Network


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

Hotspot


Dive into the research topics where Cristina Royo is active.

Publication


Featured researches published by Cristina Royo.


Cancer Research | 2010

Genomic and Gene Expression Profiling Defines Indolent Forms of Mantle Cell Lymphoma

Verònica Fernàndez; Olga Salamero; Blanca Espinet; Francesc Solé; Cristina Royo; Alba Navarro; Francisca I. Camacho; Sílvia Beà; Elena Hartmann; Virginia Amador; Luis Hernández; Claudio Agostinelli; Rachel L. Sargent; María Rozman; Marta Aymerich; Dolors Colomer; Neus Villamor; Steven H. Swerdlow; Stefano Pileri; Francesc Bosch; Miguel A. Piris; Emili Montserrat; German Ott; Andreas Rosenwald; Armando López-Guillermo; Pedro Jares; Sergi Serrano; Elias Campo

Mantle cell lymphoma (MCL) is typically a very aggressive disease with poor outcomes, but some cases display an indolent behavior that might not necessitate treatment at diagnosis. To define molecular criteria that might permit recognition of such cases, we compared the clinicopathologic features, gene expression, and genomic profile of patients who had indolent or conventional disease (iMCL or cMCL). Patients with iMCL displayed nonnodal leukemic disease with predominantly hypermutated IGVH and noncomplex karyotypes. iMCL and cMCL shared a common gene expression profile that differed from other leukemic lymphoid neoplasms. However, we identified a signature of 13 genes that was highly expressed in cMCL but underexpressed in iMCL. SOX11 was notable in this signature and we confirmed a restriction of SOX11 protein expression to cMCL. To validate the potential use of SOX11 as a biomarker for cMCL, we evaluated SOX11 protein expression in an independent series of 112 cases of MCL. Fifteen patients with SOX11-negative tumors exhibited more frequent nonnodal presentation and better survival compared with 97 patients with SOX11-positive MCL (5-year overall survival of 78% versus 36%, respectively; P = 0.001). In conclusion, we defined nonnodal presentation, predominantly hypermutated IGVH, lack of genomic complexity, and absence of SOX11 expression as qualities of a specific subtype of iMCL with excellent outcomes that might be managed more conservatively than cMCL.


Nature | 2015

Non-coding recurrent mutations in chronic lymphocytic leukaemia.

Xose S. Puente; Sílvia Beà; Rafael Valdés-Mas; Neus Villamor; Jesús Gutiérrez-Abril; José I. Martín-Subero; Marta Munar; Carlota Rubio-Perez; Pedro Jares; Marta Aymerich; Tycho Baumann; Renée Beekman; Laura Belver; Anna Carrió; Giancarlo Castellano; Guillem Clot; Enrique Colado; Dolors Colomer; Dolors Costa; Julio Delgado; Anna Enjuanes; Xavier Estivill; Adolfo A. Ferrando; Josep Lluís Gelpí; Blanca González; S. Gonzalez; Marcos González; Marta Gut; Jesús María Hernández-Rivas; Mónica López-Guerra

Chronic lymphocytic leukaemia (CLL) is a frequent disease in which the genetic alterations determining the clinicobiological behaviour are not fully understood. Here we describe a comprehensive evaluation of the genomic landscape of 452 CLL cases and 54 patients with monoclonal B-lymphocytosis, a precursor disorder. We extend the number of CLL driver alterations, including changes in ZNF292, ZMYM3, ARID1A and PTPN11. We also identify novel recurrent mutations in non-coding regions, including the 3′ region of NOTCH1, which cause aberrant splicing events, increase NOTCH1 activity and result in a more aggressive disease. In addition, mutations in an enhancer located on chromosome 9p13 result in reduced expression of the B-cell-specific transcription factor PAX5. The accumulative number of driver alterations (0 to ≥4) discriminated between patients with differences in clinical behaviour. This study provides an integrated portrait of the CLL genomic landscape, identifies new recurrent driver mutations of the disease, and suggests clinical interventions that may improve the management of this neoplasia.


Haematologica | 2009

SOX11 expression is highly specific for mantle cell lymphoma and identifies the cyclin D1-negative subtype

Ana Mozos; Cristina Royo; Elena Hartmann; Daphne de Jong; Cristina Baró; Alexandra Valera; Kai Fu; Dennis D. Weisenburger; Jan Delabie; Shih Sung Chuang; Elaine S. Jaffe; Carmen Ruíz-Marcellán; Sandeep S. Dave; Lisa M. Rimsza; Rita M. Braziel; Randy D. Gascoyne; Francisco Solé; Armando López-Guillermo; Dolors Colomer; Louis M. Staudt; Andreas Rosenwald; German Ott; Pedro Jares; Elias Campo

Cyclin D1-negative mantle cell lymphoma is difficult to distinguish from other small B-cell lymphomas. This study shows that SOX11 mRNA and nuclear protein expression is a highly specific marker for both cyclin D1-positive and negative mantle cell lymphoma. See related perspective article on page 1488. Background Cyclin D1-negative mantle cell lymphoma is difficult to distinguish from other small B-cell lymphomas. The clinical and pathological characteristics of patients with this form of lymphoma have not been well defined. Overexpression of the transcription factor SOX11 has been observed in conventional mantle cell lymphoma. The aim of this study was to determine whether this gene is expressed in cyclin D1-negative mantle cell lymphoma and whether its detection may be useful to identify these tumors. Design and Methods The microarray database of 238 mature B-cell neoplasms was re-examined. SOX11 protein expression was investigated immunohistochemically in 12 cases of cyclin D1-negative mantle cell lymphoma, 54 cases of conventional mantle cell lymphoma, and 209 additional lymphoid neoplasms. Results SOX11 mRNA was highly expressed in conventional and cyclin D1-negative mantle cell lymphoma and in 33% of the cases of Burkitt’s lymphoma but not in any other mature lymphoid neoplasm. SOX11 nuclear protein was detected in 50 cases (93%) of conventional mantle cell lymphoma and also in the 12 cyclin D1-negative cases of mantle cell lymphoma, the six cases of lymphoblastic lymphomas, in two of eight cases of Burkitt’s lymphoma, and in two of three T-prolymphocytic leukemias but was negative in the remaining lymphoid neoplasms. Cyclin D2 and D3 mRNA levels were significantly higher in cyclin D1-negative mantle cell lymphoma than in conventional mantle cell lymphoma but the protein expression was not discriminative. The clinico-pathological features and outcomes of the patients with cyclin D1-negative mantle cell lymphoma identified by SOX11 expression were similar to those of patients with conventional mantle cell lymphoma. Conclusions SOX11 mRNA and nuclear protein expression is a highly specific marker for both cyclin D1-positive and negative mantle cell lymphoma.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Landscape of somatic mutations and clonal evolution in mantle cell lymphoma

Sílvia Beà; Rafael Valdés-Mas; Alba Navarro; Itziar Salaverria; David Martín-García; Pedro Jares; Eva Giné; Magda Pinyol; Cristina Royo; Ferran Nadeu; Laura Conde; Manel Juan; Guillem Clot; Pedro Vizán; Luciano Di Croce; Diana A. Puente; Mónica López-Guerra; Alexandra Moros; Gaël Roué; Marta Aymerich; Neus Villamor; Lluis Colomo; Antonio Martínez; Alexandra Valera; José I. Martín-Subero; Virginia Amador; Luis Hernández; María Rozman; Anna Enjuanes; Pilar Forcada

Significance This is a comprehensive whole-genome/whole-exome analysis of mantle cell lymphoma (MCL). We sequenced 29 MCL cases and validated the findings by target sequencing of 172 additional tumors. We identified recurrent mutations in genes regulating chromatin modification and genes such as NOTCH2 that have a major impact on clinical outcome. Additionally, we demonstrated the subclonal heterogeneity of the tumors already at diagnosis and the modulation of the mutational architecture in the progression of the disease. The identification of new molecular mechanisms may open perspectives for the management of MCL patients. Mantle cell lymphoma (MCL) is an aggressive tumor, but a subset of patients may follow an indolent clinical course. To understand the mechanisms underlying this biological heterogeneity, we performed whole-genome and/or whole-exome sequencing on 29 MCL cases and their respective matched normal DNA, as well as 6 MCL cell lines. Recurrently mutated genes were investigated by targeted sequencing in an independent cohort of 172 MCL patients. We identified 25 significantly mutated genes, including known drivers such as ataxia-telangectasia mutated (ATM), cyclin D1 (CCND1), and the tumor suppressor TP53; mutated genes encoding the anti-apoptotic protein BIRC3 and Toll-like receptor 2 (TLR2); and the chromatin modifiers WHSC1, MLL2, and MEF2B. We also found NOTCH2 mutations as an alternative phenomenon to NOTCH1 mutations in aggressive tumors with a dismal prognosis. Analysis of two simultaneous or subsequent MCL samples by whole-genome/whole-exome (n = 8) or targeted (n = 19) sequencing revealed subclonal heterogeneity at diagnosis in samples from different topographic sites and modulation of the initial mutational profile at the progression of the disease. Some mutations were predominantly clonal or subclonal, indicating an early or late event in tumor evolution, respectively. Our study identifies molecular mechanisms contributing to MCL pathogenesis and offers potential targets for therapeutic intervention.


Cancer Research | 2012

Molecular subsets of mantle cell lymphoma defined by the IGHV mutational status and SOX11 expression have distinct biologic and clinical features

Alba Navarro; Guillem Clot; Cristina Royo; Pedro Jares; Anastasia Hadzidimitriou; Andreas Agathangelidis; Vasilis Bikos; Nikos Darzentas; Theodora Papadaki; Itziar Salaverria; Magda Pinyol; Xavier Puig; Jara Palomero; Maria Carmela Vegliante; Virgina Amador; Alejandra Martínez-Trillos; Lenka Stefancikova; Adrian Wiestner; Wyndham H. Wilson; Christiane Pott; María José Calasanz; Nicola Trim; Wendy N. Erber; Birgitta Sander; German Ott; Andreas Rosenwald; Dolors Colomer; Eva Giné; Reiner Siebert; Armando López-Guillermo

Mantle cell lymphoma (MCL) is a heterogeneous disease with most patients following an aggressive clinical course, whereas others having an indolent behavior. We conducted an integrative and multidisciplinary analysis of 177 MCL to determine whether the immunogenetic features of the clonotypic B-cell receptors (BcR) may identify different subsets of tumors. Truly unmutated (100% identity) IGHV genes were found in 24% cases, 40% were minimally/borderline mutated (99.9%-97%), 19% significantly mutated (96.9%-95%), and 17% hypermutated (<95%). Tumors with high or low mutational load used different IGHV genes, and their gene expression profiles were also different for several gene pathways. A gene set enrichment analysis showed that MCL with high and low IGHV mutations were enriched in memory and naive B-cell signatures, respectively. Furthermore, the highly mutated tumors had less genomic complexity, were preferentially SOX11-negative, and showed more frequent nonnodal disease. The best cut-off of germline identity of IGHV genes to predict survival was 97%. Patients with high and low mutational load had significant different outcome with 5-year overall survival (OS) of 59% and 40%, respectively (P = 0.004). Nodal presentation and SOX11 expression also predicted for poor OS. In a multivariate analysis, IGHV gene status and SOX11 expression were independent risk factors. In conclusion, these observations suggest the idea that MCL with mutated IGHV, SOX11-negativity, and nonnodal presentation correspond to a subtype of the disease with more indolent behavior.


Genome Research | 2014

Transcriptome characterization by RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia

Pedro G. Ferreira; Pedro Jares; Daniel Rico; Gonzalo Gómez-López; Alejandra Martínez-Trillos; Neus Villamor; Simone Ecker; Abel Gonzalez-Perez; David G. Knowles; Jean Monlong; Rory Johnson; Víctor Quesada; Sarah Djebali; Panagiotis Papasaikas; Mónica López-Guerra; Dolors Colomer; Cristina Royo; Maite Cazorla; Magda Pinyol; Guillem Clot; Marta Aymerich; María Rozman; Marta Kulis; David Tamborero; Anaı̈s Gouin; Julie Blanc; Marta Gut; Ivo Gut; Xose S. Puente; David G. Pisano

Chronic lymphocytic leukemia (CLL) has heterogeneous clinical and biological behavior. Whole-genome and -exome sequencing has contributed to the characterization of the mutational spectrum of the disease, but the underlying transcriptional profile is still poorly understood. We have performed deep RNA sequencing in different subpopulations of normal B-lymphocytes and CLL cells from a cohort of 98 patients, and characterized the CLL transcriptional landscape with unprecedented resolution. We detected thousands of transcriptional elements differentially expressed between the CLL and normal B cells, including protein-coding genes, noncoding RNAs, and pseudogenes. Transposable elements are globally derepressed in CLL cells. In addition, two thousand genes-most of which are not differentially expressed-exhibit CLL-specific splicing patterns. Genes involved in metabolic pathways showed higher expression in CLL, while genes related to spliceosome, proteasome, and ribosome were among the most down-regulated in CLL. Clustering of the CLL samples according to RNA-seq derived gene expression levels unveiled two robust molecular subgroups, C1 and C2. C1/C2 subgroups and the mutational status of the immunoglobulin heavy variable (IGHV) region were the only independent variables in predicting time to treatment in a multivariate analysis with main clinico-biological features. This subdivision was validated in an independent cohort of patients monitored through DNA microarrays. Further analysis shows that B-cell receptor (BCR) activation in the microenvironment of the lymph node may be at the origin of the C1/C2 differences.


Blood | 2013

CCND2 rearrangements are the most frequent genetic events in cyclin D1(-) mantle cell lymphoma.

Itziar Salaverria; Cristina Royo; Alejandra Carvajal-Cuenca; Guillem Clot; Alba Navarro; Alejandra Valera; Joo Y. Song; Renata Woroniecka; Grzegorz Rymkiewicz; Wolfram Klapper; Elena Hartmann; Pierre Sujobert; Iwona Wlodarska; Judith A. Ferry; Philippe Gaulard; German Ott; Andreas Rosenwald; Armando López-Guillermo; Leticia Quintanilla-Martinez; Nancy Lee Harris; Elaine S. Jaffe; Reiner Siebert; Elias Campo; Sílvia Beà

Cyclin D1(-) mantle cell lymphomas (MCLs) are not well characterized, in part because of the difficulties in their recognition. SOX11 has been identified recently as a reliable biomarker of MCL that is also expressed in the cyclin D1(-) variant. We investigated 40 lymphomas with MCL morphology and immunophenotype that were negative for cyclin D1 expression/t(11;14)(q13;q32) but positive for SOX11. These tumors presented clinically with generalized lymphadenopathy, advanced stage, and poor outcome (5-year overall survival, 48%). Chromosomal rearrangements of the CCND2 locus were detected in 55% of the cases, with an IG gene as partner in 18 of 22, in particular with light chains (10 IGK@ and 5 IGL@). No mutations in the phosphorylation motifs of CCND1, CCND2, or CCND3 were detected. The global genomic profile and the high complexity of the 32 cyclin D1(-) SOX11(+) MCL patients analyzed by copy number arrays were similar to the conventional cyclin D1/SOX11 MCL. 17p deletions and high Ki67 expression conferred a significantly worse outcome for the patients. This comprehensive characterization of a large series of cyclin D1(-) MCL patients indicates that these tumors are clinically and biologically similar to the conventional cyclin D1(+) MCL and provides a basis for the proper identification and clinical management of these patients.


Haematologica | 2012

In situ mantle cell lymphoma: clinical implications of an incidental finding with indolent clinical behavior.

Alejandra Carvajal-Cuenca; Luz F. Sua; Nhora M. Silva; Stefania Pittaluga; Cristina Royo; Joo Y. Song; Rachel L. Sargent; Blanca Espinet; Fina Climent; Samuel A. Jacobs; Jan Delabie; Kikkeri N. Naresh; Adam Bagg; Pierre Brousset; Roger A. Warnke; Sergi Serrano; Nancy Lee Harris; Steven H. Swerdlow; Elaine S. Jaffe; Elias Campo

Background Cyclin D1-positive B cells are occasionally found in the mantle zones of reactive lymphoid follicles, a condition that has been called “in situ mantle cell lymphoma”. The clinical significance of this lesion remains uncertain. Design and Methods The clinical and pathological characteristics, including SOX11 expression, of 23 cases initially diagnosed as in situ mantle cell lymphoma were studied. Results Seventeen of the 23 cases fulfilled the criteria for in situ mantle cell lymphoma. In most cases, the lesions were incidental findings in reactive lymph nodes. The t(11;14) was detected in all eight cases examined. SOX11 was positive in seven of 16 cases (44%). Five cases were associated with other small B-cell lymphomas. In two cases, both SOX11-positive, the in situ mantle cell lymphoma lesions were discovered after the diagnosis of overt lymphoma; one 4 years earlier, and one 3 years later. Twelve of the remaining 15 patients had a follow-up of at least 1 year (median 2 years; range, 1–19.5), of whom 11 showed no evidence of progression, including seven who were not treated. Only one of 12 patients with an in situ mantle cell lymphoma lesion and no diagnosis of mantle cell lymphoma at the time developed an overt lymphoma, 4 years later; this case was also SOX11-positive. The six remaining cases were diagnosed as mantle cell lymphoma with a mantle zone pattern. Five were SOX11-positive and four of them were associated with lymphoma without a mantle zone pattern. Conclusions In situ mantle cell lymphoma lesions are usually an incidental finding with a very indolent behavior. These cases must be distinguished from mantle cell lymphoma with a mantle zone pattern and overt mantle cell lymphoma because they may not require therapeutic intervention.


Blood | 2016

Clinical impact of clonal and subclonal TP53, SF3B1, BIRC3, NOTCH1, and ATM mutations in chronic lymphocytic leukemia

Ferran Nadeu; Julio Delgado; Cristina Royo; Tycho Baumann; Tatjana Stankovic; Magda Pinyol; Pedro Jares; Alba Navarro; David Martín-García; Sílvia Beà; Itziar Salaverria; Ceri Oldreive; Marta Aymerich; Helena Suárez-Cisneros; María Rozman; Neus Villamor; Dolors Colomer; Armando López-Guillermo; Marcos González; Miguel Alcoceba; María José Terol; Enrique Colado; Xose S. Puente; Carlos López-Otín; Anna Enjuanes; Elias Campo

Genomic studies have revealed the complex clonal heterogeneity of chronic lymphocytic leukemia (CLL). The acquisition and selection of genomic aberrations may be critical to understanding the progression of this disease. In this study, we have extensively characterized the mutational status of TP53, SF3B1, BIRC3, NOTCH1, and ATM in 406 untreated CLL cases by ultra-deep next-generation sequencing, which detected subclonal mutations down to 0.3% allele frequency. Clonal dynamics were examined in longitudinal samples of 48 CLL patients. We identified a high proportion of subclonal mutations, isolated or associated with clonal aberrations. TP53 mutations were present in 10.6% of patients (6.4% clonal, 4.2% subclonal), ATM mutations in 11.1% (7.8% clonal, 1.3% subclonal, 2% germ line mutations considered pathogenic), SF3B1 mutations in 12.6% (7.4% clonal, 5.2% subclonal), NOTCH1 mutations in 21.8% (14.2% clonal, 7.6% subclonal), and BIRC3 mutations in 4.2% (2% clonal, 2.2% subclonal). ATM mutations, clonal SF3B1, and both clonal and subclonal NOTCH1 mutations predicted for shorter time to first treatment irrespective of the immunoglobulin heavy-chain variable-region gene (IGHV) mutational status. Clonal and subclonal TP53 and clonal NOTCH1 mutations predicted for shorter overall survival together with the IGHV mutational status. Clonal evolution in longitudinal samples mainly occurred in cases with mutations in the initial samples and was observed not only after chemotherapy but also in untreated patients. These findings suggest that the characterization of the subclonal architecture and its dynamics in the evolution of the disease may be relevant for the management of CLL patients.


Seminars in Cancer Biology | 2011

The complex landscape of genetic alterations in mantle cell lymphoma.

Cristina Royo; Itziar Salaverria; Elena Hartmann; Andreas Rosenwald; Elias Campo; Sílvia Beà

Mantle cell lymphoma (MCL) is genetically characterized by the t(11;14)(q13;q32) which deregulates cyclin D1. Small subsets of cases have been identified with variant CCND1 translocations with the immunoglobulin light chain genes or with alternative translocations involving CCND2 and CCND3. Additionally, double-hit MCL with MYC rearrangements with a highly aggressive clinical course have been reported, but no other frequent recurrent translocations have been identified. In recent years, genome-wide screening of copy number alterations by comparative genomic hybridization and genomic microarray platforms have revealed a characteristic MCL profile of multiple secondary gains and losses as well as regions of copy number neutral loss of heterozygosity that target mainly genes involved in cell cycle regulation, DNA damage response, and cell survival pathways. Several aberrations have been found to be associated with worse prognosis, 3q gains and losses of 8p, 9p, and 17p. An increased number of secondary alterations and blastoid morphology have also been shown to be associated with cases with short survival. On the contrary, indolent MCL cases carry only the primary t(11;14) and few or no other additional genomic alterations. Altogether these observations suggest that the genetic background of MCL is an important factor that dictates their different clinical behavior. This review will focus on MCL from a genetic perspective and will present next-generation sequencing technology as a new potential tool to complement the study of complex genomes. The better understanding of genetic alterations of MCL may offer new approaches for more patient-tailored, risk-adapted treatment options.

Collaboration


Dive into the Cristina Royo's collaboration.

Top Co-Authors

Avatar

Alba Navarro

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Elias Campo

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Sílvia Beà

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Guillem Clot

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Pedro Jares

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Magda Pinyol

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge