Network


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

Hotspot


Dive into the research topics where Marta Larrayoz is active.

Publication


Featured researches published by Marta Larrayoz.


Leukemia | 2015

Recurrent mutations refine prognosis in chronic lymphocytic leukemia

Panagiotis Baliakas; Anastasia Hadzidimitriou; La. Sutton; Davide Rossi; E. Minga; Neus Villamor; Marta Larrayoz; Jana Kmínková; Andreas Agathangelidis; Zadie Davis; Eugen Tausch; Evangelia Stalika; Barbara Kantorová; Larry Mansouri; Lydia Scarfò; Diego Cortese; Veronika Navrkalová; Mj. Rose-Zerilli; Karin E. Smedby; Gunnar Juliusson; Achilles Anagnostopoulos; A. Makris; Alba Navarro; Julio Delgado; David Oscier; Chrysoula Belessi; Stephan Stilgenbauer; Paolo Ghia; Šárka Pospíšilová; G. Gaidano

Through the European Research Initiative on chronic lymphocytic leukemia (CLL) (ERIC), we screened 3490 patients with CLL for mutations within the NOTCH1 (n=3334), SF3B1 (n=2322), TP53 (n=2309), MYD88 (n=1080) and BIRC3 (n=919) genes, mainly at diagnosis (75%) and before treatment (>90%). BIRC3 mutations (2.5%) were associated with unmutated IGHV genes (U-CLL), del(11q) and trisomy 12, whereas MYD88 mutations (2.2%) were exclusively found among M-CLL. NOTCH1, SF3B1 and TP53 exhibited variable frequencies and were mostly enriched within clinically aggressive cases. Interestingly, as the timespan between diagnosis and mutational screening increased, so too did the incidence of SF3B1 mutations; no such increase was observed for NOTCH1 mutations. Regarding the clinical impact, NOTCH1 mutations, SF3B1 mutations and TP53 aberrations (deletion/mutation, TP53ab) correlated with shorter time-to-first-treatment (P<0.0001) in 889 treatment-naive Binet stage A cases. In multivariate analysis (n=774), SF3B1 mutations and TP53ab along with del(11q) and U-CLL, but not NOTCH1 mutations, retained independent significance. Importantly, TP53ab and SF3B1 mutations had an adverse impact even in U-CLL. In conclusion, we support the clinical relevance of novel recurrent mutations in CLL, highlighting the adverse impact of SF3B1 and TP53 mutations, even independent of IGHV mutational status, thus underscoring the need for urgent standardization/harmonization of the detection methods.


Leukemia | 2016

The SF3B1 inhibitor spliceostatin A (SSA) elicits apoptosis in chronic lymphocytic leukaemia cells through downregulation of Mcl-1

Marta Larrayoz; Stuart Blakemore; R C Dobson; M D Blunt; Matthew Jj Rose-Zerilli; Renata Walewska; Andrew S Duncombe; David Oscier; K Koide; Francesco Forconi; Graham Packham; Minoru Yoshida; Mark S. Cragg; Jon C. Strefford; Andrew Steele

The pro-survival Bcl-2 family member Mcl-1 is expressed in chronic lymphocytic leukaemia (CLL), with high expression correlated with progressive disease. The spliceosome inhibitor spliceostatin A (SSA) is known to regulate Mcl-1 and so here we assessed the ability of SSA to elicit apoptosis in CLL. SSA induced apoptosis of CLL cells at low nanomolar concentrations in a dose- and time-dependent manner, but independently of SF3B1 mutational status, IGHV status and CD38 or ZAP70 expression. However, normal B and T cells were less sensitive than CLL cells (P=0.006 and P<0.001, respectively). SSA altered the splicing of anti-apoptotic MCL-1L to MCL-1s in CLL cells coincident with induction of apoptosis. Overexpression studies in Ramos cells suggested that Mcl-1 was important for SSA-induced killing since its expression inversely correlated with apoptosis (P=0.001). IL4 and CD40L, present in patient lymph nodes, are known to protect tumour cells from apoptosis and significantly inhibited SSA, ABT-263 and ABT-199 induced killing following administration to CLL cells (P=0.008). However, by combining SSA with the Bcl-2/Bcl-xL antagonists ABT-263 or ABT-199, we were able to overcome this pro-survival effect. We conclude that SSA combined with Bcl-2/Bcl-xL antagonists may have therapeutic utility for CLL.


Blood | 2016

Surface IgM expression and function are associated with clinical behavior, genetic abnormalities, and DNA methylation in CLL

Annalisa D'Avola; Samantha Drennan; Ian Tracy; Isla Henderson; Laura Chiecchio; Marta Larrayoz; Matthew J. Rose-Zerilli; Jonathan C. Strefford; Christoph Plass; Peter Johnson; Andrew Steele; Graham Packham; Freda K. Stevenson; Christopher C. Oakes; Francesco Forconi

Chronic lymphocytic leukemia (CLL) with unmutated (U-CLL) or mutated (M-CLL) immunoglobulin gene heavy-chain variable region (IGHV) displays different states of anergy, indicated by reduced surface immunoglobulin M (sIgM) levels and signaling, consequent to chronic (super)antigen exposure. The subsets also differ in the incidence of high-risk genetic aberrations and in DNA methylation profile, preserved from the maturational status of the original cell. We focused on sIgM expression and function, measured as intracellular Ca(2+) mobilization following stimulation, and probed correlations with clinical outcome. The relationship with genetic features and maturation status defined by DNA methylation of an 18-gene panel signature was then investigated. sIgM levels/signaling were higher and less variable in U-CLL than in M-CLL and correlated with disease progression between and within U-CLL and M-CLL. In U-CLL, increased levels/signaling associated with +12, del(17p) or NOTCH1 mutations. In M-CLL, there were fewer genetic lesions, although the methylation maturation status, generally higher than in U-CLL, varied and was increased in cases with lower sIgM levels/signaling. These features revealed heterogeneity in M-CLL and U-CLL with clear clinical correlations. Multivariate analyses with phenotype, genetic lesions, or DNA methylation maturation status identified high sIgM levels as a new potential independent factor for disease progression. Multiple influences on sIgM include the cell of origin, the clonal history of antigen encounter in vivo, and genetic damage. This simple marker compiles these different factors into an indicator worthy of further investigations for prediction of clinical behavior, particularly within the heterogeneous M-CLL subset.


Blood | 2015

The PI3K/mTOR inhibitor PF-04691502 induces apoptosis and inhibits microenvironmental signaling in CLL and the Eµ-TCL1 mouse model

Matthew D. Blunt; Matthew J. Carter; Marta Larrayoz; Lindsay D. Smith; Maria M. Aguilar-Hernandez; Kerry L. Cox; Thomas R.W. Tipton; Mark Reynolds; Sarah Murphy; Elizabeth Lemm; Samantha Dias; Andrew S Duncombe; Jonathan C. Strefford; Peter Johnson; Francesco Forconi; Freda K. Stevenson; Graham Packham; Mark S. Cragg; Andrew Steele

Current treatment strategies for chronic lymphocytic leukemia (CLL) involve a combination of conventional chemotherapeutics, monoclonal antibodies, and targeted signaling inhibitors. However, CLL remains largely incurable, with drug resistance and treatment relapse a common occurrence, leading to the search for novel treatments. Mechanistic target of rapamycin (mTOR)-specific inhibitors have been previously assessed but their efficacy is limited due to a positive feedback loop via mTOR complex 2 (mTORC2), resulting in activation of prosurvival signaling. In this study, we show that the dual phosphatidylinositol 3-kinase (PI3K)/mTOR inhibitor PF-04691502 does not induce an mTORC2 positive feedback loop similar to other PI3K inhibitors but does induce substantial antitumor effects. PF-04691502 significantly reduced survival coincident with the induction of Noxa and Puma, independently of immunoglobulin heavy chain variable region mutational status, CD38, and ZAP-70 expression. PF-04691502 inhibited both anti-immunoglobulin M-induced signaling and overcame stroma-induced survival signals and migratory stimuli from CXCL12. Equivalent in vitro activity was seen in the Eμ-TCL1 murine model of CLL. In vivo, PF-04691502 treatment of tumor-bearing animals resulted in a transient lymphocytosis, followed by a clear reduction in tumor in the blood, bone marrow, spleen, and lymph nodes. These data indicate that PF-04691502 or other dual PI3K/mTOR inhibitors in development may prove efficacious for the treatment of CLL, increasing our armamentarium to successfully manage this disease.


Leukemia | 2016

Longitudinal copy number, whole exome and targeted deep sequencing of 'good risk' IGHV-mutated CLL patients with progressive disease.

Matthew J. Rose-Zerilli; Jane Gibson; Jun Wang; Wi Tapper; Zadie Davis; Helen Parker; Marta Larrayoz; Helen McCarthy; Renata Walewska; Jade Forster; Anne Gardiner; Andrew Steele; Claude Chelala; Sarah Ennis; Andrew Collins; Christopher C. Oakes; David Oscier; Jonathan C. Strefford

The biological features of IGHV-M chronic lymphocytic leukemia responsible for disease progression are still poorly understood. We undertook a longitudinal study close to diagnosis, pre-treatment and post relapse in 13 patients presenting with cMBL or Stage A disease and good-risk biomarkers (IGHV-M genes, no del(17p) or del(11q) and low CD38 expression) who nevertheless developed progressive disease, of whom 10 have required therapy. Using cytogenetics, fluorescence in situ hybridisation, genome-wide DNA methylation and copy number analysis together with whole exome, targeted deep- and Sanger sequencing at diagnosis, we identified mutations in established chronic lymphocytic leukemia driver genes in nine patients (69%), non-coding mutations (PAX5 enhancer region) in three patients and genomic complexity in two patients. Branching evolutionary trajectories predominated (n=9/13), revealing intra-tumoural epi- and genetic heterogeneity and sub-clonal competition before therapy. Of the patients subsequently requiring treatment, two had sub-clonal TP53 mutations that would not be detected by standard methodologies, three qualified for the very-low-risk category defined by integrated mutational and cytogenetic analysis and yet had established or putative driver mutations and one patient developed progressive, therapy-refractory disease associated with the emergence of an IGHV-U clone. These data suggest that extended genomic and immunogenetic screening may have clinical utility in patients with apparent good-risk disease.


Haematologica | 2016

Different spectra of recurrent gene mutations in subsets of chronic lymphocytic leukemia harboring stereotyped B-cell receptors

Lesley Ann Sutton; Emma Young; Panagiotis Baliakas; Anastasia Hadzidimitriou; Theodoros Moysiadis; Karla Plevová; Davide Rossi; Jana Kmínková; Evangelia Stalika; Lone Bredo Pedersen; Jitka Malčíková; Andreas Agathangelidis; Zadie Davis; Larry Mansouri; Lydia Scarfò; Myriam Boudjoghra; Alba Navarro; Alice F. Muggen; Xiao Jie Yan; Marta Larrayoz; Panagiotis Panagiotidis; Nicholas Chiorazzi; Carsten U. Niemann; Chrysoula Belessi; Elias Campo; Jonathan C. Strefford; Anton W. Langerak; David Oscier; Gianluca Gaidano; Šárka Pospíšilová

We report on markedly different frequencies of genetic lesions within subsets of chronic lymphocytic leukemia patients carrying mutated or unmutated stereotyped B-cell receptor immunoglobulins in the largest cohort (n=565) studied for this purpose. By combining data on recurrent gene mutations (BIRC3, MYD88, NOTCH1, SF3B1 and TP53) and cytogenetic aberrations, we reveal a subset-biased acquisition of gene mutations. More specifically, the frequency of NOTCH1 mutations was found to be enriched in subsets expressing unmutated immunoglobulin genes, i.e. #1, #6, #8 and #59 (22–34%), often in association with trisomy 12, and was significantly different (P<0.001) to the frequency observed in subset #2 (4%, aggressive disease, variable somatic hypermutation status) and subset #4 (1%, indolent disease, mutated immunoglobulin genes). Interestingly, subsets harboring a high frequency of NOTCH1 mutations were found to carry few (if any) SF3B1 mutations. This starkly contrasts with subsets #2 and #3 where, despite their immunogenetic differences, SF3B1 mutations occurred in 45% and 46% of cases, respectively. In addition, mutations within TP53, whilst enriched in subset #1 (16%), were rare in subsets #2 and #8 (both 2%), despite all being clinically aggressive. All subsets were negative for MYD88 mutations, whereas BIRC3 mutations were infrequent. Collectively, this striking bias and skewed distribution of mutations and cytogenetic aberrations within specific chronic lymphocytic leukemia subsets implies that the mechanisms underlying clinical aggressiveness are not uniform, but rather support the existence of distinct genetic pathways of clonal evolution governed by a particular stereotyped B-cell receptor selecting a certain molecular lesion(s).


Leukemia | 2017

Non-coding NOTCH1 mutations in chronic lymphocytic leukemia; their clinical impact in the UK CLL4 trial

Marta Larrayoz; Matthew J. Rose-Zerilli; Latha Kadalayil; Helen L. Parker; Stuart Blakemore; Jade Forster; Zadie Davis; Andrew Steele; Andrew Collins; Monica Else; Daniel Catovsky; David Oscier; Jonathan C. Strefford

Non-coding NOTCH1 mutations in chronic lymphocytic leukemia; their clinical impact in the UK CLL4 trial


Clinical Cancer Research | 2017

The dual Syk/JAK inhibitor cerdulatinib antagonizes B-cell receptor and microenvironmental signaling in chronic lymphocytic leukemia

Matthew D. Blunt; Stefan Koehrer; Rachel Dobson; Marta Larrayoz; Sarah Wilmore; Alice Hayman; Jack Parnell; Lindsay D. Smith; Andrew Davies; Peter Johnson; Pamela B. Conley; Anjali Pandey; Jonathan C. Strefford; Freda K. Stevenson; Graham Packham; Francesco Forconi; Greg Coffey; Jan A. Burger; Andrew Steele

Purpose: B-cell receptor (BCR)–associated kinase inhibitors, such as ibrutinib, have revolutionized the treatment of chronic lymphocytic leukemia (CLL). However, these agents are not curative, and resistance is already emerging in a proportion of patients. IL4, expressed in CLL lymph nodes, can augment BCR signaling and reduce the effectiveness of BCR kinase inhibitors. Therefore, simultaneous targeting of the IL4- and BCR signaling pathways by cerdulatinib, a novel dual Syk/JAK inhibitor currently in clinical trials (NCT01994382), may improve treatment responses in patients. Experimental Design: PBMCs from patients with CLL were treated in vitro with cerdulatinib alone or in combination with venetoclax. Cell death, chemokine, and cell signaling assay were performed and analyzed by flow cytometry, immunoblotting, q-PCR, and ELISA as indicated. Results: At concentrations achievable in patients, cerdulatinib inhibited BCR- and IL4-induced downstream signaling in CLL cells using multiple readouts and prevented anti-IgM- and nurse-like cell (NLC)–mediated CCL3/CCL4 production. Cerdulatinib induced apoptosis of CLL cells, in a time- and concentration-dependent manner, and particularly in IGHV-unmutated samples with greater BCR signaling capacity and response to IL4, or samples expressing higher levels of sIgM, CD49d+, or ZAP70+. Cerdulatinib overcame anti-IgM, IL4/CD40L, or NLC-mediated protection by preventing upregulation of MCL-1 and BCL-XL; however, BCL-2 expression was unaffected. Furthermore, in samples treated with IL4/CD40L, cerdulatinib synergized with venetoclax in vitro to induce greater apoptosis than either drug alone. Conclusions: Cerdulatinib is a promising therapeutic for the treatment of CLL either alone or in combination with venetoclax, with the potential to target critical survival pathways in this currently incurable disease. Clin Cancer Res; 23(9); 2313–24. ©2016 AACR.


Haematologica | 2018

Tailored approaches grounded on immunogenetic features for refined prognostication in chronic lymphocytic leukemia

Panagiotis Baliakas; Theodoros Moysiadis; Anastasia Hadzidimitriou; Aliki Xochelli; Sabine Jeromin; Andreas Agathangelidis; Mattias Mattsson; Lesley-Ann Sutton; Eva Minga; Lydia Scarfò; Davide Rossi; Zadie Davis; Neus Villamor; Helen Parker; Jana Kotašková; Evangelia Stalika; Karla Plevová; Larry Mansouri; Diego Cortese; Alba Navarro; Julio Delgado; Marta Larrayoz; Emma Young; Achilles Anagnostopoulos; Karin E. Smedby; Gunnar Juliusson; Oonagh Sheehy; Mark A. Catherwood; Jonathan C. Strefford; Niki Stavroyianni

Chronic lymphocytic leukemia (CLL) patients with differential somatic hypermutation status of the immunoglobulin heavy variable genes, namely mutated or unmutated, display fundamental clinico-biological differences. Considering this, we assessed prognosis separately within mutated (M-CLL) and unmutated (U-CLL) CLL in 3015 patients, hypothesizing that the relative significance of relevant indicators may differ between these two categories. Within Binet A M-CLL patients, besides TP53 abnormalities, trisomy 12 and stereotyped subset #2 membership were equivalently associated with the shortest time-to-first-treatment and a treatment probability at five and ten years after diagnosis of 40% and 55%, respectively; the remaining cases exhibited 5-year and 10-year treatment probability of 12% and 25%, respectively. Within Binet A U-CLL patients, besides TP53 abnormalities, del(11q) and/or SF3B1 mutations were associated with the shortest time-to-first-treatment (5- and 10-year treatment probability: 78% and 98%, respectively); in the remaining cases, males had a significantly worse prognosis than females. In conclusion, the relative weight of indicators that can accurately risk stratify early-stage CLL patients differs depending on the somatic hypermutation status of the immunoglobulin heavy variable genes of each patient. This finding highlights the fact that compartmentalized approaches based on immunogenetic features are necessary to refine and tailor prognostication in CLL.


Blood | 2016

IL-4 enhances expression and function of surface IgM in CLL cells

Maria M. Aguilar-Hernandez; Matthew D. Blunt; Rachel Dobson; Alison Yeomans; Stephen M. Thirdborough; Marta Larrayoz; Lindsay D. Smith; Adam Linley; Jonathan C. Strefford; Andrew Davies; Peter M. W. Johnson; Natalia Savelyeva; Mark S. Cragg; Francesco Forconi; Graham Packham; Freda K. Stevenson; Andrew Steele

Collaboration


Dive into the Marta Larrayoz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Steele

University of Southampton

View shared research outputs
Top Co-Authors

Avatar

David Oscier

Royal Bournemouth Hospital

View shared research outputs
Top Co-Authors

Avatar

Zadie Davis

Royal Bournemouth Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Davide Rossi

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Graham Packham

University of Southampton

View shared research outputs
Top Co-Authors

Avatar

Mark S. Cragg

University of Southampton

View shared research outputs
Top Co-Authors

Avatar

Evangelia Stalika

Aristotle University of Thessaloniki

View shared research outputs
Researchain Logo
Decentralizing Knowledge