37 results on '"Crisafulli, Giovanni"'
Search Results
2. Post-surgery sequelae unrelated to disease progression and chemotherapy revealed in follow-up of patients with stage III colon cancer
- Author
-
Mirandola, Alexia, Kudriavtsev, Andrei, Cofre Muñoz, Catalina Isabel, Navarro, Raquel Comas, Macagno, Marco, Daoud, Saidi, Sanchez, Cynthia, Pastor, Brice, Pisareva, Ekaterina, Marin, Mireia Sanchis, Ruiz, Javier Gonzalo, Piris, Alejandro, Rodriguez, Ariadna Garcia, Gonzalez, Nadia Saoudi, Vivancos, Ana, Quarà, Virginia, Mellano, Alfredo, Borghi, Felice, Corti, Giorgio, Marchiò, Caterina, Sapino, Anna, Bartolini, Alice, Crisafulli, Giovanni, Bardelli, Alberto, Di Maio, Massimo, Lossaint, Gerald, Frayssinoux, Florence, Crapez, Evelyne, Ychou, Marc, Soler, Ramon Salazar, Fenocchio, Elisabetta, Fernandez Calotti, Paula X., Mazard, Thibault, Vivas, Cristina Santos, Elez, Elena, Di Nicolantonio, Federica, and Thierry, Alain R.
- Published
- 2024
- Full Text
- View/download PDF
3. Concurrent RB1 and P53 pathway disruption predisposes to the development of a primitive neuronal component in high-grade gliomas depending on MYC-driven EBF3 transcription
- Author
-
Pagani, Francesca, Orzan, Francesca, Lago, Sara, De Bacco, Francesca, Prelli, Marta, Cominelli, Manuela, Somenza, Elena, Gryzik, Magdalena, Balzarini, Piera, Ceresa, Davide, Marubbi, Daniela, Isella, Claudio, Crisafulli, Giovanni, Poli, Maura, Malatesta, Paolo, Galli, Rossella, Ronca, Roberto, Zippo, Alessio, Boccaccio, Carla, and Poliani, Pietro Luigi
- Published
- 2025
- Full Text
- View/download PDF
4. Coexisting cancer stem cells with heterogeneous gene amplifications, transcriptional profiles, and malignancy are isolated from single glioblastomas
- Author
-
De Bacco, Francesca, Orzan, Francesca, Crisafulli, Giovanni, Prelli, Marta, Isella, Claudio, Casanova, Elena, Albano, Raffaella, Reato, Gigliola, Erriquez, Jessica, D’Ambrosio, Antonio, Panero, Mara, Dall’Aglio, Carmine, Casorzo, Laura, Cominelli, Manuela, Pagani, Francesca, Melcarne, Antonio, Zeppa, Pietro, Altieri, Roberto, Morra, Isabella, Cassoni, Paola, Garbossa, Diego, Cassisa, Anna, Bartolini, Alice, Pellegatta, Serena, Comoglio, Paolo M., Finocchiaro, Gaetano, Poliani, Pietro L., and Boccaccio, Carla
- Published
- 2023
- Full Text
- View/download PDF
5. Liquid biopsies to monitor and direct cancer treatment in colorectal cancer
- Author
-
Mauri, Gianluca, Vitiello, Pietro Paolo, Sogari, Alberto, Crisafulli, Giovanni, Sartore-Bianchi, Andrea, Marsoni, Silvia, Siena, Salvatore, and Bardelli, Alberto
- Published
- 2022
- Full Text
- View/download PDF
6. Circulating tumor DNA to guide rechallenge with panitumumab in metastatic colorectal cancer: the phase 2 CHRONOS trial
- Author
-
Sartore-Bianchi, Andrea, Pietrantonio, Filippo, Lonardi, Sara, Mussolin, Benedetta, Rua, Francesco, Crisafulli, Giovanni, Bartolini, Alice, Fenocchio, Elisabetta, Amatu, Alessio, Manca, Paolo, Bergamo, Francesca, Tosi, Federica, Mauri, Gianluca, Ambrosini, Margherita, Daniel, Francesca, Torri, Valter, Vanzulli, Angelo, Regge, Daniele, Cappello, Giovanni, Marchiò, Caterina, Berrino, Enrico, Sapino, Anna, Marsoni, Silvia, Siena, Salvatore, and Bardelli, Alberto
- Published
- 2022
- Full Text
- View/download PDF
7. A modified fluctuation-test framework characterizes the population dynamics and mutation rate of colorectal cancer persister cells
- Author
-
Russo, Mariangela, Pompei, Simone, Sogari, Alberto, Corigliano, Mattia, Crisafulli, Giovanni, Puliafito, Alberto, Lamba, Simona, Erriquez, Jessica, Bertotti, Andrea, Gherardi, Marco, Di Nicolantonio, Federica, Bardelli, Alberto, and Cosentino Lagomarsino, Marco
- Published
- 2022
- Full Text
- View/download PDF
8. Liquid Biopsy and Challenge of Assay Heterogeneity for Minimal Residual Disease Assessment in Colon Cancer Treatment.
- Author
-
Crisafulli, Giovanni
- Subjects
- *
COLON cancer , *ADJUVANT chemotherapy , *COLORECTAL cancer , *SURVIVAL rate , *COLON diseases - Abstract
This review provides a comprehensive overview of the evolving role of minimal residual disease (MRD) for patients with Colon Cancer (CC). Currently, the standard of care for patients with non-metastatic CC is adjuvant chemotherapy (ACT) for all patients with stage III and high-risk stage II CC following surgical intervention. Despite a 5–20% improvement in long-term survival outcomes, this approach also results in a significant proportion of patients receiving ACT without any therapeutic benefit and being unnecessarily exposed to the risks of secondary side effects. This underscores an unmet clinical need for more precise stratification to distinguish patients who necessitate ACT from those who can be treated with surgery alone. By employing liquid biopsy, it is possible to discern MRD enabling the categorization of patients as MRD-positive or MRD-negative, potentially revolutionizing the management of ACT. This review aimed to examine the heterogeneity of methodologies currently available for MRD detection, encompassing the state-of-the-art technologies, their respective advantages, limitations, and the technological challenges and multi-omic approaches that can be utilized to enhance assay performance. Furthermore, a discussion was held regarding the clinical trials that employ an MRD assay focusing on the heterogeneity of the assays used. These differences in methodology, target selection, and performance risk producing inconsistent results that may not solely reflect biological/clinical differences but may be the consequence of the preferential use of particular products in studies conducted in different countries. Standardization and harmonization of MRD assays will be crucial to ensure the liquid revolution delivers reliable and clinically actionable outcomes for patients. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
9. Mutational Signatures in Colorectal Cancer: Translational Insights, Clinical Applications, and Limitations.
- Author
-
Crisafulli, Giovanni
- Subjects
- *
MEDICAL protocols , *MEDICAL quality control , *GENOMICS , *COLORECTAL cancer , *DNA , *DECISION making in clinical medicine , *CANCER chemotherapy , *GENES , *DNA damage , *GENETIC mutation , *MEDICAL practice - Abstract
Simple Summary: This review provides a comprehensive overview of the current knowledge on mutational signatures in colorectal cancer (CRC), highlighting their potential clinical applications. It discusses the challenges and limitations in translating these analyses into clinical practice and proposes strategies to overcome these obstacles. Additionally, it provides insights into future directions and emerging proof-of-concept studies that highlight the translational potential of mutational signature analysis in improving patient care and outcomes in CRC. A multitude of exogenous and endogenous processes have the potential to result in DNA damage. While the repair mechanisms are typically capable of correcting this damage, errors in the repair process can result in mutations. The findings of research conducted in 2012 indicate that mutations do not occur randomly but rather follow specific patterns that can be attributed to known or inferred mutational processes. The process of mutational signature analysis allows for the inference of the predominant mutational process for a given cancer sample, with significant potential for clinical applications. A deeper comprehension of these mutational signatures in CRC could facilitate enhanced prevention strategies, facilitate the comprehension of genotoxic drug activity, predict responses to personalized treatments, and, in the future, inform the development of targeted therapies in the context of precision oncology. The efforts of numerous researchers have led to the identification of several mutational signatures, which can be categorized into different mutational signature references. In CRC, distinct mutational signatures are identified as correlating with mismatch repair deficiency, polymerase mutations, and chemotherapy treatment. In this context, a mutational signature analysis offers considerable potential for enhancing minimal residual disease (MRD) tests in stage II (high-risk) and stage III CRC post-surgery, stratifying CRC based on the impacts of genetic and epigenetic alterations for precision oncology, identifying potential therapeutic vulnerabilities, and evaluating drug efficacy and guiding therapy, as illustrated in a proof-of-concept clinical trial. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Adaptive mutability of colorectal cancers in response to targeted therapies
- Author
-
Russo, Mariangela, Crisafulli, Giovanni, Sogari, Alberto, Reilly, Nicole M., Arena, Sabrina, Lamba, Simona, Bartolini, Alice, Amodio, Vito, Magrì, Alessandro, Novara, Luca, Sarotto, Ivana, Nagel, Zachary D., Piett, Cortt G., Amatu, Alessio, Sartore-Bianchi, Andrea, Siena, Salvatore, Bertotti, Andrea, Trusolino, Livio, Corigliano, Mattia, Gherardi, Marco, Lagomarsino, Marco Cosentino, Di Nicolantonio, Federica, and Bardelli, Alberto
- Published
- 2019
11. A Genomic Analysis Workflow for Colorectal Cancer Precision Oncology
- Author
-
Corti, Giorgio, Bartolini, Alice, Crisafulli, Giovanni, Novara, Luca, Rospo, Giuseppe, Montone, Monica, Negrino, Carola, Mussolin, Benedetta, Buscarino, Michela, Isella, Claudio, Barault, Ludovic, Siravegna, Giulia, Siena, Salvatore, Marsoni, Silvia, Di Nicolantonio, Federica, Medico, Enzo, and Bardelli, Alberto
- Published
- 2019
- Full Text
- View/download PDF
12. Mutational signatures of colorectal cancers according to distinct computational workflows.
- Author
-
Battuello, Paolo, Corti, Giorgio, Bartolini, Alice, Lorenzato, Annalisa, Sogari, Alberto, Russo, Mariangela, Nicolantonio, Federica Di, Bardelli, Alberto, and Crisafulli, Giovanni
- Subjects
COLORECTAL cancer ,WHOLE genome sequencing ,WORKFLOW software ,LUNG cancer ,COMPARATIVE genomics ,CANCER patients ,ENDOMETRIAL cancer ,WORKFLOW ,NUCLEOTIDE sequencing - Abstract
Tumor mutational signatures have gained prominence in cancer research, yet the lack of standardized methods hinders reproducibility and robustness. Leveraging colorectal cancer (CRC) as a model, we explored the influence of computational parameters on mutational signature analyses across 230 CRC cell lines and 152 CRC patients. Results were validated in three independent datasets: 483 endometrial cancer patients stratified by mismatch repair (MMR) status, 35 lung cancer patients by smoking status and 12 patient-derived organoids (PDOs) annotated for colibactin exposure. Assessing various bioinformatic tools, reference datasets and input data sizes including whole genome sequencing, whole exome sequencing and a pan-cancer gene panel, we demonstrated significant variability in the results. We report that the use of distinct algorithms and references led to statistically different results, highlighting how arbitrary choices may induce variability in the mutational signature contributions. Furthermore, we found a differential contribution of mutational signatures between coding and intergenic regions and defined the minimum number of somatic variants required for reliable mutational signature assignment. To facilitate the identification of the most suitable workflows, we developed Comparative Mutational Signature analysis on Coding and Extragenic Regions (CoMSCER), a bioinformatic tool which allows researchers to easily perform comparative mutational signature analysis by coupling the results from several tools and public reference datasets and to assess mutational signature contributions in coding and non-coding genomic regions. In conclusion, our study provides a comparative framework to elucidate the impact of distinct computational workflows on mutational signatures. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Transcriptome‐wide gene expression outlier analysis pinpoints therapeutic vulnerabilities in colorectal cancer.
- Author
-
Mariella, Elisa, Grasso, Gaia, Miotto, Martina, Buzo, Kristi, Reilly, Nicole Megan, Andrei, Pietro, Vitiello, Pietro Paolo, Crisafulli, Giovanni, Arena, Sabrina, Rospo, Giuseppe, Corti, Giorgio, Lorenzato, Annalisa, Cancelliere, Carlotta, Barault, Ludovic, Gionfriddo, Giulia, Linnebacher, Michael, Russo, Mariangela, Di Nicolantonio, Federica, and Bardelli, Alberto
- Abstract
Multiple strategies are continuously being explored to expand the drug target repertoire in solid tumors. We devised a novel computational workflow for transcriptome‐wide gene expression outlier analysis that allows the systematic identification of both overexpression and underexpression events in cancer cells. Here, it was applied to expression values obtained through RNA sequencing in 226 colorectal cancer (CRC) cell lines that were also characterized by whole‐exome sequencing and microarray‐based DNA methylation profiling. We found cell models displaying an abnormally high or low expression level for 3533 and 965 genes, respectively. Gene expression abnormalities that have been previously associated with clinically relevant features of CRC cell lines were confirmed. Moreover, by integrating multi‐omics data, we identified both genetic and epigenetic alternations underlying outlier expression values. Importantly, our atlas of CRC gene expression outliers can guide the discovery of novel drug targets and biomarkers. As a proof of concept, we found that CRC cell lines lacking expression of the MTAP gene are sensitive to treatment with a PRMT5‐MTA inhibitor (MRTX1719). Finally, other tumor types may also benefit from this approach. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Whole exome sequencing analysis of urine trans-renal tumour DNA in metastatic colorectal cancer patients
- Author
-
Crisafulli, Giovanni, Mussolin, Benedetta, Cassingena, Andrea, Montone, Monica, Bartolini, Alice, Barault, Ludovic, Martinetti, Antonia, Morano, Federica, Pietrantonio, Filippo, Sartore-Bianchi, Andrea, Siena, Salvatore, Di Nicolantonio, Federica, Marsoni, Silvia, Bardelli, Alberto, and Siravegna, Giulia
- Published
- 2019
- Full Text
- View/download PDF
15. Radiologic and Genomic Evolution of Individual Metastases during HER2 Blockade in Colorectal Cancer
- Author
-
Siravegna, Giulia, Lazzari, Luca, Crisafulli, Giovanni, Sartore-Bianchi, Andrea, Mussolin, Benedetta, Cassingena, Andrea, Martino, Cosimo, Lanman, Richard B., Nagy, Rebecca J., Fairclough, Stephen, Rospo, Giuseppe, Corti, Giorgio, Bartolini, Alice, Arcella, Pamela, Montone, Monica, Lodi, Francesca, Lorenzato, Annalisa, Vanzati, Alice, Valtorta, Emanuele, Cappello, Giovanni, Bertotti, Andrea, Lonardi, Sara, Zagonel, Vittorina, Leone, Francesco, Russo, Mariangela, Balsamo, Antonella, Truini, Mauro, Di Nicolantonio, Federica, Amatu, Alessio, Bonazzina, Erica, Ghezzi, Silvia, Regge, Daniele, Vanzulli, Angelo, Trusolino, Livio, Siena, Salvatore, Marsoni, Silvia, and Bardelli, Alberto
- Published
- 2018
- Full Text
- View/download PDF
16. Genotyping tumour DNA in cerebrospinal fluid and plasma of a HER2-positive breast cancer patient with brain metastases
- Author
-
Siravegna, Giulia, Geuna, Elena, Mussolin, Benedetta, Crisafulli, Giovanni, Bartolini, Alice, Galizia, Danilo, Casorzo, Laura, Sarotto, Ivana, Scaltriti, Maurizio, Sapino, Anna, Bardelli, Alberto, and Montemurro, Filippo
- Published
- 2017
- Full Text
- View/download PDF
17. Loss of AXIN1 drives acquired resistance to WNT pathway blockade in colorectal cancer cells carrying RSPO3 fusions
- Author
-
Picco, Gabriele, Petti, Consalvo, Centonze, Alessia, Torchiaro, Erica, Crisafulli, Giovanni, Novara, Luca, Acquaviva, Andrea, Bardelli, Alberto, and Medico, Enzo
- Published
- 2017
- Full Text
- View/download PDF
18. An extended multi-locus molecular typing schema for Streptococcus pneumoniae demonstrates that a limited number of capsular switch events is responsible for serotype heterogeneity of closely related strains from different countries
- Author
-
Crisafulli, Giovanni, Guidotti, Silvia, Muzzi, Alessandro, Torricelli, Giulia, Moschioni, Monica, Masignani, Vega, Censini, Stefano, and Donati, Claudio
- Published
- 2013
- Full Text
- View/download PDF
19. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth
- Author
-
Germano, Giovanni, Lamba, Simona, Rospo, Giuseppe, Barault, Ludovic, Magrì, Alessandro, Maione, Federica, Russo, Mariangela, Crisafulli, Giovanni, Bartolini, Alice, Lerda, Giulia, Siravegna, Giulia, Mussolin, Benedetta, Frapolli, Roberta, Montone, Monica, Morano, Federica, de Braud, Filippo, Amirouchene-Angelozzi, Nabil, Marsoni, Silvia, D’Incalci, Maurizio, Orlandi, Armando, Giraudo, Enrico, Sartore-Bianchi, Andrea, Siena, Salvatore, Pietrantonio, Filippo, Di Nicolantonio, Federica, and Bardelli, Alberto
- Published
- 2017
- Full Text
- View/download PDF
20. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients
- Author
-
Siravegna, Giulia, Mussolin, Benedetta, Buscarino, Michela, Corti, Giorgio, Cassingena, Andrea, Crisafulli, Giovanni, Ponzetti, Agostino, Cremolini, Chiara, Amatu, Alessio, Lauricella, Calogero, Lamba, Simona, Hobor, Sebastijan, Avallone, Antonio, Valtorta, Emanuele, Rospo, Giuseppe, Medico, Enzo, Motta, Valentina, Antoniotti, Carlotta, Tatangelo, Fabiana, Bellosillo, Beatriz, Veronese, Silvio, Budillon, Alfredo, Montagut, Clara, Racca, Patrizia, Marsoni, Silvia, Falcone, Alfredo, Corcoran, Ryan B., Nicolantonio, Federica Di, Loupakis, Fotios, Siena, Salvatore, Sartore-Bianchi, Andrea, and Bardelli, Alberto
- Subjects
Care and treatment ,Physiological aspects ,Development and progression ,Genetic aspects ,Research ,Epidermal growth factor receptors -- Physiological aspects -- Genetic aspects -- Research ,Colorectal cancer -- Development and progression -- Genetic aspects -- Care and treatment -- Research - Abstract
An improved understanding of the underlying molecular pathology of CRC has enabled tailored treatment regimens and helped optimize outcomes. CRC tissue is currently used to define the molecular status of [...], 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 (1). 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.
- Published
- 2015
21. Editorial: The impact of genetics on CRC therapy: from adaptive mutability to drug resistance.
- Author
-
Crisafulli, Giovanni and Siravegna, Giulia
- Subjects
DRUG resistance ,GENETICS ,COLORECTAL cancer - Published
- 2023
- Full Text
- View/download PDF
22. Case report: Preclinical efficacy of NEDD8 and proteasome inhibitors in patient-derived models of signet ring high-grade mucinous colorectal cancer from a Lynch syndrome patient.
- Author
-
Torchiaro, Erica, Petti, Consalvo, Arena, Sabrina, Sassi, Francesco, Migliardi, Giorgia, Mellano, Alfredo, Porporato, Roberta, Basiricò, Marco, Gammaitoni, Loretta, Berrino, Enrico, Montone, Monica, Corti, Giorgio, Crisafulli, Giovanni, Marchiò, Caterina, Bardelli, Alberto, and Medico, Enzo
- Subjects
COLORECTAL cancer ,MUCINOUS adenocarcinoma ,PROTEASOME inhibitors ,HEREDITARY nonpolyposis colorectal cancer ,THERAPEUTICS ,DRUG efficacy ,TUMOR growth - Abstract
High-grade mucinous colorectal cancer (HGM CRC) is particularly aggressive, prone to metastasis and treatment resistance, frequently accompanied by "signet ring" cancer cells. A sizeable fraction of HGM CRCs (20-40%) arises in the context of the Lynch Syndrome, an autosomal hereditary syndrome that predisposes to microsatellite instable (MSI) CRC. Development of patient-derived preclinical models for this challenging subtype of colorectal cancer represents an unmet need in oncology. We describe here successful propagation of preclinical models from a case of early-onset, MSIpositive metastatic colorectal cancer in a male Lynch syndrome patient, refractory to standard care (FOLFOX6, FOLFIRI-Panitumumab) and, surprisingly, also to immunotherapy. Surgical material from a debulking operation was implanted in NOD/ SCID mice, successfully yielding one patient-derived xenograft (PDX). PDX explants were subsequently used to generate 2D and 3D cell cultures. Histologically, all models resembled the tumor of origin, displaying a high-grade mucinous phenotype with signet ring cells. For preclinical exploration of alternative treatments, in light of recent findings, we considered inhibition of the proteasome by bortezomib and of the related NEDD8 pathway by pevonedistat. Indeed, sensitivity to bortezomib was observed in mucinous adenocarcinoma of the lung, and we previously found that HGM CRC is preferentially sensitive to pevonedistat in models with low or absent expression of cadherin 17 (CDH17), a differentiationmarker. Wetherefore performed IHC on the tumor and models, and observed no CDH17 expression, suggesting sensitivity to pevonedistat. Both bortezomib and pevonedistat showed strong activity on 2D cells at 72 hours and on 3D organoids at 7 days, thus providing valid options for in vivo testing. Accordingly, three PDX cohorts were treated for four weeks, respectively with vehicle, bortezomib and pevonedistat. Both drugs significantly reduced tumor growth, as compared to the vehicle group. Interestingly, while bortezomib was more effective in vitro, pevonedistat was more effective in vivo. Drug efficacy was further substantiated by a reduction of cellularity and of Ki67-positive cells in the treated tumors. These results highlight proteasome and NEDD8 inhibition as potentially effective therapeutic approaches against Lynch syndrome-associated HGM CRC, also when the disease is refractory to all available treatment options. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. DNA damage response and repair genes in advanced bone and soft tissue sarcomas: An 8- gene signature as a candidate predictive biomarker of response to trabectedin and olaparib combination.
- Author
-
Merlini, Alessandra, Centomo, Maria Laura, Ferrero, Giulio, Chiabotto, Giulia, Miglio, Umberto, Berrino, Enrico, Giordano, Giorgia, Brusco, Silvia, Pisacane, Alberto, Maldi, Elena, Sarotto, Ivana, Capozzi, Federica, Lano, Cristina, Isella, Claudio, Crisafulli, Giovanni, Aglietta, Massimo, Dei Tos, Angelo Paolo, Sbaraglia, Marta, Sangiolo, Dario, and D’Ambrosio, Lorenzo
- Subjects
SARCOMA ,DNA damage ,NUCLEIC acid hybridization ,TRABECTEDIN ,OLAPARIB - Abstract
Background: Advanced and unresectable bone and soft tissue sarcomas (BSTS) still represent an unmet medical need. We demonstrated that the alkylating agent trabectedin and the PARP1-inhibitor olaparib display antitumor activity in BSTS preclinical models. Moreover, in a phase Ib clinical trial (NCT02398058), feasibility, tolerability and encouraging results have been observed and the treatment combination is currently under study in a phase II trial (NCT03838744). Methods: Differential expression of genes involved in DNA Damage Response and Repair was evaluated by Nanostring® technology, extracting RNA from pretreatment tumor samples of 16 responder (≥6-month progression free survival) and 16 non-responder patients. Data validation was performed by quantitative real-time PCR, RNA in situ hybridization, and immunohistochemistry. The correlation between the identified candidate genes and both progression-free survival and overall survival was investigated in the publicly available dataset “Sarcoma (TCGA, The Cancer Genome Atlas)”. Results: Differential RNA expression analysis revealed an 8-gene signature (CDKN2A, PIK3R1, SLFN11, ATM, APEX2, BLM, XRCC2, MAD2L2) defining patients with better outcome upon trabectedin+olaparib treatment. In responder vs. non-responder patients, a significant differential expression of these genes was further confirmed by RNA in situ hybridization and by qRTPCR and immunohistochemistry in selected experiments. Correlation between survival outcomes and genetic alterations in the identified genes was shown in the TCGA sarcoma dataset. Conclusions: This work identified an 8-gene expression signature to improve prediction of response to trabectedin+olaparib combination in BSTS. The predictive role of these potential biomarkers warrants further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
24. Genetic Evolution of Glioblastoma Stem-Like Cells From Primary to Recurrent Tumor.
- Author
-
Orzan, Francesca, De Bacco, Francesca, Crisafulli, Giovanni, Pellegatta, Serena, Mussolin, Benedetta, Siravegna, Giulia, D'Ambrosio, Antonio, Comoglio, Paolo M., Finocchiaro, Gaetano, and Boccaccio, Carla
- Abstract
Glioblastoma (GBM) is a lethal tumor that displays remarkable genetic heterogeneity. It is also known that GBM contains a cell hierarchy driven by GBM stem-like cells (GSCs), responsible for tumor generation, therapeutic resistance, and relapse. An important and still open issue is whether phylogenetically related GSCs can be found in matched primary and recurrent GBMs, and reflect tumor genetic evolution under therapeutic pressure. To address this, we analyzed the mutational profile of GSCs isolated from either human primary GBMs (primary GSCs) or their matched tumors recurring after surgery and chemoradiotherapy (recurrent GSCs). We found that recurrent GSCs can accumulate temozolomide-related mutations over primary GSCs, following both linear and branched patterns. In the latter case, primary and recurrent GSCs share a common set of lesions, but also harbor distinctive mutations indicating that primary and recurrent GSCs derive from a putative common ancestor GSC by divergent genetic evolution. Interestingly, TP53 mutations distinctive of recurrent GSCs were detectable at low frequency in the corresponding primary tumors and likely marked pre-existent subclones that evolved under therapeutic pressure and expanded in the relapsing tumor. Consistently, recurrent GSCs displayed in vitro greater therapeutic resistance than primary GSCs. Overall, these data indicate that (a) phylogenetically related GSCs are found in matched primary and recurrent GBMs and (b) recurrent GSCs likely pre-exist in the untreated primary tumor and are both mutagenized and positively selected by chemoradiotherapy. Stem Cells 2017;35:2218-2228 [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
25. Emergence of MET hyper-amplification at progression to MET and BRAF inhibition in colorectal cancer.
- Author
-
Oddo, Daniele, Siravegna, Giulia, Gloghini, Annunziata, Vernieri, Claudio, Mussolin, Benedetta, Morano, Federica, Crisafulli, Giovanni, Berenato, Rosa, Corti, Giorgio, Volpi, Chiara Costanza, Buscarino, Michela, Niger, Monica, Dunne, Philip D, Rospo, Giuseppe, Valtorta, Emanuele, Bartolini, Alice, Fucà, Giovanni, Lamba, Simona, Martinetti, Antonia, and Di Bartolomeo, Maria
- Abstract
Background:Combined MET and BRAF inhibition showed clinical benefit in a patient with rectal cancer carrying BRAF
V600E 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. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
26. Sequence Analysis of 96 Genomic Regions Identifies Distinct Evolutionary Lineages within CC156, the Largest Streptococcus pneumoniae Clonal Complex in the MLST Database.
- Author
-
Moschioni, Monica, Lo Sapio, Morena, Crisafulli, Giovanni, Torricelli, Giulia, Guidotti, Silvia, Muzzi, Alessandro, Barocchi, Michèle A., and Donati, Claudio
- Subjects
BIOLOGICAL evolution ,GENOMICS ,STREPTOCOCCUS pneumoniae ,CLONING ,NUCLEOTIDE sequence ,EPIDEMIOLOGICAL research ,POPULATION biology - Abstract
Multi-Locus Sequence Typing (MLST) of Streptococcus pneumoniae is based on the sequence of seven housekeeping gene fragments. The analysis of MLST allelic profiles by eBURST allows the grouping of genetically related strains into Clonal Complexes (CCs) including those genotypes with a common descent from a predicted ancestor. However, the increasing use of MLST to characterize S. pneumoniae strains has led to the identification of a large number of new Sequence Types (STs) causing the merger of formerly distinct lineages into larger CCs. An example of this is the CC156, displaying a high level of complexity and including strains with allelic profiles differing in all seven of the MLST loci, capsular type and the presence of the Pilus Islet-1 (PI-1). Detailed analysis of the CC156 indicates that the identification of new STs, such as ST4945, induced the merging of formerly distinct clonal complexes. In order to discriminate the strain diversity within CC156, a recently developed typing schema, 96-MLST, was used to analyse 66 strains representative of 41 different STs. Analysis of allelic profiles by hierarchical clustering and a minimum spanning tree identified ten genetically distinct evolutionary lineages. Similar results were obtained by phylogenetic analysis on the concatenated sequences with different methods. The identified lineages are homogenous in capsular type and PI-1 presence. ST4945 strains were unequivocally assigned to one of the lineages. In conclusion, the identification of new STs through an exhaustive analysis of pneumococcal strains from various laboratories has highlighted that potentially unrelated subgroups can be grouped into a single CC by eBURST. The analysis of additional loci, such as those included in the 96-MLST schema, will be necessary to accurately discriminate the clonal evolution of the pneumococcal population. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
27. PARP1 Inhibitor and Trabectedin Combination Does Not Increase Tumor Mutational Burden in Advanced Sarcomas—A Preclinical and Translational Study.
- Author
-
Pignochino, Ymera, Crisafulli, Giovanni, Giordano, Giorgia, Merlini, Alessandra, Berrino, Enrico, Centomo, Maria Laura, Chiabotto, Giulia, Brusco, Silvia, Basiricò, Marco, Maldi, Elena, Pisacane, Alberto, Leuci, Valeria, Sangiolo, Dario, D'Ambrosio, Lorenzo, Aglietta, Massimo, Kasper, Bernd, Bardelli, Alberto, and Grignani, Giovanni
- Subjects
- *
THERAPEUTIC use of antineoplastic agents , *GENETIC mutation , *XENOGRAFTS , *CONFIDENCE intervals , *CANCER patients , *DESCRIPTIVE statistics , *TRANSLATIONAL research , *CELL lines , *DATA analysis software , *ODDS ratio , *ENZYME inhibitors , *SARCOMA - Abstract
Simple Summary: Immunotherapy has revolutionized cancer treatment, but not for all tumor types. Indeed, sarcomas are considered "immune-cold" tumors, which are relatively unresponsive to immunotherapy. One strategy to potentiate immunotherapy efficacy is to increase tumor immunogenicity, for instance by boosting the number of candidate targets (neoantigens) to be recognized by the immune system. Tumor mutational burden indicates the number of somatic mutations identified in the tumor and normalized per megabase. Tumor mutational burden is considered as an acceptable, measurable surrogate of tumor neoantigens. Here, we explored whether the combination of two DNA-damaging agents, trabectedin and olaparib, could increase tumor mutational burden in sarcomas, to prime subsequent immunotherapy. We found no variation in tumor mutational burden after trabectedin + olaparib in preclinical and clinical samples. Therefore, other aspects should be considered to increase sarcoma immunogenicity, by exploiting different pathways such as the potential modulation of the tumor microenvironment induced by trabectedin + olaparib. Drug-induced tumor mutational burden (TMB) may contribute to unleashing the immune response in relatively "immune-cold" tumors, such as sarcomas. We previously showed that PARP1 inhibition perpetuates the DNA damage induced by the chemotherapeutic agent trabectedin in both preclinical models and sarcoma patients. In the present work, we explored acquired genetic changes in DNA repair genes, mutational signatures, and TMB in a translational platform composed of cell lines, xenografts, and tumor samples from patients treated with trabectedin and olaparib combination, compared to cells treated with temozolomide, an alkylating agent that induces hypermutation. Whole-exome and targeted panel sequencing data analyses revealed that three cycles of trabectedin and olaparib combination neither affected the mutational profiles, DNA repair gene status, or copy number alterations, nor increased TMB both in homologous recombinant-defective and proficient cells or in xenografts. Moreover, TMB was not increased in tumor specimens derived from trabectedin- and olaparib-treated patients (5–6 cycles) when compared to pre-treatment biopsies. Conversely, repeated treatments with temozolomide induced a massive TMB increase in the SJSA-1 osteosarcoma model. In conclusion, a trabectedin and olaparib combination did not show mutagenic effects and is unlikely to prime subsequent immune-therapeutic interventions based on TMB increase. On the other hand, these findings are reassuring in the increasing warning of treatment-induced hematologic malignancies correlated to PARP1 inhibitor use. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
28. Acquired RAS or EGFR mutations and duration of response to EGFR blockade in colorectal cancer.
- Author
-
Van Emburgh, Beth O., Arena, Sabrina, Siravegna, Giulia, Lazzari, Luca, Crisafulli, Giovanni, Corti, Giorgio, Mussolin, Benedetta, Baldi, Federica, Buscarino, Michela, Bartolini, Alice, Valtorta, Emanuele, Vidal, Joana, Bellosillo, Beatriz, Germano, Giovanni, Pietrantonio, Filippo, Ponzetti, Agostino, Albanell, Joan, Siena, Salvatore, Sartore-Bianchi, Andrea, and Di Nicolantonio, Federica
- Published
- 2016
- Full Text
- View/download PDF
29. Erratum: Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients.
- Author
-
Siravegna, Giulia, Mussolin, Benedetta, Buscarino, Michela, Corti, Giorgio, Cassingena, Andrea, Crisafulli, Giovanni, Ponzetti, Agostino, Cremolini, Chiara, Amatu, Alessio, Lauricella, Calogero, Lamba, Simona, Hobor, Sebastijan, Avallone, Antonio, Valtorta, Emanuele, Rospo, Giuseppe, Medico, Enzo, Motta, Valentina, Antoniotti, Carlotta, Tatangelo, Fabiana, and Bellosillo, Beatriz
- Subjects
EPIDERMAL growth factor receptors ,COLON cancer patients - Abstract
A correction to the article "Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients" that was published in a previous issue of 2015.
- Published
- 2015
- Full Text
- View/download PDF
30. Circulating Tumor DNA to Drive Treatment in Metastatic Colorectal Cancer.
- Author
-
Patelli G, Mauri G, Tosi F, Amatu A, Bencardino K, Bonazzina E, Pizzutilo EG, Villa F, Calvanese G, Agostara AG, Stabile S, Ghezzi S, Crisafulli G, Di Nicolantonio F, Marsoni S, Bardelli A, Siena S, and Sartore-Bianchi A
- Subjects
- Humans, Biomarkers, Tumor genetics, Biomarkers, Tumor therapeutic use, Circulating Tumor DNA genetics, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Colonic Neoplasms drug therapy, Rectal Neoplasms drug therapy, Antineoplastic Agents therapeutic use
- Abstract
In the evolving molecular treatment landscape of metastatic colorectal cancer (mCRC), the identification of druggable alterations is pivotal to achieve the best therapeutic opportunity for each patient. Because the number of actionable targets is expanding, there is the need to timely detect their presence or emergence to guide the choice of different available treatment options. Liquid biopsy, through the analysis of circulating tumor DNA (ctDNA), has proven safe and effective as a complementary method to address cancer evolution while overcoming the limitations of tissue biopsy. Even though data are accumulating regarding the potential for ctDNA-guided treatments applied to targeted agents, still major gaps in knowledge exist as for their application to different areas of the continuum of care. In this review, we recapitulate how ctDNA information could be exploited to drive different targeted treatment strategies in mCRC patients, by refining molecular selection before treatment by addressing tumor heterogeneity beyond tumor tissue biopsy; longitudinally monitoring early-tumor response and resistance mechanisms to targeted agents, potentially leading to tailored, molecular-driven, therapeutic options; guiding the molecular triage towards rechallenge strategies with anti-EGFR agents, suggesting the best time for retreatment; and providing opportunities for an "enhanced rechallenge" through additional treatments or combos aimed at overcoming acquired resistance. Besides, we discuss future perspectives concerning the potential role of ctDNA to fine-tune investigational strategies such as immuno-oncology., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
31. Liquid Biopsy of Cerebrospinal Fluid Enables Selective Profiling of Glioma Molecular Subtypes at First Clinical Presentation.
- Author
-
Orzan F, De Bacco F, Lazzarini E, Crisafulli G, Gasparini A, Dipasquale A, Barault L, Macagno M, Persico P, Pessina F, Bono B, Giordano L, Zeppa P, Melcarne A, Cassoni P, Garbossa D, Santoro A, Comoglio PM, Indraccolo S, Simonelli M, and Boccaccio C
- Subjects
- Humans, Mutation, Prognosis, Liquid Biopsy, DNA, Neoplasm, Biomarkers, Tumor genetics, Glioma diagnosis, Glioma genetics, Glioma pathology, Brain Neoplasms diagnosis, Brain Neoplasms genetics
- Abstract
Purpose: Current glioma diagnostic guidelines call for molecular profiling to stratify patients into prognostic and treatment subgroups. In case the tumor tissue is inaccessible, cerebrospinal fluid (CSF) has been proposed as a reliable tumor DNA source for liquid biopsy. We prospectively investigated the use of CSF for molecular characterization of newly diagnosed gliomas., Experimental Design: We recruited two cohorts of newly diagnosed patients with glioma, one (n = 45) providing CSF collected in proximity of the tumor, the other (n = 39) CSF collected by lumbar puncture (LP). Both cohorts provided tumor tissues by surgery concomitant with CSF sampling. DNA samples retrieved from CSF and matched tumors were systematically characterized and compared by comprehensive (NGS, next-generation sequencing) or targeted (ddPCR, droplet digital PCR) methodologies. Conventional and molecular diagnosis outcomes were compared., Results: We report that tumor DNA is abundant in CSF close to the tumor, but scanty and mostly below NGS sensitivity threshold in CSF from LP. Indeed, tumor DNA is mostly released by cells invading liquoral spaces, generating a gradient that attenuates by departing from the tumor. Nevertheless, in >60% of LP CSF samples, tumor DNA is sufficient to assess a selected panel of genetic alterations (IDH and TERT promoter mutations, EGFR amplification, CDKN2A/B deletion: ITEC protocol) and MGMT methylation that, combined with imaging, enable tissue-agnostic identification of main glioma molecular subtypes., Conclusions: This study shows potentialities and limitations of CSF liquid biopsy in achieving molecular characterization of gliomas at first clinical presentation and proposes a protocol to maximize diagnostic information retrievable from CSF DNA., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
32. Temozolomide Treatment Alters Mismatch Repair and Boosts Mutational Burden in Tumor and Blood of Colorectal Cancer Patients.
- Author
-
Crisafulli G, Sartore-Bianchi A, Lazzari L, Pietrantonio F, Amatu A, Macagno M, Barault L, Cassingena A, Bartolini A, Luraghi P, Mauri G, Battuello P, Personeni N, Zampino MG, Pessei V, Vitiello PP, Tosi F, Idotta L, Morano F, Valtorta E, Bonoldi E, Germano G, Di Nicolantonio F, Marsoni S, Siena S, and Bardelli A
- Subjects
- Antineoplastic Agents, Alkylating pharmacology, Antineoplastic Agents, Alkylating therapeutic use, Cell Line, Tumor, DNA Mismatch Repair, DNA-Binding Proteins genetics, Dacarbazine therapeutic use, Humans, Mutation, O(6)-Methylguanine-DNA Methyltransferase genetics, O(6)-Methylguanine-DNA Methyltransferase metabolism, Temozolomide pharmacology, Temozolomide therapeutic use, Brain Neoplasms drug therapy, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics
- Abstract
The majority of metastatic colorectal cancers (mCRC) are mismatch repair (MMR) proficient and unresponsive to immunotherapy, whereas MMR-deficient (MMRd) tumors often respond to immune-checkpoint blockade. We previously reported that the treatment of colorectal cancer preclinical models with temozolomide (TMZ) leads to MMR deficiency, increased tumor mutational burden (TMB), and sensitization to immunotherapy. To clinically translate these findings, we designed the ARETHUSA clinical trial whereby O6-methylguanine-DNA-methyltransferase (MGMT)-deficient, MMR-proficient, RAS-mutant mCRC patients received priming therapy with TMZ. Analysis of tissue biopsies and circulating tumor DNA (ctDNA) revealed the emergence of a distinct mutational signature and increased TMB after TMZ treatment. Multiple alterations in the nucleotide context favored by the TMZ signature emerged in MMR genes, and the p.T1219I MSH6 variant was detected in ctDNA and tissue of 94% (16/17) of the cases. A subset of patients whose tumors displayed the MSH6 mutation, the TMZ mutational signature, and increased TMB achieved disease stabilization upon pembrolizumab treatment., Significance: MMR-proficient mCRCs are unresponsive to immunotherapy. We provide the proof of concept that inactivation of MMR genes can be achieved pharmacologically with TMZ and molecularly monitored in the tissue and blood of patients with mCRC. This strategy deserves additional evaluation in mCRC patients whose tumors are no longer responsive to standard-of-care treatments. See related commentary by Willis and Overman, p. 1612. This article is highlighted in the In This Issue feature, p. 1599., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
33. Werner Helicase Is a Synthetic-Lethal Vulnerability in Mismatch Repair-Deficient Colorectal Cancer Refractory to Targeted Therapies, Chemotherapy, and Immunotherapy.
- Author
-
Picco G, Cattaneo CM, van Vliet EJ, Crisafulli G, Rospo G, Consonni S, Vieira SF, Rodríguez IS, Cancelliere C, Banerjee R, Schipper LJ, Oddo D, Dijkstra KK, Cinatl J, Michaelis M, Yang F, Di Nicolantonio F, Sartore-Bianchi A, Siena S, Arena S, Voest EE, Bardelli A, and Garnett MJ
- Subjects
- Colorectal Neoplasms genetics, Drug Therapy, Humans, Immunotherapy, Molecular Targeted Therapy, Colorectal Neoplasms therapy, DNA Mismatch Repair, Werner Syndrome Helicase genetics
- Abstract
Targeted therapies, chemotherapy, and immunotherapy are used to treat patients with mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal cancer. The clinical effectiveness of targeted therapy and chemotherapy is limited by resistance and drug toxicities, and about half of patients receiving immunotherapy have disease that is refractory to immune checkpoint inhibitors. Loss of Werner syndrome ATP-dependent helicase (WRN) is a synthetic lethality in dMMR/MSI-H cells. To inform the development of WRN as a therapeutic target, we performed WRN knockout or knockdown in 60 heterogeneous dMMR colorectal cancer preclinical models, demonstrating that WRN dependency is an almost universal feature and a robust marker for patient selection. Furthermore, models of resistance to clinically relevant targeted therapy, chemotherapy, and immunotherapy retain WRN dependency. These data show the potential of therapeutically targeting WRN in patients with dMMR/MSI-H colorectal cancer and support WRN as a therapeutic option for patients with dMMR/MSI-H cancers refractory to current treatment strategies. SIGNIFICANCE: We found that a large, diverse set of dMMR/MSI-H colorectal cancer preclinical models, including models of treatment-refractory disease, are WRN-dependent. Our results support WRN as a promising synthetic-lethal target in dMMR/MSI-H colorectal cancer tumors as a monotherapy or in combination with targeted agents, chemotherapy, or immunotherapy. This article is highlighted in the In This Issue feature, p. 1861 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
34. Molecular Landscape of Acquired Resistance to Targeted Therapy Combinations in BRAF-Mutant Colorectal Cancer.
- Author
-
Oddo D, Sennott EM, Barault L, Valtorta E, Arena S, Cassingena A, Filiciotto G, Marzolla G, Elez E, van Geel RM, Bartolini A, Crisafulli G, Boscaro V, Godfrey JT, Buscarino M, Cancelliere C, Linnebacher M, Corti G, Truini M, Siravegna G, Grasselli J, Gallicchio M, Bernards R, Schellens JH, Tabernero J, Engelman JA, Sartore-Bianchi A, Bardelli A, Siena S, Corcoran RB, and Di Nicolantonio F
- Subjects
- Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Gene Amplification, Humans, Signal Transduction, Colorectal Neoplasms genetics, Drug Resistance, Neoplasm genetics, Gene Dosage genetics, Proto-Oncogene Proteins B-raf genetics
- Abstract
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., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
35. Tumor Heterogeneity and Lesion-Specific Response to Targeted Therapy in Colorectal Cancer.
- Author
-
Russo M, Siravegna G, Blaszkowsky LS, Corti G, Crisafulli G, Ahronian LG, Mussolin B, Kwak EL, Buscarino M, Lazzari L, Valtorta E, Truini M, Jessop NA, Robinson HE, Hong TS, Mino-Kenudson M, Di Nicolantonio F, Thabet A, Sartore-Bianchi A, Siena S, Iafrate AJ, Bardelli A, and Corcoran RB
- Subjects
- Antibodies, Monoclonal therapeutic use, Cell Line, Tumor, Cetuximab therapeutic use, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, DNA, Neoplasm blood, Humans, Liver Neoplasms genetics, Liver Neoplasms secondary, Molecular Targeted Therapy, Panitumumab, Precision Medicine, Treatment Outcome, Colorectal Neoplasms drug therapy, Drug Resistance, Neoplasm, Liver Neoplasms drug therapy, MAP Kinase Kinase 1 genetics, Mutation, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
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 patient's 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., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
36. Acquired Resistance to the TRK Inhibitor Entrectinib in Colorectal Cancer.
- Author
-
Russo M, Misale S, Wei G, Siravegna G, Crisafulli G, Lazzari L, Corti G, Rospo G, Novara L, Mussolin B, Bartolini A, Cam N, Patel R, Yan S, Shoemaker R, Wild R, Di Nicolantonio F, Bianchi AS, Li G, Siena S, and Bardelli A
- Subjects
- Animals, Catalytic Domain, Cell Line, Tumor, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Gene Rearrangement, Humans, Mice, Mutation, Neoplasm Transplantation, Neoplastic Cells, Circulating pathology, Receptor, trkA chemistry, Benzamides administration & dosage, Colorectal Neoplasms drug therapy, Drug Resistance, Neoplasm, Indazoles administration & dosage, Protein Kinase Inhibitors administration & dosage, Receptor, trkA genetics
- Abstract
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 patient's 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., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
37. Emergence of Multiple EGFR Extracellular Mutations during Cetuximab Treatment in Colorectal Cancer.
- Author
-
Arena S, Bellosillo B, Siravegna G, Martínez A, Cañadas I, Lazzari L, Ferruz N, Russo M, Misale S, González I, Iglesias M, Gavilan E, Corti G, Hobor S, Crisafulli G, Salido M, Sánchez J, Dalmases A, Bellmunt J, De Fabritiis G, Rovira A, Di Nicolantonio F, Albanell J, Bardelli A, and Montagut C
- Subjects
- Antineoplastic Agents therapeutic use, Blotting, Western, Cell Line, Tumor, Cetuximab therapeutic use, Colorectal Neoplasms drug therapy, DNA Mutational Analysis, Extracellular Space genetics, Flow Cytometry, Humans, Real-Time Polymerase Chain Reaction, Colorectal Neoplasms genetics, Drug Resistance, Neoplasm genetics, Genes, erbB-1 genetics, Mutation
- Abstract
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., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.