Network


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

Hotspot


Dive into the research topics where Francesco Sclafani is active.

Publication


Featured researches published by Francesco Sclafani.


Journal of Hepatology | 2012

Usefulness of alpha-fetoprotein response in patients treated with sorafenib for advanced hepatocellular carcinoma

Nicola Personeni; Silvia Bozzarelli; Tiziana Pressiani; Lorenza Rimassa; Maria Chiara Tronconi; Francesco Sclafani; Carlo Carnaghi; Vittorio Pedicini; Laura Giordano; Armando Santoro

BACKGROUND & AIMS Tumor shrinkage has been considered a fundamental surrogate efficacy measure for new cancer treatments. However, in patients treated with sorafenib for advanced hepatocellular carcinoma (HCC), tumor shrinkage rarely accompanies increased survival, thereby questioning the prognostic value of imaging-based Response Evaluation Criteria in Solid Tumors (RECIST). We investigated the prognostic usefulness of a decrease in serum alpha-fetoprotein (AFP) and compared it to RECIST. METHODS In HCC patients treated with sorafenib with baseline AFP >20 ng/ml, AFP response was defined as a >20% decrease in AFP during 8weeks of treatment. Patients were also assessed by RECIST and were categorized as having radiologically proven progressive disease or disease control (consisting of complete or partial responses and stable disease). Comparisons of survival by RECIST and AFP response were corrected for guarantee-time bias by the landmark method. RESULTS We evaluated 85 patients for AFP response, among them, 82 were also evaluated by RECIST. In the analysis of AFP response, 32 out of 85 patients (37.6%) were responders, whereas 58 out of 82 patients (70.7%) achieved disease control. In landmark analysis, the hazard ratios (HR) for survival according to AFP response and disease control were 0.59 (p=0.040) and 1.03 (p=0.913), respectively. In multivariate analysis, only AFP response (HR=0.52; p=0.009) and Cancer of the Liver Italian Program dichotomized stage (HR=0.42; p=0.002) were prognostic factors of survival. CONCLUSIONS Assessment of AFP response may be considered as an alternative to RECIST to capture sorafenib activity in HCC.


OncoTargets and Therapy | 2014

Emerging molecular targets in oncology: clinical potential of MET/hepatocyte growth-factor inhibitors

Elizabeth C. Smyth; Francesco Sclafani; David Cunningham

The MET/hepatocyte growth-factor (HGF) signaling pathway plays a key role in the processes of embryogenesis, wound healing, and organ regeneration. Aberrant activation of MET/HGF occurs through multiple mechanisms including gene amplification, mutation, protein overexpression, and abnormal gene splicing interrupting autocrine and paracrine regulatory feedback mechanisms. In many cancers including non-small-cell lung cancer, colorectal, gastric, renal, and hepatocellular cancer, dysregulation of MET may lead to a more aggressive cancer phenotype and may be a negative prognostic indicator. Successful therapeutic targeting of the MET/HGF pathway has been achieved using monoclonal antibodies against the MET receptor and its ligand HGF in addition to MET-specific and multitargeted small-molecule tyrosine-kinase inhibitors with several drugs in late-phase clinical trials including onartuzumab, rilotumumab, tivantinib, and cabozantinib. MET frequently interacts with other key oncogenic tyrosine kinases including epidermal growth-factor receptor (EGFR) and HER-3 and these interactions may be responsible for resistance to anti-EGFR therapies. Similarly, resistance to MET inhibition may be mediated through EGFR activation, or alternatively by increasing levels of MET amplification or acquisition of novel “gatekeeper” mutations. In order to optimize development of effective inhibitors of the MET/HGF pathway clinical trials must be enriched for patients with demonstrable MET-pathway dysregulation for which robustly standardized and validated assays are required.


European Journal of Cancer | 2015

Clinical trial designs for rare diseases: Studies developed and discussed by the International Rare Cancers Initiative

Jan Bogaerts; Matthew R. Sydes; Nicola Keat; Andrea McConnell; Al B. Benson; Alan Ho; Arnaud Roth; Catherine Fortpied; Cathy Eng; Clare Peckitt; Corneel Coens; Curtis A. Pettaway; Dirk Arnold; Emma Hall; Ernie Marshall; Francesco Sclafani; Helen Hatcher; Helena M. Earl; Isabelle Ray-Coquard; James Paul; Jean Yves Blay; Jeremy Whelan; K. S. Panageas; Keith Wheatley; Kevin J. Harrington; L. Licitra; Lucinda Billingham; Martee L. Hensley; Martin McCabe; Poulam M. Patel

Background The past three decades have seen rapid improvements in the diagnosis and treatment of most cancers and the most important contributor has been research. Progress in rare cancers has been slower, not least because of the challenges of undertaking research. Settings The International Rare Cancers Initiative (IRCI) is a partnership which aims to stimulate and facilitate the development of international clinical trials for patients with rare cancers. It is focused on interventional – usually randomised – clinical trials with the clear goal of improving outcomes for patients. The key challenges are organisational and methodological. A multi-disciplinary workshop to review the methods used in ICRI portfolio trials was held in Amsterdam in September 2013. Other as-yet unrealised methods were also discussed. Results The IRCI trials are each presented to exemplify possible approaches to designing credible trials in rare cancers. Researchers may consider these for use in future trials and understand the choices made for each design. Interpretation Trials can be designed using a wide array of possibilities. There is no ‘one size fits all’ solution. In order to make progress in the rare diseases, decisions to change practice will have to be based on less direct evidence from clinical trials than in more common diseases.


Journal of the National Cancer Institute | 2014

TP53 Mutational Status and Cetuximab Benefit in Rectal Cancer: 5-Year Results of the EXPERT-C Trial

Francesco Sclafani; David Gonzalez; David Cunningham; Sanna Hulkki Wilson; Clare Peckitt; Josep Tabernero; Bengt Glimelius; A. Cervantes; Alice Dewdney; Andrew Wotherspoon; Gina Brown; D. Tait; Jacqueline Oates; Ian Chau

In this updated analysis of the EXPERT-C trial we show that, in magnetic resonance imaging-defined, high-risk, locally advanced rectal cancer, adding cetuximab to a treatment strategy with neoadjuvant CAPOX followed by chemoradiotherapy, surgery, and adjuvant CAPOX is not associated with a statistically significant improvement in progression-free survival (PFS) and overall survival (OS) in both KRAS/BRAF wild-type and unselected patients. In a retrospective biomarker analysis, TP53 was not prognostic but emerged as an independent predictive biomarker for cetuximab benefit. After a median follow-up of 65.0 months, TP53 wild-type patients (n = 69) who received cetuximab had a statistically significant better PFS (89.3% vs 65.0% at 5 years; hazard ratio [HR] = 0.23; 95% confidence interval [CI] = 0.07 to 0.78; two-sided P = .02 by Cox regression) and OS (92.7% vs 67.5% at 5 years; HR = 0.16; 95% CI = 0.04 to 0.70; two-sided P = .02 by Cox regression) than TP53 wild-type patients who were treated in the control arm. An interaction between TP53 status and cetuximab effect was found (P < .05) and remained statistically significant after adjusting for statistically significant prognostic factors and KRAS.


European Journal of Cancer | 2010

Phase II study of NGR-hTNF, a selective vascular targeting agent, in patients with metastatic colorectal cancer after failure of standard therapy

Armando Santoro; Lorenza Rimassa; Alberto Sobrero; Giovanni Citterio; Francesco Sclafani; Carlo Carnaghi; A. Pessino; Francesco Caprioni; Valeria Andretta; Maria Chiara Tronconi; Giovanna Finocchiaro; Gloria Rossoni; Angela Zanoni; Chiara Miggiano; Gian Paolo Rizzardi; Catia Traversari; Federico Caligaris-Cappio; A. Lambiase; Claudio Bordignon

BACKGROUND NGR-hTNF consists of human tumour necrosis factor (hTNF) fused with the tumour-homing peptide Asp-Gly-Arg (NGR), which is able to selectively bind an aminopeptidase N overexpressed on tumour blood vessels. Preclinical antitumour activity was observed even at low doses. We evaluated the activity and safety of low-dose NGR-hTNF in colorectal cancer (CRC) patients failing standard therapies. PATIENTS AND METHODS Thirty-three patients with progressive disease at study entry received NGR-hTNF 0.8 μg/m(2) given intravenously every 3 weeks. The median number of prior treatment regimens was three (range, 2-5). One-quarter of patients had previously received four or more regimens and two-thirds targeted agents. Progression-free survival (PFS) was the primary study objective. RESULTS NGR-hTNF was well tolerated. No treatment-related grade 3 to 4 toxicities were detected, most common grade 1 to 2 adverse events being short-lived, infusion-time related chills (50.0%). One partial response and 12 stable diseases were observed, yielding a disease control rate of 39.4% (95% CI, 22.9-57.8%). Median PFS and overall survival were 2.5 months (95% CI, 2.1-2.8) and 13.1 months (95% CI, 8.9-17.3), respectively; whereas in patients who achieved disease control the median PFS and overall survival were 3.8 and 15.4 months, respectively. In an additional cohort of 13 patients treated at same dose with a weekly schedule, there was no increased toxicity and 2 patients experienced PFS longer than 10 months. CONCLUSION Based on tolerability and preliminary evidence of disease control in heavily pretreated CRC patients, NGR-hTNF deserves further evaluation in combination with standard chemotherapy.


Critical Reviews in Oncology Hematology | 2013

BRAF mutations in melanoma and colorectal cancer: A single oncogenic mutation with different tumour phenotypes and clinical implications

Francesco Sclafani; Giuseppe Gullo; Kieran Sheahan; John Crown

BRAF is an oncogene encoding a serine-threonine protein kinase involved in the MAPK signalling cascade. BRAF acts as direct effector of RAS and through the activation of MEK, promotes tumour growth and survival. Approximately, 8% of cancers carry a BRAF mutation. However, the prevalence of this mutation varies significantly across different tumour types. There has been increasing interest in the specific role of BRAF mutations in cancer growth and progression over the last few years, especially since the clinical introduction of therapeutic BRAF inhibitors. In this paper we review the published literature on the role of BRAF mutations in melanoma and colorectal cancer, focusing on similarities and differences of BRAF mutations with respect to frequency, demographics, risk factors, mutation-associated clinico-pathologic and molecular features and clinical implications between these two diseases.


Pancreatology | 2016

International Association of Pancreatology (IAP)/European Pancreatic Club (EPC) consensus review of guidelines for the treatment of pancreatic cancer

Kyoichi Takaori; Claudio Bassi; Andrew V. Biankin; Thomas Brunner; Ivana Cataldo; Fiona Campbell; David Cunningham; Massimo Falconi; Adam E. Frampton; Junji Furuse; Marc Giovannini; Richard J. Jackson; Akira Nakamura; William H. Nealon; John P. Neoptolemos; Francisco X. Real; Aldo Scarpa; Francesco Sclafani; John A. Windsor; Koji Yamaguchi; Christopher L. Wolfgang; C. D. Johnson

BACKGROUND Pancreatic cancer is one of the most devastating diseases with an extremely high mortality. Medical organizations and scientific societies have published a number of guidelines to address active treatment of pancreatic cancer. The aim of this consensus review was to identify where there is agreement or disagreement among the existing guidelines and to help define the gaps for future studies. METHODS A panel of expert pancreatologists gathered at the 46th European Pancreatic Club Meeting combined with the 18th International Association of Pancreatology Meeting and collaborated on critical reviews of eight English language guidelines for the clinical management of pancreatic cancer. Clinical questions (CQs) of interest were proposed by specialists in each of nine areas. The recommendations for the CQs in existing guidelines, as well as the evidence on which these were based, were reviewed and compared. The evidence was graded as sufficient, mediocre or poor/absent. RESULTS Only 4 of the 36 CQs, had sufficient evidence for agreement. There was also agreement in five additional CQs despite the lack of sufficient evidence. In 22 CQs, there was disagreement regardless of the presence or absence of evidence. There were five CQs that were not addressed adequately by existing guidelines. CONCLUSION The existing guidelines provide both evidence- and consensus-based recommendations. There is also considerable disagreement about the recommendations in part due to the lack of high level evidence. Improving the clinical management of patients with pancreatic cancer, will require continuing efforts to undertake research that will provide sufficient evidence to allow agreement.


Journal of the National Cancer Institute | 2015

A Randomized Phase II/III Study of Dalotuzumab in Combination With Cetuximab and Irinotecan in Chemorefractory, KRAS Wild-Type, Metastatic Colorectal Cancer

Francesco Sclafani; Tae Y. Kim; David Cunningham; Tae W. Kim; Josep Tabernero; Hans J. Schmoll; Jae K. Roh; Sun Young Kim; Young Suk Park; Tormod Kyrre Guren; Eliza A. Hawkes; Steven J. Clarke; David Ferry; Jan-Erik Frödin; Mark Ayers; Michael Nebozhyn; Clare Peckitt; Andrey Loboda; David J. Mauro; David Watkins

BACKGROUND Insulin-like growth factor type 1 receptor (IGF-1R) mediates resistance to epidermal growth factor receptor (EGFR) inhibition and may represent a therapeutic target. We conducted a multicenter, randomized, double blind, phase II/III trial of dalotuzumab, an anti-IGF-1R monoclonal antibody, with standard therapy in chemo-refractory, KRAS wild-type metastatic colorectal cancer. METHODS Eligible patients were randomly assigned to dalotuzumab 10mg/kg weekly (arm A), dalotuzumab 7.5mg/kg every alternate week (arm B), or placebo (arm C) in combination with cetuximab and irinotecan. Primary endpoints were progression-free survival (PFS) and overall survival (OS). Secondary endpoints included exploratory biomarker analyses. All statistical tests were two-sided. RESULTS The trial was prematurely discontinued for futility after 344 eligible KRAS wild-type patients were included in the primary efficacy population (arm A = 116, arm B = 117, arm C = 111). Median PFS was 3.9 months in arm A (hazard ratio [HR] = 1.33, 95% confidence interval [CI] = 0.98 to 1.83, P = .07) and 5.4 months in arm B (HR = 1.13, 95% CI = 0.83 to 1.55, P = .44) compared with 5.6 months in arm C. Median OS was 10.8 months in arm A (HR = 1.41, 95% CI = 0.99 to 2.00, P = .06) and 11.6 months in arm B (HR = 1.26, 95% CI = 0.89 to 1.79, P = .18) compared with 14.0 months in arm C. Grade 3 or higher asthenia and hyperglycaemia occurred more frequently with dalotuzumab compared with placebo. In exploratory biomarker analyses, patients with high IGF-1 mRNA tumors in arm A had numerically better PFS (5.6 vs 3.6 months, HR = 0.59, 95% CI = 0.28 to 1.23, P = .16) and OS (17.9 vs 9.4 months, HR = 0.67, 95% CI = 0.31 to 1.45, P = .31) compared with those with high IGF-1 mRNA tumors in arm C. In contrast, in arm C high IGF-1 mRNA expression predicted lower response rate (17.6% vs 37.3%, P = .04), shorter PFS (3.6 vs 6.6 months, HR = 2.15, 95% CI = 1.15 to 4.02, P = .02), and shorter OS (9.4 vs 15.5 months, HR = 2.42, 95% CI = 1.21 to 4.82, P = .01). CONCLUSIONS Adding dalotuzumab to irinotecan and cetuximab was feasible but did not improve survival outcome. IGF-1R ligands are promising biomarkers for differential response to anti-EGFR and anti-IGF-1R therapies.


European Journal of Cancer | 2014

RAS mutations and cetuximab in locally advanced rectal cancer: Results of the EXPERT-C trial

Francesco Sclafani; David Gonzalez; David Cunningham; S. Hulkki Wilson; Clare Peckitt; Jordi Giralt; Bengt Glimelius; S. Roselló Keränen; A. Wotherspoon; Gina Brown; D. Tait; J. Oates; I. Chau

BACKGROUND RAS mutations predict resistance to anti-epidermal growthfactor receptor (EGFR) monoclonal antibodies in metastatic colorectal cancer. We analysed RAS mutations in 30 non-metastatic rectal cancer patients treated with or without cetuximab within the 31 EXPERT-C trial. METHODS Ninety of 149 patients with tumours available for analysis were KRAS/BRAF wild-type, and randomly assigned to capecitabine plus oxaliplatin (CAPOX) followed by chemoradiotherapy, surgery and adjuvant CAPOX or the same regimen plus cetuximab (CAPOX-C). Of these, four had a mutation of NRAS exon 3, and 84 were retrospectively analysed for additional KRAS (exon 4) and NRAS (exons 2/4) mutations by using bi-directional Sanger sequencing. The effect of cetuximab on study end-points in the RAS wild-type population was analysed. RESULTS Eleven (13%) of 84 patients initially classified as KRAS/BRAF wild-type were found to have a mutation in KRAS exon 4 (11%) or NRAS exons 2/4 (2%). Overall, 78/149 (52%) assessable patients were RAS wild-type (CAPOX, n=40; CAPOX-C, n=38). In this population, after a median follow-up of 63.8months, in line with the initial analysis, the addition of cetuximab was associated with numerically higher, but not statistically significant, rates of complete response (15.8% versus 7.5%, p=0.31), 5-year progression-free survival (75.5% versus 67.5%, hazard ratio (HR) 0.61, p=0.25) and 5-year overall survival (83.8% versus 70%, HR 0.54, p=0.20). CONCLUSIONS RAS mutations beyond KRAS exon 2 and 3 were identified in 17% of locally advanced rectal cancer patients. Given the small sample size, no definitive conclusions on the effect of additional RAS mutations on cetuximab treatment in this setting can be drawn and further investigation of RAS in larger studies is warranted.


Annals of Oncology | 2012

Durable complete response following chemotherapy and trastuzumab for metastatic HER2-positive breast cancer

G. Gullo; Monica Zuradelli; Francesco Sclafani; Armando Santoro; John Crown

Individual cases of prolonged complete response (CR) of HER2-positive metastatic breast cancer (MBC) have been reported following treatment with trastuzumab/chemotherapy, but the frequency of durable remission is unknown [1, 2]. We carried out a retrospective study of long-term outcome of all patients with HER2-positive MBC treated in our institutions with chemotherapy and trastuzumab before March 2007. All patients had histology-proven, HER2-positive (3+ on immunohistochemistry and/or HER2/neu gene amplification on FISH) breast cancer. None had received adjuvant trastuzumab. Eighty-four patients were treated from May 2000 to March 2007 (Table 1). Thirteen (15%) achieved CR as defined according to RECIST 1.0 criteria [3]. As part of different institutional practices, patients in Dublin continued on trastuzumab until progression or at least for five years. In Milan trastuzumab was generally stopped in CR patients within two years of achieving remission. As of March 17, 2012, (median follow up 7 years, range 2.5–11.8 years), six of these patients remain alive and continuously cancer free at 142, 139, 122, 101, 84, and 84 months. Two additional patients are alive and continuously free of metastatic cancer at 107 and 105 months, having received curative locoregional therapy for new primary breast cancers. Five patients who achieved CR have developed relapsed MBC, at 44, 37, 35, 30, and 15 months, two while receiving maintenance trastuzumab (at 44 and 37 months, respectively). Three others had discontinued trastuzumab (21, 8, and 4 months after cessation). All of the eight DCR patients received trastuzumab together with their first chemotherapy for metastatic disease. Five (63%) DCR patients had estrogen receptor (ER) negative disease, and five had metastases limited to liver. All but one received a taxane-containing regimen with trastuzumab (docetaxel and carboplatin-4 and single agent taxane-3). The median duration of trastuzumab for CR patients in the two institutions was 67 months (range: 49 to 107+) in Dublin and 14 months (range 5–26) in Milan. Interestingly, although the frequency of CR was very similar in the two institutions (Milan 16% and Dublin 15%), the proportion of patients with DCR was higher in Dublin than Milan (11% versus 6%, respectively), prompting speculation that the duration of trastuzumab therapy might be important. This is the first reported series of long-term follow-up of patients with HER2-positive MBC who achieved CR following chemotherapy and trastuzumab. Our data suggest that a meaningful minority of patients achieve very prolonged complete remissions. Although the small numbers and the retrospective nature of the study preclude definitive statistics, the data also suggest that the impact of trastuzumab might be greater in patients with ER-negative disease (14% DCR—an

Collaboration


Dive into the Francesco Sclafani's collaboration.

Top Co-Authors

Avatar

David Cunningham

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Clare Peckitt

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Ian Chau

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

D. Tait

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gina Brown

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Ruwaida Begum

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Andrew Wotherspoon

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

I. Chau

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge