Network


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

Hotspot


Dive into the research topics where Filippo Pietrantonio is active.

Publication


Featured researches published by Filippo Pietrantonio.


European Journal of Cancer | 2015

Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: A meta-analysis

Filippo Pietrantonio; Fausto Petrelli; Andrea Coinu; Maria Di Bartolomeo; Karen Borgonovo; Claudia Maggi; Mary Cabiddu; Roberto Iacovelli; Ilaria Bossi; Veronica Lonati; Mara Ghilardi; Filippo de Braud; Sandro Barni

BACKGROUND Wild type RAS (RAS-wt) status is predictive of the activities of the anti-epidermal growth factor receptor (EGFR) monoclonal antibodies cetuximab (C) and panitumumab (P). We examined the impact of C and P on progression-free survival (PFS), overall survival (OS) and overall response rate (ORR) in advanced colorectal cancer (CRC) patients who have RAS-wt/BRAF-mutant (BRAF-mut) status. METHODS Randomised trials that compared C or P plus chemotherapy (or C or P monotherapy) with standard therapy or best supportive care (BSC) were included. We used published hazard ratios (HRs) if they were available, or we derived treatment estimates from other survival data. Pooled estimates of the treatment efficacy of anti-EGFR-based therapy with C or P for the RAS-wt/BRAF-mut subgroup were calculated with the random-effect inverse variance weighted method. All statistical tests were two-sided. RESULTS Nine phase III trials and one phase II trial (six first-line and two second-line trials, plus two trials involving chemorefractory patients), that included 463 RAS-wt/BRAF-mut CRC patients, were analysed. Overall, the addition of C or P treatment in the BRAF-mut subgroup did not significantly improve PFS (HR, 0.88; 95% confidence interval (CI), 0.67-1.14; p=0.33), OS (HR, 0.91; 95% CI, 0.62-1.34; p=0.63) and ORR (relative risk, 1.31; 95% CI 0.83-2.08, p=0.25) compared with control regimens. CONCLUSIONS C- or P-based therapy did not increase the benefit of standard therapy or the BSC in RAS-wt/BRAF-mut CRC patients. These findings support BRAF mutation assessment before initiation of treatment with anti-EGFR monoclonal antibodies.


Annals of Oncology | 2015

BRAF codons 594 and 596 mutations identify a new molecular subtype of metastatic colorectal cancer at favorable prognosis

Chiara Cremolini; M. Di Bartolomeo; Alessio Amatu; Carlotta Antoniotti; Roberto Moretto; Rosa Berenato; F. Perrone; Elena Tamborini; Giuseppe Aprile; Sara Lonardi; Andrea Sartore-Bianchi; Gabriella Fontanini; Massimo Milione; C. Lauricella; S. Siena; Alfredo Falcone; F. de Braud; Fotios Loupakis; Filippo Pietrantonio

BACKGROUND While the negative prognostic role of BRAF V600E mutation in metastatic colorectal cancer (mCRC) is well established, the impact of BRAF codons 594 and 596 mutations, occurring in <1% of CRCs, is completely unknown. The present work aims to describe clinical, pathological and molecular features and prognosis of BRAF codons 594 and 596 mutant mCRCs, compared with BRAF V600E mutant and wild-type ones. PATIENTS AND METHODS Patients treated for mCRC at three Italian Institutions between October 2006 and October 2014, with available KRAS and NRAS codon 12, 13, 59, 61, 117 and 146 and BRAF codon 594, 596 and 600 mutational status, as detected by means of direct sequencing or matrix assisted laser desorption ionization time-of-flight MassArray, were included. RESULTS Ten patients bearing BRAF codons 594 or 596 mutated tumors were identified and compared with 77 and 542 patients bearing BRAF V600E mutated and BRAF wild-type tumors, respectively. While BRAF V600E mutated tumors were more frequently right-sided, mucinous and with peritoneal spread, BRAF 594 or 596 mutated were more frequently rectal, nonmucinous and with no peritoneal spread. All BRAF 594 or 596 mutated tumors were microsatellite stable. Patients with BRAF codons 594 or 596 mutated tumors had markedly longer overall survival (OS) when compared with BRAF V600E mutated [median OS: 62.0 versus 12.6 months; hazard ratio: 0.36 (95% confidence interval 0.20-0.64), P = 0.002], both at univariate and multivariate analyses. CONCLUSIONS BRAF codon 594 or 596 mutated mCRCs are different from BRAF V600E ones in terms of molecular features, pathological characteristics and clinical outcome. This is consistent with preclinical evidences of a kinase inactivating effect of these mutations. The role of CRAF in transducing the intracellular signal downstream BRAF 594 or 596 mutated proteins opens the way to further preclinical investigation.


Annals of Oncology | 2012

Reply to FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer-subgroup analysis of patients with KRAS-mutated tumours in the randomised German AIO study KRK-0306.

Filippo Pietrantonio; Marina Chiara Garassino; Valter Torri; F. de Braud

We read with great interest the article of Stintzing et al. published in Annals of Oncology on July 2012 [1] and would make a few remarks. The results of this unplanned retrospective analysis, carried out on 96 KRAS-mutated colorectal cancer patients, did not show any substantial difference in terms of response rate, progression-free and overall survival between anti-epidermal growth factor receptorand anti-vascular endothelial growth factor-based treatments. These results may be just related to operative bias: for example, the similar response rate observed in the cetuximab and bevacizumab arms is explainable by the lack of independent radiological review and the limitation of standard radiological assessment for anti-angiogenic treatments, which may delay disease progression mainly through disease stabilization: in fact, biological response is often characterized by a reduction of lesions’ density, without a substantial RECIST response [2]. Overall survival seemed to favour cetuximab over bevacizumab (22.7 versus 18.7 months), although this difference was not statistically significant (HR = 0.86, 95% CI, 0.55–1.35; P = 0.55). Since cetuximab arm was affected by a higher and earlier dropout rate, it is possible that lower median treatment duration in the cetuximab arm may have led to earlier initiation of effective bevacizumab-based second-line treatment. In fact, half of the patients treated with FOLFIRI plus cetuximab received subsequent bevacizumab-based regimens. Not surprisingly, a combination of bevacizumab and oxaliplatin-based regimens was shown to prolong overall survival in the second-line setting [3]. Even though the addition of cetuximab to FOLFIRI regimen seems to be at least not detrimental for progression-free survival in KRAS-mutated patients, [4] the apparent lack of benefit from the addition of bevacizumab could also derive from confounding factors and imbalance between prognostic factors rather than KRAS mutation itself. For example, the rate of patients undergoing surgery was twice as high in the cetuximab arm: although this is clearly not attributable to treatment (given the identical response rate in both the arms), a multimodality strategy may have improved progression-free and overall survival in patients treated with the cetuximabbased combination. The authors stated that KRAS G13D mutations, found in 20% of cases, may be associated with poorer prognosis independently of the treatment arm. However, in the recently published pooled analysis of CRYSTAL and OPUS trials, KRAS G13D-mutated patients receiving first-line chemotherapy alone failed to show a statistically significant difference in terms of progression-free survival and overall survival, when compared with other KRAS-mutated subtypes [5]. We believe that the study of Stintzing et al. is particularly valuable because it stimulates the investigation on the predictive role of KRAS mutation in bevacizumab-based treatment. However, given the small sample and evidence of great benefit of bevacizumab independently of KRAS status, [6] the preliminary nature of these results add a few more to what was previously known and any conclusions about the present analysis should be drawn very carefully.


Annals of Oncology | 2015

Digital PCR quantification of MGMT methylation refines prediction of clinical benefit from alkylating agents in glioblastoma and metastatic colorectal cancer

Ludovic Barault; Alessio Amatu; Fonnet E. Bleeker; Catia Moutinho; Chiara Falcomatà; V. Fiano; Andrea Cassingena; Giulia Siravegna; Massimo Milione; Paola Cassoni; F. de Braud; R. Rudà; Riccardo Soffietti; Tiziana Venesio; Alberto Bardelli; Pieter Wesseling; P. C. de Witt Hamer; Filippo Pietrantonio; S. Siena; Manel Esteller; Andrea Sartore-Bianchi; F Di Nicolantonio

BACKGROUND O(6)-methyl-guanine-methyl-transferase (MGMT) silencing by promoter methylation may identify cancer patients responding to the alkylating agents dacarbazine or temozolomide. PATIENTS AND METHODS We evaluated the prognostic and predictive value of MGMT methylation testing both in tumor and cell-free circulating DNA (cfDNA) from plasma samples using an ultra-sensitive two-step digital PCR technique (methyl-BEAMing). Results were compared with two established techniques, methylation-specific PCR (MSP) and Bs-pyrosequencing. RESULTS Thresholds for MGMT methylated status for each technique were established in a training set of 98 glioblastoma (GBM) patients. The prognostic and the predictive value of MGMT methylated status was validated in a second cohort of 66 GBM patients treated with temozolomide in which methyl-BEAMing displayed a better specificity than the other techniques. Cutoff values of MGMT methylation specific for metastatic colorectal cancer (mCRC) tissue samples were established in a cohort of 60 patients treated with dacarbazine. In mCRC, both quantitative assays methyl-BEAMing and Bs-pyrosequencing outperformed MSP, providing better prediction of treatment response and improvement in progression-free survival (PFS) (P < 0.001). Ability of methyl-BEAMing to identify responding patients was validated in a cohort of 23 mCRC patients treated with temozolomide and preselected for MGMT methylated status according to MSP. In mCRC patients treated with dacarbazine, exploratory analysis of cfDNA by methyl-BEAMing showed that MGMT methylation was associated with better response and improved median PFS (P = 0.008). CONCLUSIONS Methyl-BEAMing showed high reproducibility, specificity and sensitivity and was applicable to formalin-fixed paraffin-embedded tissues and cfDNA. This study supports the quantitative assessment of MGMT methylation for clinical purposes since it could refine prediction of response to alkylating agents.


Nature | 2017

Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth

Giovanni Germano; Simona Lamba; Giuseppe Rospo; Ludovic Barault; Alessandro Magri; Federica Maione; Mariangela Russo; Giovanni Crisafulli; Alice Bartolini; Giulia Lerda; Giulia Siravegna; Benedetta Mussolin; Roberta Frapolli; Monica Montone; Federica Morano; Filippo de Braud; Nabil Amirouchene-Angelozzi; Silvia Marsoni; Maurizio D’Incalci; Armando Orlandi; Enrico Giraudo; Andrea Sartore-Bianchi; Salvatore Siena; Filippo Pietrantonio; Federica Di Nicolantonio; Alberto Bardelli

Molecular alterations in genes involved in DNA mismatch repair (MMR) promote cancer initiation and foster tumour progression. Cancers deficient in MMR frequently show favourable prognosis and indolent progression. The functional basis of the clinical outcome of patients with tumours that are deficient in MMR is not clear. Here we genetically inactivate MutL homologue 1 (MLH1) in colorectal, breast and pancreatic mouse cancer cells. The growth of MMR-deficient cells was comparable to their proficient counterparts in vitro and on transplantation in immunocompromised mice. By contrast, MMR-deficient cancer cells grew poorly when transplanted in syngeneic mice. The inactivation of MMR increased the mutational burden and led to dynamic mutational profiles, which resulted in the persistent renewal of neoantigens in vitro and in vivo, whereas MMR-proficient cells exhibited stable mutational load and neoantigen profiles over time. Immune surveillance improved when cancer cells, in which MLH1 had been inactivated, accumulated neoantigens for several generations. When restricted to a clonal population, the dynamic generation of neoantigens driven by MMR further increased immune surveillance. Inactivation of MMR, driven by acquired resistance to the clinical agent temozolomide, increased mutational load, promoted continuous renewal of neoantigens in human colorectal cancers and triggered immune surveillance in mouse models. These results suggest that targeting DNA repair processes can increase the burden of neoantigens in tumour cells; this has the potential to be exploited in therapeutic approaches.


Cancer Discovery | 2016

MET-Driven Resistance to Dual EGFR and BRAF Blockade May Be Overcome by Switching from EGFR to MET Inhibition in BRAF-Mutated Colorectal Cancer

Filippo Pietrantonio; Daniele Oddo; Annunziata Gloghini; Emanuele Valtorta; Rosa Berenato; Ludovic Barault; Marta Caporale; Adele Busico; Federica Morano; Ambra Vittoria Gualeni; Alessandra Alessi; Giulia Siravegna; Federica Perrone; Maria Di Bartolomeo; Alberto Bardelli; Filippo de Braud; Federica Di Nicolantonio

UNLABELLED A patient with metastatic BRAF-mutated colorectal cancer initially responded to combined EGFR and BRAF inhibition with panitumumab plus vemurafenib. Pre-existing cells with increased MET gene copy number in the archival tumor tissue likely underwent clonal expansion during treatment, leading to the emergence of MET amplification in the rebiopsy taken at progression. In BRAF-mutated colorectal cancer cells, ectopic expression of MET conferred resistance to panitumumab and vemurafenib, which was overcome by combining BRAF and MET inhibition. Based on tumor genotyping and functional in vitro data, the patient was treated with the dual ALK-MET inhibitor crizotinib plus vemurafenib, thus switching to dual MET and BRAF blockade, with rapid and marked effectiveness of such strategy. Although acquired resistance is a major limitation to the clinical efficacy of anticancer agents, the identification of molecular targets emerging during the first treatment may afford the opportunity to design the next line of targeted therapies, maximizing patient benefit. SIGNIFICANCE MET amplification is here identified-clinically and preclinically-as a new mechanism of resistance to EGFR and BRAF dual/triple block combinations in BRAF-mutated colorectal cancer. Switching from EGFR to MET inhibition, while maintaining BRAF inhibition, resulted in clinical benefit after the occurrence of MET-driven acquired resistance. Cancer Discov; 6(9); 963-71. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 932.


Annals of Oncology | 2014

Activity of temozolomide in patients with advanced chemorefractory colorectal cancer and MGMT promoter methylation

Filippo Pietrantonio; F. Perrone; F. de Braud; Alessandra Castano; Claudia Maggi; Ilaria Bossi; Arpine Gevorgyan; Pamela Biondani; Monica Pacifici; Adele Busico; Manuela Gariboldi; Fabrizio Festinese; Elena Tamborini; M. Di Bartolomeo

BACKGROUND No evidence-based treatment options are available for patients with advanced colorectal cancer (CRC) progressing after standard therapies. MGMT is involved in repair of DNA damage and MGMT promoter methylation may predict benefit from alkylating agents such as temozolomide. The aim of our study was to evaluate the activity of temozolomide in terms of response rate in patients with metastatic CRC and MGMT methylation, after failure of approved treatments. PATIENTS AND METHODS Patients were enrolled in a monocentre, open-label, phase II study and treated with temozolomide at a dose of 150 mg/m2/day for 5 consecutive days in 4-weekly cycles. The treatment was continued for at least six cycles or until progressive disease. RESULTS Thirty-two patients were enrolled from August 2012 to July 2013. Treatment was well tolerated with one grade 4 thrombocytopenia and no other grade≥3 toxicities. No complete response occurred. The objective response rate was 12%, reaching the pre-specified level for promising activity. Median progression-free survival and overall survival were 1.8 and 8.4 months, respectively. Patients with KRAS, BRAF and NRAS wild-type CRC showed significantly higher response when compared with those with any RAS or BRAF mutation (44% versus 0%; P=0.004). TP53 status had no influence on the primary end point. CONCLUSIONS Temozolomide is tolerable and active in heavily pre-treated patients with advanced CRC and MGMT promoter methylation. Further studies in biomolecularly enriched populations or in a randomized setting are necessary to demonstrate the efficacy of temozolomide after failure of standard treatments.


PLOS ONE | 2014

Chemotherapy or Targeted Therapy as Second-Line Treatment of Advanced Gastric Cancer. A Systematic Review and Meta-Analysis of Published Studies

Roberto Iacovelli; Filippo Pietrantonio; Alessio Farcomeni; Claudia Maggi; Antonella Palazzo; Francesca Ricchini; Filippo de Braud; Maria Di Bartolomeo

Chemotherapy is a cornerstone in treatments of gastric cancer, but despite its benefit, less than 60% of patients receive salvage therapy in clinical practice. We performed a systematic review and meta-analysis based on trial data on the role of second-line treatment of advanced gastric cancer. MEDLINE/PubMed and Cochrane Library were searched for randomized phase III trials that compared active therapy to best supportive care in advanced gastric cancer. Data extraction was conducted according to the PRISMA statement. Summary HR for OS was calculated using a hierarchical Bayesian model and subgroup analysis was performed based on baseline Eastern Cooperative Oncology Group Performance Status (ECOG) performance status (0 vs. 1 or more). A total of 1,407 patients were evaluable for efficacy, 908 were treated in the experimental arms, with chemotherapy (231 pts) or with targeted therapies (677 pts). The risk of death was decreased by 18% (HR = 0.82; 95% CI, 0.79–0.85; posterior probability HR≥1: <0.00001) with active therapies. Chemotherapy and ramucirumab were able to decrease this risk by 27% and 22%, respectively. No differences were found between chemotherapy and ramucirumab. In patients with ECOG = 0 a greater benefit was found for chemotherapy with a reduction of the risk of death by 43% and no benefits were found for ramucirumab or everolimus. In patients with ECOG = 1 or more a significant reduction of the risk of death by 32% was reported in patients treated with ramucirumab, even if no significant difference was reported between chemotherapy and ramucirumab. This analysis reports that active and available therapies are able to prolong survival in patients with advanced gastric cancer with a different outcome based on initial patient’s performance status. New trials based on a better patient stratification are awaited.


Clinical Cancer Research | 2017

Heterogeneity of acquired resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer

Filippo Pietrantonio; Claudio Vernieri; Giulia Siravegna; Alessia Mennitto; Rosa Berenato; Federica Perrone; Annunziata Gloghini; Elena Tamborini; Sara Lonardi; Federica Morano; Benedetta Picciani; Adele Busico; Chiara C. Volpi; Antonia Martinetti; Francesca Battaglin; Ilaria Bossi; Alessio Pellegrinelli; Massimo Milione; Chiara Cremolini; Maria Di Bartolomeo; Alberto Bardelli; Filippo de Braud

Purpose: Even if RAS-BRAF wild-type and HER2/MET–negative metastatic colorectal cancer (mCRC) patients frequently respond to anti-EGFR mAbs, acquired resistance almost invariably occurs. Mechanisms of resistance to EGFR blockade include the emergence of KRAS, NRAS, and EGFR extracellular domain mutations as well as HER2/MET alterations. However, these findings derive from retrospective studies that analyzed one single resistance mechanism at a time; moreover, it is still unclear how molecular heterogeneity affects clonal evolution in patients. In this work, we aimed at extensively characterizing and correlating the molecular characteristics of tissue- and blood-based data in a prospective cohort of patients with mCRC who received anti-EGFR antibodies. Experimental design: Twenty-two RAS-BRAF wild-type, HER2/MET–negative mCRC patients progressing on anti-EGFR therapy after initial response underwent rebiopsy. Next-generation sequencing and silver in situ hybridization (SISH)/IHC analyses were performed both on archival tumors and postprogression samples. Circulating tumor (ctDNA) molecular profiles were obtained in matched tissue–plasma samples. Results: RAS mutations and HER2/MET amplification were the most frequently detected resistance mechanisms in both tissue and blood sample analysis. On the other hand, BRAF and EGFR ectodomain mutations were much rarer. Patients with acquired MET amplification showed worse PFS on anti-EGFRs. We detected both intralesion heterogeneity, as suggested by co-occurrence of different resistance mechanisms in the same sample, and interlesion heterogeneity. The combined analysis of tissue and blood (ctDNA) results highlights the complexity of clonal evolution triggered by EGFR blockade. Conclusions: Our results indicate that it may be extremely challenging to target the complex landscape of molecular heterogeneity associated with emergence of resistance to targeted therapies in patients with mCRC. Clin Cancer Res; 23(10); 2414–22. ©2016 AACR.


Nature Communications | 2016

Acquired RAS or EGFR mutations and duration of response to EGFR blockade in colorectal cancer

Beth O. Van Emburgh; Sabrina Arena; Giulia Siravegna; Luca Lazzari; Giovanni Crisafulli; Giorgio Corti; Benedetta Mussolin; Federica Baldi; Michela Buscarino; Alice Bartolini; Emanuele Valtorta; Joana Vidal; Beatriz Bellosillo; Giovanni Germano; Filippo Pietrantonio; Agostino Ponzetti; Joan Albanell; Salvatore Siena; Andrea Sartore-Bianchi; Federica Di Nicolantonio; Clara Montagut; Alberto Bardelli

Blockade of the epidermal growth factor receptor (EGFR) with the monoclonal antibodies cetuximab or panitumumab is effective in a subset of colorectal cancers (CRCs), but the emergence of resistance limits the efficacy of these therapeutic agents. At relapse, the majority of patients develop RAS mutations, while a subset acquires EGFR extracellular domain (ECD) mutations. Here we find that patients who experience greater and longer responses to EGFR blockade preferentially develop EGFR ECD mutations, while RAS mutations emerge more frequently in patients with smaller tumour shrinkage and shorter progression-free survival. In circulating cell-free tumour DNA of patients treated with anti-EGFR antibodies, RAS mutations emerge earlier than EGFR ECD variants. Subclonal RAS but not EGFR ECD mutations are present in CRC samples obtained before exposure to EGFR blockade. These data indicate that clonal evolution of drug-resistant cells is associated with the clinical outcome of CRC patients treated with anti-EGFR antibodies.

Collaboration


Dive into the Filippo Pietrantonio'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

Federica Morano

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claudia Maggi

Sapienza University of Rome

View shared research outputs
Researchain Logo
Decentralizing Knowledge