Network


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

Hotspot


Dive into the research topics where Amaya Viros is active.

Publication


Featured researches published by Amaya Viros.


The New England Journal of Medicine | 2012

RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors.

Fei Su; Amaya Viros; Carla Milagre; Kerstin Trunzer; Gideon Bollag; Olivia Spleiss; Jorge S. Reis-Filho; Xiangju Kong; Richard C. Koya; Keith T. Flaherty; Paul B. Chapman; Min Jung Kim; Robert Hayward; Matthew Martin; Hong Yang; Qiongqing Wang; Holly Hilton; Julie S. Hang; Johannes Noe; Maryou B. Lambros; Felipe C. Geyer; Nathalie Dhomen; Ion Niculescu-Duvaz; Alfonso Zambon; Dan Niculescu-Duvaz; Natasha Preece; Lidia Robert; Nicholas Otte; Stephen Mok; Damien Kee

BACKGROUND Cutaneous squamous-cell carcinomas and keratoacanthomas are common findings in patients treated with BRAF inhibitors. METHODS We performed a molecular analysis to identify oncogenic mutations (HRAS, KRAS, NRAS, CDKN2A, and TP53) in the lesions from patients treated with the BRAF inhibitor vemurafenib. An analysis of an independent validation set and functional studies with BRAF inhibitors in the presence of the prevalent RAS mutation was also performed. RESULTS Among 21 tumor samples, 13 had RAS mutations (12 in HRAS). In a validation set of 14 samples, 8 had RAS mutations (4 in HRAS). Thus, 60% (21 of 35) of the specimens harbored RAS mutations, the most prevalent being HRAS Q61L. Increased proliferation of HRAS Q61L-mutant cell lines exposed to vemurafenib was associated with mitogen-activated protein kinase (MAPK)-pathway signaling and activation of ERK-mediated transcription. In a mouse model of HRAS Q61L-mediated skin carcinogenesis, the vemurafenib analogue PLX4720 was not an initiator or a promoter of carcinogenesis but accelerated growth of the lesions harboring HRAS mutations, and this growth was blocked by concomitant treatment with a MEK inhibitor. CONCLUSIONS Mutations in RAS, particularly HRAS, are frequent in cutaneous squamous-cell carcinomas and keratoacanthomas that develop in patients treated with vemurafenib. The molecular mechanism is consistent with the paradoxical activation of MAPK signaling and leads to accelerated growth of these lesions. (Funded by Hoffmann-La Roche and others; ClinicalTrials.gov numbers, NCT00405587, NCT00949702, NCT01001299, and NCT01006980.).


PLOS Medicine | 2008

Improving Melanoma Classification by Integrating Genetic and Morphologic Features

Amaya Viros; Jane Fridlyand; Juergen Bauer; Konstantin Lasithiotakis; Claus Garbe; Daniel Pinkel; Boris C. Bastian

Background In melanoma, morphology-based classification systems have not been able to provide relevant information for selecting treatments for patients whose tumors have metastasized. The recent identification of causative genetic alterations has revealed mutations in signaling pathways that offer targets for therapy. Identifying morphologic surrogates that can identify patients whose tumors express such alterations (or functionally equivalent alterations) would be clinically useful for therapy stratification and for retrospective analysis of clinical trial data. Methodology/Principal Findings We defined and assessed a panel of histomorphologic measures and correlated them with the mutation status of the oncogenes BRAF and NRAS in a cohort of 302 archival tissues of primary cutaneous melanomas from an academic comprehensive cancer center. Melanomas with BRAF mutations showed distinct morphological features such as increased upward migration and nest formation of intraepidermal melanocytes, thickening of the involved epidermis, and sharper demarcation to the surrounding skin; and they had larger, rounder, and more pigmented tumor cells (all p-values below 0.0001). By contrast, melanomas with NRAS mutations could not be distinguished based on these morphological features. Using simple combinations of features, BRAF mutation status could be predicted with up to 90.8% accuracy in the entire cohort as well as within the categories of the current World Health Organization (WHO) classification. Among the variables routinely recorded in cancer registries, we identified age < 55 y as the single most predictive factor of BRAF mutation in our cohort. Using age < 55 y as a surrogate for BRAF mutation in an independent cohort of 4,785 patients of the Southern German Tumor Registry, we found a significant survival benefit (p < 0.0001) for patients who, based on their age, were predicted to have BRAF mutant melanomas in 69% of the cases. This group also showed a different pattern of metastasis, more frequently involving regional lymph nodes, compared to the patients predicted to have no BRAF mutation and who more frequently displayed satellite, in-transit metastasis, and visceral metastasis (p < 0.0001). Conclusions Refined morphological classification of primary melanomas can be used to improve existing melanoma classifications by forming subgroups that are genetically more homogeneous and likely to differ in important clinical variables such as outcome and pattern of metastasis. We expect this information to improve classification and facilitate stratification for therapy as well as retrospective analysis of existing trial data.


Cancer Discovery | 2013

Inhibiting EGF receptor or SRC family kinase signaling overcomes BRAF inhibitor resistance in melanoma

Maria Romina Girotti; Malin Pedersen; Berta Sanchez-Laorden; Amaya Viros; Samra Turajlic; Dan Niculescu-Duvaz; Alfonso Zambon; John Sinclair; Andrew Hayes; Martin Gore; Paul Lorigan; Caroline J. Springer; James Larkin; Claus Jørgensen; Richard Marais

UNLABELLED We generated cell lines resistant to BRAF inhibitors and show that the EGF receptor (EGFR)-SRC family kinase (SFK)-STAT3 signaling pathway was upregulated in these cells. In addition to driving proliferation of resistant cells, this pathway also stimulated invasion and metastasis. EGFR inhibitors cooperated with BRAF inhibitors to block the growth of the resistant cells in vitro and in vivo, and monotherapy with the broad specificity tyrosine kinase inhibitor dasatinib blocked growth and metastasis in vivo. We analyzed tumors from patients with intrinsic or acquired resistance to vemurafenib and observed increased EGFR and SFK activity. Furthermore, dasatinib blocked the growth and metastasis of one of the resistant tumors in immunocompromised mice. Our data show that BRAF inhibitor-mediated activation of EGFR-SFK-STAT3 signaling can mediate resistance in patients with BRAF-mutant melanoma. We describe 2 treatments that seem to overcome this resistance and could deliver therapeutic efficacy in patients with drug-resistant BRAF-mutant melanoma. SIGNIFICANCE Therapies that target the driver oncogenes in cancer can achieve remarkable responses if patients are stratified for treatment. However, as with conventional therapies, patients often develop acquired resistance to targeted therapies, and a proportion of patients are intrinsically resistant and fail to respond despite the presence of an appropriate driver oncogene mutation. We found that the EGFR/SFK pathway mediated resistance to vemurafenib in BRAF -mutant melanoma and that BRAF and EGFR or SFK inhibition blocked proliferation and invasion of these resistant tumors, providing potentially effective therapeutic options for these patients.


Cancer Cell | 2015

Intravital Imaging Reveals How BRAF Inhibition Generates Drug-Tolerant Microenvironments with High Integrin β1/FAK Signaling

Eishu Hirata; Maria Romina Girotti; Amaya Viros; Steven Hooper; Bradley Spencer-Dene; Michiyuki Matsuda; James Larkin; Richard Marais; Erik Sahai

Summary Intravital imaging of BRAF-mutant melanoma cells containing an ERK/MAPK biosensor reveals how the tumor microenvironment affects response to BRAF inhibition by PLX4720. Initially, melanoma cells respond to PLX4720, but rapid reactivation of ERK/MAPK is observed in areas of high stromal density. This is linked to “paradoxical” activation of melanoma-associated fibroblasts by PLX4720 and the promotion of matrix production and remodeling leading to elevated integrin β1/FAK/Src signaling in melanoma cells. Fibronectin-rich matrices with 3–12 kPa elastic modulus are sufficient to provide PLX4720 tolerance. Co-inhibition of BRAF and FAK abolished ERK reactivation and led to more effective control of BRAF-mutant melanoma. We propose that paradoxically activated MAFs provide a “safe haven” for melanoma cells to tolerate BRAF inhibition.


European Journal of Endocrinology | 2011

Analysis of the efficacy and toxicity of sorafenib in thyroid cancer: a phase II study in a UK based population.

Merina Ahmed; Yolanda Barbachano; Angela M. Riddell; Jen Hickey; K. Newbold; Amaya Viros; Kevin J. Harrington; Richard Marais; Christopher M. Nutting

AIM To evaluate the tolerability and efficacy of sorafenib in patients with thyroid carcinoma. METHODS Patients with progressive locally advanced/metastatic medullary thyroid carcinoma (MTC), or differentiated thyroid carcinoma (DTC) with non-radioiodine-avid disease, were treated with sorafenib 400 mg twice daily until disease progression. The primary endpoint was the radiological response rate (RR) at 6 months. Secondary endpoints were RR at 3, 9 and 12 months, biochemical responses, toxicity, biomarker analyses and progression free and overall survival (OS). RESULTS A total of 34 patients were recruited to the study (15 medullary and 19 differentiated). After 6 months, the RR rate was 15% and a further 74% of patients achieved stable disease in the first 6 months. After 12 months of treatment, the RR was 21%. In the MTC patients, the RR at 12 months was 25% and OS was 100%. In DTC patients corresponding rates were 18 and 79% respectively. Median overall and progression-free survival points were not reached at 19 months. Commonest adverse events included hand-foot syndrome, other skin toxicities, diarrhoea and alopecia. Dose reduction was required in 79% patients. Median time on treatment was 16.5 months. CONCLUSION This study demonstrates that sorafenib is tolerable at reduced doses over prolonged periods of time in patients with thyroid cancer. Sorafenib leads to radiological and biochemical stabilisation of disease in the majority of these patients despite dose reductions.


Science Translational Medicine | 2010

Gatekeeper Mutations Mediate Resistance to BRAF-Targeted Therapies

Steven Whittaker; Ruth S. Kirk; Robert Hayward; Alfonso Zambon; Amaya Viros; Neus Cantarino; Annette Affolter; Arnaud Nourry; Dan Niculescu-Duvaz; Caroline J. Springer; Richard Marais

Gatekeeper mutations in BRAF confer drug resistance. Getting Back on Target The identification of clinically useful small-molecule inhibitors of dysregulated proteins that trigger a cascade of detrimental downstream effects during the process of tumorigenesis requires a highly precise understanding of the specific residues responsible for the damage. However, although this information can elucidate promising candidate targets, clinical data have revealed that the utility of anticancer drugs within tumors of similar origin and mutational status is variable. It has been postulated that the haphazard effects of such drugs are the result of off-targeting effects—that is, the ability of a drug to inhibit tumor growth in a manner less specific than that which it was optimally designed for. In an era that now seeks to further personalize medicine, these off-targeting effects reveal how distant we remain from truly achieving an individualized cancer cocktail. In melanoma, the failure of the FDA-approved drug sorafenib to consistently curb disease in individuals with tumors that harbor cancer-driving mutations in BRAF—the overactive kinase presumed to be the target of the drug on the basis of its efficacy against CRAF—raised doubts as to the utility of BRAF as true melanoma target, despite its more successful application as a liver and kidney cancer therapeutic. Now, Marais and colleagues have designed a system by which to tease apart whether BRAF-mediated therapeutics such as sorafenib and PLX4720 inhibit tumor growth by homing in on the gatekeeper mutations that cause aberrant signaling in BRAF or by other nonspecific tumor-inhibiting mechanisms. Their compelling results reveal that BRAF is indeed a tractable target for melanoma, and while a plethora of tractable cancer targets are continuously being resolved, the need for proper preclinical validation—where the effects can easily be discerned as being mechanistic or off-target—is paramount to developing the right drug for the specific target. BRAF is a serine-threonine–specific protein kinase that is mutated in 2% of human cancers. Oncogenic BRAF is a validated therapeutic target that constitutively activates mitogen-activated protein kinase kinase (MEK)–extracellular signal–regulated kinase (ERK) signaling, driving tumor cell proliferation and survival. Drugs designed to target BRAF have been developed, but it is difficult to prove that they mediate their antitumor effects by inhibiting BRAF rather than by working through off-target effects. We generated drug-resistant versions of oncogenic BRAF by mutating the gatekeeper residue. Signaling by the mutant proteins was resistant to the small-molecule inhibitor sorafenib, but sorafenib still inhibited the growth of tumors driven by the mutant protein. In contrast, both BRAF signaling and tumor growth were resistant to another RAF drug, PLX4720. These data provide unequivocal evidence that sorafenib mediates its antitumor effects in a manner that is independent of its ability to target oncogenic BRAF, whereas PLX4720 inhibits tumor growth by targeting oncogenic BRAF directly.


Cancer Cell | 2015

Paradox-Breaking RAF Inhibitors that Also Target SRC Are Effective in Drug-Resistant BRAF Mutant Melanoma

Maria Romina Girotti; Filipa Lopes; Natasha Preece; Dan Niculescu-Duvaz; Alfonso Zambon; Lawrence Davies; Steven Whittaker; Grazia Saturno; Amaya Viros; Malin Pedersen; Bart M. J. M. Suijkerbuijk; Delphine Menard; Robert McLeary; Louise Johnson; Laura Fish; Sarah Ejiama; Berta Sanchez-Laorden; Juliane Hohloch; Neil O. Carragher; Kenneth G MacLeod; Garry Ashton; Anna A. Marusiak; Alberto Fusi; John Brognard; Margaret C. Frame; Paul Lorigan; Richard Marais; Caroline J. Springer

Summary BRAF and MEK inhibitors are effective in BRAF mutant melanoma, but most patients eventually relapse with acquired resistance, and others present intrinsic resistance to these drugs. Resistance is often mediated by pathway reactivation through receptor tyrosine kinase (RTK)/SRC-family kinase (SFK) signaling or mutant NRAS, which drive paradoxical reactivation of the pathway. We describe pan-RAF inhibitors (CCT196969, CCT241161) that also inhibit SFKs. These compounds do not drive paradoxical pathway activation and inhibit MEK/ERK in BRAF and NRAS mutant melanoma. They inhibit melanoma cells and patient-derived xenografts that are resistant to BRAF and BRAF/MEK inhibitors. Thus, paradox-breaking pan-RAF inhibitors that also inhibit SFKs could provide first-line treatment for BRAF and NRAS mutant melanomas and second-line treatment for patients who develop resistance.


Nature | 2014

Ultraviolet radiation accelerates BRAF-driven melanomagenesis by targeting TP53

Amaya Viros; Berta Sanchez-Laorden; Malin Pedersen; Simon J. Furney; Joel Rae; Kate Hogan; Sarah Ejiama; Maria Romina Girotti; Martin G. Cook; Nathalie Dhomen; Richard Marais

Cutaneous melanoma is epidemiologically linked to ultraviolet radiation (UVR), but the molecular mechanisms by which UVR drives melanomagenesis remain unclear. The most common somatic mutation in melanoma is a V600E substitution in BRAF, which is an early event. To investigate how UVR accelerates oncogenic BRAF-driven melanomagenesis, we used a BRAF(V600E) mouse model. In mice expressing BRAF(V600E) in their melanocytes, a single dose of UVR that mimicked mild sunburn in humans induced clonal expansion of the melanocytes, and repeated doses of UVR increased melanoma burden. Here we show that sunscreen (UVA superior, UVB sun protection factor (SPF) 50) delayed the onset of UVR-driven melanoma, but only provided partial protection. The UVR-exposed tumours showed increased numbers of single nucleotide variants and we observed mutations (H39Y, S124F, R245C, R270C, C272G) in the Trp53 tumour suppressor in approximately 40% of cases. TP53 is an accepted UVR target in human non-melanoma skin cancer, but is not thought to have a major role in melanoma. However, we show that, in mice, mutant Trp53 accelerated BRAF(V600E)-driven melanomagenesis, and that TP53 mutations are linked to evidence of UVR-induced DNA damage in human melanoma. Thus, we provide mechanistic insight into epidemiological data linking UVR to acquired naevi in humans. Furthermore, we identify TP53/Trp53 as a UVR-target gene that cooperates with BRAF(V600E) to induce melanoma, providing molecular insight into how UVR accelerates melanomagenesis. Our study validates public health campaigns that promote sunscreen protection for individuals at risk of melanoma.


Cancer Discovery | 2012

Metformin accelerates the growth of BRAF V600E-driven melanoma by upregulating VEGF-A.

Matthew Martin; Robert Hayward; Amaya Viros; Richard Marais

UNLABELLED The antidiabetic drug metformin has antitumor activity in a variety of cancers because it blocks cell growth by inhibiting TORC1. Here, we show that melanoma cells that are driven by oncogenic BRAF are resistant to the growth-inhibitory effects of metformin because RSK sustains TORC1 activity even when AMP-activated protein kinase (AMPK) is activated. We further show that AMPK targets the dual-specificity protein phosphatase DUSP6 for degradation and this increases ERK activity, which then upregulates the VEGF-A protein. Critically, this drives angiogenesis and accelerates the growth of BRAF-driven tumors in mice. Unexpectedly, however, when VEGF signaling is inhibited, instead of accelerating tumor growth, metformin inhibits tumor growth. Thus, we show that BRAF-driven melanoma cells are resistant to the antigrowth effects of AMPK and that AMPK mediates cell-autonomous and cell-nonautonomous effects that accelerate the growth of these cells in vivo. SIGNIFICANCE Metformin inhibits the growth of most tumor cells, but BRAF-mutant melanoma cells are resistant to metformin in vitro, and metformin accelerates their growth in vivo. Unexpectedly, VEGF inhibitors and metformin synergize to suppress the growth of BRAF-mutant tumors, revealing a combination of drugs that may be effective in these patients.


Cancer Discovery | 2016

Application of Sequencing, Liquid Biopsies, and Patient-Derived Xenografts for Personalized Medicine in Melanoma

Maria Romina Girotti; Gabriela Gremel; Rebecca Lee; E. Galvani; Dominic G. Rothwell; Amaya Viros; Amit Kumar Mandal; Kok Haw Jonathan Lim; Grazia Saturno; Simon J. Furney; Franziska Baenke; Malin Pedersen; Jane Rogan; Jacqueline Swan; Matthew R. Smith; Alberto Fusi; Deemesh Oudit; Nathalie Dhomen; Ged Brady; Paul Lorigan; Caroline Dive; Richard Marais

UNLABELLED Targeted therapies and immunotherapies have transformed melanoma care, extending median survival from ∼9 to over 25 months, but nevertheless most patients still die of their disease. The aim of precision medicine is to tailor care for individual patients and improve outcomes. To this end, we developed protocols to facilitate individualized treatment decisions for patients with advanced melanoma, analyzing 364 samples from 214 patients. Whole exome sequencing (WES) and targeted sequencing of circulating tumor DNA (ctDNA) allowed us to monitor responses to therapy and to identify and then follow mechanisms of resistance. WES of tumors revealed potential hypothesis-driven therapeutic strategies for BRAF wild-type and inhibitor-resistant BRAF-mutant tumors, which were then validated in patient-derived xenografts (PDX). We also developed circulating tumor cell-derived xenografts (CDX) as an alternative to PDXs when tumors were inaccessible or difficult to biopsy. Thus, we describe a powerful technology platform for precision medicine in patients with melanoma. SIGNIFICANCE Although recent developments have revolutionized melanoma care, most patients still die of their disease. To improve melanoma outcomes further, we developed a powerful precision medicine platform to monitor patient responses and to identify and validate hypothesis-driven therapies for patients who do not respond, or who develop resistance to current treatments.

Collaboration


Dive into the Amaya Viros's collaboration.

Top Co-Authors

Avatar

Richard Marais

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Malin Pedersen

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nathalie Dhomen

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Caroline J. Springer

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Paul Lorigan

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Dan Niculescu-Duvaz

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Alfonso Zambon

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Grazia Saturno

University of Manchester

View shared research outputs
Researchain Logo
Decentralizing Knowledge