Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Francesca Simionato is active.

Publication


Featured researches published by Francesca Simionato.


PLOS ONE | 2012

True 3q Chromosomal Amplification in Squamous Cell Lung Carcinoma by FISH and aCGH Molecular Analysis: Impact on Targeted Drugs

Matteo Brunelli; Emilio Bria; Alessia Nottegar; Sara Cingarlini; Francesca Simionato; Anna Caliò; Albino Eccher; Claudia Parolini; Antonio Iannucci; Eliana Gilioli; Serena Pedron; Francesco Massari; Giampaolo Tortora; Ioana Borze; Sakari Knuutila; Stefano Gobbo; Antonio Santo; Luca Tondulli; Francesco Calabrò; Guido Martignoni; Marco Chilosi

Squamous lung carcinoma lacks specific “ad hoc” therapies. Amplification of chromosome 3q is the most common genomic aberration and this region harbours genes having role as novel targets for therapeutics. There is no standard definition on how to score and report 3q amplification. False versus true 3q chromosomal amplification in squamous cell lung carcinoma may have tremendous impact on trials involving drugs which target DNA zones mapping on 3q. Forty squamous lung carcinomas were analyzed by FISH to assess chromosome 3q amplification. aCGH was performed as gold-standard to avoid false positive amplifications. Three clustered patterns of fluorescent signals were observed. Eight cases out of 40 (20%) showed ≥8 3q signals. Twenty out of 40 (50%) showed from 3 to 7 signals. The remaining showed two fluorescent signals (30%). When corrected by whole chromosome 3 signals, only cases with ≥8 signals maintained a LSI 3q/CEP3 ratio >2. Only the cases showing 3q amplification by aCGH (+3q25.3−3q27.3) showed ≥8 fluorescent signals at FISH evidencing a 3q/3 ratio >2. The remaining cases showed flat genomic portrait at aCGH on chromosome 3. We concluded that: 1) absolute copy number of 3q chromosomal region may harbour false positive interpretation of 3q amplification in squamous cell carcinoma; 2) a case results truly “amplified for chromosome 3q” when showing ≥8 fluorescent 3q signals; 3) trials involving drugs targeting loci on chromosome 3q in squamous lung carcinoma therapy have to consider false versus true 3q chromosomal amplification.


Journal of Thoracic Oncology | 2014

ALK/EML4 Fusion Gene May Be Found in Pure Squamous Carcinoma of the Lung

Anna Caliò; Alessia Nottegar; Eliana Gilioli; Emilio Bria; Sara Pilotto; Umberto Peretti; Stefania Kinspergher; Francesca Simionato; Serena Pedron; Sakari Knuutila; Giampaolo Tortora; Albino Eccher; Antonio Santo; Luca Tondulli; Giorgio Inghirami; Fabrizio Tabbò; Guido Martignoni; Marco Chilosi; Aldo Scarpa; Matteo Brunelli

Introduction: The report of cases of lung squamous cell cancers harboring anaplastic lymphoma kinase (ALK) gene rearrangements raises the question whether this histologic subtype should be also evaluated for such molecular predictive test. Methods: A consecutive series of 40 lung pure squamous cell carcinomas were analyzed for ALK gene status by fluorescence in situ hybridization. Squamous differentiation was validated using an immunohistochemical panel including n-p63 (p40), cytokeratin (CK) 5/6, sex-determining region Y (SRY)-Box2 (SOX2), thyroid transcription factor 1, CK7, and Napsin-A. Results: Squamous differentiation was confirmed in all tumors as they stained positive for n-p63 and CK5/6 and negative for thyroid transcription factor 1 and Napsin-A. One of 40 cases (2.5%) showed an ALK rearrangement on fluorescence in situ hybridization analysis. Conclusions: ALK translocation may be found in lung pure squamous cell carcinomas. Our data suggest the opportunity to test ALK rearrangements on biopsy samples harboring squamous cell cancer differentiation.


Clinical Cancer Research | 2016

An FGFR3 autocrine loop sustains acquired resistance to trastuzumab in gastric cancer patients

Geny Piro; Carmine Carbone; Ivana Cataldo; Federica Di Nicolantonio; Simone Giacopuzzi; Giuseppe Aprile; Francesca Simionato; Federico Boschi; Marco Zanotto; Maria Mihaela Mina; Raffaela Santoro; Valeria Merz; Andrea Sbarbati; Giovanni de Manzoni; Aldo Scarpa; Giampaolo Tortora; Davide Melisi

Purpose: The majority of gastric cancer patients who achieve an initial response to trastuzumab-based regimens develop resistance within 1 year of treatment. This study was aimed at identifying the molecular mechanisms responsible for resistance. Experimental Design: A HER2+-trastuzumab sensitive NCI-N87 gastric cancer orthotopic nude mouse model was treated with trastuzumab until resistance emerged. Differentially expressed transcripts between trastuzumab-resistant and sensitive gastric cancer cell lines were annotated for functional interrelatedness by Ingenuity Pathway Analysis software. Immunohistochemical analyses were performed in pretreatment versus posttreatment biopsies from gastric cancer patients receiving trastuzumab-based treatments. All statistical tests were two-sided. Results: Four NCI-N87 trastuzumab-resistant (N87-TR) cell lines were established. Microarray analysis showed HER2 downregulation, induction of epithelial-to-mesenchymal transition, and indicated fibroblast growth factor receptor 3 (FGFR3) as one of the top upregulated genes in N87-TR cell lines. In vitro, N87-TR cell lines demonstrated a higher sensitivity than did trastuzumab-sensitive parental cells to the FGFR3 inhibitor dovitinib, which reduced expression of pAKT, ZEB1, and cell migration. Oral dovitinib significantly (P = 0.0006) reduced tumor burden and prolonged mice survival duration in N87-TR mouse models. A higher expression of FGFR3, phosphorylated AKT, and ZEB1 were observed in biopsies from patients progressing under trastuzumab-based therapies if compared with matched pretreatment biopsies. Conclusions: This study identified the FGFR3/AKT axis as an escape pathway responsible for trastuzumab resistance in gastric cancer, thus indicating the inhibition of FGFR3 as a potential strategy to modulate this resistance. Clin Cancer Res; 22(24); 6164–75. ©2016 AACR.


OncoImmunology | 2017

A circulating TH2 cytokines profile predicts survival in patients with resectable pancreatic adenocarcinoma

Geny Piro; Francesca Simionato; Carmine Carbone; Melissa Frizziero; Giuseppe Malleo; Silvia Zanini; Raffaella Casolino; Raffaela Santoro; Maria Mihaela Mina; Camilla Zecchetto; Valeria Merz; Aldo Scarpa; Claudio Bassi; Giampaolo Tortora; Davide Melisi

ABSTRACT Surgery is the only potentially curative option for patients with pancreatic ductal adenocarcinoma (PDAC), but metastatic relapse remains common. We hypothesized that the expression levels of inflammatory cytokines could predict recurrence of PDAC, thus allowing to select patients who most likely could benefit from surgical resection. We prospectively collected plasma at diagnosis from 287 patients with pancreatic resectable neoplasms. The expression levels of 23 cytokines were measured in 90 patients with PDAC by using a multiplex analyte profiling assay. Levels higher than cutoff identified of the TH2 cytokines interleukin (IL)4, IL5, IL6 of macrophage inflammatory protein (MIP)1α, granulocyte-macrophage colony-stimulating factor (GM-CSF), and monocyte chemoattractant protein (MCP)1, and of IL17α, IFNγ-induced protein (IP)10, and IL1b were significantly associated with a shorter median OS. In particular, levels of IL4 and IP10 higher than cutoff identified, and level of TH1 cytokines TNFα and INFγ, and of IL9 and IL1Rα lower than cutoff identified were significantly associated with a shorter DFS. In the multivariate analysis, high IP10 was confirmed as negatively associated with OS (HR = 3.097, p = 0.014) and IL4 and TNFα remain negatively (HR = 2.75, p = 0.002) and positively (HR = 0.224, p = 0.049) associated with DFS, respectively. Simultaneous expression of low IL4 and high TNFα identified patients with best prognosis (HR = 0.313, p < 0.0001). In conclusion, we demonstrated that, among a series of cytokines, IL4 is the most significant independent prognostic factor for DFS in resectable PDAC patients, and it could be useful to select patients with high risk of early recurrence who may avoid an unnecessary resection.


Clinical Cancer Research | 2017

Homeobox B9 mediates resistance to anti-VEGF therapy in colorectal cancer patients

Carmine Carbone; Geny Piro; Francesca Simionato; Francesca Ligorio; Chiara Cremolini; Fotios Loupakis; Greta Alì; Daniele Rossini; Valeria Merz; Raffaela Santoro; Camilla Zecchetto; Marco Zanotto; Federica Di Nicolantonio; Alberto Bardelli; Gabriella Fontanini; Giampaolo Tortora; Davide Melisi

Purpose: The identification of predictive biomarkers for antiangiogenic therapies remains an unmeet need. We hypothesized that the transcription factor Homeobox B9 (HOXB9) could be responsible for the tumor resistance to the anti-VEGF agent bevacizumab. Experimental Design: HOXB9 expression and activation were measured in eight models of colorectal and pancreatic cancer with different resistance to bevacizumab. Serum levels of Angiopoietin-like Protein (Angptl)2, CXC receptor ligand (CXCL)1, IL8, and TGFβ1 in tumor-bearing mice were measured by multiplex xMAP technology. HOXB9 expression was measured by immunohistochemical analysis in 81 pretreatment specimens from metastatic colorectal cancer patients. Differences in progression-free survival (PFS) were determined using a log-rank test. Results: HOXB9-positive tumors were resistant to bevacizumab, whereas mice bearing HOXB9-negative tumors were cured by this agent. Silencing HOXB9 in bevacizumab-resistant models significantly (P < 0.05) reduced Angptl2, CXCL1, IL8, and TGFβ1 levels, reverted their mesenchymal phenotype, reduced CD11b+ cells infiltration, and restored, in turn, sensitivity to bevacizumab. HOXB9 had no prognostic value in patients treated with a first-line chemotherapeutic regimen noncontaining bevacizumab. However, patients affected by an HOXB9-negative tumor had a significantly longer PFS compared with those with an HOXB9-positive tumor if treated with a first-line regimen containing bevacizumab (18.0 months vs. 10.4 months; HR 2.037; 95% confidence interval, 1.006–4.125; P = 0.048). Conclusions: These findings integrate the complexity of numerous mechanisms of anti-VEGF resistance into the single transcription factor HOXB9. Silencing HOXB9 could be a promising approach to modulate this resistance. Our results candidate HOXB9 as predictive biomarker for selecting colorectal cancer patients for antiangiogenic therapy. Clin Cancer Res; 23(15); 4312–22. ©2017 AACR.


International Journal of Obesity | 2018

Adipocytes sustain pancreatic cancer progression through a non-canonical WNT paracrine network inducing ROR2 nuclear shuttling.

Carmine Carbone; Geny Piro; N Gaianigo; Francesca Ligorio; Raffaela Santoro; Valeria Merz; Francesca Simionato; Camilla Zecchetto; Geppino Falco; G Conti; P T Kamga; M Krampera; F Di Nicolantonio; L De Franceschi; Aldo Scarpa; Giampaolo Tortora; Davide Melisi

Background:Solid epidemiological evidences connect obesity with incidence, stage and survival in pancreatic cancer. However, the underlying mechanistic basis linking adipocytes to pancreatic cancer progression remain largely elusive. We hypothesized that factors secreted by adipocytes could be responsible for epithelial-to-mesenchymal transition (EMT) induction and, in turn, a more aggressive phenotype in models of pancreatic preneoplastic lesions.Methods:We studied the role of factors secreted by two adipogenic model systems from primary human bone marrow stromal cells (hBMSCs) in an in vitro experimental cell transformation model system of human pancreatic ductal epithelial (HPDE) cell stably expressing activated KRAS (HPDE/KRAS),Results:We measured a significant induction of EMT and aggressiveness in HPDE and HPDE/KRAS cell lines when cultured with medium conditioned by fully differentiated adipocytes (ADIPOCM) if compared with the same cells cultured with medium conditioned by hBMSC (hBMSCCM) from two different healthy donors. Several genes coding for soluble modulators of the non-canonical WNT signaling pathway, including FRZB, SFRP2, RSPO1, WNT5A and 5B were significantly overexpressed in fully differentiated adipocytes than in their respective in hBMSC. ADIPOCM induced the overexpression and the nuclear translocation of the Frizzled family member receptor tyrosine kinase-like orphan receptor (Ror) 2 in HPDE and HPDE/KRAS cells. Vantictumab, an anti-Frizzled monoclonal antibody, reduced ROR2 nuclear translocation and in turn the EMT and aggressiveness in HPDE and HPDE/KRAS cells.Conclusions:We demonstrated that adipocytes could induce EMT and aggressiveness in models of pancreatic preneoplastic lesions by orchestrating a complex paracrine signaling of soluble modulators of the non-canonical WNT signaling pathway that determine, in turn, the activation and nuclear translocation of ROR2. This signaling pathway could represent a novel target for pancreatic cancer chemoprevention. Most importantly, these factors could serve as novel biomarkers to select a risk population among obese subjects for screening and, thus, early diagnosis of pancreatic cancer.


International Journal of Molecular Sciences | 2018

Angiopoietin-Like Proteins in Angiogenesis, Inflammation and Cancer

Carmine Carbone; Geny Piro; Valeria Merz; Francesca Simionato; Raffaela Santoro; Camilla Zecchetto; Giampaolo Tortora; Davide Melisi

Altered expression of secreted factors by tumor cells or cells of the tumor microenvironment is a key event in cancer development and progression. In the last decade, emerging evidences supported the autocrine and paracrine activity of the members of the Angiopoietin-like (ANGPTL) protein family in angiogenesis, inflammation and in the regulation of different steps of carcinogenesis and metastasis development. Thus, ANGPTL proteins become attractive either as prognostic or predictive biomarkers, or as novel target for cancer treatment. Here, we outline the current knowledge about the functions of the ANGPTL proteins in angiogenesis, cancer progression and metastasis. Moreover, we discuss the most recent evidences sustaining their role as prognostic or predictive biomarkers for cancer therapy. Although the role of ANGPTL proteins in cancer has not been fully elucidated, increasing evidence suggest their key effects in the proliferative and invasive properties of cancer cells. Moreover, given the common overexpression of ANGPTL proteins in several aggressive solid tumors, and their role in tumor cells and cells of the tumor microenvironment, the field of research about ANGPTL proteins network may highlight new potential targets for the development of future therapeutic strategies.


Expert Review of Anticancer Therapy | 2018

The development of PARP as a successful target for cancer therapy

Roberto Ferrara; Francesca Simionato; Chiara Ciccarese; Elisabetta Grego; Sara Cingarlini; Roberto Iacovelli; Emilio Bria; Giampaolo Tortora; Davide Melisi

ABSTRACT Introduction: PARP1 and BRCA genes are essential genome caretakers and their interaction has been the first example of synthetic lethality, a genetic concept proposed in the early 20th century, but deeply explored in cancer patients only in the last decade. Areas covered: This review describes PARP1 and BRCA main functions and different roles in genome protection. Furthermore, an overview of the principle mechanisms of action and resistance to PARP inhibitors (PARPi) is presented. This review illustrates the concept of BRCAness, and how this discovery has broadened the routes of PARPi to several different malignancies such as ovarian, breast and prostate cancer. Finally, an insight is provided into the key data of PARPi in these distinctive clinical settings. Expert commentary: PARP inhibition could be a new therapeutic option for a number of tumors in the near future. However, several aspects will be of paramount interest for future investigations, including the molecular bases for PARPi synthetic lethality, the DNA repair independent functions of PARP and BRCA genes, the resistance and biomarkers of response to PARP inhibition, and the mechanisms of interaction between PARPi and antiangiogenic or immunotherapeutic agents.


Anticancer Research | 2018

CT Texture Analysis of Ductal Adenocarcinoma Downstaged After Chemotherapy

Valentina Ciaravino; Nicolò Cardobi; Riccardo De Robertis; Paola Capelli; Davide Melisi; Francesca Simionato; Giovanni Marchegiani; Roberto Salvia; Mirko D'Onofrio

Background/Aim: Re-staging of ductal adenocarcinoma using computed tomography (CT) scan can be problematic so new imaging techniques and evaluation parameters are required. The aim of the study was to evaluate the added value of CT texture analysis in estimation of tissue changes in ductal adenocarcinoma downsized and resected after chemotherapy. Materials and Methods: Patients with ductal adenocarcinoma downstaged after neoadjuvant treatment, and resected, were included. A pre- and post-treatment CT was obtained. In comparison, patients with disease progression were included for texture analysis evaluation at CT pre- and post-treatment. CT texture analysis results were compared. Results: A total of 17 patients affected by un-resectable or borderline ductal adenocarcinoma reached the resectable stage after treatment. The comparison between Kurtosis pre- and Kurtosis post-treatment showed a statistically significant difference. On the contrary, in the comparison group composed of 14 patients with disease progression there was no statistical difference regarding this parameter. Conclusion: This evaluation may represent an added value in tumor tissue changes judgment and can be extremely useful to diagnose downstaging in those cases with no evident downsizing after chemotherapy.


Archive | 2016

Resistance to ALK Inhibitors

Francesca Simionato; Carmine Carbone; Giampaolo Tortora; Davide Melisi

The discovery of specific driver genetic alterations has led to the development of more tailored approaches for advanced human malignancies, moving short steps forward in the cure of these lethal diseases. Among them, rearrangements of the anaplastic lymphoma kinase (ALK) gene are key drivers in the carcinogenesis of a portion of anaplastic large cell lymphomas (ALCL) and non-small cell lung cancer (NSCLC). Many molecules targeting these specific rearrangements have been developed in preclinical models and clinical studies. Among these, crizotinib, an oral small-molecule tyrosine kinase inhibitor targeting ALK, MET, and ROS1 tyrosine kinases demonstrated a significant clinical activity in patients with ALK-positive tumors and, thus, achieved the US Food and Drug Administration (FDA) approval for the treatment of advanced NSCLC harboring ALK-rearrangements. Despite initially responses in most patients, acquired resistance to crizotinib arises unavoidably often within the first year of treatment. To overcome the acquired resistance more potent ALK inhibitors have been developed and tested in clinical trials with encouraging activity results. In this review, we discuss new findings about molecular mechanisms of crizotinib resistance and novel therapeutic strategies to address crizotinib resistance.

Collaboration


Dive into the Francesca Simionato's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emilio Bria

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge