Network


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

Hotspot


Dive into the research topics where Laia Paré is active.

Publication


Featured researches published by Laia Paré.


Bioinformatics | 2016

Genefu: an R/Bioconductor package for computation of gene expression-based signatures in breast cancer.

Deena M.A. Gendoo; Natchar Ratanasirigulchai; Markus S. Schröder; Laia Paré; Joel S. Parker; Aleix Prat; Benjamin Haibe-Kains

UNLABELLED Breast cancer is one of the most frequent cancers among women. Extensive studies into the molecular heterogeneity of breast cancer have produced a plethora of molecular subtype classification and prognosis prediction algorithms, as well as numerous gene expression signatures. However, reimplementation of these algorithms is a tedious but important task to enable comparison of existing signatures and classification models between each other and with new models. Here, we present the genefu R/Bioconductor package, a multi-tiered compendium of bioinformatics algorithms and gene signatures for molecular subtyping and prognostication in breast cancer. AVAILABILITY AND IMPLEMENTATION The genefu package is available from Bioconductor. http://www.bioconductor.org/packages/devel/bioc/html/genefu.html Source code is also available on Github https://github.com/bhklab/genefu CONTACT [email protected] SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.


Cancer Research | 2017

Immune-related gene expression profiling after PD-1 blockade in non–small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma

Aleix Prat; Alejandro Navarro; Laia Paré; Noemi Reguart; Patricia Galván; Tomás Pascual; Alex Martinez; Paolo Nuciforo; Laura Comerma; Llucia Alos; Nuria Pardo; Susana Cedrés; Cheng Fan; Joel S. Parker; Lydia Gaba; Iván Victoria; Nuria Viñolas; Ana Vivancos; Ana Arance; Enriqueta Felip

Antibody targeting of the immune checkpoint receptor PD1 produces therapeutic activity in a variety of solid tumors, but most patients exhibit partial or complete resistance to treatment for reasons that are unclear. In this study, we evaluated tumor specimens from 65 patients with melanoma, lung nonsquamous, squamous cell lung or head and neck cancers who were treated with the approved PD1-targeting antibodies pembrolizumab or nivolumab. Tumor RNA before anti-PD1 therapy was analyzed on the nCounter system using the PanCancer 730-Immune Panel, and we identified 23 immune-related genes or signatures linked to response and progression-free survival (PFS). In addition, we evaluated intra- and interbiopsy variability of PD1, PD-L1, CD8A, and CD4 mRNAs and their relationship with tumor-infiltrating lymphocytes (TIL) and PD-L1 IHC expression. Among the biomarkers examined, PD1 gene expression along with 12 signatures tracking CD8 and CD4 T-cell activation, natural killer cells, and IFN activation associated significantly with nonprogressive disease and PFS. These associations were independent of sample timing, drug used, or cancer type. TIL correlated moderately (∼0.50) with PD1 and CD8A mRNA levels and weakly (∼0.35) with CD4 and PD-L1. IHC expression of PD-L1 correlated strongly with PD-L1 (0.90), moderately with CD4 and CD8A, and weakly with PD1. Reproducibility of gene expression in intra- and interbiopsy specimens was very high (total SD <3%). Overall, our results support the hypothesis that identification of a preexisting and stable adaptive immune response as defined by mRNA expression pattern is reproducible and sufficient to predict clinical outcome, regardless of the type of cancer or the PD1 therapeutic antibody administered to patients. Cancer Res; 77(13); 3540-50. ©2017 AACR.


Cancer Research | 2017

Intrinsic subtypes and gene expression profiles in primary and metastatic breast cancer

Juan Miguel Cejalvo; Eduardo Martínez de Dueñas; Patricia Galván; Susana García-Recio; Octavio Burgués Gasión; Laia Paré; Silvia Antolín; Rossella Martinello; Isabel Blancas; Barbara Adamo; Angel Guerrero-Zotano; Montserrat Muñoz; Paolo Nuciforo; Maria Vidal; Rm Pérez; Jose Ignacio Chacon Lopez-Muniz; Rosalia Caballero; Vicente Peg; Eva Carrasco; Federico Rojo; Charles M. Perou; Javier Cortes; Vincenzo Adamo; Joan Albanell; Roger R. Gomis; Ana Lluch; Aleix Prat

Biological changes that occur during metastatic progression of breast cancer are still incompletely characterized. In this study, we compared intrinsic molecular subtypes and gene expression in 123 paired primary and metastatic tissues from breast cancer patients. Intrinsic subtype was identified using a PAM50 classifier and χ2 tests determined the differences in variable distribution. The rate of subtype conversion was 0% in basal-like tumors, 23.1% in HER2-enriched (HER2-E) tumors, 30.0% in luminal B tumors, and 55.3% in luminal A tumors. In 40.2% of cases, luminal A tumors converted to luminal B tumors, whereas in 14.3% of cases luminal A and B tumors converted to HER2-E tumors. We identified 47 genes that were expressed differentially in metastatic versus primary disease. Metastatic tumors were enriched for proliferation-related and migration-related genes and diminished for luminal-related genes. Expression of proliferation-related genes were better at predicting overall survival in metastatic disease (OSmet) when analyzed in metastatic tissue rather than primary tissue. In contrast, a basal-like gene expression signature was better at predicting OSmet in primary disease compared with metastatic tissue. We observed correlations between time to tumor relapse and the magnitude of changes of proliferation, luminal B, or HER2-E signatures in metastatic versus primary disease. Although the intrinsic subtype was largely maintained during metastatic progression, luminal/HER2-negative tumors acquired a luminal B or HER2-E profile during metastatic progression, likely reflecting tumor evolution or acquisition of estrogen independence. Overall, our analysis revealed the value of stratifying gene expression by both cancer subtype and tissue type, providing clinicians more refined tools to evaluate prognosis and treatment. Cancer Res; 77(9); 2213-21. ©2017 AACR.


JAMA Oncology | 2016

Prognostic Value of Intrinsic Subtypes in Hormone Receptor-Positive Metastatic Breast Cancer Treated With Letrozole With or Without Lapatinib.

Aleix Prat; Maggie Cheang; Patricia Galván; Paolo Nuciforo; Laia Paré; Barbara Adamo; Montserrat Muñoz; Margarida Viladot; Michael F. Press; Robert Gagnon; Catherine E. Ellis; Stephen R. D. Johnston

Importance The value of the intrinsic subtypes of breast cancer (luminal A, luminal B, human epidermal growth factor receptor 2 [currently known as ERBB2, but referred to as HER2 in this study]-enriched, and basal-like) in the metastatic setting is currently unknown. Objective To evaluate the association of the intrinsic subtypes of breast cancer with outcome and/or benefit in hormone receptor (HR)-positive metastatic breast cancer. Design, Setting, and Participants Unplanned retrospective analysis of 821 tumor samples (85.7% primary and 14.3% metastatic) from the EGF30008 phase 3 clinical trial (NCT00073528), in which postmenopausal women with HR-positive invasive breast cancer and no prior therapy for advanced or metastatic disease were randomized to letrozole with or without lapatinib, an epidermal growth factor receptor (EGFR)/HER2 tyrosine kinase inhibitor. Tumor samples were classified into each subtype using the research-based PAM50 classifier. Prior neoadjuvant/adjuvant antiestrogen therapy was allowed. Patients with extensive symptomatic visceral disease were excluded. Treatment effects were evaluated using interaction tests. Main Outcomes and Measures Primary and secondary end points were progression-free survival and overall survival. Results The median (range) age was 62 (31-94) years. Intrinsic subtype was the strongest prognostic factor independently associated with progression-free survival and overall survival in all patients, and in patients with HER2-negative (n = 644) or HER2-positive (n = 157) diseases. Median progression-free survival differed across the intrinsic subtypes of clinically HER2-negative disease: luminal A (16.9 [95% CI, 14.1-19.9] months), luminal B (11.0 [95% CI, 9.6-13.6] months), HER2-enriched (4.7 [95% CI, 2.7-10.8] months), and basal-like (4.1 [95% CI, 2.5-13.8] months). Median OS also differed across the intrinsic subtypes: luminal A (45 [95% CI, 41-not applicable {NA}] months), luminal B (37 [95% CI, 31-42] months), HER2-enriched (16 [95% CI, 10-NA] months), and basal-like (23 [95% CI, 12-NA] months). Patients with HER2-negative/HER2-enriched disease benefited from lapatinib therapy (median PFS, 6.49 vs 2.60 months; progression-free survival hazard ratio, 0.238 [95% CI, 0.066-0.863]; interaction P = .02). Conclusions and Relevance This is the first study to reveal an association between intrinsic subtype and outcome in first-line HR-positive metastatic breast cancer. Patients with HR-positive/HER2-negative disease with a HER2-enriched profile may benefit from lapatinib in combination with endocrine therapy. The clinical value of intrinsic subtyping in hormone receptor-positive metastatic breast cancer warrants further investigation, but patients with luminal A/HER2-negative metastatic breast cancer might be good candidates for letrozole monotherapy in the first-line setting regardless of visceral disease and number of metastases.


Clinical Chemistry | 2017

Identification of ALK, ROS1 and RET Fusions by a Multiplexed mRNA-Based Assay in Formalin-Fixed, Paraffin-Embedded Samples from Advanced Non–Small-Cell Lung Cancer Patients

Noemi Reguart; Cristina Teixidó; Ana Giménez-Capitán; Laia Paré; Patricia Galván; Santiago Viteri; Sonia Rodríguez; Vicente Peg; Erika Aldeguer; Nuria Viñolas; Jordi Remon; Niki Karachaliou; Esther Conde; Fernando López-Ríos; Ernest Nadal; Sabine Merkelbach-Bruse; Reinhard Büttner; Rafael Rosell; Miguel Angel Molina-Vila; Aleix Prat

BACKGROUND Anaplastic lymphoma receptor tyrosine kinase (ALK), ROS proto-oncogene 1, receptor tyrosine kinase (ROS1), and ret proto-oncogene (RET) fusions are present in 5%-7% of patients with advanced non-small-cell lung cancer (NSCLC); their accurate identification is critical to guide targeted therapies. FISH and immunohistochemistry (IHC) are considered the gold standards to determine gene fusions, but they have limitations. The nCounter platform is a potentially useful genomic tool for multiplexed detection of gene fusions, but has not been validated in the clinical setting. METHODS Formalin-fixed, paraffin embedded (FFPE) samples from 108 patients with advanced NSCLC were analyzed with an nCounter-based assay and the results compared with FISH, IHC, and reverse transcription PCR (RT-PCR). Data on response to fusion kinase inhibitors was retrospectively collected in a subset of 29 patients. RESULTS Of 108 FFPE samples, 98 were successfully analyzed by nCounter (91%), which identified 55 fusion-positive cases (32 ALK, 21 ROS1, and 2 RET). nCounter results were highly concordant with IHC for ALK (98.5%, CI = 91.8-99.7), while 11 discrepancies were found compared with FISH (87.5% concordance, CI = 79.0-92.9). For ROS1, nCounter showed similar agreement with IHC and FISH (87.2% and 85.9%), but a substantial number of samples were positive only by 1 or 2 techniques. Of the 25 patients deriving clinical benefit from fusion kinase inhibitors, 24 were positive by nCounter and 22 by FISH. CONCLUSIONS nCounter compares favorably with IHC and FISH and can be used for identifying patients with advanced NSCLC positive for ALK/ROS1/RET fusion genes.


Stem cell reports | 2017

Resistance to Taxanes in Triple-Negative Breast Cancer Associates with the Dynamics of a CD49f+ Tumor-Initiating Population.

Jorge Gómez-Miragaya; Marta Palafox; Laia Paré; Guillermo Yoldi; Irene Ferrer; Sergi Vila; Patricia Galván; Pasquale Pellegrini; Héctor Pérez-Montoyo; Ana Igea; Purificación Muñoz; Manel Esteller; Angel R. Nebreda; Ander Urruticoechea; Idoia Morilla; Sonia Pernas; Fina Climent; María Teresa Soler-Monso; Ana Petit; Violeta Serra; Aleix Prat; Eva González-Suárez

Summary Taxanes are a mainstay of treatment for breast cancer, but resistance often develops followed by metastatic disease and mortality. Aiming to reveal the mechanisms underlying taxane resistance, we used breast cancer patient-derived orthoxenografts (PDX). Mimicking clinical behavior, triple-negative breast tumors (TNBCs) from PDX models were more sensitive to docetaxel than luminal tumors, but they progressively acquired resistance upon continuous drug administration. Mechanistically, we found that a CD49f+ chemoresistant population with tumor-initiating ability is present in sensitive tumors and expands during the acquisition of drug resistance. In the absence of the drug, the resistant CD49f+ population shrinks and taxane sensitivity is restored. We describe a transcriptional signature of resistance, predictive of recurrent disease after chemotherapy in TNBC. Together, these findings identify a CD49f+ population enriched in tumor-initiating ability and chemoresistance properties and evidence a drug holiday effect on the acquired resistance to docetaxel in triple-negative breast cancer.


Clinical Cancer Research | 2017

A PAM50-based chemoendocrine score for hormone receptor-positive breast cancer with an intermediate risk of relapse

Aleix Prat; Ana Lluch; Ak Turnbull; Anita K. Dunbier; Lourdes Calvo; Joan Albanell; Juan de la Haba-Rodriguez; Angels Arcusa; Jose Ignacio Chacon; Pedro Sánchez-Rovira; Arrate Plazaola; Montserrat Muñoz; Laia Paré; Joel S. Parker; Nuria Ribelles; Begoña Jimenez; Abdul Aziz Bin Aiderus; Rosalia Caballero; Barbara Adamo; Mitch Dowsett; Eva Carrasco; Miguel Martin; J. Michael Dixon; Charles M. Perou; Emilio Alba

Purpose: Hormone receptor–positive (HR+) breast cancer is clinically and biologically heterogeneous, and subgroups with different prognostic and treatment sensitivities need to be identified. Experimental Design: Research-based PAM50 subtyping and expression of additional genes was performed on 63 patients with HR+/HER2− disease randomly assigned to neoadjuvant multiagent chemotherapy versus endocrine therapy in a phase II trial. The biology associated with treatment response was used to derive a PAM50-based chemoendocrine score (CES). CESs predictive ability was evaluated in 4 independent neoadjuvant data sets (n = 675) and 4 adjuvant data sets (n = 1,505). The association of CES, intrinsic biology, and PAM50 risk of relapse (ROR) was explored across 6,007 tumors. Results: Most genes associated with endocrine sensitivity were also found associated with chemotherapy resistance. In the chemotherapy test/validation data sets, CES was independently associated with pathologic complete response (pCR), even after adjusting for intrinsic subtype. pCR rates of the CES endocrine–sensitive (CES-E), uncertain (CES-U), and chemotherapy-sensitive (CES-C) groups in both data sets combined were 25%, 11%, and 2%, respectively. In the endocrine test/validation data sets, CES was independently associated with response. Compared with ROR, >90% of ROR-low and ROR-high tumors were identified as CES-E and CES-C, respectively; however, each CES group represented >25% of ROR-intermediate disease. In terms of survival outcome, CES-C was associated with poor relapse-free survival in patients with ROR-intermediate disease treated with either adjuvant endocrine therapy only or no adjuvant systemic therapy, but not in patients treated with (neo)adjuvant chemotherapy. Conclusions: CES is a genomic signature capable of estimating chemoendocrine sensitivity in HR+ breast cancer beyond intrinsic subtype and risk of relapse. Clin Cancer Res; 23(12); 3035–44. ©2016 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Coamplification of miR-4728 protects HER2-amplified breast cancers from targeted therapy

Konstantinos V. Floros; Timothy L. Lochmann; Bin Hu; Carles Monterrubio; Mark T. Hughes; Jason D. Wells; Cristina Morales; Maninderjit S. Ghotra; Carlotta Costa; Andrew J. Souers; Sosipatros A. Boikos; Joel D. Leverson; Ming Tan; Violeta Serra; Jennifer Koblinski; J. Arribas; Aleix Prat; Laia Paré; Todd W. Miller; Mikhail G. Dozmorov; Hisashi Harada; Brad Windle; Maurizio Scaltriti; Anthony C. Faber

Significance In HER2-amplified breast cancers, HER2 inhibitors have been very successful as adjuvant therapy but not as monotherapy. Here, we demonstrate that coamplification of a HER2 intronic miRNA causes intrinsic resistance to HER2 inhibitors by indirectly down-regulating the pro-apoptotic NOXA. Importantly, coinhibition with MCL-1 inhibitors overcomes this resistance. HER2 (ERBB2) amplification is a driving oncogenic event in breast cancer. Clinical trials have consistently shown the benefit of HER2 inhibitors (HER2i) in treating patients with both local and advanced HER2+ breast cancer. Despite this benefit, their efficacy as single agents is limited, unlike the robust responses to other receptor tyrosine kinase inhibitors like EGFR inhibitors in EGFR-mutant lung cancer. Interestingly, the lack of HER2i efficacy occurs despite sufficient intracellular signaling shutdown following HER2i treatment. Exploring possible intrinsic causes for this lack of response, we uncovered remarkably depressed levels of NOXA, an endogenous inhibitor of the antiapoptotic MCL-1, in HER2-amplified breast cancer. Upon investigation of the mechanism leading to low NOXA, we identified a micro-RNA encoded in an intron of HER2, termed miR-4728, that targets the mRNA of the Estrogen Receptor α (ESR1). Reduced ESR1 expression in turn prevents ERα-mediated transcription of NOXA, mitigating apoptosis following treatment with the HER2i lapatinib. Importantly, resistance can be overcome with pharmacological inhibition of MCL-1. More generally, while many cancers like EGFR-mutant lung cancer are driven by activated kinases that when drugged lead to robust monotherapeutic responses, we demonstrate that the efficacy of targeted therapies directed against oncogenes active through focal amplification may be mitigated by coamplified genes.


Annals of Oncology | 2018

A predictive model of pathologic response based on tumor cellularity and tumor-infiltrating lymphocytes (CelTIL) in HER2-positive breast cancer treated with chemo-free dual HER2 blockade

Paolo Nuciforo; T Pascual; Javier Cortes; Antonio Llombart-Cussac; Roberta Fasani; Laia Paré; M.M. Oliveira; Patricia Galván; Noelia Martínez; Begoña Bermejo; Marcela A. Vidal; Sonia Pernas; Ramon Lopez; Montserrat Muñoz; I Garau; Luis Manso; J Alarcón; E Martínez; V Rodrik-Outmezguine; Jan C. Brase; P Villagrasa; Aleix Prat; E Holgado

Background The presence of stromal tumor-infiltrating lymphocytes (TILs) is associated with increased pathologic complete response (pCR) and improved outcomes in HER2-positive early-breast cancer (BC) treated with anti-HER2-based chemotherapy. In the absence of chemotherapy, the association of TILs with pCR following anti-HER2 therapy-only is largely unknown. Patients and methods The PAMELA neoadjuvant trial treated 151 women with HER2-positive BC with lapatinib and trastuzumab [and hormonal therapy if hormone receptor (HR)-positive] for 18 weeks. Percentage of TILs and tumor cellularity were determined at baseline (N = 148) and at day 15 (D15) of treatment (N = 134). Associations of TILs and tumor cellularity with pCR in the breast were evaluated. A combined score based on tumor cellularity and TILs (CelTIL) measured at D15 was derived in PAMELA, and validated in D15 samples from 65 patients with HER2-positive disease recruited in the LPT109096 neoadjuvant trial, where anti-HER2 therapy-only was administer for 2 weeks, then standard chemotherapy was added for 24 weeks. Results In PAMELA, baseline and D15 TILs were significantly associated with pCR in univariate analysis. In multivariable analysis, D15 TILs, but not baseline TILs, were significantly associated with pCR. At D15, TILs and tumor cellularity were found independently associated with pCR. A combined score (CelTIL) taking into account both variables was derived. CelTIL at D15 as a continuous variable was significantly associated with pCR, and patients with CelTIL-low and CelTIL-high scores had a pCR rate of 0% and 33%, respectively. In LPT109096, CelTIL at D15 was found associated with pCR both as a continuous variable and as group categories using a pre-defined cut-off (75.0% versus 33.3%). Conclusions On-treatment TILs, but not baseline TILs, are independently associated with response following anti-HER2 therapy-only. A combined score of TILs and tumor cellularity measured at D15 provides independent predictive information upon completion of neoadjuvant anti-HER2-based therapy. Clinical trial number NCT01973660.


Cancer Research | 2017

Abstract S3-03: PAM50 intrinsic subtype as a predictor of pathological complete response following neoadjuvant dual HER2 blockade without chemotherapy in HER2-positive breast cancer: First results of the PAMELA clinical trial

A Prat Aparicio; J. Cortes Castan; Laia Paré; Patricia Galván; Begoña Bermejo; Noelia Martínez; María N. Vidal; Sonia Pernas; Rafael López; Montserrat Muñoz; Paolo Nuciforo; Roberta Fasani; Serafin Morales; Mafalda Oliveira; L de la Pena; A Peláez; Antonio Llombart

Background: Prior neoadjuvant studies in HER2+ breast cancer have shown that dual HER2 blockade without chemotherapy achieves pathological complete responses (pCR) rates of 6-36% (TBCRC006/TBCRC023/NeoSphere). However, a major challenge today is how to select prospectively patients who will derive the maximum benefit from dual anti-HER2 therapies without chemotherapy. In this context, we and others have previously shown that HER2+ disease is biologically heterogeneous and composed of all the intrinsic molecular subtypes (Luminal A, Luminal B, HER2-enriched [HER2-E] and Basal-like). Among them, the HER2-E subtype shows the highest activation of the EGFR/HER2 pathway. Methods: PAMELA (NCT01973660) is a non-randomized, open-label, multicentric, prospective translational research study in stage I-IIIA HER2+ breast cancer designed to evaluate the ability of the PAM50 intrinsic subtypes to predict pCR in the breast (pCRB; in situ allowed) following 18 weeks of neoadjuvant lapatinib and trastuzumab). Patients with HR+ disease received letrozole (if postmenopausal) or tamoxifen (if pre-menopausal). The primary objective was to compare the pCRB rates of the HER2-E versus the non-HER2-E subtypes in the intent-to-treat population. The study was planned with a power of 95% at a significance level of 0.05 to detect an absolute relative difference in pCRB rates between the two groups of 27% (i.e. 35% in HER2E and 8% in non-HER2-E). Day-15 formalin-fixed, paraffin-embedded tissue samples were prospectively collected and gene expression profiled using the nCounter platform. The intrinsic subtypes were identified using the research-based PAM50 predictor (Parker JCO 2009). Results: A total of 151 patients were recruited (n=77 HR+ and n=74 HR-). Patient characteristics were: mean age (55 years), mean tumor size (2.84 cm), negative axilla (63.5%) and postmenopausal (60.2%). At baseline, intrinsic subtype distribution was: HER2-E (n=101, 66.9%), Luminal A (n=22; 14.6%), Luminal B (n=16; 10.6%), Basal-like (n=9; 6%) and Normal-Like (n=3; 2%). The overall pCRB was 30.5% (46/151), 18.2% in HR+ disease and 43.2% in HR- disease. Five patients (3.3%) presented progressive disease. Rates of pCRB in HER2-E and non-HER2-E subtypes were 40.6% and 10.0% (p Conclusions: The PAMELA trial met its primary endpoint. PAM50 HER2-E subtype identifies patients with HER2+ disease likely to derive a large benefit from dual anti-HER2 therapies +/- endocrine therapy, especially in HER2+/HR+ disease. In addition, early changes in gene expression indicative of a reduction of tumor cellularity are predictive of pathological complete response at surgery. Citation Format: Prat Aparicio A, Cortes Castan J, Pare L, Galvan P, Bermejo B, Martinez N, Vidal M, Pernas S, Lopez R, Munoz M, Nuciforo P, Fasani R, Morales S, Oliveira M, de La Pena L, Pelaez A, Llombart A. PAM50 intrinsic subtype as a predictor of pathological complete response following neoadjuvant dual HER2 blockade without chemotherapy in HER2-positive breast cancer: First results of the PAMELA clinical trial [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr S3-03.

Collaboration


Dive into the Laia Paré's collaboration.

Top Co-Authors

Avatar

Aleix Prat

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana Lluch

University of Valencia

View shared research outputs
Top Co-Authors

Avatar

Antonio Llombart

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ramon Lopez

University of Texas at Arlington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles M. Perou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge