Network


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

Hotspot


Dive into the research topics where Alessandra Curioni-Fontecedro is active.

Publication


Featured researches published by Alessandra Curioni-Fontecedro.


Journal of Thoracic Oncology | 2015

Targeted Therapy for Patients with BRAF-Mutant Lung Cancer Results from the European EURAF Cohort

Oliver Gautschi; Julie Milia; Bastien Cabarrou; Marie-Virginia Bluthgen; Benjamin Besse; Egbert F. Smit; Juergen Wolf; Solange Peters; Martin Früh; Dieter Koeberle; Youssouf Oulkhouir; Martin Schuler; Alessandra Curioni-Fontecedro; Benjamin Huret; Mallorie Kerjouan; Sebastian Michels; Georg Pall; Sacha I. Rothschild; Gerald Schmid-Bindert; Matthias Scheffler; Rémi Veillon; Luciano Wannesson; Joachim Diebold; G. Zalcman; Thomas Filleron; Julien Mazieres

Introduction: Approximately 2% of lung adenocarcinomas have BRAF (v-Raf murine sarcoma viral oncogene homolog B) mutations, including V600E and other types. Vemurafenib, dabrafenib, and sorafenib as BRAF inhibitors are currently tested in clinical trials, but access for patients is limited. The aim of this study was to document the clinical course of patients treated outside of clinical trials. Methods: We conducted a retrospective multicenter cohort study in Europe of patients with advanced BRAF-mutant lung cancer treated with known BRAF inhibitors. Data were anonymized and centrally assessed for age, gender, smoking, histology, stage, local molecular diagnostic results, systemic therapies, and survival. Best response was assessed locally by RECIST1.1. Results: We documented 35 patients treated in 17 centers with vemurafenib, dabrafenib, or sorafenib. Median age was 63 years (range 42–85); gender was balanced; 14 (40%) were never smokers; all (100%) had adenocarcinoma; 29 (83%) had V600E; 6 (17%) had other mutations; one of them had a concomitant KRAS mutation. Thirty (86%) patients had chemotherapy in the first line. Overall survival with first-line therapy was 25.3 months for V600E and 11.8 months for non-V600E. Thirty-one patients received one BRAF inhibitor, and four received a second inhibitor. Overall response rate with BRAF therapy was 53%, and disease control rate was 85%. Median progression-free survival with BRAF therapy was 5.0 months, and overall survival was 10.8 months. Conclusions: These results confirm the activity of targeted therapy in patients with BRAF-mutant lung adenocarcinoma. Further trials are warranted to study combination therapies and drug resistance mechanisms.


British Journal of Cancer | 2015

Frequent PD-L1 expression in testicular germ cell tumors.

Christian Fankhauser; Alessandra Curioni-Fontecedro; V Allmann; Joerg Beyer; Verena Tischler; Tullio Sulser; Holger Moch; Peter Bode

Background:Many testicular germ cell cancers are curable despite metastatic disease, but about 10–15% of patients fail cisplatin-based first-line treatment. Immunotherapy is considered as additional treatment approach for these patients. Inhibition of the interaction between Programmed Death Receptor 1 (PD-1) and Programmed Death Receptor Ligand 1 (PD-L1) enhances T-cell responses in vitro and mediates clinical antitumour activity. We analysed the expression of PD-L1 in testicular germ cell tumours to evaluate its potential as target for immunotherapeutic strategies.Methods:Immunohistochemistry was performed in 479 formalin-fixed paraffin-embedded specimens using a rabbit monoclonal antibody (E1L3N). The tissue microarray consisted of 208 pure seminomas, 121 non-seminomas, 20 intratubular germ cell neoplasia unclassified (IGCNU) and 20 specimens of non-neoplastic testicular tissue.Results:Programmed Death Receptor Ligand-1 expression was found in 73% of all seminomas and in 64% of all non-seminomas. None of 20 IGCNU and none of 20 normal tissue specimens exhibited PD-L1 expression. PD-L1 positive stromal cells were only detected in seminomas, but not in non-seminomas. The anti PD-L1 antibody showed a pre-dominantly membranous staining pattern in testicular tumour cells, as well as expression in stromal cells.Conclusions:This frequent expression of PD-L1 in human testicular germ cell tumours suggests that patients with testicular germ cell tumours could profit from immunotherapeutic strategies using anti-PD1 and anti-PDL1 antibodies.


The Lancet Respiratory Medicine | 2017

Erlotinib and bevacizumab in patients with advanced non-small-cell lung cancer and activating EGFR mutations (BELIEF): an international, multicentre, single-arm, phase 2 trial.

Rafael Rosell; Urania Dafni; Enriqueta Felip; Alessandra Curioni-Fontecedro; Oliver Gautschi; Solange Peters; Bartomeu Massuti; Ramon Palmero; Santiago Ponce Aix; Enric Carcereny; Martin Früh; Miklos Pless; Sanjay Popat; Athanasios Kotsakis; Sinead Cuffe; P. Bidoli; Adolfo Favaretto; Patrizia Froesch; Noemi Reguart; Javier Puente; Linda Coate; Fabrice Barlesi; Daniel Rauch; Mike Thomas; Carlos Camps; José Gómez-Codina; Margarita Majem; Rut Porta; Riyaz Shah; Emer Hanrahan

BACKGROUND The tyrosine kinase inhibitor erlotinib improves the outcomes of patients with advanced non-small-cell lung carcinoma (NSCLC) harbouring epidermal growth factor receptor (EGFR) mutations. The coexistence of the T790M resistance mutation with another EGFR mutation in treatment-naive patients has been associated with a shorter progression-free survival to EGFR inhibition than in the absence of the T790M mutation. To test this hypothesis clinically, we developed a proof-of-concept study, in which patients with EGFR-mutant NSCLC were treated with the combination of erlotinib and bevacizumab, stratified by the presence of the pretreatment T790M mutation. METHODS BELIEF was an international, multicentre, single-arm, phase 2 trial done at 29 centres in eight European countries. Eligible patients were aged 18 years or older and had treatment-naive, pathologically confirmed stage IIIB or stage IV lung adenocarcinoma with a confirmed, activating EGFR mutation (exon 19 deletion or L858R mutation). Patients received oral erlotinib 150 mg per day and intravenous bevacizumab 15 mg/kg every 21 days and were tested centrally for the pretreatment T790M resistance mutation with a peptide nucleic acid probe-based real-time PCR. The primary endpoint was progression-free survival. The primary efficacy analysis was done in the intention-to-treat population and was stratified into two parallel substudies according to the centrally confirmed pretreatment T790M mutation status of enrolled patients (T790M positive or negative). The safety analysis was done in all patients that have received at least one dose of trial treatment. This trial was registered with ClinicalTrials.gov, number NCT01562028. FINDINGS Between June 11, 2012, and Oct 28, 2014, 109 patients were enrolled and included in the efficacy analysis. 37 patients were T790M mutation positive and 72 negative. The overall median progression-free survival was 13·2 months (95% CI 10·3-15·5), with a 12 month progression-free survival of 55% (95% CI 45-64). The primary endpoint was met only in substudy one (T790M-positive patients). In the T790M-positive group, median progression-free survival was 16·0 months (12·7 to not estimable), with a 12 month progression-free survival of 68% (50-81), whereas in the T790M-negative group, median progression-free survival was 10·5 months (9·4-14·2), with a 12 month progression-free survival of 48% (36-59). Of 106 patients included in the safety analysis, five had grade 4 adverse events (one acute coronary syndrome, one biliary tract infection, one other neoplasms, and two colonic perforations) and one died due to sepsis. INTERPRETATION The BELIEF trial provides further evidence of benefit for the combined use of erlotinib and bevacizumab in patients with NSCLC harbouring activating EGFR mutations. FUNDING European Thoracic Oncology Platform, Roche.


PLOS ONE | 2011

Expression of MAGE-C1/CT7 and MAGE-C2/CT10 predicts lymph node metastasis in melanoma patients.

Alessandra Curioni-Fontecedro; Natko Nuber; Daniela Mihic-Probst; Burkhardt Seifert; Davide Soldini; Reinhard Dummer; Alexander Knuth; Maries van den Broek; Holger Moch

MAGE-C1/CT7 and MAGE-C2/CT10 are members of the large MAGE family of cancer-testis (CT) antigens. CT antigens are promising targets for immunotherapy in cancer because their expression is restricted to cancer and germ line cells and a proportion of cancer patients presents with immune responses against CT antigens, which clearly demonstrates their immunogenicity. This study investigates the expression of MAGE-C1/CT7 and MAGE-C2/CT10 in primary and metastatic melanoma. Immunohistochemical staining of tissue microarrays that consisted of 59 primary malignant melanomas of the skin, 163 lymph node and distant melanoma metastases and 68 melanoma cell lines was performed. We found MAGE-C1/CT7 expression in 15 out of 50 (24%) primary melanomas and 15 out of 50 (24%) cell lines, whereas MAGE-C2/CT10 was detected in 17 out of 51 (33%) primary melanomas and 14 out of 68 (17%) cell lines. MAGE-C1/CT7 and MAGE-C2/CT10 were both detected in 40% of melanoma metastases. Patients with MAGE-C1/CT7 or MAGE-C2/CT10 positive primary melanoma had significantly more lymph node metastases (p = 0.005 and p<0.001, resp.). Prediction of lymph node metastasis by MAGE-C1/CT7 and MAGE-C2/CT10 was independent of tumor cell proliferation rate (Ki67 labeling index) in a multivariate analysis (p = 0.01). Our results suggest that the expression of MAGE-C1/CT7 and MAGE-C2/CT10 in primary melanoma is a potent predictor of sentinel lymph node metastasis.


Leukemia | 2008

MAGE-C1/CT7 is the dominant cancer-testis antigen targeted by humoral immune responses in patients with multiple myeloma

Alessandra Curioni-Fontecedro; A. Knights; Marianne Tinguely; Natko Nuber; Christoph Schneider; C W Thomson; L von Boehmer; W Bossart; Steffen Pahlich; Heinz Gehring; Holger Moch; Christoph Renner; Alexander Knuth; Alfred Zippelius

MAGE-C1/CT7 is the dominant cancer-testis antigen targeted by humoral immune responses in patients with multiple myeloma


Proceedings of the National Academy of Sciences of the United States of America | 2010

Fine analysis of spontaneous MAGE-C1/CT7–specific immunity in melanoma patients

Natko Nuber; Alessandra Curioni-Fontecedro; Claudia Matter; Davide Soldini; Jean Marie Tiercy; Lotta von Boehmer; Holger Moch; Reinhard Dummer; Alexander Knuth; Maries van den Broek

Cancer/testis (CT) antigens represent prime candidates for immunotherapy in cancer patients, because their expression is restricted to cancer cells and germ cells of the testis. MAGE-C1/CT7 is a CT antigen that is highly expressed in several types of cancers. Spontaneous occurrence of CT7-specific antibodies was previously detected by SEREX screen in a melanoma patient. However, naturally occurring CT7-specific T-cell responses have thus far not been detected. Peripheral blood mononuclear cells (PBMCs) from 26 metastatic melanoma patients expressing CT7 in their tumor lesions (CT7+) were analyzed for CT7-specific T-cell responses using overlapping peptides. CT7-specific CD4+ T-cell responses were detected in three patients (11.5%). These CT7-specific CD4+ T-cell responses were detectable in melanoma patients’ PBMCs exclusively from preexisting CD45RA− memory CD4+ T-cell pool. Additional CT7-specific memory CD4+ T-cell responses were detected in CT7+ melanoma patients after depletion of CD4+CD25high Treg cells showing that Treg cells impact on CT7-specific CD4+ T cells in melanoma patients. CT7-specific CD4+ T-cell clones were generated and used to define minimal epitopes, restriction elements, and confirm the recognition of naturally processed antigen. Surprisingly, these clones were able to secrete perforin and exert cytotoxicity. This study shows that CT7 can induce specific cellular immunity in melanoma patients. Based on these findings, CT7 will be further explored as a potential vaccine for melanoma immunotherapy.


Annals of Oncology | 2015

Searching for targets for the systemic therapy of mesothelioma

Rolf A. Stahel; Walter Weder; E. Felley-Bosco; Ulf Petrausch; Alessandra Curioni-Fontecedro; I. Schmitt-Opitz; Solange Peters

Malignant mesothelioma is an incurable disease associated with asbestos exposure arising in the pleural cavity and less frequently in the peritoneal cavity. Platinum-based combination chemotherapy with pemetrexed is the established standard of care. Multimodality approaches including surgery and radiotherapy are being investigated. Increasing knowledge about the molecular characteristics of mesothelioma had led to the identification of novel potential targets for systemic therapy. Current evidence suggests pathways activated in response to merlin deficiency, including Pi3K/mTOR and the focal adhesion kinase, as well as immunotherapeutic approaches to be most promising. This review elaborates on the rationale behind targeted approaches that have been and are undergoing exploration in mesothelioma and summarizes available clinical results and ongoing efforts to improve the systemic therapy of mesothelioma.


PLOS ONE | 2016

Targeting the mTOR Complex by Everolimus in NRAS Mutant Neuroblastoma

Michael K. Kiessling; Alessandra Curioni-Fontecedro; Panagiotis Samaras; Silvia Lang; Michael Scharl; Adriano Aguzzi; Derek A. Oldrige; John M. Maris; Gerhard Rogler

High-risk neuroblastoma remains lethal in about 50% of patients despite multimodal treatment. Recent attempts to identify molecular targets for specific therapies have shown that Neuroblastoma RAS (NRAS) is significantly mutated in a small number of patients. However, few inhibitors for the potential treatment for NRAS mutant neuroblastoma have been investigated so far. In this in-vitro study, we show that MEK inhibitors AZD6244, MEK162 and PD0325901 block cell growth in NRAS mutant neuroblastoma cell lines but not in NRAS wild-type cell lines. Several studies show that mutant NRAS leads to PI3K pathway activation and combined inhibitors of PI3K/mTOR effectively block cell growth. However, we observed the combination of MEK inhibitors with PI3K or AKT inhibitors did not show synergestic effects on cell growth. Thus, we tested single mTOR inhibitors Everolimus and AZD8055. Interestingly, Everolimus and AZD8055 alone were sufficient to block cell growth in NRAS mutant cell lines but not in wild-type cell lines. We found that Everolimus alone induced apoptosis in NRAS mutant neuroblastoma. Furthermore, the combination of mTOR and MEK inhibitors resulted in synergistic growth inhibition. Taken together, our results show that NRAS mutant neuroblastoma can be targeted by clinically available Everolimus alone or in combination with MEK inhibitors which could impact future clinical studies.


Annals of Oncology | 2015

A novel germline mutation of PDGFR-β might be associated with clinical response of colorectal cancer to regorafenib

Markus Rechsteiner; Peter Wild; M. K. Kiessling; Annette Bohnert; Qing Zhong; Rolf A. Stahel; Holger Moch; Alessandra Curioni-Fontecedro

ABSTRACT This is the first description of a germline mutation of the PDGFR-β gene, that correlates with response to regorafenib in a patient with metastatic colorectal cancer.This is the first description of a germline mutation of the PDGFR-β gene, that correlates with response to regorafenib in a patient with metastatic colorectal cancer.


Journal of Thoracic Oncology | 2017

Randomized phase III trial of erlotinib versus docetaxel in patients with advanced squamous cell non-small cell lung cancer failing first line platinum-based doublet chemotherapy stratified by Veristrat good versus Veristrat poor. The European Thoracic Oncology Platform (ETOP) EMPHASIS-lung trial.

Solange Peters; Rolf A. Stahel; Urania Dafni; Santiago Ponce Aix; Bartomeu Massuti; Oliver Gautschi; Linda Coate; Ana López Martín; Robbert van Heemst; Thierry Berghmans; Peter Meldgaard; Manuel Cobo Dols; Javier Garde Noguera; Alessandra Curioni-Fontecedro; Daniel Rauch; Michael Mark; Sinead Cuffe; Bonne Biesma; Arjen M.J. van Henten; Oscar Juan Vidal; Ramón Palmero Sanchez; José Carlos Villa Guzmán; Ricardo Collado Martin; Sergio Peralta; Amelia Insa; Yvonne Summers; István Láng; Anne M. Horgan; Fortunato Ciardiello; Sander de Hosson

Introduction Docetaxel and erlotinib are registered second‐line treatments for wild‐type EGFR NSCLC. Previous studies suggested a predictive value of the VeriStrat test in second‐line therapy of NSCLC, classifying patients as either VeriStrat good or VeriStrat poor. EMPHASIS‐lung aimed at exploring this predictive effect in patients with squamous cell NSCLC. The trial closed prematurely because of low accrual and results from other trials. Our analysis includes an exploratory combined analysis with results from the PROSE trial. Methods EMPHASIS‐lung was a randomized phase III multicenter trial exploring the differential effect of second‐line erlotinib versus docetaxel on progression‐free survival (PFS) in VeriStrat good versus VeriStrat poor patients with squamous cell NSCLC. Results A total of 80 patients were randomized, with 72.5% categorized as VeriStrat good. Patient characteristics were balanced between VeriStrat status and treatment groups. The median PFS times with docetaxel and erlotinib treatment in the VeriStrat good cohort were 4.1 and 1.6 months, respectively, versus 1.9 and 2.1 months, respectively, in the VeriStrat poor cohort. The median overall survival (OS) times with docetaxel and erlotinib treatment in the VeriStrat good cohort were 7.8 and 8.4 months, respectively, and 4.4 and 5.2 months, respectively, in the VeriStrat poor cohort. An additional exploratory analysis was performed; in it, 47 patients from the squamous cell subgroup of PROSE were included in a combined analysis, contributing with 45 PFS and 41 OS events. Conclusions The final analysis of EMPHASIS‐lung did not show a differential effect on PFS for erlotinib versus docetaxel stratified by VeriStrat status. Similarly, in the combined analysis, no significant treatment by VeriStrat status interaction was observed (interaction p = 0.24 for PFS and 0.45 for OS, stratified by study).

Collaboration


Dive into the Alessandra Curioni-Fontecedro's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Früh

University of St. Gallen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge