Network


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

Hotspot


Dive into the research topics where Álvaro Taus is active.

Publication


Featured researches published by Álvaro Taus.


Clinical Cancer Research | 2014

Targeting Epithelial-to-Mesenchymal Transition with Met Inhibitors Reverts Chemoresistance in Small Cell Lung Cancer

Israel Cañadas; Federico Rojo; Álvaro Taus; Oriol Arpí; Montserrat Arumi-Uria; Lara Pijuan; Silvia Menendez; Sandra Zazo; Manuel Domine; Marta Salido; Sergio Mojal; Antonio García de Herreros; Ana Rovira; Joan Albanell; Edurne Arriola

Purpose: Met receptor phosphorylation is associated with poor prognosis in human small cell lung cancer (SCLC). The aim of our work was to investigate the effects of hepatocyte growth factor (HGF)/Met–mediated epithelial-to-mesenchymal transition (EMT) in SCLC and to evaluate the role of Met inhibition in mesenchymal/chemorefractory SCLC models. Experimental Design: SCLC models of HGF-induced EMT were evaluated in vitro and in vivo (subcutaneous xenografts in BALB/c nude mice) for chemosensitivity and response to Met inhibition with PF-2341066 (crizotinib). Human SCLC samples at diagnosis (N = 87) and relapse (N = 5) were evaluated by immunohistochemistry and immunofluorescence for EMT markers and Met status and these were correlated with patient outcome. Results: We identified that the activation of the Met receptor through HGF induced expression of mesenchymal markers, an aggressive phenotype, and chemoresistance. Blockade of this process with the Met inhibitor resensitized cells to chemotherapy in vitro and in vivo. Moreover, mesenchymal markers in human SCLC specimens were associated with Met activation, predicted worse survival, and were upregulated in chemorefractory disease. Conclusion: These results provide novel evidence on an important role of Met-dependent EMT in the adverse clinical behavior of SCLC and support clinical trials of Met inhibitors and chemotherapy in this fatal disease. Clin Cancer Res; 20(4); 938–50. ©2013 AACR.


Clinical Cancer Research | 2016

First-in-Human Phase I Study of Lumretuzumab, a Glycoengineered Humanized Anti-HER3 Monoclonal Antibody, in Patients with Metastatic or Advanced HER3-Positive Solid Tumors.

Didier Meulendijks; Wolfgang Jacob; Maria Martinez-Garcia; Álvaro Taus; Martijn P. Lolkema; Emile E. Voest; Marlies H.G. Langenberg; Tania Fleitas Kanonnikoff; A. Cervantes; Maja J.A. de Jonge; Stefan Sleijfer; Morten Mau Soerensen; Marlene Thomas; Maurizio Ceppi; Georgina Meneses-Lorente; Ian James; Celine Adessi; Francesca Michielin; Keelara Abiraj; Birgit Bossenmaier; Jan H. M. Schellens; Martin Weisser; Ulrik Niels Lassen

Purpose: A first-in-human phase I study was conducted to characterize safety, efficacy, and pharmacokinetic (PK) and pharmacodynamic (PD) properties of lumretuzumab, a humanized and glycoengineered anti-HER3 monoclonal antibody, in patients with advanced cancer. Experimental Design: Twenty-five patients with histologically confirmed HER3-expressing tumors received lumretuzumab (100, 200, 400, 800, 1,600, and 2,000 mg) every two weeks (q2w) in 3+3 dose-escalation phase. In addition, 22 patients were enrolled into an extension cohort at 2,000 mg q2w. Results: There were no dose-limiting toxicities. Common adverse events (any grade) included diarrhea (22 patients, 46.8%), fatigue (21 patients, 44.7%), decreased appetite (15 patients, 31.9%), infusion-related reactions (13 patients, 27.7%), and constipation (10 patients, 21.3%). The peak concentration (Cmax) and area under the concentration–time curve up to the last measurable concentration (AUClast) of lumretuzumab increased more than dose proportionally from 100 mg up to 400 mg. Linear PK was observed with doses ≥400 mg q2w indicating target-mediated drug disposition saturation. Downregulation of HER3 membranous protein was observed in on-treatment tumor biopsies from 200 mg, and was maximal at and above 400 mg. An ex vivo assay demonstrated increased activation potential of peripheral NK lymphocytes with lumretuzumab compared with a non-glycoengineered anti-HER3 antibody. Ten patients (21.3%) had stable disease and remained on study at a median of 111 days (range, 80–225 days). Conclusions: Lumretuzumab was well tolerated and showed evidence of clinical activity. Linear serum PK properties and plateauing of PD effects in serial tumor biopsies indicate optimal biologically active doses of lumretuzumab from 400 mg onwards. Clin Cancer Res; 22(4); 877–85. ©2015 AACR.


Clinical Lung Cancer | 2017

Heterogeneity of Tumor and Immune Cell PD-L1 Expression and Lymphocyte Counts in Surgical NSCLC Samples

David Casadevall; Sergi Clavé; Álvaro Taus; Max Hardy-Werbin; Pedro Rocha; Marta Lorenzo; Silvia Menendez; Marta Salido; Joan Albanell; Lara Pijuan; Edurne Arriola

Background Immune‐checkpoint inhibitors against programmed cell death protein 1 (PD‐1)/programmed death‐ligand 1 (PD‐L1) have shown remarkable therapeutic activity in non–small‐cell lung cancer (NSCLC). However, biomarker‐based patient selection remains a challenge. Our aim was to assess the heterogeneity of various immune markers between different tumor areas of surgically resected NSCLC specimens. Materials and Methods We included 94 adenocarcinoma (ADC) and 50 squamous cell carcinoma (SCC) specimens. Two distinct tumor areas of each tumor sample were selected and incorporated into tissue microarrays. PD‐L1 expression in tumor cells (TCs) and immune cells (ICs) was assessed using clone SP142 (Ventana). PDL1 gene amplification was assessed using fluorescence in situ hybridization. CD3 and CD8 densities were quantified using digital image‐based analysis. Heterogeneity was assessed using kappa agreement index (KI) and intraclass correlation coefficient. Results Prevalence of PD‐L1 expression was 16.8% in TCs and 27.8% in ICs. Eleven tumors (7.6%) showed PDL1 amplification. In ADC, KI of PD‐L1 TC and IC expression between cores was 0.465 and 0.260, compared with 0.274 and 0.124 in SCC, respectively. Higher concordance was observed for PDL1 amplification (KI, 0.647 in ADC and KI, 1 in SCC). Eleven (61.1%) of 18 amplified cores showed PD‐L1 staining in < 5% of TCs. Intraclass correlation coefficients for CD3 and CD8 were 0.293 and 0.186 in ADC and 0.489 and 0.610 in SCC samples, respectively. Conclusions We found significant heterogeneity of PD‐L1 expression in both ADC and SCC samples, especially in the IC compartment. Heterogeneous expression of PD‐L1 could misclassify patients for PD‐1/PD‐L1‐directed therapies. Micro‐Abstract Expression of programmed death‐ligand 1 (PD‐L1) in tumor cells and infiltrating immune cells was retrospectively analyzed in a cohort of surgically‐treated patients with non–small‐cell lung cancer. There was significant discordance of PD‐L1 expression between different tumor areas, especially in the immune cell compartment. Heterogeneous PD‐L1 expression represents a challenge for adequate biomarker‐based selection of patients for programmed cell death protein 1/PD‐L1‐directed therapies.


Clinical Cancer Research | 2017

Phase Ib study of lumretuzumab plus cetuximab or erlotinib in solid tumor patients and evaluation of HER3 and heregulin as potential biomarkers of clinical activity

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.


Journal of Thoracic Oncology | 2014

Assessment of ALK Status by FISH on 1000 Spanish Non-Small Cell Lung Cancer Patients

Joana Vidal; Sergi Clavé; Silvia de Muga; Iria González; Lara Pijuan; Javier Gimeno; Jordi Remon; Noemi Reguart; Nuria Viñolas; Regina Gironés; Laia Bernet; Margarita Majem; Joaquim Bosch-Barrera; Rut Porta; Nieves Alonso; Ramon Palmero; Álvaro Taus; Joan Albanell; Blanca Espinet; Marta Salido; Edurne Arriola

Introduction: Patients with non-small cell lung cancer (NSCLC) harboring anaplastic lymphoma kinase (ALK) rearrangement selectively respond to ALK inhibitors. Thus, identification of ALK rearrangements has become a standard diagnostic test in advanced NSCLC patients. Our institution has been a referral center in Spain for ALK determination by Fluorescent in situ hybridization (FISH). The aim of our study was to assess the feasibility and the FISH patterns of the ALK gene and to evaluate the clinical and pathological features of patients with ALK alterations. Methods: Between 2010 and 2014, 1092 samples were evaluated for ALK using FISH technique (927 histological samples, 165 cytological samples). Correlation with available clinical-pathological information was assessed. Results: ALK rearrangement was found in 35 patients (3.2%). Cytological samples (using either direct smears or cell blocks), were more frequently non-assessable than histological samples (69% versus 89%, respectively) (p < 0.001). Within the ALK-rearranged cases the majority were female, non-smokers, and stage IV. Conclusions: Although assessable in cytological samples, biopsies are preferred when available for ALK evaluation by FISH. The ALK translocation prevalence and the associated clinico-pathological features in Spanish NSCLC patients are similar to those previously reported.


Archivos De Bronconeumologia | 2014

Impacto del 18F-FDG PET/TC en el abordaje terapéutico del cáncer de pulmón no microcítico

Álvaro Taus; Rafael Aguiló; Víctor Curull; Marina Suárez-Piñera; Alberto Rodríguez-Fuster; Nuria Rodríguez de Dios; Lara Pijuan; Flavio Zuccarino; Ivan Vollmer; Albert Sánchez-Font; José Belda-Sanchis; Edurne Arriola

INTRODUCTION Disease stage is the most important prognostic factor in lung cancer, and optimal staging is important to determine the best therapeutic option. FDG-PET/CT has demonstrated its value in early stage non-small cell lung cancer (NSCLC) but there is still insufficient data to define its role in other stages. HYPOTHESIS Information provided by FDG-PET/CT has an impact on the therapeutic management of patients with NSCLC. METHODS A retrospective review was made of patients who underwent FDG-PET/CT between January 2008 and December 2010 for the diagnosis of NSCLC. Clinical stage before and after FDG-PET/CT and information about any change in therapeutic decision due to information provided by FDG-PET/CT were collected. Using pathologic evaluation as the gold standard, sensitivity, specificity, and positive and negative predictive values for CT and FDG-PET/CT were calculated. RESULTS Of the 522 patients diagnosed of NSCLC, FDG-PET/CT was performed in 246 (47.1%). In 85 cases (34.6%) FDG-PET/CT led to stage migration. Treatment was modified in 60 patients (24.4% of all FDG-PET/CT performed), avoiding a futile thoracotomy in 13 cases (5.2%), and allowing treatment with curative intent in 26 (10.5%). Out of 90 patients (36.5%) evaluated as stage iii by CT staging, FDG-PET/CT modified the therapeutic approach in 36 (40%). For the 133 cases (54%) with pathological assessment of the mediastinal lymph nodes, sensitivity, specificity, positive predictive value and negative predictive value were 0.57, 0.64, 0.48 and 0.72 for CT, and 0.68, 0.86, 0.75 and 0.81 for FDG-PET/CT. DISCUSSION Our data support previous reports that FDG-PET/CT is essential in the staging process not only for patients with potentially operable NSCLC but also for stage iii patients, as demonstrated by our data.


Archivos De Bronconeumologia | 2011

Activating and Resistance Mutations of the Epidermal Growth Factor Receptor (EGFR) Gene and Non-Small Cell Lung Cancer: A Clinical Reality

Álvaro Taus; Ivan Vollmer; Edurne Arriola

Abstract In non-small cell lung cancer, EGFR gene mutations identify a patient sub-population with different clinical characteristics and treatment responses than those who do not present these mutations. There are mutations that lead to increased sensitivity to EGFR-targeted therapy, as well as mutations that result in resistance. The determination of EGFR mutations involves a change in the therapeutic approach to lung cancer patients in current clinical practice. In this article we present a case of a patient suffering from a metastatic lung adenocarcinoma with an activating mutation on diagnosis, initially responding to treatment with erlotinib, who subsequently developed a secondary resistance due to acquiring the T790M mutation in exon 20 of the EGFR gene.


Epigenetics | 2014

Methylation status of IGFBP-3 as a useful clinical tool for deciding on a concomitant radiotherapy.

Olga Pernía; Cristóbal Belda-Iniesta; Veronica Pulido; María Cortes-Sempere; Carlos J. Rodriguez; Olga Vera; Javier Soto; Julia Jimenez; Álvaro Taus; Federico Rojo; Edurne Arriola; Ana Rovira; Joan Albanell; M. Teresa Macías; Javier Castro; Rosario Perona; Inmaculada Ibanez de Caceres

The methylation status of the IGFBP-3 gene is strongly associated with cisplatin sensitivity in patients with non-small cell lung cancer (NSCLC). In this study, we found in vitro evidence that linked the presence of an unmethylated promoter with poor response to radiation. Our data also indicate that radiation might sensitize chemotherapy-resistant cells by reactivating IGFBP-3-expression through promoter demethylation, inactivating the PI3K/AKT pathway. We also explored the IGFBP-3 methylation effect on overall survival (OS) in a population of 40 NSCLC patients who received adjuvant therapy after R0 surgery. Our results indicate that patients harboring an unmethylated promoter could benefit more from a chemotherapy schedule alone than from a multimodality therapy involving radiotherapy and platinum-based treatments, increasing their OS by 2.5 y (p = .03). Our findings discard this epi-marker as a prognostic factor in a patient population without adjuvant therapy, indicating that radiotherapy does not improve survival for patients harboring an unmethylated IGFBP-3 promoter.


Archivos De Bronconeumologia | 2011

Mutaciones de sensibilidad y resistencia del gen epidermal growth factor receptor (EGFR) en el cáncer de pulmón de célula no pequeña: una realidad clínica

Álvaro Taus; Ivan Vollmer; Edurne Arriola

In non-small cell lung cancer, the EGFR gene mutations identify a patient sub-population with different clinical characteristics and treatment responses to those that do not have these mutations. There are mutations that derive in increased sensitive to EGFR targeted therapy, as well as mutations that result in resistance. The determination of EGFR mutations involves a change in the therapeutic approach to lung cancer patients in current clinical practice. In this article we present a case of a patient suffering from a metastatic lung adenocarcinoma with an activating mutation on diagnosis, initially responding to treatment with erlotinib, who subsequently developed a secondary resistance due to acquiring the T790M mutation in exon 20 of the EGFR gene.


Histopathology | 2018

CD274 (PDL1) and JAK2 genomic amplifications in pulmonary squamous-cell and adenocarcinoma patients

Sergi Clavé; Lara Pijuan; David Casadevall; Álvaro Taus; Javier Gimeno; Silvia Hernández-Llodrà; María Rodríguez-Rivera; Marta Lorenzo; Silvia Menendez; Joan Albanell; Blanca Espinet; Edurne Arriola; Marta Salido

CD274 (PDL1) and JAK2 (9p24.1) gene amplifications have been recently described in pulmonary carcinomas in association with programmed death‐ligand 1 (PD‐L1) expression. Furthermore, PTEN loss has been explored preclinically in relation to PD‐L1 expression. Our aim was to determine whether these genomic alterations affect PD‐L1 expression levels in non‐small‐cell lung cancer.

Collaboration


Dive into the Álvaro Taus's collaboration.

Top Co-Authors

Avatar

Edurne Arriola

University of Southampton

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lara Pijuan

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Marta Salido

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Blanca Espinet

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

David Casadevall

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Silvia Menendez

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ana Rovira

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Javier Gimeno

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge