Tania Fleitas
University of Valencia
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Tania Fleitas.
Oncotarget | 2016
Sarah Derks; Xiaoyun Liao; Anna Maria Chiaravalli; Xinsen Xu; M. Constanza Camargo; Enrico Solcia; Fausto Sessa; Tania Fleitas; Gordon J. Freeman; Scott J. Rodig; Charles S. Rabkin; Adam J. Bass
Gastric cancer (GC) is a deadly disease with limited treatment options. Recent studies with PD-1 inhibition have shown promising results in GC, but key questions remain regarding which GC subclass may respond best. In other cancers, expression of the PD-1 ligand PD-L1 has been shown to identify cancers with greater likelihood of response to PD-1 blockade. We here show with immunohistochemistry that Epstein-Barr Virus (EBV)+ GCs (n = 32) have robust PD-L1 expression not seen in other GCs. In EBV+ GC, we observed PD-L1 staining in tumor cells in 50% (16/32) and immune cells in 94% (30/32) of cases. Among EBV-negative GCs, PD-L1 expression within tumors cells was observed only in cases with microsatellite instability (MSI), although 35% of EBV-/MSS GCs possessed PD-L1 expression of inflammatory cells. Moreover, distinct classes of GC showed different patterns of PD-L1+ immune cell infiltrations. In both EBV+ and MSI tumors, PD-L1+ inflammatory cells were observed to infiltrate the tumor. By contrast, such cells remained at the tumor border of EBV-/MSS GCs. Consistent with these findings, we utilized gene expression profiling of GCs from The Cancer Genome Atlas study to demonstrate that an interferon-γ driven gene signature, an additional proposed marker of sensitivity to PD-1 therapy, were enriched in EBV+ and MSI GC. These data suggest that patients with EBV+ and MSI GC may have greater likelihood of response to PD-1 blockade and that EBV and MSI status should be evaluated as variables in clinical trials of these emerging inhibitors.
Oncotarget | 2015
Maider Ibarrola-Villava; Marta J. Llorca-Cardeñosa; Noelia Tarazona; Cristina Mongort; Tania Fleitas; Jose Alejandro Perez-Fidalgo; Susana Roselló; Samuel Navarro; Gloria Ribas; A. Cervantes
Genetic and epigenetic alterations play an important role in gastric cancer (GC) pathogenesis. Aberrations of the phosphatidylinositol-3-kinase signaling pathway are well described. However, emerging genes have been described such as, the chromatin remodeling gene ARID1A. Our aim was to determine the expression levels of four GC-related genes, ARID1A, CDH1, cMET and PIK3CA, and 14 target-related microRNAs (miRNAs). We compared mRNA and miRNA expression levels among 66 gastric tumor and normal adjacent mucosa samples using quantitative real-time reverse transcription PCR. Moreover, ARID1A, cMET and PIK3CA protein levels were assessed by immunohistochemistry (IHC). Finally, gene and miRNAs associations with clinical characteristics and outcome were also evaluated. An increased cMET and PIK3CA mRNA expression was found in 78.0% (P = 2.20 × 10−5) and 73.8% (P = 1.00 × 10−3) of the tumors, respectively. Moreover, IHC revealed that cMET and PIK3CA expression was positive in 63.6% and 87.8% of the tumors, respectively. Six miRNAs had significantly different expression between paired-samples, finding five up-regulated [miR-223-3p (P = 1.65 × 10−6), miR-19a-3p (P = 1.23 × 10−4), miR-128-3p (P = 3.49 × 10−4), miR-130b-3p (P = 1.00 × 10−3) and miR-34a-5p (P = 4.00 × 10−3)] and one down-regulated [miR-124-3p (P = 0.03)]. Our data suggest that cMET, PIK3CA and target-related miRNAs play an important role in GC and may serve as potential targets for therapy.
Clinical Cancer Research | 2017
Didier Meulendijks; Wolfgang Jacob; Emile E. Voest; Morten Mau-Sørensen; Maria Martinez-Garcia; Álvaro Taus; Tania Fleitas; A. Cervantes; Martijn P. Lolkema; Marlies H.G. Langenberg; Maja J.A. de Jonge; Stefan Sleijfer; Ji-Youn Han; Antonio Calles; Enriqueta Felip; Sang-We Kim; Jan H. M. Schellens; Sabine Wilson; Marlene Thomas; Maurizio Ceppi; Georgina Meneses-Lorente; I. James; Suzana Vega-Harring; Rajiv Dua; Maitram Nguyen; Lori Steiner; Celine Adessi; Francesca Michielin; Birgit Bossenmaier; Martin Weisser
Purpose: This study investigated the safety, clinical activity, and target-associated biomarkers of lumretuzumab, a humanized, glycoengineered, anti-HER3 monoclonal antibody (mAb), in combination with the EGFR-blocking agents erlotinib or cetuximab in patients with advanced HER3-positive carcinomas. Experimental Design: The study included two parts: dose escalation and dose extension phases with lumretuzumab in combination with either cetuximab or erlotinib, respectively. In both parts, patients received lumretuzumab doses from 400 to 2,000 mg plus cetuximab or erlotinib according to standard posology, respectively. The effect of HRG mRNA and HER3 mRNA and protein expression were investigated in a dedicated extension cohort of squamous non–small cell lung cancer (sqNSCLC) patients treated with lumretuzumab and erlotinib. Results: Altogether, 120 patients were treated. One dose-limiting toxicity (DLT) in the cetuximab part and two DLTs in the erlotinib part were reported. The most frequent adverse events were gastrointestinal and skin toxicities, which were manageable. The objective response rate (ORR) was 6.1% in the cetuximab part and 4.2% in the erlotinib part. In the sqNSCLC extension cohort of the erlotinib part, higher tumor HRG and HER3 mRNA levels were associated with a numerically higher disease control rate but not ORR. Conclusions: The toxicity profile of lumretuzumab in combination with cetuximab and erlotinib was manageable, but only modest clinical activity was observed across tumor types. In the sqNSCLC cohort, there was no evidence of meaningful clinical benefit despite enriching for tumors with higher HRG mRNA expression levels. Clin Cancer Res; 23(18); 5406–15. ©2017 AACR.
Oncotarget | 2016
Maider Ibarrola-Villava; Tania Fleitas; Marta J. Llorca-Cardeñosa; Cristina Mongort; Elisa Alonso; Samuel Navarro; Octavio Burgués; Ana Vivancos; Juan Miguel Cejalvo; Jose Alejandro Perez-Fidalgo; Susana Roselló; Gloria Ribas; A. Cervantes
Somatic mutation analysis represents a useful tool in selecting personalized therapy. The aim of our study was to determine the presence of common genetic events affecting actionable oncogenes using a MassARRAY technology in patients with advanced solid tumors who were potential candidates for target-based therapies. The analysis of 238 mutations across 19 oncogenes was performed in 197 formalin-fixed paraffin-embedded samples of different tumors using the OncoCarta Panel v1.0 (Sequenom Hamburg, Germany). Of the 197 specimens, 97 (49.2%) presented at least one mutation. Forty-nine different oncogenic mutations in 16 genes were detected. Mutations in KRAS and PIK3CA were detected in 40/97 (41.2%) and 30/97 (30.9%) patients respectively. Thirty-one patients (32.0%) had mutations in two genes, 20 of them (64.5%) initially diagnosed with colorectal cancer. The co-occurrence of mutation involved mainly KRAS, PIK3CA, KIT and RET. Mutation profiles were validated using a customized panel and the Junior Next-Generation Sequencing technology (GS-Junior 454, Roche). Twenty-eight patients participated in early clinical trials or received specific treatments according to the molecular characterization (28.0%). MassARRAY technology is a rapid and effective method for identifying key cancer-driving mutations across a large number of samples, which allows for a more appropriate selection for personalized therapies.
Oncotarget | 2016
Marta J. Llorca-Cardeñosa; Tania Fleitas; Maider Ibarrola-Villava; Maria Peña-Chilet; Cristina Mongort; Carolina Martínez-Ciarpaglini; Lara Navarro; Valentina Gambardella; Josefa Castillo; Susana Roselló; Samuel Navarro; Gloria Ribas; A. Cervantes
Gastric cancer (GC) pathogenesis involves genetic, epigenetic and environmental factors. Epigenetic alterations, such as DNA methylation are considered pivotal in the inactivation of tumor-related genes. We assessed a methylation panel of 5 genes to study their association to GC progression and microsatellite instability (MSI), and studied the role of RUNX3 in GC pathogenesis and the tumor immune microenvironment. The methylation status of 47 promoter-CpG islands was studied through MALDI-TOF mass spectrometry analysis in 35 Microsatellite stable (MSS) GC, 26 MSI, and 18 cancer-free samples (CFS), and 6 MSS GC and 4 MSI GC cell lines. We also studied RUNX3 expression by immunohistochemistry (IHC) in 40 samples, and validated differences in methylation levels between tumor, normal, and immune tissue in 14 additional samples. Unsupervised hierarchical clustering of methylation levels revealed no distinct subgroups between MSI and MSS samples or cell lines. CFSs clustered together showing higher levels of RUNX3 methylation compared to GC samples. RUNX3 showed protein silencing in cancer and normal mucosa, compared to inflammatory peritumoural infiltrate in almost all cases, showing a non-lymphocytic predominant pattern and being correlated with epigenetic silencing. Our results show aberrant promoters methylation in APC, CDH1, CDKN2A, MLH1 and RUNX3 associated with GC, as well as a non-lymphocytic predominant infiltrate with high expression of RUNX3. Deep study of RUNX3 inflammation signaling could help in understanding inflammation and immune activation in the tumor microenvironment.
Annals of Oncology | 2014
U N Lassen; A. Cervantes Ruipérez; Tania Fleitas; Didier Meulendijks; Jan H. M. Schellens; M. Lolkemar; M.J.A. de Jonge; Stefan Sleijfer; Morten Mau-Soerensen; Álvaro Taus; Celine Adessi; A. Keelara; Francesca Michielin; Birgit Bossenmaier; Georgina Meneses-Lorente; Ian James; Wolfgang Jacob; Martin Weisser; Maria Martinez-Garcia
ABSTRACT Aim: To evaluate the safety profile of RG7116 in combination with cetuximab or erlotinib. Methods: Patients (pts) with advanced or metastatic carcinomas with centrally confirmed HER3 protein expression were included. RG7116 plus cetuximab (400 mg/m2 followed by 250 mg/m2 qw IV) and RG7116 plus erlotinib (150 mg/day p.o.) combinations were evaluated in a dose escalation study with “3 + 3” design at a starting dose of 400 mg IV of RG7116 in a q2w regimen. Results: Twenty-seven pts were enrolled in 5 cohorts (400 to 2000 mg) in the cetuximab arm. One dose-limiting toxicity (DLT) of grade 3 dehydration was reported in the 800-mg cohort. Twenty-seven pts were enrolled in 4 cohorts (400 to 2000 mg) in the erlotinib arm. One DLT was reported in the 1600-mg cohort (grade 3 diarrhea and grade 3 hypokalemia) and one DLT was reported in the 2000-mg cohort (grade 3 blood bilirubin increase). No maximum tolerated dose was reached. The most frequently reported adverse events of any grade were diarrhea (78%) and rash (59%) for the cetuximab arm and diarrhea (82%) and decreased appetite (48%) for the erlotinib arm. In the erlotinib arm treatment-related grade 3 diarrhea was observed more frequently at higher doses of RG7116 (400 mg: 0%; 800 mg: 17%; 1600 mg: 43%; 2000 mg: 33%). Overall, infusion-related reactions related to RG7116 occurred in 11% of pts. Two of these were grade 3 (4%). The pharmacokinetic profile of RG7116 in combination with cetuximab and erlotinib was comparable to that in the monotherapy setting (Meulendijks et al. J Clin Oncol 31, 2013 suppl; abstr 2522). HER3 membranous protein down-regulation was observed from 400 mg onwards in on-treatment tumor and skin tissue. In the cetuximab arm, two pts with colorectal carcinoma had a confirmed partial response (PR). In the erlotinib arm, one patient with ovarian carcinoma had a confirmed PR. Metabolic PR on FDG-PET occurred in 42% of pts in the cetuximab arm and in 28% of pts in the erlotinib arm. Conclusions: RG7116 combination with cetuximab or erlotinib was well tolerated, and demonstrated preliminary signs of clinical activity. Disclosure: U.N. Lassen: discloses Research grants from Roche; A. Cervantes Ruiperez: discloses Research grants, advisory boards and lectures for Roche; C. Adessi, A. Keelara, F. Michielin, B. Bossenmaier, G. Meneses-Lorente, I. James, W. Jacob and M. Weisser: Employee of Roche. All other authors have declared no conflicts of interest.
Cancer Treatment Reviews | 2016
Tania Fleitas; Maider Ibarrola-Villava; Gloria Ribas; A. Cervantes
This article will review the impact of the recently developed MassARRAY technology on our understanding of cancer biology and treatment. Analysis of somatic mutations is a useful tool in selecting personalized therapy, and for predicting the outcome of many solid tumors. Here, we review the literature on the application of MassARRAY technology (Sequenom Hamburg, Germany) to determine the mutation profile of solid tumors from patients. We summarize the use of commercially available panels of mutations - such as OncoCarta™ or other combinations - and their concordance with results obtained by using other technologies, such as next generation sequencing.
European Journal of Cancer | 2015
Maider Ibarrola-Villava; Marta J. Llorca-Cardeñosa; Noelia Tarazona; Cristina Mongort; Tania Fleitas; Jose Alejandro Perez-Fidalgo; Susana Roselló; Samuel Navarro; Gloria Ribas; A. Cervantes
Genetic and epigenetic alterations play an important role in gastric cancer (GC) pathogenesis. Aberrations of the phosphatidylinositol-3-kinase signaling pathway are well described. However, emerging genes have been described such as, the chromatin remodeling gene ARID1A. Our aim was to determine the expression levels of four GCrelated genes, ARID1A, CDH1, cMET and PIK3CA, and 14 target-related microRNAs (miRNAs). We compared mRNA and miRNA expression levels among 66 gastric tumor and normal adjacent mucosa samples using quantitative real-time reverse transcription PCR. Moreover, ARID1A, cMET and PIK3CA protein levels were assessed by immunohistochemistry (IHC). Finally, gene and miRNAs associations with clinical characteristics and outcome were also evaluated. An increased cMET and PIK3CA mRNA expression was found in 78.0% (P = 2.20 × 10 −5 ) and 73.8% (P = 1.00 × 10 −3 ) of the tumors, respectively. Moreover, IHC revealed that cMET and PIK3CA expression was positive in 63.6% and 87.8% of the tumors, respectively. Six miRNAs had significantly different expression between paired-samples, finding five up-regulated [miR-223-3p ( P = 1.65 × 10 −6 ), miR-19a3p (P = 1.23 × 10 −4 ), miR-128-3p (P = 3.49 × 10 −4 ), miR-130b-3p (P = 1.00 × 10 −3
Annals of Oncology | 2014
Tania Fleitas; M. Ibarrola-Villava; N. Tarazona; R. Cervera; C. Mongort; S. Navarro; Susana Roselló; G. Ribas; Jose Alejandro Perez-Fidalgo; A. Cervantes Ruipérez
ABSTRACT Aim: Gene expression analysis has gained interest as a tool for classifying gastric cancer. We studied patterns of expression of ARID1A, CDH1, cMET and PIK3CA in primary gastric tumors. A correlation of these genes with HER2 and HER3 was also analyzed. Methods: A retrospective case-control study was done with normal gastric tissue as control. Real-time-quantitative PCR was used to examine gene expression. HER2 and HER3 status were determined by immunohistochemistry. Results: 48 gastric patients were recruited of whom 22 had normal gastric tissue available. cMET and PIK3CA were significantly overexpressed in tumor versus normal tissue. ARID1A and CDH1 expression showed no differences. In tumor tissue, expression of cMET and PIK3CA were inversely correlated with ARID1A (Odds ratio (OR)= 0.52, p = 0.002 and OR = 0.45, p = 0.008 respectively), as well as cMET and CDH1 (OR = 0.59, p Conclusions: cMET and PIK3CA genes are overexpressed in gastric tumors independently of HER2 and HER3 overexpression. *Dr. Ibarrola-Villava M. contributed equally to this work as the presenter author. **Dr. Tarazona N.is currently working at the Gastrointestinal Cancer Unit of the Royal Marsden Hospital as an ESMO Traslational Reasearch Fellow. Disclosure: All authors have declared no conflicts of interest.
Journal of Clinical Oncology | 2016
Sarah Derks; Xiaoyun Liao; Xinsen Xu; M. Constanza Camargo; Anna Maria Chiaravalli; Enrico Solcia; Tania Fleitas; Gordon J. Freeman; Scott J. Rodig; Charles S. Rabkin; Adam J. Bass