Network


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

Hotspot


Dive into the research topics where Maria Romina Girotti is active.

Publication


Featured researches published by Maria Romina Girotti.


Cell | 2015

Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity

Santiago Zelenay; Annemarthe G. van der Veen; Jan P. Böttcher; Kathryn J. Snelgrove; Neil C. Rogers; Sophie E. Acton; Probir Chakravarty; Maria Romina Girotti; Richard Marais; Sergio A. Quezada; Erik Sahai; Caetano Reis e Sousa

Summary The mechanisms by which melanoma and other cancer cells evade anti-tumor immunity remain incompletely understood. Here, we show that the growth of tumors formed by mutant BrafV600E mouse melanoma cells in an immunocompetent host requires their production of prostaglandin E2, which suppresses immunity and fuels tumor-promoting inflammation. Genetic ablation of cyclooxygenases (COX) or prostaglandin E synthases in BrafV600E mouse melanoma cells, as well as in NrasG12D melanoma or in breast or colorectal cancer cells, renders them susceptible to immune control and provokes a shift in the tumor inflammatory profile toward classic anti-cancer immune pathways. This mouse COX-dependent inflammatory signature is remarkably conserved in human cutaneous melanoma biopsies, arguing for COX activity as a driver of immune suppression across species. Pre-clinical data demonstrate that inhibition of COX synergizes with anti-PD-1 blockade in inducing eradication of tumors, implying that COX inhibitors could be useful adjuvants for immune-based therapies in cancer patients.


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.


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 | 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.


Science Signaling | 2014

BRAF Inhibitors Induce Metastasis in RAS Mutant or Inhibitor-Resistant Melanoma Cells By Reactivating MEK and ERK Signaling

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

When therapy leads to cancer metastasis, knowing where else to target in the pathway may be the key to successful treatment. Blocking Melanoma Metastasis Although inhibitors of the mutant BRAF kinase are effective in some melanoma patients, intrinsic or acquired resistance to the drug is common. Furthermore, the growth of melanoma tumors with concomitant mutations in guanosine triphosphatase RAS, which activated kinases in the RAF family, is paradoxically accelerated by BRAF inhibition. RAF is the first kinase in a three-kinase cascade [the RAF–MEK (mitogen-activated protein kinase kinase)–ERK (extracellular signal–regulated kinase) pathway] that is involved in cell proliferation. Using proteomics, patient material, and mouse models, Sanchez-Laorden et al. found that BRAF inhibition paradoxically stimulated MEK and ERK signaling to induce metastasis of melanoma cells with mutant BRAF, resistance to a BRAF inhibitor, or mutant RAS. Combined treatment with a MEK inhibitor prevented BRAF inhibitor–induced metastasis in mice. Thus, combination therapies may be best to prevent both primary tumor growth and drug-induced metastasis. Melanoma is a highly metastatic and lethal form of skin cancer. The protein kinase BRAF is mutated in about 40% of melanomas, and BRAF inhibitors improve progression-free and overall survival in these patients. However, after a relatively short period of disease control, most patients develop resistance because of reactivation of the RAF–ERK (extracellular signal–regulated kinase) pathway, mediated in many cases by mutations in RAS. We found that BRAF inhibition induces invasion and metastasis in RAS mutant melanoma cells through a mechanism mediated by the reactivation of the MEK (mitogen-activated protein kinase kinase)–ERK pathway, increased expression and secretion of interleukin 8, and induction of protease-dependent invasion. These events were accompanied by a cell morphology switch from predominantly rounded to predominantly elongated cells. We also observed similar responses in BRAF inhibitor–resistant melanoma cells. These data show that BRAF inhibitors can induce melanoma cell invasion and metastasis in tumors that develop resistance to these drugs.


Cancer Cell | 2016

Inhibiting Drivers of Non-mutational Drug Tolerance Is a Salvage Strategy for Targeted Melanoma Therapy.

Holly Brunton; Emily J. Rowling; Jennifer Ferguson; Imanol Arozarena; Zsofia Miskolczi; Jessica L. Lee; Maria Romina Girotti; Richard Marais; Mitchell P. Levesque; Reinhard Dummer; Dennie T. Frederick; Keith T. Flaherty; Zachary A. Cooper; Jennifer A. Wargo; Claudia Wellbrock

Summary Once melanomas have progressed with acquired resistance to mitogen-activated protein kinase (MAPK)-targeted therapy, mutational heterogeneity presents a major challenge. We therefore examined the therapy phase before acquired resistance had developed and discovered the melanoma survival oncogene MITF as a driver of an early non-mutational and reversible drug-tolerance state, which is induced by PAX3-mediated upregulation of MITF. A drug-repositioning screen identified the HIV1-protease inhibitor nelfinavir as potent suppressor of PAX3 and MITF expression. Nelfinavir profoundly sensitizes BRAF and NRAS mutant melanoma cells to MAPK-pathway inhibitors. Moreover, nelfinavir is effective in BRAF and NRAS mutant melanoma cells isolated from patients progressed on MAPK inhibitor (MAPKi) therapy and in BRAF/NRAS/PTEN mutant tumors. We demonstrate that inhibiting a driver of MAPKi-induced drug tolerance could improve current approaches of targeted melanoma therapy.


Molecular Oncology | 2014

No longer an untreatable disease: How targeted and immunotherapies have changed the management of melanoma patients

Maria Romina Girotti; Grazia Saturno; Paul Lorigan; Richard Marais

The discovery that BRAF is a driver oncogene in cancer, and complementary improvements in our understanding of the immune system have resulted in new targeted and immune‐therapies for metastatic melanoma. Targeted therapies achieve impressive clinical results in carefully selected patients but the development of resistance seems inevitable in most cases. Conversely, immune‐checkpoints inhibitors can achieve long‐term remission and cures, but in a smaller proportion of patients, and biomarkers to predict which patients will respond are not available. Nevertheless, melanoma has led the evolution of cancer treatment from relatively nonspecific cytotoxic agents to highly selective therapies and here we review the lessons from this paradigm shift in treatment and the opportunities for further improvements in outcomes for melanoma patients.


Nature Medicine | 2015

Epigenetic activation of a cryptic TBC1D16 transcript enhances melanoma progression by targeting EGFR

Miguel Vizoso; Humberto J. Ferreira; Paula Lopez-Serra; F. Javier Carmona; Anna Martínez-Cardús; Maria Romina Girotti; Alberto Villanueva; Sonia Guil; Catia Moutinho; Julia Liz; Anna Portela; Holger Heyn; Sebastian Moran; August Vidal; María Martínez-Iniesta; Jose Luis Manzano; Maria Teresa Fernandez-Figueras; Elena Elez; Eva Muñoz-Couselo; Rafael Botella-Estrada; A. Berrocal; Fredrik Pontén; Joost van den Oord; William M. Gallagher; Dennie T. Frederick; Keith T. Flaherty; Ultan McDermott; Paul Lorigan; Richard Marais; Manel Esteller

Metastasis is responsible for most cancer-related deaths, and, among common tumor types, melanoma is one with great potential to metastasize. Here we study the contribution of epigenetic changes to the dissemination process by analyzing the changes that occur at the DNA methylation level between primary cancer cells and metastases. We found a hypomethylation event that reactivates a cryptic transcript of the Rab GTPase activating protein TBC1D16 (TBC1D16-47 kDa; referred to hereafter as TBC1D16-47KD) to be a characteristic feature of the metastatic cascade. This short isoform of TBC1D16 exacerbates melanoma growth and metastasis both in vitro and in vivo. By combining immunoprecipitation and mass spectrometry, we identified RAB5C as a new TBC1D16 target and showed that it regulates EGFR in melanoma cells. We also found that epigenetic reactivation of TBC1D16-47KD is associated with poor clinical outcome in melanoma, while conferring greater sensitivity to BRAF and MEK inhibitors.

Collaboration


Dive into the Maria Romina Girotti's collaboration.

Top Co-Authors

Avatar

Richard Marais

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Paul Lorigan

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Amaya Viros

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Malin Pedersen

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Rebecca Lee

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Nathalie Dhomen

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amit Kumar Mandal

Council of Scientific and Industrial Research

View shared research outputs
Top Co-Authors

Avatar

Grazia Saturno

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge