Network


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

Hotspot


Dive into the research topics where Giovanni Crisafulli is active.

Publication


Featured researches published by Giovanni Crisafulli.


Nature Medicine | 2015

Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients

Giulia Siravegna; Benedetta Mussolin; Michela Buscarino; Giorgio Corti; Andrea Cassingena; Giovanni Crisafulli; Agostino Ponzetti; Chiara Cremolini; Alessio Amatu; Calogero Lauricella; Simona Lamba; Sebastijan Hobor; Antonio Avallone; Emanuele Valtorta; Giuseppe Rospo; Enzo Medico; Valentina Motta; Carlotta Antoniotti; Fabiana Tatangelo; Beatriz Bellosillo; Silvio Veronese; Alfredo Budillon; Clara Montagut; Patrizia Racca; Silvia Marsoni; Alfredo Falcone; Ryan B. Corcoran; Federica Di Nicolantonio; Fotios Loupakis; Salvatore Siena

Colorectal cancers (CRCs) evolve by a reiterative process of genetic diversification and clonal evolution. The molecular profile of CRC is routinely assessed in surgical or bioptic samples. Genotyping of CRC tissue has inherent limitations; a tissue sample represents a single snapshot in time, and it is subjected to spatial selection bias owing to tumor heterogeneity. Repeated tissue samples are difficult to obtain and cannot be used for dynamic monitoring of disease progression and response to therapy. We exploited circulating tumor DNA (ctDNA) to genotype colorectal tumors and track clonal evolution during treatment with the epidermal growth factor receptor (EGFR)-specific antibodies cetuximab or panitumumab. We identified alterations in ctDNA of patients with primary or acquired resistance to EGFR blockade in the following genes: KRAS, NRAS, MET, ERBB2, FLT3, EGFR and MAP2K1. Mutated KRAS clones, which emerge in blood during EGFR blockade, decline upon withdrawal of EGFR-specific antibodies, indicating that clonal evolution continues beyond clinical progression. Pharmacogenomic analysis of CRC cells that had acquired resistance to cetuximab reveals that upon antibody withdrawal KRAS clones decay, whereas the population regains drug sensitivity. ctDNA profiles of individuals who benefit from multiple challenges with anti-EGFR antibodies exhibit pulsatile levels of mutant KRAS. These results indicate that the CRC genome adapts dynamically to intermittent drug schedules and provide a molecular explanation for the efficacy of rechallenge therapies based on EGFR blockade.


Cancer Discovery | 2016

Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer

Mariangela Russo; Giulia Siravegna; Lawrence S. Blaszkowsky; Giorgio Corti; Giovanni Crisafulli; Leanne G. Ahronian; Benedetta Mussolin; Eunice L. Kwak; Michela Buscarino; Luca Lazzari; Emanuele Valtorta; Mauro Truini; Nicholas A. Jessop; Hayley Robinson; Theodore S. Hong; Mari Mino-Kenudson; Federica Di Nicolantonio; Ashraf Thabet; Andrea Sartore-Bianchi; Salvatore Siena; A. John Iafrate; Alberto Bardelli; Ryan B. Corcoran

UNLABELLED How genomic heterogeneity associated with acquired resistance to targeted agents affects response to subsequent therapy is unknown. We studied EGFR blockade in colorectal cancer to assess whether tissue and liquid biopsies can be integrated with radiologic imaging to monitor the impact of individual oncogenic alterations on lesion-specific responses. Biopsy of a patients progressing liver metastasis following prolonged response to cetuximab revealed a MEK1(K57T) mutation as a novel mechanism of acquired resistance. This lesion regressed upon treatment with panitumumab and the MEK inhibitor trametinib. In circulating tumor DNA (ctDNA), mutant MEK1 levels declined with treatment, but a previously unrecognized KRAS(Q61H) mutation was also identified that increased despite therapy. This same KRAS mutation was later found in a separate nonresponding metastasis. In summary, parallel analyses of tumor biopsies and serial ctDNA monitoring show that lesion-specific radiographic responses to subsequent targeted therapies can be driven by distinct resistance mechanisms arising within separate tumor lesions in the same patient. SIGNIFICANCE Molecular heterogeneity ensuing from acquired resistance drives lesion-specific responses to subsequent targeted therapies. Analysis of a single-lesion biopsy is inadequate to guide selection of subsequent targeted therapies. ctDNA profiles allow the detection of concomitant resistance mechanisms residing in separate metastases and assessment of the effect of therapies designed to overcome resistance.


Clinical Cancer Research | 2015

Emergence of Multiple EGFR Extracellular Mutations during Cetuximab Treatment in Colorectal Cancer

Sabrina Arena; Beatriz Bellosillo; Giulia Siravegna; Alejandro Martinez; Israel Cañadas; Luca Lazzari; Noelia Ferruz; Mariangela Russo; Sandra Misale; Iria González; Mar Iglesias; Elena Gavilan; Giorgio Corti; Sebastijan Hobor; Giovanni Crisafulli; Marta Salido; Juan Sánchez; Alba Dalmases; Joaquim Bellmunt; Gianni De Fabritiis; Ana Rovira; Federica Di Nicolantonio; Joan Albanell; Alberto Bardelli; Clara Montagut

Purpose: Patients with colorectal cancer who respond to the anti-EGFR antibody cetuximab often develop resistance within several months of initiating therapy. To design new lines of treatment, the molecular landscape of resistant tumors must be ascertained. We investigated the role of mutations in the EGFR signaling axis on the acquisition of resistance to cetuximab in patients and cellular models. Experimental Design: Tissue samples were obtained from 37 patients with colorectal cancer who became refractory to cetuximab. Colorectal cancer cells sensitive to cetuximab were treated until resistant derivatives emerged. Mutational profiling of biopsies and cell lines was performed. Structural modeling and functional analyses were performed to causally associate the alleles to resistance. Results: The genetic profile of tumor specimens obtained after cetuximab treatment revealed the emergence of a complex pattern of mutations in EGFR, KRAS, NRAS, BRAF, and PIK3CA genes, including two novel EGFR ectodomain mutations (R451C and K467T). Mutational profiling of cetuximab-resistant cells recapitulated the molecular landscape observed in clinical samples and revealed three additional EGFR alleles: S464L, G465R, and I491M. Structurally, these mutations are located in the cetuximab-binding region, except for the R451C mutant. Functionally, EGFR ectodomain mutations prevent binding to cetuximab but a subset is permissive for interaction with panitumumab. Conclusions: Colorectal tumors evade EGFR blockade by constitutive activation of downstream signaling effectors and through mutations affecting receptor–antibody binding. Both mechanisms of resistance may occur concomitantly. Our data have implications for designing additional lines of therapy for patients with colorectal cancer who relapse upon treatment with anti-EGFR antibodies. Clin Cancer Res; 21(9); 2157–66. ©2015 AACR.


Cancer Discovery | 2016

Acquired Resistance to the TRK Inhibitor Entrectinib in Colorectal Cancer

Mariangela Russo; Sandra Misale; Ge Wei; Giulia Siravegna; Giovanni Crisafulli; Luca Lazzari; Giorgio Corti; Giuseppe Rospo; Luca Novara; Benedetta Mussolin; Alice Bartolini; Nicholas Cam; Roopal Patel; Shunqi Yan; Robert Shoemaker; Robert Wild; Federica Di Nicolantonio; Andrea Sartore-Bianchi; Gang Li; Salvatore Siena; Alberto Bardelli

UNLABELLED Entrectinib is a first-in-class pan-TRK kinase inhibitor currently undergoing clinical testing in colorectal cancer and other tumor types. A patient with metastatic colorectal cancer harboring an LMNA-NTRK1 rearrangement displayed a remarkable response to treatment with entrectinib, which was followed by the emergence of resistance. To characterize the molecular bases of the patients relapse, circulating tumor DNA (ctDNA) was collected longitudinally during treatment, and a tissue biopsy, obtained before entrectinib treatment, was transplanted in mice (xenopatient), which then received the same entrectinib regimen until resistance developed. Genetic profiling of ctDNA and xenopatient samples showed acquisition of two point mutations in the catalytic domain of NTRK1, p.G595R and p.G667C. Biochemical and pharmacologic analysis in multiple preclinical models confirmed that either mutation renders the TRKA kinase insensitive to entrectinib. These findings can be immediately exploited to design next-generation TRKA inhibitors. SIGNIFICANCE We provide proof of principle that analyses of xenopatients (avatar) and liquid biopsies allow the identification of drug resistance mechanisms in parallel with clinical treatment of an individual patient. We describe for the first time that p.G595R and p.G667C TRKA mutations drive acquired resistance to entrectinib in colorectal cancers carrying NTRK1 rearrangements.


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 Research | 2016

Molecular landscape of acquired resistance to targeted therapy combinations in BRAF mutant colorectal cancer

Daniele Oddo; Erin M. Sennott; Ludovic Barault; Emanuele Valtorta; Sabrina Arena; Andrea Cassingena; Genny Filiciotto; Giulia Marzolla; Elena Elez; Robin Van Geel; Alice Bartolini; Giovanni Crisafulli; Valentina Boscaro; Jason T. Godfrey; Michela Buscarino; Carlotta Cancelliere; Giorgio Corti; Mauro Truini; Giulia Siravegna; Julieta Grasselli; Margherita Gallicchio; René Bernards; Jan H. M. Schellens; Josep Tabernero; Jeffrey A. Engelman; Andrea Sartore-Bianchi; Alberto Bardelli; Salvatore Siena; Ryan B. Corcoran; Federica Di Nicolantonio

Although recent clinical trials of BRAF inhibitor combinations have demonstrated improved efficacy in BRAF-mutant colorectal cancer, emergence of acquired resistance limits clinical benefit. Here, we undertook a comprehensive effort to define mechanisms underlying drug resistance with the goal of guiding development of therapeutic strategies to overcome this limitation. We generated a broad panel of BRAF-mutant resistant cell line models across seven different clinically relevant drug combinations. Combinatorial drug treatments were able to abrogate ERK1/2 phosphorylation in parental-sensitive cells, but not in their resistant counterparts, indicating that resistant cells escaped drug treatments through one or more mechanisms leading to biochemical reactivation of the MAPK signaling pathway. Genotyping of resistant cells identified gene amplification of EGFR, KRAS, and mutant BRAF, as well as acquired mutations in KRAS, EGFR, and MAP2K1 These mechanisms were clinically relevant, as we identified emergence of a KRAS G12C mutation and increase of mutant BRAF V600E allele frequency in the circulating tumor DNA of a patient at relapse from combined treatment with BRAF and MEK inhibitors. To identify therapeutic combinations capable of overcoming drug resistance, we performed a systematic assessment of candidate therapies across the panel of resistant cell lines. Independent of the molecular alteration acquired upon drug pressure, most resistant cells retained sensitivity to vertical MAPK pathway suppression when combinations of ERK, BRAF, and EGFR inhibitors were applied. These therapeutic combinations represent promising strategies for future clinical trials in BRAF-mutant colorectal cancer. Cancer Res; 76(15); 4504-15. ©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.


Embo Molecular Medicine | 2017

Loss of AXIN1 drives acquired resistance to WNT pathway blockade in colorectal cancer cells carrying RSPO3 fusions

Gabriele Picco; Consalvo Petti; Alessia Centonze; Erica Torchiaro; Giovanni Crisafulli; Luca Novara; Andrea Acquaviva; Alberto Bardelli; Enzo Medico

In colorectal cancer (CRC), WNT pathway activation by genetic rearrangements of RSPO3 is emerging as a promising target. However, its low prevalence severely limits availability of preclinical models for in‐depth characterization. Using a pipeline designed to suppress stroma‐derived signal, we find that RSPO3 “outlier” expression in CRC samples highlights translocation and fusion transcript expression. Outlier search in 151 CRC cell lines identified VACO6 and SNU1411 cells as carriers of, respectively, a canonical PTPRK(e1)‐RSPO3(e2) fusion and a novel PTPRK(e13)‐RSPO3(e2) fusion. Both lines displayed marked in vitro and in vivo sensitivity to WNT blockade by the porcupine inhibitor LGK974, associated with transcriptional and morphological evidence of WNT pathway suppression. Long‐term treatment of VACO6 cells with LGK974 led to the emergence of a resistant population carrying two frameshift deletions of the WNT pathway inhibitor AXIN1, with consequent protein loss. Suppression of AXIN1 in parental VACO6 cells by RNA interference conferred marked resistance to LGK974. These results provide the first mechanism of secondary resistance to WNT pathway inhibition.


British Journal of Cancer | 2017

Emergence of MET hyper-amplification at progression to MET and BRAF inhibition in colorectal cancer

Daniele Oddo; Giulia Siravegna; Annunziata Gloghini; Claudio Vernieri; Benedetta Mussolin; Federica Morano; Giovanni Crisafulli; Rosa Berenato; Giorgio Corti; Chiara C. Volpi; Michela Buscarino; Monica Niger; Philip D. Dunne; Giuseppe Rospo; Emanuele Valtorta; Alice Bartolini; Giovanni Fucà; Simona Lamba; Antonia Martinetti; Maria Di Bartolomeo; Filippo de Braud; Alberto Bardelli; Filippo Pietrantonio; Federica Di Nicolantonio

Background:Combined MET and BRAF inhibition showed clinical benefit in a patient with rectal cancer carrying BRAFV600E and MET amplification. However after 4 months, acquired resistance emerged and the patient deceased shortly after disease progression. The mechanism of resistance to this drug combination is unknown.Methods:We analysed plasma circulating tumour DNA obtained at progression by exome sequencing and digital PCR. MET gene and mRNA in situ hybridisation analyses in two bioptic specimens obtained at progression were used to confirm the plasma data.Results:We identified in plasma MET gene hyper-amplification as a potential mechanism underlying therapy resistance. Increased MET gene copy and transcript levels were detected in liver and lymph node metastatic biopsies. Finally, transduction of MET in BRAF mutant colorectal cancer cells conferred refractoriness to BRAF and MET inhibition.Conclusions:We identified in a rectal cancer patient MET gene hyper-amplification as mechanism of resistance to dual BRAF and MET inhibition.


Annals of Oncology | 2017

Tracking a CAD-ALK gene rearrangement in urine and blood of a colorectal cancer patient treated with an ALK inhibitor

Giulia Siravegna; Andrea Sartore-Bianchi; Benedetta Mussolin; Andrea Cassingena; Alessio Amatu; Luca Novara; Michela Buscarino; Giorgio Corti; Giovanni Crisafulli; Alice Bartolini; Federica Tosi; Mark G. Erlander; F Di Nicolantonio; S. Siena; Alberto Bardelli

Background Monitoring response and resistance to kinase inhibitors is essential to precision cancer medicine, and is usually investigated by molecular profiling of a tissue biopsy obtained at progression. However, tumor heterogeneity and tissue sampling bias limit the effectiveness of this strategy. In addition, tissue biopsies are not always feasible and are associated with risks due to the invasiveness of the procedure. To overcome these limitations, blood-based liquid biopsy analysis has proven effective to non-invasively follow tumor clonal evolution. Patients and methods We exploited urine cell-free, trans-renal DNA (tr-DNA) and matched plasma circulating tumor DNA (ctDNA) to monitor a metastatic colorectal cancer patient carrying a CAD-ALK translocation during treatment with an ALK inhibitor. Results Using a custom next generation sequencing panel we identified the genomic CAD-ALK rearrangement and a TP53 mutation in plasma ctDNA. Sensitive assays were developed to detect both alterations in urine tr-DNA. The dynamics of the CAD-ALK rearrangement in plasma and urine were concordant and paralleled the patients clinical course. Detection of the CAD-ALK gene fusion in urine tr-DNA anticipated radiological confirmation of disease progression. Analysis of plasma ctDNA identified ALK kinase mutations that emerged during treatment with the ALK inhibitor entrectinib. Conclusion We find that urine-based genetic testing allows tracing of tumor-specific oncogenic rearrangements. This strategy could be effectively applied to non-invasively monitor tumor evolution during therapy. The same approach could be exploited to monitor minimal residual disease after surgery with curative intent in patients whose tumors carry gene fusions. The latter could be implemented without the need of patient hospitalization since urine tr-DNA can be self-collected, is stable over time and can be shipped at specified time-points to central labs for testing.

Collaboration


Dive into the Giovanni Crisafulli'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
Top Co-Authors

Avatar

Salvatore Siena

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge