Liquid dynamic medicine" approach is best captured by N-of-1 trials where each patient acts as tester and control of truly personalized therapies.


Subject(s)
Biomarkers, Tumor/blood , Neoplasms/pathology , Biomarkers, Tumor/genetics , Clinical Trials as Topic , Genomics , Humans , Liquid Biopsy , Medical Oncology , Mutation , Neoplasms/genetics , Precision Medicine
10.
Ecancermedicalscience ; 11: 747, 2017.
Article in English | MEDLINE | ID: mdl-28690678

ABSTRACT

The beginning of our understanding of the molecular basis of cancer and the discovery in the 1980s of cancer associated genes, oncogenes, and tumour suppressor genes has led to cancer becoming a treatable condition rather than an unspeakable disease. In 1971, the then USA President, Richard Nixon, declared 'war against cancer' with a far too optimistic perspective of winning in just a few years. This tactic failed because our knowledge of the disease was still very limited and even its origin-viral or due to exposure to external agents-was still highly debated. A better understanding of the cause(s) of the origin of cancer led to its definition as a genetic disease at the somatic level and heralded a new era for molecular diagnosis and the development of more mechanistic evidence-based, targeted cancer therapies. However, the initial positive results were soon overshadowed by a major limitation of targeted agents, namely resistance mechanisms, which still represent an obstacle for the full eradication of the disease. More recently, effective therapeutic approaches have been developed in the field of 'immunotherapy'. The combination of novel therapies will hopefully result in effective cancer growth control and make the disease 'chronic'. The launch of the 'Moonshot Cancer Program' by President Barack Obama aims to significantly reduce cancer deaths in the next decade-let us see.

11.
J Exp Clin Cancer Res ; 36(1): 16, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28114961

ABSTRACT

BACKGROUND: Strategies aimed at obtaining a complete cytoreduction are needed to improve long-term survival for patients with colorectal cancer peritoneal carcinomatosis (CRC-pc). METHODS: We established organoid models from peritoneal metastases of two naïve CRC patients. A standard paraffin inclusion was conducted to compare their 3D structure and immunohistochemical profile with that of the corresponding surgical samples. RNA expression levels of the CRC stem cell marker LGR5 was measured by in situ hybridization. The secretome of organoids was profiled by mass spectrometry. Energy homeostasis of organoids was interfered with 4-IPP and metformin. Biochemical and metabolic changes after drug treatments were investigated by western blot and mass spectrometry. Mitochondria impairment was evaluated by electron microscopy and mitotraker staining. RESULTS: The two organoids recapitulated their corresponding clinical samples in terms of 3D structure and immmunoistochemical profile and were positive for the cancer stem cells marker LGR5. Proteomic analyses of organoids highlighted their strong dependence on energy producing pathways, which suggest that their targeting could be an effective therapeutic approach. To test this hypothesis, we treated organoids with two drugs that target metabolism acting on AMP-activated protein kinase (AMPK), the main regulator of cellular energy homeostasis, which may act as metabolic tumour suppressor in CRC. Organoids were treated with 4-IPP, an inhibitor of MIF/CD74 signalling axis which activates AMPK function, or metformin that inhibits mitochondrial respiratory chain complex I. As a new finding we observed that treatment with 4-IPP downregulated AMPK signalling activity, reduced AKT phosphorylation and activated a JNK-mediated stress-signalling response, thus generating mitochondrial impairment and cell death. Metformin treatment enhanced AMPK activation, decreasing the activity of the anabolic factors ribosomal protein S6 and p4EBP-1 and inducing mitochondrial depolarization. CONCLUSION: We provide evidence that the modulation of AMPK activity may be a strategy for targeting metabolism of CRC-pc organoids.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Colonic Neoplasms/metabolism , Histocompatibility Antigens Class II/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Metformin/pharmacology , Peritoneal Neoplasms/secondary , Pyrimidines/pharmacology , AMP-Activated Protein Kinases/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Energy Metabolism/drug effects , Humans , Molecular Targeted Therapy , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/genetics , Proteomics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured
12.
Oncotarget ; 7(29): 45015-45026, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27304187

ABSTRACT

Sunitinib improves the outcomes of patients with solitary fibrous tumours (SFTs). The aim of this study was to investigate and contextualise sunitinib-induced morpho-functional changes in order to gain insights into the drug's mechanism of action.To this end, four surgical specimens obtained from two sunitinib-responsive patients with malignant SFT, and one primary cell culture obtained from fresh tumoral tissue and its stabilised cell line, were studied by means of immunohistochemistry, bright field in situ hybridisation, immunofluorescence/confocal microscopy, and biochemistry.The post-sunitinib surgical samples were characterised by two biologically relevant morpho-functional changes: clear areas and necrotic foci. The first were associated with the attenuation/loss of PDGFRB expression and decreased mTOR signalling, and corresponded to a pathological response. The second were associated with the over-expression of PDGFRB and VEGFA, strong mTOR signalling activation, and the appearance of HIF1α expression, hallmarks of pathological progression. The analysis clearly showed that sunitinib reduces the vascular supply network and inhibits tumoral cells. It also either induces autophagy, thus favouring drug response, or impairs autophagy as a result of lysosome sequestration, thus favouring disease progression. These distinct autophagic events were associated with different myeloid immune contextures. Finally, we also found that PDGFRB is one of the components of a complex that includes Beclin 1 and VPS34.The results of these tissue-based analyses provide new insights into sunitinib's mechanism of action in SFT patients.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Indoles/pharmacology , Pyrroles/pharmacology , Solitary Fibrous Tumors/pathology , Angiogenesis Inhibitors/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Humans , Male , Solitary Fibrous Tumors/drug therapy , Sunitinib , Transcriptome/drug effects
13.
Chin J Cancer ; 35(1): 51, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27267075

ABSTRACT

BACKGROUND: Androgen insensitivity syndrome (AIS), a disorder of sexual development in 46, XY individuals, is caused by loss-of-function mutations in the androgen receptor (AR) gene. A variety of tumors have been reported in association with AIS, but no cases with colorectal cancer (CRC) have been described. CASE PRESENTATION: Here, we present a male patient with AIS who developed multiple early-onset CRCs and his pedigree. His first cousin was diagnosed with AIS and harbored the same AR gene mutation, but with no signs of CRC. The difference in clinical management for the two patients was that testosterone treatment was given to the proband for a much longer time compared with the cousin. The CRC family history was negative, and no germline mutations in well-known CRC-related genes were identified. A single nucleotide polymorphism array revealed a microduplication on chromosome 22q11.22 that encompassed a microRNA potentially related to CRC pathogenesis. In the proband, whole exome sequencing identified a polymorphism in an oncogene and 13 rare loss-of-function variants, of which two were in CRC-related genes and four were in genes associated with other human cancers. CONCLUSIONS: By pathway analysis, all inherited germline genetic events were connected in a unique network whose alteration in the proband, together with continuous testosterone stimulation, may have played a role in CRC pathogenesis.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Colorectal Neoplasms/genetics , Polymorphism, Single Nucleotide , Testosterone/genetics , Adolescent , Adult , Age of Onset , Androgen-Insensitivity Syndrome/complications , Androgen-Insensitivity Syndrome/drug therapy , Chromosomes, Human, Pair 22 , Colorectal Neoplasms/etiology , DNA Copy Number Variations , Exome , Female , Germ-Line Mutation , Humans , Male , Pedigree , Sequence Analysis, DNA , Testosterone/metabolism , Testosterone/pharmacology
14.
Oncotarget ; 7(14): 18594-604, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26919240

ABSTRACT

Expression of miR-342 has been strongly correlated with estrogen receptor (ER) status in breast cancer, where it is highest in ER-positive and lowest in triple-negative tumors. We investigated the effects of miR-342 transfection in the triple-negative breast cancer cell lines MDA-MB-231 and HCC1937, the latter carrying a germ-line BRCA1 mutation. Reconstitution of miR-342 led to caspase-dependent induction of apoptosis only in HCC1937 cells, while overexpression of wild-type BRCA1 in HCC1937 cells counteracted miR-342-mediated induction of apoptosis, suggesting that miR-342 overexpression and the lack of functional BRCA1 result in a synthetic lethal phenotype. Moreover, siRNA-mediated depletion of BRCA1 in MDA-MB-231 cells expressing the wild-type protein led to apoptosis upon transfection with miR-342. Using an in silico approach and a luciferase reporter system, we identified and functionally validated the Baculoviral IAP repeat-containing 6 gene (BIRC6), which encodes the anti-apoptotic factor Apollon/BRUCE, as a target of miR-342. In our model, BIRC6 likely acts as a determinant of the miRNA-dependent induction of apoptosis in BRCA1-mutant HCC1937 cells. Together, our findings suggest a tumor-suppressive function of miR-342 that could be exploited in the treatment of a subset of BRCA1-mutant hereditary breast cancers.


Subject(s)
BRCA1 Protein/genetics , MicroRNAs/biosynthesis , Triple Negative Breast Neoplasms/genetics , Apoptosis/genetics , BRCA1 Protein/metabolism , Cell Line, Tumor , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Phenotype , Transfection , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
16.
Oncotarget ; 6(33): 34629-48, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26431489

ABSTRACT

The incidence of thyroid carcinoma is rapidly increasing. Although generally associated with good prognosis, a fraction of thyroid tumors are not cured by standard therapy and progress to aggressive forms for which no effective treatments are currently available. In order to identify novel therapeutic targets for thyroid carcinoma, we focused on the discovery of genes essential for sustaining the oncogenic phenotype of thyroid tumor cells, but not required to the same degree for the viability of normal cells (non-oncogene addiction paradigm). We screened a siRNA oligonucleotide library targeting the human druggable genome in thyroid cancer BCPAP cell line in comparison with immortalized normal human thyrocytes (Nthy-ori 3-1). We identified a panel of hit genes whose silencing interferes with the growth of tumor cells, while sparing that of normal ones. Further analysis of three selected hit genes, namely Cyclin D1, MASTL and COPZ1, showed that they represent common vulnerabilities for thyroid tumor cells, as their inhibition reduced the viability of several thyroid tumor cell lines, regardless the histotype or oncogenic lesion. This work identified non-oncogenes essential for sustaining the phenotype of thyroid tumor cells, but not of normal cells, thus suggesting that they might represent promising targets for new therapeutic strategies.


Subject(s)
Carcinoma/genetics , Coatomer Protein/genetics , Genes, bcl-1/genetics , High-Throughput Nucleotide Sequencing/methods , Microtubule-Associated Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Thyroid Neoplasms/genetics , Blotting, Western , Carcinoma/pathology , Carcinoma, Papillary , Cell Line, Tumor , Cell Proliferation/genetics , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Humans , Polymerase Chain Reaction , RNA, Small Interfering , Sequence Analysis, RNA/methods , Thyroid Cancer, Papillary , Thyroid Neoplasms/pathology , Transcriptome , Transfection
17.
Oncotarget ; 6(16): 14139-52, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25944621

ABSTRACT

Non-small cell lung cancer (NSCLC) is a leading cause of death worldwide. Targeted monotherapies produce high regression rates, albeit for limited patient subgroups, who inevitably succumb. We present a novel strategy for identifying customized combinations of triplets of targeted agents, utilizing a simplified interventional mapping system (SIMS) that merges knowledge about existent drugs and their impact on the hallmarks of cancer. Based on interrogation of matched lung tumor and normal tissue using targeted genomic sequencing, copy number variation, transcriptomics, and miRNA expression, the activation status of 24 interventional nodes was elucidated. An algorithm was developed to create a scoring system that enables ranking of the activated interventional nodes for each patient. Based on the trends of co-activation at interventional points, combinations of drug triplets were defined in order to overcome resistance. This methodology will inform a prospective trial to be conducted by the WIN consortium, aiming to significantly impact survival in metastatic NSCLC and other malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Precision Medicine/methods , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Molecular Targeted Therapy/methods , Transcriptome
18.
Mod Pathol ; 28(8): 1074-83, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26022454

ABSTRACT

Solitary fibrous tumors, which are characterized by their broad morphological spectrum and unpredictable behavior, are rare mesenchymal neoplasias that are currently divided into three main variants that have the NAB2-STAT6 gene fusion as their unifying molecular lesion: usual, malignant and dedifferentiated solitary fibrous tumors. The aims of this study were to validate molecular and immunohistochemical/biochemical approaches to diagnose the range of solitary fibrous tumors by focusing on the dedifferentiated variant, and to reveal the genetic events associated with dedifferentiation by integrating the findings of array comparative genomic hybridization. We studied 29 usual, malignant and dedifferentiated solitary fibrous tumors from 24 patients (including paired samples from five patients whose tumors progressed to the dedifferentiated form) by means of STAT6 immunohistochemistry and (when frozen material was available) reverse-transcriptase polymerase chain reaction and biochemistry. In addition, the array comparative genomic hybridization findings were used to profile 12 tumors from nine patients. The NAB2/STAT6 fusion was detected in all of the tumors, but immunohistochemistry and western blotting indicated that chimeric protein expression was atypical or absent in 9 out of 11 dedifferentiated tumors. The comparative genomic hybridization results revealed that the usual and malignant solitary fibrous tumors had a simple profile, whereas the genome of the dedifferentiated tumors was complex and unstable, and suggested that 13q and 17p deletions and TP53 mutations may be present in malignant lesions before the full expression of a dedifferentiated phenotype. Solitary fibrous tumor dedifferentiation is associated with the loss of chimeric oncoprotein expression, genomic instability, and cell decommitment and reprogramming. The assessment of dedifferentiated solitary fibrous tumors is based on the presence of the fusion transcripts and, in principle, negative STAT6 immunohistochemistry should not rule out a diagnosis of solitary fibrous tumor.


Subject(s)
Biomarkers, Tumor , Gene Fusion , Genomic Instability , Molecular Diagnostic Techniques , Repressor Proteins , STAT6 Transcription Factor , Solitary Fibrous Tumors/diagnosis , Adult , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Blotting, Western , Chromosome Deletion , Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 17 , Comparative Genomic Hybridization , DNA Mutational Analysis , Diagnosis, Differential , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Male , Middle Aged , Mutation , Phenotype , Predictive Value of Tests , Repressor Proteins/analysis , Repressor Proteins/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , STAT6 Transcription Factor/analysis , STAT6 Transcription Factor/genetics , Solitary Fibrous Tumors/chemistry , Solitary Fibrous Tumors/genetics , Solitary Fibrous Tumors/pathology , Tumor Suppressor Protein p53/genetics , Young Adult
19.
J Cell Physiol ; 230(12): 3105-14, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26012475

ABSTRACT

Loss of response to TGF-ß is a central event in the genesis of colorectal cancer (CRC), a disease that, in the majority cases, is refractory to growth inhibition induced by this cytokine. However, inactivating mutations at receptors and transducers from the TGF-ß cascade occur only in approximately half of CRCs, suggesting the involvement of additional mechanisms altering the response to the cytokine. We have recently described the amplification of the 13q31 locus, where the miR-17-92 cluster maps, associated with overexpression of its members. In this study, we address the potential role of miR-20a, from the miR-17-92 cluster, in the suppression of TGF-ß cytostatic response in CRC. Using the poorly tumorigenic and TGF-ß-sensitive FET cell line that expresses low miR-20a levels, we first confirmed that miR-20a downmodulated CDKN1A expression, both at mRNA and protein level, through direct binding to its 3'-UTR. We demonstrated that miR-20a significantly diminished cell response to TGF-ß by preventing its delay of G1/S transition and promoting progression into cell cycle. Moreover, besides modulating CDKN1A, miR-20a blocked TGF-ß-induced transactivation of its promoter without affecting the post-receptor activation of Smad3/4 effectors directly. Finally, miR-20a abrogated the TGF-ß-mediated c-Myc repression, a direct inhibitor of the CDKN1A promoter activation, most likely by reducing the expression of specific MYC-regulating genes from the Smad/E2F-based core repressor complex. Our experiments indicate that miR-20a interferes with the colonic epithelium homeostasis by disrupting the regulation of Myc/p21 by TGF-ß, which is essential for its malignant transformation.


Subject(s)
Carcinoma/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , MicroRNAs/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , 3' Untranslated Regions , Binding Sites , Caco-2 Cells , Carcinoma/genetics , Carcinoma/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , MicroRNAs/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Transcriptional Activation , Transfection
20.
Orphanet J Rare Dis ; 10: 25, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25887804

ABSTRACT

BACKGROUND: Hereditary medullary thyroid carcinoma (MTC) is caused by germ-line gain of function mutations in the RET proto-oncogene, and a phenotypic variability among carriers of the same mutation has been reported. We recently observed this phenomenon in a large familial MTC (FMTC) family carrying the RET-S891A mutation. Among genetic modifiers affecting RET-driven MTC, a role has been hypothesized for RET-G691S non-synonymous polymorphism, though the issue remains controversial. Aim of this study was to define the in vitro contribution of RET-G691S to the oncogenic potential of the RET-S891A, previously shown to harbour low transforming activity. METHODS: The RET-S891A and RET-G691S/S891A mutants were generated by site-directed mutagenesis, transiently transfected in HEK293T cells and stably expressed in NIH3T3 cells. Their oncogenic potential was defined by assessing the migration ability by wound healing assay and the anchorage-independent growth by soft agar assay in NIH3T3 cells stably expressing either the single or the double mutants. Two RET-S891A families were characterised for the presence of RET-G691S. RESULTS: The functional studies demonstrated that RET-G691S/S891A double mutant displays a higher oncogenic potential than RET-S891A single mutant, assessed by focus formation and migration ability. Moreover, among the 25 RET-S891A carriers, a trend towards an earlier age of diagnosis was found in the MTC patients harboring RET-S891A in association with RET-G691S. CONCLUSIONS: We demonstrate that the RET-G691S non-synonymous polymorphism enhances in vitro the oncogenic activity of RET-S891A. Moreover, an effect on the phenotype was observed in the RET-G691S/S891A patients, thus suggesting that the analysis of this polymorphism could contribute to the decision on the more appropriate clinical and follow-up management.


Subject(s)
Carcinoma, Medullary/congenital , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Animals , Carcinoma, Medullary/genetics , Carcinoma, Medullary/metabolism , Carcinoma, Medullary/pathology , Gene Expression Regulation/physiology , Genomics , HEK293 Cells , Humans , Mice , Multiple Endocrine Neoplasia Type 2a/pathology , Mutation , NIH 3T3 Cells , Penetrance , Polymorphism, Genetic , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...