Network


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

Hotspot


Dive into the research topics where Francesco Facchinetti is active.

Publication


Featured researches published by Francesco Facchinetti.


Anti-Cancer Drugs | 2013

Chemotherapy in metastatic renal cell carcinoma today? A systematic review.

Sebastiano Buti; Melissa Bersanelli; Angelica Sikokis; Francesca Maines; Francesco Facchinetti; Emilio Bria; Andrea Ardizzoni; Giampaolo Tortora; Francesco Massari

The prognosis of patients affected by metastatic renal cell carcinoma (mRCC) has improved markedly with targeted therapies. Unfortunately, 20–25% of the patients are refractory to treatment at the first response assessment and most patients will acquire drug resistance during the treatment. Moreover, current data on the clinical activity of targeted agents in poor risk or non-clear-cell mRCC patients are inconclusive because of the absence of prospective trials. Therefore, there are still several patients in need of new therapeutic approaches to improve clinical outcomes. Kidney cancer is historically considered resistant to chemotherapy on the basis that the results of phase II trials have not always been promising. We carried out a systematic review of both monochemotherapy and polychemotherapy alone or combined with immunotherapy or targeted agents in mRCC to define the state of the art and to evaluate further clinical research fields. All retrospectives, phase I/dose finding, phase II and phase III studies on chemotherapy in mRCC, published in the literature from January 2003 to November 2012, with at least 20 patients enrolled, were evaluated. Although the results of clinical trials have often been disappointing, in selected cases of mRCC, chemotherapy may have a promising antitumor activity, particularly when there are sarcomatoid differentiation features, or in highly progressive disease where the combination of doxorubicine plus gemcitabine or capecitabine has yielded interesting results. Chemotherapy may play a role in mRCC, whereas targeted agents and immunotherapy have not yielded durable and satisfactory results; further studies are needed.


Clinical Cancer Research | 2016

Crizotinib-Resistant ROS1 Mutations Reveal a Predictive Kinase Inhibitor Sensitivity Model for ROS1- and ALK-Rearranged Lung Cancers

Francesco Facchinetti; Yohann Loriot; Mei-Shiue Kuo; Linda Mahjoubi; Ludovic Lacroix; David Planchard; Benjamin Besse; Françoise Farace; Nathalie Auger; Jordi Remon; Jean-Yves Scoazec; F. Andre; Jean-Charles Soria; Luc Friboulet

Background: The identification of molecular mechanisms conferring resistance to tyrosine kinase inhibitor (TKI) is a key step to improve therapeutic results for patients with oncogene addiction. Several alterations leading to EGFR and anaplastic lymphoma kinase (ALK) resistance to TKI therapy have been described in non–small cell lung cancer (NSCLC). Only two mutations in the ROS1 kinase domain responsible for crizotinib resistance have been described in patients thus far. Methods: A patient suffering from a metastatic NSCLC harboring an ezrin (EZR)–ROS1 fusion gene developed acquired resistance to the ALK/ROS1 inhibitor crizotinib. Molecular analysis (whole-exome sequencing, CGH) and functional studies were undertaken to elucidate the mechanism of resistance. Based on this case, we took advantage of the structural homology of ROS1 and ALK to build a predictive model for drug sensitivity regarding future ROS1 mutations. Results: Sequencing revealed a dual mutation, S1986Y and S1986F, in the ROS1 kinase domain. Functional in vitro studies demonstrated that ROS1 harboring either the S1986Y or the S1986F mutation, while conferring resistance to crizotinib and ceritinib, was inhibited by lorlatinib (PF-06463922). The patients clinical response confirmed the potency of lorlatinib against S1986Y/F mutations. The ROS1 S1986Y/F and ALK C1156Y mutations are homologous and displayed similar sensitivity patterns to ALK/ROS1 TKIs. We extended this analogy to build a model predicting TKI efficacy against potential ROS1 mutations. Conclusions: Clinical evidence, in vitro validation, and homology-based prediction provide guidance for treatment decision making for patients with ROS1-rearranged NSCLC who progressed on crizotinib. Clin Cancer Res; 22(24); 5983–91. ©2016 AACR.


Translational lung cancer research | 2016

Tackling ALK in non-small cell lung cancer: the role of novel inhibitors

Francesco Facchinetti; Marcello Tiseo; Massimo Di Maio; Paolo Graziano; Emilio Bria; Giulio Rossi; Silvia Novello

Crizotinib is an oral inhibitor of anaplastic lymphoma kinase (ALK) with remarkable clinical activity in patients suffering from ALK-rearranged non-small cell lung cancer (NSCLC), accounting to its superiority compared to chemotherapy. Unfortunately, virtually all ALK-rearranged tumors acquire resistance to crizotinib, frequently within one year since the treatment initiation. To date, therapeutic strategies to overcome crizotinib resistance have focused on the use of more potent and structurally different compounds. Second-generation ALK inhibitors such as ceritinib (LDK378), alectinib (CH5424802/RO5424802) and brigatinib (AP26113) have shown relevant clinical activity, consequently fostering their rapid clinical development and their approval by health agencies. The third-generation inhibitor lorlatinib (PF-06463922), selectively active against ALK and ROS1, harbors impressive biological potency; its efficacy in reversing resistance to crizotinib and to other ALK inhibitors is being proven by early clinical trials. The NTRK1-3 and ROS1 inhibitor entrectinib (RXDX-101) has been reported to act against NSCLC harboring ALK fusion proteins too. Despite the quick development of these novel agents, several issues remain to be discussed in the treatment of patients suffering from ALK-rearranged NSCLC. This position paper will discuss the development, the current evidence and approvals, as long as the future perspectives of new ALK inhibitors beyond crizotinib. Clinical behaviors of ALK-rearranged NSCLC vary significantly among patients and differential molecular events responsible of crizotinib resistance account for the most important quote of this heterogeneity. The precious availability of a wide range of active anti-ALK compounds should be approached in a critical and careful perspective, in order to develop treatment strategies tailored on the disease evolution of every single patient.


Clinical Lung Cancer | 2014

Activity of the EGFR-HER2 dual inhibitor afatinib in EGFR-mutant lung cancer patients with acquired resistance to reversible EGFR tyrosine kinase inhibitors.

Lorenza Landi; Marcello Tiseo; Rita Chiari; Serena Ricciardi; Elisa Rossi; Domenico Galetta; Silvia Novello; Michele Milella; Armida D'Incecco; Gabriele Minuti; Carmelo Tibaldi; Jessica Salvini; Francesco Facchinetti; Eva Regina Haspinger; Diego Cortinovis; Antonio Santo; Giuseppe Luigi Banna; Annamaria Catino; Matteo GiajLevra; Lucio Crinò; Filippo De Marinis; Federico Cappuzzo

BACKGROUND The purpose of this study was to evaluate the efficacy of afatinib in EGFR-mutant metastatic NSCLC patients with acquired resistance to erlotinib or gefitinib. MATERIALS AND METHODS We retrospectively analyzed the outcome of patients with EGFR-mutant advanced NSCLC treated with afatinib after failure of chemotherapy and EGFR TKIs. RESULTS A total of 96 individuals were included in the study. According to EGFR status, most patients (n = 63; 65.6%) harbored a deletion in exon 19, and de novo T790M mutation was detected in 2 cases (T790M and exon 19). Twenty-four (25%) patients underwent repeated biopsy immediately before starting afatinib and secondary T790M was detected in 8 (33%) samples. Among the 86 patients evaluable for efficacy, response rate was 11.6%, with a median progression free-survival (PFS) and overall survival (OS) of 3.9 and 7.3 months, respectively. No significant difference in PFS and OS was observed according to type of last therapy received before afatinib, type of EGFR mutation or adherence to Jackman criteria, and patients benefiting from afatinib therapy had longer PFS and OS (P < .001). Outcome results for repeated biopsy patients were similar to the whole population, with no evidence of response in T790M-positive patients. All patients were evaluable for toxicity, and 81% experienced an AE of any grade, with grade 3 to 4 AEs, mainly diarrhea and skin toxicity, occurring in 19 (20%) patients. CONCLUSION Our results showed that afatinib has only modest efficacy in a real life population of EGFR mutant NSCLC patients with acquired resistance to erlotinib or gefitinib.


Annals of Oncology | 2017

A prospective examination of circulating tumor cell profiles in non-small-cell lung cancer molecular subgroups

C. R. Lindsay; V. Faugeroux; S. Michiels; Emma Pailler; Francesco Facchinetti; D. Ou; M. V. Bluthgen; C. Pannet; M. Ngocamus; G. Bescher; C. Caramella; F. Billiot; Jordi Remon; David Planchard; Benjamin Besse; Françoise Farace

Background We report the first study examining the clinical, numerical and biological properties of circulating tumor cells according to molecular subtypes of non-small-cell lung cancer. Patients and methods 125 patients with treatment-naïve stage IIIb-IV NSCLC were prospectively recruited for CellSearch analysis. Anti-vimentin antibody was included for examination of CTCs to assess their mesenchymal character. Associations of total CTCs and vimentin-positive (vim +) CTCs with clinical characteristics, tumor genotype, and survival were assessed. Results 51/125 patients (40.8%) were total CTC+ and 26/125 (20.8%) were vim CTC+ at baseline. Multivariate analysis showed patients with ≥5 total CTCs had significantly reduced OS (HR 0.55, 95% CI 0.33-0.92, P = 0.022) but not PFS (HR 0.68, 95% CI 0.42-1.1, P = 0.118) compared to patients with <5 total CTCs. No OS difference was evident between vim+ CTC and vim-negative CTC patients overall (HR 1.24, 95% CI 0.67-2.28, P = 0.494), but after subdivision according to NSCLC driver mutation, we found an increase of vim+ CTCs in the EGFR-mutated subgroup (N = 21/94 patients; mean 1.24 vs 1.22 vim+ CTCs, P = 0.013), a reduction of total CTCs in the ALK-rearranged subgroup (N = 13/90 patients; mean 1.69 vs 5.82 total CTCs, P = 0.029), and a total absence of vim+ CTCs in KRAS-mutated adenocarcinomas (N = 19/78 patients; mean 0 vs 1.4 vim+ CTCs, P = 0.006). Conclusions We validate that the baseline presence of ≥5 total CTCs in advanced NSCLC confers a poor prognosis. CTCs from EGFR-mutant NSCLC express epithelial-mesenchymal transition characteristics, not seen in CTCs from patients with KRAS-mutant adenocarcinoma.


Lung Cancer: Targets and Therapy | 2017

Detection of ROS1 rearrangement in non-small cell lung cancer: current and future perspectives

Giulio Rossi; Genny Jocollé; Antonia Conti; Marcello Tiseo; Federica Zito Marino; Giovanni Donati; Renato Franco; Francesca Bono; Francesca Barbisan; Francesco Facchinetti

ROS1 rearrangement characterizes a small subset (1%–2%) of non-small cell lung cancer and is associated with slight/never smoking patients and adenocarcinoma histology. Identification of ROS1 rearrangement is mandatory to permit targeted therapy with specific inhibitors, demonstrating a significantly better survival when compared with conventional chemotherapy. Detection of ROS1 rearrangement is based on in situ (immunohistochemistry, fluorescence in situ hybridization) and extractive non-in situ assays. While fluorescence in situ hybridization still represents the gold standard in clinical trials, this technique may fail to recognize rearrangements of ROS1 with some gene fusion partner. On the other hand, immunohistochemistry is the most cost-effective screening technique, but it seems to be characterized by low specificity. Extractive molecular assays are expensive and laborious methods, but they specifically recognize almost all ROS1 fusions using a limited amount of mRNA even from formalin-fixed, paraffin-embedded tumor tissues. This review is a discussion on the present and futuristic diagnostic scenario of ROS1 identification in lung cancer.


Tumori | 2016

Crizotinib primary resistance overcome by ceritinib in a patient with ALK-rearranged non-small cell lung cancer.

Francesco Facchinetti; C. Caramella; Nathalie Auger; David Planchard; Julien Adam; Ludovic Lacroix; Jordi Remon; Christophe Massard; Jean-Charles Soria; Luc Friboulet; Benjamin Besse

We report on the case of a patient affected by advanced non-small cell lung cancer (NSCLC) harboring an anaplastic lymphoma kinase (ALK) gene rearrangement who did not respond to crizotinib but subsequently benefited from treatment with ceritinib (LDK378). Although second-generation ALK inhibitors have shown activity in patients pretreated with crizotinib who experienced secondary resistance, this is the first report to date describing their efficacy in a case of primary resistance. Of note, none of the previously described molecular mechanisms explaining resistance to crizotinib was detected on either the initial or post-crizotinib biopsies. We hypothesize that crizotinib was powerless in controlling disease progression due to its inadequate inhibition of ALK signaling. Although we lack any molecular evidence elucidating the primary crizotinib resistance, we believe that ceritinib treatment led to tumor regression thanks to its superior biological potency.


Cancer Treatment Reviews | 2018

BRAF in non-small cell lung cancer (NSCLC): Pickaxing another brick in the wall

Alessandro Leonetti; Francesco Facchinetti; Giulio Rossi; Roberta Minari; Antonia Conti; Luc Friboulet; Marcello Tiseo; David Planchard

Molecular characterization of non-small cell lung cancer (NSCLC) marked an historical turning point for the treatment of lung tumors harboring kinase alterations suitable for specific targeted drugs inhibition, translating into major clinical improvements. Besides EGFR, ALK and ROS1, BRAF represents a novel therapeutic target for the treatment of advanced NSCLC. BRAF mutations, found in 1.5-3.5% of NSCLC, are responsible of the constitutive activation of mitogen activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway. Clinical trials evaluating the efficacy of the BRAF inhibitor dabrafenib in combination with the downstream MEK inhibitor trametinib in metastatic BRAFV600E-mutated NSCLC guaranteed FDA and EMA rapid approval of the combination regimen in this clinical setting. In line with the striking results observed in metastatic melanoma harboring the same molecular alteration, BRAF and MEK inhibition should be considered a new standard of care in this molecular subtype of NSCLC. In the present review, we propose an overview of the available evidence about BRAF in NSCLC mutations (V600E and non-V600E), from biological significance to emerging clinical implications of BRAF mutations detection. Focusing on the current strategies to act against the mutated kinase, we moreover approach additional strategies to overcome treatment resistance.


Lung Cancer | 2013

NSE level in combined neuroendocrine and adenocarcinoma EGFR mutated lung cancer resistant to EGFR-TKI

Francesco Facchinetti; Marcello Tiseo; Letizia Gnetti; Enrico Maria Silini; Andrea Ardizzoni

In a recent issue of Lung Cancer, Zhang et al. [1] reported patient with EGFR-mutated lung adenocarcinoma resistant to GFR-TKI treatment. Considered the rapid disease progression and he increase of neuron specific enolase (NSE) blood level, a biopsy n a paravertebral mass was performed, showing a tumor with mall cell carcinoma histology harboring the same EGFR mutaion of the primary. This kind of transformation was thought to be esponsible for EGFR-TKI resistance and subsequent chemotherpy treatment with cisplatin-etoposide and local radiotherapy led o tumor response. We wish to contribute to this report concerning he role of tumor markers as early predictors of SCLC transformaion, by reporting a similar case seen in our Institution. A 74-year-old Caucasian non-smoker woman had a cytological iagnosis of stage II lung adenocarcinoma from a transhoracic needle aspiration in July 2010. She received neoadjuvant hemotherapy with carboplatin-gemcitabine and, in November 010, underwent lobectomy with hylo-mediastinal lymphadenecomy. The surgical specimen showed a single tumor lesion with ombined histology (Fig. 1A), 80% adenocarcinoma (Fig. 1B) and 20% oorly differentiated neuroendocrine (NE) carcinoma, both small nd large cells (Fig. 1C). The pathological staging was pT2aN1 due to ymphnode metastasis by the NE component. Adjuvant chemotherpy was not performed because of patient slow recovery after urgery. DNA sequencing revealed exon 21 (L858R) EGFR gene utation in both adenocarcinoma and NE components (Fig. 1D and ). The patient relapsed 5 months later with mediastinal node and


Tumori | 2017

Osteoblastic progression during EGFR tyrosine kinase inhibitor therapy in mutated non-small cell lung cancer: a potential blunder.

Melissa Bersanelli; Paola Bini; Enrico Rabaiotti; Francesco Facchinetti; Massimo De Filippo; Beatrice Bortesi; Sebastiano Buti; Marcello Tiseo

Aims and background Bone flare reaction as a sign of response to antineoplastic treatment has been redefined, including the onset of new osteoblastic lesions. If misunderstood as skeletal progression, this finding could lead to erroneous therapy discontinuation, changing the disease clinical course. We aim to describe this clinical phenomenon in patients with advanced non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) gene-activating mutations treated with tyrosine kinase inhibitor (TKI). Methods We retrospectively reviewed the computed tomography scans of 43 EGFR-mutated patients with NSCLC treated with EGFR-TKI, analyzing the bone response in terms of increase in the quantity and/or density of lesions, and assessing objective tumor response to treatment. Results Osteoblastic reaction was detected in 10 cases (23%), showing different patterns: dimensional or density increase of known osteosclerotic metastases (pattern A, n = 4); response of previously lytic lesions (pattern B, n = 2); onset of new osteosclerotic lesions (pattern C, n = 4). Seven patients had partial response to TKI treatment, with response rate of 70%, vs 50% of patients with bone metastases without this reaction. No difference in terms of median overall survival or progression-free survival emerged between patients with or without osteoblastic reaction. Conclusions The correct clinico-radiologic interpretation of osteoblastic reaction is crucial to avoid waste of therapeutic lines when TKI treatment has not yet exhausted its potential effectiveness. Clinical implications of ambiguous radiologic findings as described in this study deserve further discussion.

Collaboration


Dive into the Francesco Facchinetti's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sebastiano Buti

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar

Jordi Remon

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Caramella

Institut Gustave Roussy

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge