568 results on '"Filippo de Braud"'
Search Results
2. A Phase Ib/II Study of WNT974 + Encorafenib + Cetuximab in Patients With BRAF V600E-Mutant KRAS Wild-Type Metastatic Colorectal Cancer
- Author
-
Josep Tabernero, Eric Van Cutsem, Elena Garralda, David Tai, Filippo De Braud, Ravit Geva, Mark T J van Bussel, Katia Fiorella Dotti, Elena Elez, María J de Miguel, Kevin Litwiler, Danielle Murphy, Michelle Edwards, Van Karlyle Morris, Institut Català de la Salut, [Tabernero J] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. UVic-UCC, IOB-Quiron, Barcelona, Spain. [Van Cutsem E] University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium. [Garralda E] START Madrid, Hospital Universitario HM Sanchinarro, Madrid, Spain. [Tai D] Division of Medical Oncology, National Cancer Centre Singapore, Singapore. [De Braud F] Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy. [Geva R] Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel. [Elez E] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Cancer Research ,Genetic Phenomena::Genetic Variation::Mutation [PHENOMENA AND PROCESSES] ,terapéutica::terapéutica::farmacoterapia::protocolos antineoplásicos::terapéutica::farmacoterapia::protocolos de quimioterapia antineoplásica combinada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Medicaments antineoplàstics - Ús terapèutic ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,colorectal cancer ,encorafenib ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,neoplasias::neoplasias por localización::neoplasias del sistema digestivo::neoplasias gastrointestinales::neoplasias intestinales::neoplasias colorrectales [ENFERMEDADES] ,metastatic ,Còlon - Càncer - Tractament ,Anomalies cromosòmiques ,Neoplasms::Neoplasms by Site::Digestive System Neoplasms::Gastrointestinal Neoplasms::Intestinal Neoplasms::Colorectal Neoplasms [DISEASES] ,Oncology ,Therapeutics::Therapeutics::Drug Therapy::Antineoplastic Protocols::Therapeutics::Drug Therapy::Antineoplastic Combined Chemotherapy Protocols [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Recte - Càncer - Tractament ,cetuximab ,WNT974 ,fenómenos genéticos::variación genética::mutación [FENÓMENOS Y PROCESOS] - Abstract
Background WNT974 is a small molecule inhibitor of Wnt signaling that specifically inhibits porcupine O-acyltransferase. This phase Ib dose-escalation study evaluated the maximum tolerated dose of WNT974 in combination with encorafenib and cetuximab in patients with BRAF V600E-mutant metastatic colorectal cancer with RNF43 mutations or RSPO fusions. Patients and Methods Patients received once-daily encorafenib and weekly cetuximab, in addition to once-daily WNT974, in sequential dosing cohorts. In the first cohort, patients received 10-mg WNT974 (COMBO10), which was reduced in subsequent cohorts to 7.5-mg (COMBO7.5) or 5-mg (COMBO5) after dose–limiting toxicities (DLTs) were observed. Primary endpoints were incidence of DLTs and exposure to WNT974 and encorafenib. Secondary endpoints were anti-tumor activity and safety. Results Twenty patients were enrolled (COMBO10, n = 4; COMBO7.5, n = 6; COMBO5, n = 10). DLTs were observed in 4 patients, including grade 3 hypercalcemia (COMBO10, n = 1; COMBO7.5, n = 1), grade 2 dysgeusia (COMBO10, n = 1), and lipase increased (COMBO10, n = 1). A high incidence of bone toxicities (n = 9) was reported, including rib fracture, spinal compression fracture, pathological fracture, foot fracture, hip fracture, and lumbar vertebral fracture. Serious adverse events were reported in 15 patients, most frequently bone fracture, hypercalcemia, and pleural effusion. The overall response rate was 10% and disease control rate 85%; most patients achieved stable disease as their best response. Conclusion Concerns surrounding the safety and lack of preliminary evidence of improved anti-tumor activity of WNT974 + encorafenib + cetuximab, compared with previous encorafenib + cetuximab data, ultimately led to study discontinuation. Phase II was not initiated. Trial registration ClinicalTrials.gov, NCT02278133
- Published
- 2023
- Full Text
- View/download PDF
3. Clinical Outcomes After Progression on First-Line Therapies in IDH1 Mutated Versus Wild-Type Intrahepatic Cholangiocarcinoma Patients
- Author
-
Margherita Rimini, Carles Fabregat-Franco, Mara Persano, Valentina Burgio, Francesca Bergamo, Monica Niger, Mario Scartozzi, Ilario Giovanni Rapposelli, Giuseppe Aprile, Francesca Ratti, Federica Pedica, Helena Verdaguer, Mario Rizzato, Federico Nichetti, Eleonora Lai, Alessandro Cappetta, Teresa Macarulla, Matteo Fassan, Filippo De Braud, Andrea Pretta, Francesca Simionato, Francesco De Cobelli, Luca Aldrighetti, Lorenzo Fornaro, Stefano Cascinu, and Andrea Casadei-Gardini
- Subjects
Cancer Research ,Oncology ,Pharmacology (medical) - Published
- 2023
- Full Text
- View/download PDF
4. A multicenter phase 2 single arm study of cabozantinib in patients with advanced or unresectable renal cell carcinoma pre-treated with one immune-checkpoint inhibitor: The BREAKPOINT trial (Meet-Uro trial 03)
- Author
-
Giuseppe Procopio, Mélanie Claps, Chiara Pircher, Luca Porcu, Pierangela Sepe, Valentina Guadalupi, Ugo De Giorgi, Davide Bimbatti, Franco Nolè, Francesco Carrozza, Sebastiano Buti, Roberto Iacovelli, Chiara Ciccarese, Cristina Masini, Cinzia Baldessari, Laura Doni, Antonio Cusmai, Angela Gernone, Sarah Scagliarini, Sandro Pignata, Filippo de Braud, and Elena Verzoni
- Subjects
Cancer Research ,Oncology ,General Medicine - Abstract
Background: First-line therapies based on immune-checkpoint inhibitors (ICIs) significantly improved survival of metastatic renal cell carcinoma (mRCC) patients. Cabozantinib was shown to target kinases involved in immune-escape and to prolong survival in patients pre-treated with tyrosine-kinase-inhibitors (TKIs). The impact of ICIs combinations in first line on subsequent therapies is still unclear. Methods: This is an open label, multicenter, single arm, phase II study designed to assess activity, safety and efficacy of cabozantinib in mRCC patients progressed after an adjuvant or first line anti-Programmed Death (PD)-1/PD-Ligand (PD-L) 1-based therapy. Primary endpoint was progression free survival (PFS), secondary endpoints were overall survival (OS), objective response rate (ORR) and safety. Results: 31 patients were included in the analysis. After a median (m) follow-up of 11.9 months, mPFS was 8.3 months (90%CI 3.9-17.4) and mOS was 13.8 months (95%CI 7.7-29.0). ORR was 37.9% with an additional 13 patients achieving disease stability. Grade 3-4 adverse events occurred in 47% of patients, including more frequently creatine phosphokinase (CPK) serum level elevation, neutropenia, hyponatremia, diarrhea, hand-food syndrome, oral mucositis and hypertension. Conclusions: The BREAKPOINT trial met its primary endpoint showing that cabozantinib as second line therapy after ICIs was active in mRCC. Safety profile was manageable. Trial registration number: NCT03463681 - A Study of CaBozantinib in Patients With Advanced or Unresectable Renal cEll cArcinoma (BREAKPOINT) - https://clinicaltrials.gov/ct2/show/NCT03463681
- Published
- 2022
- Full Text
- View/download PDF
5. Next-generation sequencing analysis of cholangiocarcinoma identifies distinct IDH1-mutated clusters
- Author
-
Margherita Rimini, Eleonora Loi, Carles Fabregat-Franco, Valentina Burgio, Sara Lonardi, Monica Niger, Mario Scartozzi, Ilario G. Raposelli, Giuseppe Aprile, Francesca Ratti, Federica Pedica, Helena Verdaguer, Mario Rizzato, Federico Nichetti, Eleonora Lai, Alessandro Cappetta, Teresa Macarulla, Matteo Fassan, Filippo De Braud, Andrea Pretta, Francesca Simionato, Francesco De Cobelli, Luca Aldrighetti, Lorenzo Fornaro, Stefano Cascinu, Patrizia Zavattari, Andrea Casadei-Gardini, Rimini, Margherita, Loi, Eleonora, Fabregat-Franco, Carle, Burgio, Valentina, Lonardi, Sara, Niger, Monica, Scartozzi, Mario, Raposelli, Ilario G, Aprile, Giuseppe, Ratti, Francesca, Pedica, Federica, Verdaguer, Helena, Rizzato, Mario, Nichetti, Federico, Lai, Eleonora, Cappetta, Alessandro, Macarulla, Teresa, Fassan, Matteo, De Braud, Filippo, Pretta, Andrea, Simionato, Francesca, De Cobelli, Francesco, Aldrighetti, Luca, Fornaro, Lorenzo, Cascinu, Stefano, Patrizia, Zavattari, and Casadei-Gardini, Andrea
- Subjects
Cancer Research ,Genomic profiling ,High-Throughput Nucleotide Sequencing ,Cholangiocarcinoma ,Clustering analysis ,IDH1 mutation ,Next-generation sequencing ,Chromatin ,Isocitrate Dehydrogenase ,Phosphatidylinositol 3-Kinases ,Bile Ducts, Intrahepatic ,Bile Duct Neoplasms ,Oncology ,Mutation ,Humans - Abstract
IDH1-mutated intrahepatic cholangiocarcinomas (IDH1m iCCAs) could be treated with anti-IDH1 drugs, although the high heterogeneity in this class of tumours could limit treatment efficacy.We selected 125 IDH1m iCCAs that were treated as resectable, locally advanced, or metastatic and were screened by the NGS-based FoundationOne gene panel. We conducted a mutation-based clustering of tumours and survival analysis.Three main clusters were identified. The most altered pathways in cluster 1 were cell cycle and apoptosis, RTK/RAS, PI3K, and chromatin modification. Of note, CDKN2A/2B were mutated in 41/44 patients of this cluster. In cluster 2, the most affected pathways were as follows: Chromatin modification, DNA damage control, PI3K, and RTK/RAS. In this cluster, the most frequently mutated genes were ARID1A and PBRM1. The most altered pathways in cluster 3 were as follows: Cell cycle and apoptosis, DNA damage control, TP53, and chromatin modification. Importantly, TP53 was mutated only in cluster 3 patients. In the cohort of patients treated with surgery, cluster 2 showed statistically significant better disease-free survival (DFS) and overall survival (OS) compared with patients in cluster 3 and cluster 1 (p = 0.0014 and p = 0.0003, respectively). In the advanced setting, cluster 2 experienced a statistically significant better PFS (p = 0.0012), a tendency toward a better OS from first-line treatment, and a better OS from first-line progression compared with patients in cluster 1 and cluster 3 (p = 0.0017). We proposed an easy-to-use algorithm able to stratify patients in the three clusters on the basis of the genomic profile.We highlighted three different mutation-based clusters with prognostic significance in a cohort of IDH1m iCCAs.
- Published
- 2022
- Full Text
- View/download PDF
6. Circulating Fatty Acid Profile as a Biomarker for Immunotherapy in Advanced Non-Small Cell Lung Cancer
- Author
-
Giulia Galli, Paola Antonia Corsetto, Claudia Proto, Giuseppe Lo Russo, Monica Ganzinelli, Eliana Rulli, Lorenzo Legramandi, Daniele Morelli, Roberto Ferrara, Arsela Prelaj, Diego Signorelli, Alessandro De Toma, Marta Brambilla, Mario Occhipinti, Sara Manglaviti, Mattia Boeri, Antonia Martinetti, Andrea Vingiani, Mario Paolo Colombo, Angela Maria Rizzo, Valter Torri, Filippo de Braud, Sabina Sangaletti, Antonio Sica, and Marina Chiara Garassino
- Subjects
Pulmonary and Respiratory Medicine ,Cancer Research ,Lung Neoplasms ,Settore MED/06 - Oncologia Medica ,Fatty Acids ,Fatty acids ,Immunotherapy ,Lipid metabolism ,Membrane fluidity ,Non-small cell lung cancer ,B7-H1 Antigen ,Oncology ,Settore BIO/10 - Biochimica ,Carcinoma, Non-Small-Cell Lung ,Humans ,Inflammation Mediators ,Biomarkers - Abstract
Lipid metabolism impacts immune cell differentiation, activation, and functions, modulating inflammatory mediators, energy homeostasis, and cell membrane composition. Despite preclinical evidence, data in humans lack concerning tumors and immunotherapy (IO). We aimed at investigating the correlations between circulating lipids and the outcome of non-small cell lung cancer (NSCLC) patients treated with IO.We identified all patients with advanced NSCLC treated with IO at our Institution with available baseline plasma samples. Fatty acids (FAs) were analyzed through gas chromatography. Survival curves were estimated by the Kaplan-Meier method. Cox multivariate models were constructed through a stepwise procedure, with entry and exit P value set at .2.We identified 112 patients, mostly with performance status 1 (65.2%) and PD-L1≥1% (75.3%). Median progression-free survival (PFS) and overall survival (OS) were 2.8 and 11.0 months, respectively. Multivariable model for survival identified a positive association of circulating free (FFA) C16:0 (P .005) and esterified (EFA) C16:1 (P .030) with PFS, and a positive association of EFA C16:1 (P .001) and EFA C18:0 (P .020) with OS. EFA C16:0 was negatively associated with PFS (P .008).FFA C16:0 and FAs derived from its unsaturation (EFA C16:1) and elongation (EFA C18:0) are associated with a better outcome in NSCLC patients treated with IO. It is conceivable that the ratio among those FAs may modify membrane fluidity and receptor activity, influencing IO efficacy. These data pave the way for the investigation of lipid-modulating strategies in association with IO in NSCLC.
- Published
- 2022
- Full Text
- View/download PDF
7. Gene mutational profile of BRCAness and clinical implication in predicting response to platinum-based chemotherapy in patients with intrahepatic cholangiocarcinoma
- Author
-
Margherita Rimini, Teresa Macarulla, Valentina Burgio, Sara Lonardi, Monica Niger, Mario Scartozzi, Ilario G. Rapposelli, Giuseppe Aprile, Francesca Ratti, Federica Pedica, Helena Verdaguer, Floriana Nappo, Federico Nichetti, Eleonora Lai, Martina Valgiusti, Alessandro Cappetta, Carles Fabregat-Franco, Matteo Fassan, Filippo De Braud, Marco Puzzoni, Giovanni L. Frassineti, Francesca Simionato, Francesco De Cobelli, Luca Aldrighetti, Lorenzo Fornaro, Stefano Cascinu, Andrea Casadei-Gardini, Rimini, Margherita, Macarulla, Teresa, Burgio, Valentina, Lonardi, Sara, Niger, Monica, Scartozzi, Mario, Rapposelli, Ilario G, Aprile, Giuseppe, Ratti, Francesca, Pedica, Federica, Verdaguer, Helena, Nappo, Floriana, Nichetti, Federico, Lai, Eleonora, Valgiusti, Martina, Cappetta, Alessandro, Febregat, Carle, Fassan, Matteo, De Braud, Filippo, Puzzoni, Marco, Frassineti, Giovanni L, Simionato, Francesca, De Cobelli, Francesco, Aldrighetti, Luca, Fornaro, Lorenzo, Cascinu, Stefano, and Casadei-Gardini, Andrea
- Subjects
BRCAness phenotype ,Biliary tract cancer ,PARP inhibitors ,Platinum-based chemotherapy ,Cancer Research ,Organoplatinum Compounds ,Antineoplastic Agents ,Genetic Profile ,Prognosis ,Cholangiocarcinoma ,Bile Ducts, Intrahepatic ,Bile Duct Neoplasms ,Oncology ,Mutation ,Humans - Abstract
Biliary tract cancers are rare malignancies with a poor prognosis and scarce therapeutic strategies. The significance of BRCAness in this setting is already unknown.Tissue specimens of BTC patients treated with platinum-based chemotherapy have been analyzed through the FOUNDATIONPne assay.72/150 (48%) BRCAness mutated and 78/150 (52.0%) wild type (WT) patients were included. The most commonly mutated genes in the BRCAness mutated group were: ARID1A (N = 32, 44%), CDKN2A (N = 23, 32%), KRAS/NRAS (N = 16, 22%), CDKN2B (N = 13, 18%), BRCA2 (N = 13, 18%), PBRM1 (N = 12, 17%), ATM (N = 11, 15%), FGFR2 (N = 10, 14%), TP53 (N = 8, 11%), IRS2 (N = 7, 10%), CREBBP (N = 7, 10%) (table 3, figure 1). At the univariate analysis BRCAness mutation was associated with longer median Progression Free Survival (mPFS) (HR 0.68; 95% CI 0.49-0.95; p = 0.0254); it was not associated with longer mOS but a trend toward a benefit in survival was found (HR 0.77; 95% CI 0.50-1.19; p = 0.2388). Patients with BRCAness mutation showed a higher percentage of disease control rate (77.8 vs 67.9; p = 0.04) compared to patients WT. Multivariate analysis confirmed BRCAness mutation (HR 0.66; 95% CI: 0.45-0.98; p = 0.0422) as independent favorable prognostic factors for PFS and a positive trend was found for OS (HR 0.84; 95% CI: 0.53-1.33; p = 0.3652).BRCAness BTC patients showed a better PFS compared BRCAnessWT patients after exposure to platinum-based chemotherapy. Moreover, the OS curves' trend showed in our analysis suggests that BRCAness mutated patients could benefit from a maintenance therapy with PARPi.
- Published
- 2022
- Full Text
- View/download PDF
8. Clinical activity of <scp>CC‐90011</scp> , an oral, potent, and reversible <scp>LSD1</scp> inhibitor, in advanced malignancies
- Author
-
Antoine Hollebecque, Stefania Salvagni, Ruth Plummer, Patricia Niccoli, Jaume Capdevila, Giuseppe Curigliano, Victor Moreno, Filippo de Braud, Sonia Gonzalez de Villambrosia, Patricia Martin‐Romano, Eric Baudin, Marina Arias, Juan de Alvaro, Josep L. Parra‐Palau, Tania Sánchez‐Pérez, Ida Aronchik, Ellen H. Filvaroff, Manisha Lamba, Zariana Nikolova, Johann S. de Bono, Institut Català de la Salut, [Hollebecque A] Gustave Roussy, Département d’innovation thérapeutique et essais précoces, Villejuif, France. [Salvagni S] S. Orsola Polyclinic, Malpighi University Hospital, Bologna, Italy. [Plummer R] Clinical and Translational Research Institute Northern, Newcastle University, Newcastle, UK. [Niccoli P] Department of Medical Oncology, ENETS Center of Excellence, IPC NET Center, Institut Paoli-Calmettes, Marseille, France. [Capdevila J] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Vall d’Hebron Hospital Universitari, Barcelona, Spain. IOB-Teknon, Barcelona, Spain. [Curigliano G] European Institute of Oncology, IRCCS, University of Milan, Milan, Italy, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
enzimas y coenzimas::enzimas::oxidorreductasas::oxidorreductasas que actúan sobre donantes de grupos CH-NH::oxidorreductasas N-desmetilantes::histona desmetilasas [COMPUESTOS QUÍMICOS Y DROGAS] ,Histone Demethylases ,Cancer Research ,Maximum Tolerated Dose ,Càncer - Tractament ,Enzymes and Coenzymes::Enzymes::Oxidoreductases::Oxidoreductases Acting on CH-NH Group Donors::Oxidoreductases, N-Demethylating::Histone Demethylases [CHEMICALS AND DRUGS] ,Neoplasms::Neoplasms by Histologic Type::Lymphoma::Lymphoma, Non-Hodgkin::Lymphoma, B-Cell::Lymphoma, B-Cell, Marginal Zone [DISEASES] ,neoplasias::neoplasias por tipo histológico::linfoma::linfoma no Hodgkin::linfoma de células B::linfoma de células B de la zona marginal [ENFERMEDADES] ,Investigative Techniques::Toxicity Tests::Maximum Tolerated Dose [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Lymphoma, B-Cell, Marginal Zone ,Posologia ,Neoplasms [DISEASES] ,neoplasias [ENFERMEDADES] ,Oncology ,Neoplasms ,Medicaments - Eficàcia ,técnicas de investigación::pruebas de toxicidad::dosis máxima tolerada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Humans ,Organic Chemicals - Abstract
Lysine-specific demethylase 1 (LSD1) inhibitor; Neuroendocrine tumor; Non-Hodgkin lymphoma Tumor neuroendocrino; Linfoma no Hodgkin; Inhibidores de desmetilasa-1 específica a lisina Tumor neuroendocrí; Limfoma no Hodgkin; Inhibidor de la desmetilasa-1 específica a la lisina Background CC-90011 is an oral, potent, selective, reversible inhibitor of lysine-specific demethylase 1 (LSD1) that was well tolerated, with encouraging activity in patients who had advanced solid tumors or relapsed/refractory marginal zone lymphoma. The authors present long-term safety and efficacy and novel pharmacodynamic and pharmacokinetic data from the first-in-human study of CC-90011. Methods CC-90011-ST-001 (ClincalTrials.gov identifier NCT02875223; Eudract number 2015–005243-13) is a phase 1, multicenter study in which patients received CC-90011 once per week in 28-day cycles. The objectives were to determine the safety, maximum tolerated dose, and/or recommended phase 2 dose (primary) and to evaluate preliminary efficacy and pharmacokinetics (secondary). Results Sixty-nine patients were enrolled, including 50 in the dose-escalation arm and 19 in the dose-expansion arm. Thrombocytopenia was the most common treatment-related adverse event and was successfully managed with dose modifications. Clinical activity with prolonged, durable responses were observed, particularly in patients who had neuroendocrine neoplasms. In the dose-escalation arm, one patient with relapsed/refractory marginal zone lymphoma achieved a complete response (ongoing in cycle 58). In the dose-expansion arm, three patients with neuroendocrine neoplasms had stable disease after nine or more cycles, including one patient who was in cycle 46 of ongoing treatment. CC-90011 decreased levels of secreted neuroendocrine peptides chromogranin A, progastrin-releasing peptide, and RNA expression of the blood pharmacodynamic marker monocyte-to-macrophage differentiation–associated. Conclusions The safety profile of CC-90011 suggested that its reversible mechanism of action may provide an advantage over other irreversible LSD1 inhibitors. The favorable tolerability profile, clinical activity, durable responses, and once-per-week dosing support further exploration of CC-90011 as monotherapy and in combination with other treatments for patients with advanced solid tumors and other malignancies. This study was supported by Bristol Myers Squibb, Princeton, New Jersey, USA. Writing and editorial assistance was provided by Bio Connections, LLC, funded by Bristol Myers Squibb.
- Published
- 2022
- Full Text
- View/download PDF
9. <scp>MGMT</scp> inactivation as a new biomarker in patients with advanced biliary tract cancers
- Author
-
Monica Niger, Federico Nichetti, Andrea Casadei‐Gardini, Federica Morano, Chiara Pircher, Elena Tamborini, Federica Perrone, Matteo Canale, Daniel B. Lipka, Andrea Vingiani, Luca Agnelli, Anna Dobberkau, Jennifer Hüllein, Felix Korell, Christoph E. Heilig, Sara Pusceddu, Francesca Corti, Michele Droz, Paola Ulivi, Michele Prisciandaro, Maria Antista, Marta Bini, Laura Cattaneo, Massimo Milione, Hanno Glimm, Bruno C. Köhler, Giancarlo Pruneri, Daniel Hübschmann, Stefan Fröhling, Vincenzo Mazzaferro, Filippo Pietrantonio, Maria Di Bartolomeo, and Filippo de Braud
- Subjects
Cancer Research ,Settore MED/06 - Oncologia Medica ,MGMT ,biliary tract cancer ,biomarker ,cholangiocarcinoma ,molecular profiling ,temozolomide ,Biomarkers ,DNA Modification Methylases ,DNA Repair Enzymes ,Humans ,O(6)-Methylguanine-DNA Methyltransferase ,Precision Medicine ,Tumor Suppressor Proteins ,Bile Duct Neoplasms ,Biliary Tract Neoplasms ,General Medicine ,Oncology ,Genetics ,Molecular Medicine - Abstract
Biliary tract cancers (BTCs) have poor prognosis and limited therapeutic options. The impact of O
- Published
- 2022
- Full Text
- View/download PDF
10. Temozolomide Followed by Combination With Low-Dose Ipilimumab and Nivolumab in Patients With Microsatellite-Stable, O6-Methylguanine–DNA Methyltransferase–Silenced Metastatic Colorectal Cancer: The MAYA Trial
- Author
-
Federica Morano, Alessandra Raimondi, Filippo Pagani, Sara Lonardi, Lisa Salvatore, Chiara Cremolini, Sabina Murgioni, Giovanni Randon, Federica Palermo, Lorenzo Antonuzzo, Nicoletta Pella, Patrizia Racca, Michele Prisciandaro, Monica Niger, Francesca Corti, Francesca Bergamo, Alberto Zaniboni, Margherita Ratti, Michele Palazzo, Celeste Cagnazzo, Maria Alessandra Calegari, Federica Marmorino, Iolanda Capone, Elena Conca, Adele Busico, Silvia Brich, Elena Tamborini, Federica Perrone, Massimo Di Maio, Massimo Milione, Maria Di Bartolomeo, Filippo de Braud, and Filippo Pietrantonio
- Subjects
O(6)-Methylguanine-DNA Methyltransferase ,Cancer Research ,Nivolumab ,Settore MED/06 - ONCOLOGIA MEDICA ,Oncology ,Rectal Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Colonic Neoplasms ,Temozolomide ,Humans ,Colorectal Neoplasms ,Ipilimumab ,Microsatellite Repeats - Abstract
PURPOSE This is a multicenter, single-arm phase II trial evaluating the efficacy and safety of an immune-sensitizing strategy with temozolomide priming followed by a combination of low-dose ipilimumab and nivolumab in patients with microsatellite-stable (MSS) and O6-methylguanine–DNA methyltransferase (MGMT)–silenced metastatic colorectal cancer (mCRC). PATIENTS AND METHODS Patients with pretreated mCRC were centrally prescreened for MSS status and MGMT silencing (ie, lack of MGMT expression by immunohistochemistry plus MGMT methylation by pyrosequencing). Eligible patients received two priming cycles of oral temozolomide 150 mg/sqm once daily, days 1-5, once every 4 weeks (first treatment part) followed, in absence of progression, by its combination with ipilimumab 1 mg/kg once every 8 weeks and nivolumab 480 mg once every 4 weeks (second treatment part). The primary end point was the 8-month progression-free survival (PFS) rate calculated from enrollment in patients who started the second treatment part, with ≥ 4 out of 27 subjects progression-free by the 8-month time point as decision rule. RESULTS Among 716 prescreened patients, 204 (29%) were molecularly eligible and 135 started the first treatment part. Among these, 102 (76%) were discontinued because of death or disease progression on temozolomide priming, whereas 33 patients (24%) who achieved disease control started the second treatment part and represented the final study population. After a median follow-up of 23.1 months (interquartile range, 14.9-24.6 months), 8-month PFS rate was 36%. Median PFS and overall survival were 7.0 and 18.4 months, respectively, and overall response rate was 45%. Grade 3-4 immune-related adverse events were skin rash (6%), colitis (3%), and hypophysitis (3%). No unexpected adverse events or treatment-related deaths were reported. CONCLUSION The MAYA study provided proof-of-concept that a sequence of temozolomide priming followed by a combination of low-dose ipilimumab and nivolumab may induce durable clinical benefit in MSS and MGMT-silenced mCRC.
- Published
- 2022
- Full Text
- View/download PDF
11. BCL6 and the Notch pathway: a signaling axis leading to a novel druggable biotarget in triple negative breast cancer
- Author
-
Francesca De Santis, Sandra L. Romero-Cordoba, Lorenzo Castagnoli, Tatiana Volpari, Simona Faraci, Giovanni Fucà, Elda Tagliabue, Filippo De Braud, Serenella M. Pupa, and Massimo Di Nicola
- Subjects
Cancer Research ,Receptors, Notch ,Oncology ,Cell Line, Tumor ,Neoplastic Stem Cells ,Proto-Oncogene Proteins c-bcl-6 ,Humans ,Molecular Medicine ,Triple Negative Breast Neoplasms ,General Medicine ,Cell Proliferation ,Signal Transduction - Abstract
The transcriptional repressor B-cell lymphoma 6 (BCL6) is dysregulated in several neoplasms, but its role in triple negative breast cancer (TNBC), a highly aggressive subtype which lacks effective treatment, is unclear. The presence of intratumoral cancer stem cells (CSCs) is a main cause of tumor relapse. The Notch signaling pathway is crucial for regulating CSC self-renewal and promoting breast cancer (BC) development and resistance to anticancer therapies. Here, we investigated signaling cascades of BCL6 in the CSC compartment of TNBCs, and the mechanisms that govern its activity, mainly through Notch signaling.Gene expression, somatic copy number alterations and clinical data from the Cancer Genome Atlas and METABRIC were accessed through the Xena and cbioportal browsers. Public transcriptome profiles from TNBC datasets were retrieved from the Gene Expression Omnibus. Mammosphere formation efficiency was calculated after BCL6 knockdown via transient siRNA transfection, stable silencing or pharmacological inhibition. The effects exhibited via BCL6 inhibition in putative TNBC stem-like cells were evaluated by immunofluorescence and qRT-PCR analyses. Chromatin immunoprecipitation experiments were performed to validate a putative BCL6 responsive element located in the first intron of the Numb gene and to define the circuit of corepressors engaged by BCL6 following its inhibition. Immunoprecipitation assays were carried out to investigate a novel interaction at the basis of BCL6 control of CSC activity in TNBC.In silico analyses of benchmarked public datasets revealed a significant enrichment of BCL6 in cancer stemness related pathways, particularly of Notch signaling in TNBC. In vitro stable inhibition of BCL6 significantly reduced tumor cell growth and, accordingly, we found that the mammosphere formation efficiency of BCL6 silenced cells was significantly impaired by pharmacological inhibition of Notch signaling. BCL6 was found to be expressed at significantly higher levels in TNBC mammospheres than in their adherent counterparts, and loss of BCL6 function significantly decreased mammosphere formation with preferential targeting of CD44-positive versus ALDH-positive stem-like cells. Functional interplay between BCL6 and the chromatin remodeling factor EZH2 triggered the BCL6/Notch stemness signaling axis via inhibition of Numb transcription.Our results may be instrumental for the prospective design of combination treatment strategies that selectively target novel TNBC-associated biomarker(s) whose activity is implicated in the regulation of cancer stemness (such as BCL6) and molecules in developmentally conserved signaling pathways (such as Notch) to achieve long-lasting tumor control and improve patient outcomes.
- Published
- 2022
- Full Text
- View/download PDF
12. Paraneoplastic neurological syndromes in patients affected by SCLC: a case series
- Author
-
Caterina Zanella, Alberto Giovanni Leone, Luca Zambelli, Achille Bottiglieri, Luca Canziani, Marta Brambilla, Giuseppe Lo Russo, Marco Platania, Filippo De Braud, and Mario Occhipinti
- Subjects
Cancer Research ,Lung Neoplasms ,Paraneoplastic Syndromes ,SCLC, paraneoplastic neurological syndromes, sensory neuropathy, autonomic neuropathy, Anti-Hu antibodies, immunotherapy ,SCLC ,General Medicine ,paraneoplastic neurological syndromes ,Small Cell Lung Carcinoma ,Oncology ,sensory neuropathy ,Humans ,immunotherapy ,Anti-Hu antibodies ,autonomic neuropathy ,Paraneoplastic Syndromes, Nervous System ,Autoantibodies - Abstract
Paraneoplastic syndromes occur in about 0.1% of patients affected by neoplastic diseases. In some types of tumors, such as Small Cell Lung Cancer (SCLC), Thymoma, and particular forms of Plasmacytoma, the association with Paraneoplastic Neurological Syndromes (PNS) is much more frequent. It seems that these syndromes are triggered by tumor production of factors normally expressed only by the individual's nervous system. The immune system stimulates an autoimmune response against these factors that induce neurological symptoms. Also, in light of the latest updates on the relationship between immunotherapy and PNS as well as of the introduction of first-line immunotherapy in SCLC, it seems that the use of immunotherapy in SCLC is associated with concomitant increase in autoimmune neurological syndromes. In this article, we report our experience at Istituto Nazionale Tumori of Milan with three patients affected by SCLC and PNS. Our experience seems to confirm that the oncological treatment scarcely impacts the paraneoplastic neurological deficits. Further research is needed to improve the treatment and recovery of patients affected by PNS.
- Published
- 2022
- Full Text
- View/download PDF
13. Prognostic significance of HER2-low status in HR-positive/HER2-negative advanced breast cancer treated with CDK4/6 inhibitors
- Author
-
Emma Zattarin, Daniele Presti, Luigi Mariani, Caterina Sposetti, Rita Leporati, Alice Menichetti, Chiara Corti, Chiara Benvenuti, Giovanni Fucà, Riccardo Lobefaro, Francesca Ligorio, Leonardo Provenzano, Andrea Vingiani, Marta Del Vecchio, Gaia Griguolo, Marianna Sirico, Ottavia Bernocchi, Antonio Marra, Paola Zagami, Elisa Agostinetto, Flavia Jacobs, Pierluigi Di Mauro, Andrea Esposito, Carlo Alberto Giorgi, Luca Lalli, Laura Boldrini, Pier Paolo Berton Giacchetti, Ambra Carnevale Schianca, Valentina Guarneri, Rebecca Pedersini, Agnese Losurdo, Alberto Zambelli, Daniele Generali, Carmen Criscitiello, Giuseppe Curigliano, Giancarlo Pruneri, Filippo de Braud, Maria Vittoria Dieci, and Claudio Vernieri
- Subjects
Oncology ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging - Abstract
Whether Human Epidermal growth factor Receptor 2 (HER2)-low status has prognostic significance in HR + /HER2- advanced Breast Cancer (aBC) patients treated with first-line Endocrine Therapy plus CDK 4/6 inhibitors remains unclear. In 428 patients evaluated, HER2-low status was independently associated with significantly worse PFS and OS when compared with HER2-0 status. Based on our findings, HER2-low status could become a new prognostic biomarker in this clinical setting.
- Published
- 2023
- Full Text
- View/download PDF
14. Reinduction of an Anti-EGFR-based First-line Regimen in Patients with RAS Wild-type Metastatic Colorectal Cancer Enrolled in the Valentino Study
- Author
-
Giovanni Fucà, Alessandra Raimondi, Michele Prisciandaro, Sara Lonardi, Chiara Cremolini, Margherita Ratti, Matteo Clavarezza, Roberto Murialdo, Andrea Sartore-Bianchi, Valeria Smiroldo, Rosa Berenato, Patrizia Racca, Francesca Bergamo, Salvatore Corallo, Maria Di Bartolomeo, Filippo de Braud, Federica Morano, and Filippo Pietrantonio
- Subjects
Proto-Oncogene Proteins B-raf ,Cancer Research ,Settore MED/06 - Oncologia Medica ,Rectal Neoplasms ,metastatic colorectal cancer ,Panitumumab ,chemotherapy ,anti-EGFR ,Oncology ,reinduction ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Prospective Studies ,Colonic Neoplasms ,Colorectal Neoplasms - Abstract
Background In patients with RAS/BRAF wild-type metastatic colorectal cancer (mCRC), growing evidence supports anti-epidermal growth factor receptor (EGFR) retreatment, whereas little is known on the outcomes of anti-EGFR-based reinduction therapy during the upfront strategy. Methods We included patients enrolled in the Valentino study who had disease progression and received at least one dose of post-progression therapy. The Kaplan–Meier method and Cox proportional hazards regression were used for the survival analysis. When comparing the outcomes of anti-EGFR-based reinduction versus any second line, a propensity score–based matching was used. Results Liver-limited/single site of disease (P < .001 and P = .002), left-sidedness (P = .029), surgery of metastases (P = .003), early tumor shrinkage, and deeper responses (P = .018 and P = .036) were associated with the use of anti-EGFR-based reinduction versus any other second line. All patients treated with reinduction had an anti-EGFR-free interval of at least 3 months. In the propensity score–matched population, progression-free survival (PFS) was similar in the 2 treatment groups, the overall survival (OS) was significantly longer for patients treated with reinduction (P = .029), and the response rate was higher in patients treated with reinduction (P = .033). An oxaliplatin-free interval ≥12 months, left-sidedness, and molecular hyperselection beyond RAS/BRAF were associated with significantly better outcomes after anti-EGFR-based reinduction. Conclusions Reinduction strategies with anti-EGFR-based regimens are commonly used in clinical practice. Our data highlight the importance of clinical–molecular selection for re-treatments and the need for prospective strategy trials in selected populations.
- Published
- 2022
- Full Text
- View/download PDF
15. Selpercatinib in Patients With RET Fusion–Positive Non–Small-Cell Lung Cancer: Updated Safety and Efficacy From the Registrational LIBRETTO-001 Phase I/II Trial
- Author
-
Alexander Drilon, Vivek Subbiah, Oliver Gautschi, Pascale Tomasini, Filippo de Braud, Benjamin J. Solomon, Daniel Shao-Weng Tan, Guzmán Alonso, Jürgen Wolf, Keunchil Park, Koichi Goto, Victoria Soldatenkova, Sylwia Szymczak, Scott S. Barker, Tarun Puri, Aimee Bence Lin, Herbert Loong, Benjamin Besse, Institut Català de la Salut, [Drilon A] Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY. [Subbiah V] The University of Texas MD Anderson Cancer Center, Houston, TX. [Gautschi O] University of Berne and Cantonal Hospital of Lucerne, Lucerne, Switzerland. [Tomasini P] Hôpitaux Universitaires, de Marseille Timone, France. [de Braud F] University of Milan, Milan, Italy. [Solomon BJ] Peter MacCallum Cancer Center, Melbourne, Australia. [Alonso G] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Cancer Research ,Otros calificadores::Otros calificadores::/genética [Otros calificadores] ,Otros calificadores::/uso terapéutico [Otros calificadores] ,Medicaments antineoplàstics - Ús terapèutic ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,diagnóstico::pronóstico::resultado del tratamiento [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Pulmons - Càncer - Aspectes genètics ,Neoplasms::Neoplasms by Site::Thoracic Neoplasms::Respiratory Tract Neoplasms::Lung Neoplasms::Bronchial Neoplasms::Carcinoma, Bronchogenic::Carcinoma, Non-Small-Cell Lung [DISEASES] ,neoplasias::neoplasias por localización::neoplasias torácicas::neoplasias del tracto respiratorio::neoplasias pulmonares::neoplasias de los bronquios::carcinoma broncogénico::carcinoma de pulmón de células no pequeñas [ENFERMEDADES] ,Diagnosis::Prognosis::Treatment Outcome [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Oncology ,Avaluació de resultats (Assistència sanitària) ,Other subheadings::Other subheadings::/genetics [Other subheadings] ,Other subheadings::/therapeutic use [Other subheadings] ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] ,Pulmons - Càncer - Tractament - Abstract
Selpercatinib; Lung cancer; Safety Selpercatinib; Cáncer de pulmón; Seguridad Selpercatinib; Càncer de pulmó; Seguretat PURPOSE Selpercatinib, a first-in-class, highly selective, and potent CNS-active RET kinase inhibitor, is currently approved for the treatment of patients with RET fusion–positive non–small-cell lung cancer (NSCLC). We provide a registrational data set update in more than double (n = 316) of the original reported population (n = 144) and better characterization of long-term efficacy and safety. METHODS Patients were enrolled to LIBRETTO-001, a phase I/II, single-arm, open-label study of selpercatinib in patients with RET-altered cancers. An analysis of patients with RET fusion–positive NSCLC, including 69 treatment-naive and 247 with prior platinum-based chemotherapy, was performed. The primary end point was objective response rate (ORR; RECIST v1.1, independent review committee). Secondary end points included duration of response (DoR), progression-free survival (PFS), overall survival, and safety. RESULTS In treatment-naive patients, the ORR was 84% (95% CI, 73 to 92); 6% achieved complete responses (CRs). The median DoR was 20.2 months (95% CI, 13.0 to could not be evaluated); 40% of responses were ongoing at the data cutoff (median follow-up of 20.3 months). The median PFS was 22.0 months; 35% of patients were alive and progression-free at the data cutoff (median follow-up of 21.9 months). In platinum-based chemotherapy pretreated patients, the ORR was 61% (95% CI, 55 to 67); 7% achieved CRs. The median DoR was 28.6 months (95% CI, 20.4 to could not be evaluated); 49% of responses were ongoing (median follow-up of 21.2 months). The median PFS was 24.9 months; 38% of patients were alive and progression-free (median follow-up of 24.7 months). Of 26 patients with measurable baseline CNS metastasis by the independent review committee, the intracranial ORR was 85% (95% CI, 65 to 96); 27% were CRs. In the full safety population (n = 796), the median treatment duration was 36.1 months. The safety profile of selpercatinib was consistent with previous reports. CONCLUSION In a large cohort with extended follow-up, selpercatinib continued to demonstrate durable and robust responses, including intracranial activity, in previously treated and treatment-naive patients with RET fusion–positive NSCLC. Supported by Loxo Oncology, a wholly owned subsidiary of Eli Lilly and Company. A.D. was supported in part by funding from the National Cancer Institute of the National Institutes of Health: 1R01CA251591- 01A1 and P30 CA008748. Partial support was likewise provided by LUNGevity.
- Published
- 2023
16. Initial Evidence for the Efficacy of Naporafenib in Combination With Trametinib in NRAS-Mutant Melanoma : Results From the Expansion Arm of a Phase Ib, Open-Label Study
- Author
-
Filippo de Braud, Christophe Dooms, Rebecca S. Heist, Celeste Lebbe, Martin Wermke, Anas Gazzah, Dirk Schadendorf, Piotr Rutkowski, Jürgen Wolf, Paolo A. Ascierto, Ignacio Gil-Bazo, Shumei Kato, Maria Wolodarski, Meredith McKean, Eva Muñoz Couselo, Martin Sebastian, Armando Santoro, Vesselina Cooke, Luca Manganelli, Kitty Wan, Anil Gaur, Jaeyeon Kim, Giordano Caponigro, Xuân-Mai Couillebault, Helen Evans, Catarina D. Campbell, Sumit Basu, Michele Moschetta, and Adil Daud
- Subjects
Cancer Research ,Oncology ,Medizin - Abstract
PURPOSE No approved targeted therapy for the treatment of patients with neuroblastoma RAS viral (v-ras) oncogene homolog ( NRAS)–mutant melanoma is currently available. PATIENTS AND METHODS In this phase Ib escalation/expansion study (ClinicalTrials.gov identifier: NCT02974725 ), the safety, tolerability, and preliminary antitumor activity of naporafenib (LXH254), a BRAF/CRAF protein kinases inhibitor, were explored in combination with trametinib in patients with advanced/metastatic KRAS- or BRAF-mutant non–small-cell lung cancer (escalation arm) or NRAS-mutant melanoma (escalation and expansion arms). RESULTS Thirty-six and 30 patients were enrolled in escalation and expansion, respectively. During escalation, six patients reported grade ≥3 dose-limiting toxicities, including dermatitis acneiform (n = 2), maculopapular rash (n = 2), increased lipase (n = 1), and Stevens-Johnson syndrome (n = 1). The recommended doses for expansion were naporafenib 200 mg twice a day plus trametinib 1 mg once daily and naporafenib 400 mg twice a day plus trametinib 0.5 mg once daily. During expansion, all 30 patients experienced a treatment-related adverse event, the most common being rash (80%, n = 24), blood creatine phosphokinase increased, diarrhea, and nausea (30%, n = 9 each). In expansion, the objective response rate, median duration of response, and median progression-free survival were 46.7% (95% CI, 21.3 to 73.4; 7 of 15 patients), 3.75 (95% CI, 1.97 to not estimable [NE]) months, and 5.52 months, respectively, in patients treated with naporafenib 200 mg twice a day plus trametinib 1 mg once daily, and 13.3% (95% CI, 1.7 to 40.5; 2 of 15 patients), 3.75 (95% CI, 2.04 to NE) months, and 4.21 months, respectively, in patients treated with naporafenib 400 mg twice a day plus trametinib 0.5 mg once daily. CONCLUSION Naporafenib plus trametinib showed promising preliminary antitumor activity in patients with NRAS-mutant melanoma. Prophylactic strategies aimed to lower the incidence of skin-related events are under investigation.
- Published
- 2023
17. Impact of Diabetes and Metformin Use on Enteropancreatic Neuroendocrine Tumors: Post Hoc Analysis of the CLARINET Study
- Author
-
Sara Pusceddu, Claudio Vernieri, Massimo Di Maio, Natalie Prinzi, Martina Torchio, Francesca Corti, Jorgelina Coppa, Roberto Buzzoni, Maria Di Bartolomeo, Massimo Milione, Benjamin Regnault, Xuan-Mai Truong Thanh, Vincenzo Mazzaferro, and Filippo de Braud
- Subjects
Cancer Research ,Oncology ,diabetes mellitus ,progression-free survival ,lanreotide ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Article ,RC254-282 - Abstract
Simple Summary Lanreotide was found to be effective for preventing tumors from worsening in patients with neuroendocrine tumors of the intestines and pancreas, regardless of whether or not patients also had diabetes. Metformin, a drug for treating diabetes, also seemed to prevent tumors from worsening. Abstract The prognostic role of diabetes mellitus (DM) in advanced enteropancreatic neuroendocrine tumors (NETs) is unclear. Progression free survival (PFS) was assessed in post-hoc analyses of the 96-week, phase III, double-blind, placebo-controlled CLARINET study of lanreotide 120 mg in patients with advanced non-functional enteropancreatic NETs with DM (with/without metformin) and without DM. Of 204 patients, there were 79 with DM (lanreotide, n = 42 {metformin, n = 14}; placebo, n = 37 {metformin, n = 10}) and 125 without DM (lanreotide, n = 59; placebo, n = 66). Median PFS was 96.0 and 98.0 weeks with and without DM, respectively (hazard ratio 1.20 {95% confidence interval 0.79 to 1.82}; p = 0.380). No difference in PFS was observed in lanreotide-treated patients with/without DM (p = 0.8476). In the placebo group, median PFS was numerically shorter with versus without DM (p = 0.052) and was significantly longer in patients with DM and metformin (85.7 weeks) versus without metformin (38.7 weeks; p = 0.009). Multivariable Cox analyses showed that DM at baseline was not associated with PFS (p = 0.079); lanreotide was significantly associated with lower disease progression risk (p = 0.017). Lanreotide efficacy was confirmed in patients with advanced enteropancreatic NETs, regardless of diabetic status; DM was not a negative prognostic factor. A potential antitumor effect of metformin was observed in patients receiving placebo.
- Published
- 2022
18. Liquid Biopsy and Radiological Response Predict Outcomes Following Discontinuation of Targeted Therapy in Patients with BRAF Mutated Melanoma
- Author
-
Lorenza Di Guardo, Viviana Vallacchi, Michele Del Vecchio, Monica Rodolfo, Filippo de Braud, Giovanni Randon, Mara Cossa, Marta Bini, Roberto Patuzzo, Licia Rivoltini, Gianfrancesco Gallino, Ilaria Mattavelli, Andrea Maurichi, Mario Santinami, Barbara Valeri, Carolina Cimminiello, Roberta Ruggeri, Giovanni Fucà, Alessandra Raimondi, Martina Angi, and Francesca Corti
- Subjects
Proto-Oncogene Proteins B-raf ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Target therapy ,BRAF ,Targeted therapy ,Internal medicine ,medicine ,Humans ,Liquid biopsy ,Melanoma ,Retrospective Studies ,business.industry ,MEK inhibitor ,Liquid Biopsy ,Cancer ,Neoplasms, Second Primary ,medicine.disease ,Confidence interval ,Discontinuation ,Clinical trial ,Melanoma and Cutaneous Malignancies ,business - Abstract
Background Outcomes of patients with metastatic melanoma discontinuing BRAF‐targeted therapy for cumulative toxicity after sustained response are unknown. Materials and Methods This retrospective case series analysis conducted at a single Cancer Center in Italy included patients with BRAF mutated metastatic melanoma treated with a BRAF inhibitor as a single agent or in combination with a MEK inhibitor between June 1, 2011 and January 1, 2020 and interrupted treatment due to cumulative toxicity after achieving complete response (CR) or long‐lasting partial response (PR; i.e. >12 months). Results We included 24 patients with a median treatment duration of 59.4 months (95% confidence interval [CI], 55.4–63.4; range, 12–88). CR and PR were achieved in 71% and 29% of patients, respectively. At a median follow‐up after treatment discontinuation of 37.8 months (95% CI, 33.7–41.9), the 12‐month progression‐free survival after discontinuation (dPFS) rate was 70.8% (95% CI 54.8–91.6) and 24‐month dPFS rate was 58.3% (95% CI, 41.6–81.8). Baseline patient and tumor characteristics as well as treatment duration and best response did not significantly impact on dPFS. Patients with CR and negative circulating tumor DNA (ctDNA) at time of discontinuation had a significantly improved dPFS compared with patients with either radiological residual disease or ctDNA positivity (p = .007). No patient in CR with undetectable ctDNA experienced progression. Conclusion The risk of progression is high even in patients with sustained sensitivity to BRAF/MEK inhibitors. Integration of liquid biopsy in clinical trials investigating the optimal management of patients with sustained sensitivity to BRAF/MEK inhibitors is warranted. Implications for Practice Outcomes of patients with metastatic melanoma discontinuing BRAF‐targeted therapy for cumulative toxicity are unknown. This study analyzed patients with sustained responses (median treatment duration 59.4 months). Twelve‐ and 24‐month progression‐free survival following discontinuation were 70.8% and 58.3%, respectively. Complete response and negative circulating tumor DNA at time of discontinuation are promising prognostic biomarkers in this setting., This article reports clinical outcomes of patients with metastatic melanoma treated with BRAF/MEK inhibitors who interrupted the treatment after achieving a long‐lasting objective response, exploring the role of circulating tumor DNA assessed at time of discontinuation as a biomarker for post‐discontinuation progression‐free survival.
- Published
- 2021
- Full Text
- View/download PDF
19. The Pan-Immune-Inflammation Value in Patients with Metastatic Melanoma Receiving First-Line Therapy
- Author
-
Carolina Cimminiello, Lorenza Di Guardo, Massimo Di Nicola, Laura Mazzeo, Marta Del Vecchio, Filippo de Braud, Marta Bini, Giovanni Randon, Teresa Beninato, Michele Del Vecchio, Giulia Apollonio, Claudia Lauria Pantano, Giovanni Fucà, and Ilaria Bisogno
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Pharmacology (medical) ,Melanoma ,Aged ,Retrospective Studies ,Inflammation ,business.industry ,Hazard ratio ,Immunotherapy ,Odds ratio ,medicine.disease ,Confidence interval ,030104 developmental biology ,030220 oncology & carcinogenesis ,Absolute neutrophil count ,Biomarker (medicine) ,Female ,business - Abstract
Since a non-negligible fraction of patients with metastatic melanoma does not experience long-term disease control, even with immunotherapy and targeted therapy, new biomarkers for patient stratification and treatment tailoring are needed in this setting. We investigated the association of a novel immune-inflammatory blood-based biomarker, the Pan-Immune-Inflammation Value (PIV), with clinical outcomes of patients with metastatic melanoma receiving first-line therapy. We retrospectively included patients treated at the Fondazione IRCCS Istituto Nazionale dei Tumori of Milan and having an available baseline complete blood cell count (CBC). PIV was calculated as: [neutrophil count (103/mm3) × platelet count (103/mm3) × monocyte count (103/mm3)]/lymphocyte count (103/mm3). A total of 228 patients were included: 119 (52%) had been treated with immunotherapy and 109 (48%) with targeted therapy. PIV was significantly higher in patients with ECOG PS ≥ 1, high disease burden, synchronous metastases, and elevated baseline LDH level. High baseline PIV was independently associated with poor overall survival (adjusted hazard ratio [HR]: 2.06; 95% confidence interval [CI]: 1.30–3.29; adjusted P = 0.002) and progression-free survival (adjusted HR 1.56; 95% CI 1.01–2.41; adjusted P = 0.044). High PIV was also associated with primary resistance to both immunotherapy (odds ratio [OR]: 3.98; 95% CI 1.45–12.32; P = 0.005) and targeted therapy (OR: 8.42; 95% CI 2.50–34.5; P < 0.001). PIV showed a promising discrimination ability in terms of AIC and c-index when compared with other CBC-based biomarkers. PIV may guide the treatment decision process and the development of novel first-line treatment strategies in melanoma, but warrants further study and validation.
- Published
- 2021
- Full Text
- View/download PDF
20. The Pan-Immune-Inflammation Value in microsatellite instability–high metastatic colorectal cancer patients treated with immune checkpoint inhibitors
- Author
-
Giovanni Fucà, Margherita Ambrosini, Rossana Intini, Maria Antista, Matteo Fassan, Giuseppe Curigliano, Alessandra Anna Prete, Marta Brambilla, Andrea Spallanzani, Federica Morano, Massimiliano Salati, Carmen Belli, Filippo Pietrantonio, Elisabetta Fenocchio, Beatrice Borelli, Sara Lonardi, Vittorina Zagonel, Francesca Corti, Filippo de Braud, and Virginia Quarà
- Subjects
Blood Platelets ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Time Factors ,Neutrophils ,Colorectal cancer ,Immune checkpoint inhibitors ,Lymphocyte ,Monocytes ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Antigen ,Internal medicine ,Tumor Microenvironment ,Humans ,Medicine ,Genetic Predisposition to Disease ,Neoplasm Metastasis ,Pan-Immune-Inflammation Value ,Immune Checkpoint Inhibitors ,Aged ,Retrospective Studies ,Inflammation ,business.industry ,Microsatellite instability ,medicine.disease ,Progression-Free Survival ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Absolute neutrophil count ,Biomarker (medicine) ,Biomarkers ,Deficient mismatch repair ,Female ,Microsatellite Instability ,Colorectal Neoplasms ,business ,Immune inflammation - Abstract
Immune checkpoint inhibitors (ICIs) yielded unprecedented efficacy in patients with microsatellite instability (MSI)-high metastatic colorectal cancer (mCRC). Since the Pan-Immune-Inflammation Value (PIV) is a blood-based biomarker with prognostic usefulness in mCRC, it might predict clinical outcomes and primary resistance to ICIs.We retrospectively analysed the association of PIV and its early modulation at 3/4 weeks after treatment initiation with the outcomes of MSI-high mCRC patients receiving anti-programmed death-(ligand)1 (PD-[L]1) +/- anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) agents. PIV was calculated as follows: (neutrophil count × platelet count × monocyte count)/lymphocyte count. PIV cut-offs were determined using the maximally selected rank statistics.A total of 163 patients were included. In the multivariable models for overall survival (OS) and progression-free survival (PFS), both high (492) baseline PIV (OS: adjusted [a] HR, 3.00; 95% CI, 1.49-6.04, p = 0.002; PFS: aHR, 1.91; 95% CI, 1.06-3.44, p = 0.031) and early PIV increase ≥+30% (OS: aHR, 3.21; 95% CI, 1.65-6.23, p 0.001; PFS: aHR, 2.25; 95% CI, 1.30-3.89, p = 0.003) confirmed an independent prognostic impact. After stratifying patients according to baseline PIV and ICI regimen, OS and PFS were significantly worse in subjects with high PIV receiving anti-PD-1/PD-L1 monotherapy. Early PIV increase was an independent predictor of clinical benefit (aOR, 0.23; 95% CI, 0.08-0.66; p = 0.007), whereas a trend was observed for baseline PIV (aOR, 0.33; 95% CI, 0.10-1.07; p = 0.065).PIV is a strong predictor of outcomes in MSI-high mCRC patients receiving ICIs. Prospective validation of these results is required to establish its role as a stratification factor for personalised combination strategies.
- Published
- 2021
- Full Text
- View/download PDF
21. A combination of extracellular matrix‐ and interferon‐associated signatures identifies high‐grade breast cancers with poor prognosis
- Author
-
Serenella M. Pupa, Mara Lecchi, Elda Tagliabue, Melissa Anna Teresa Monica, Filippo de Braud, Paolo Verderio, Giovanni Fucà, Marilena V. Iorio, Massimo Di Nicola, Francesca De Santis, Giulia Bianchi, Sabina Sangaletti, Vera Cappelletti, Massimiliano Gennaro, and Biagio Paolini
- Subjects
Adult ,0301 basic medicine ,Cancer Research ,Breast Neoplasms ,Disease ,gene signature ,Cohort Studies ,Diagnosis, Differential ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Breast cancer ,Interferon ,Gene expression ,Biomarkers, Tumor ,Genetics ,medicine ,Humans ,tumor microenvironment ,high‐grade breast cancer ,Research Articles ,RC254-282 ,Aged ,Tumor microenvironment ,business.industry ,Gene Expression Profiling ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,General Medicine ,Middle Aged ,Gene signature ,Prognosis ,medicine.disease ,Extracellular Matrix ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Biomarker (medicine) ,Female ,Interferons ,Neoplasm Grading ,Transcriptome ,business ,prognostic marker ,Research Article ,medicine.drug - Abstract
Breast cancer (BC) is a heterogeneous disease in which the tumor microenvironment (TME) seems to impact the clinical outcome. Here, we investigated whether a combination of gene expression signatures relating to both the structural and immune TME aspects can help predict prognosis in women with high‐grade BC (HGBC). Thus, we focused on a combined molecular biomarker variable that involved extracellular matrix (ECM)‐associated gene expression (ECM3 signature) and interferon (IFN)‐associated metagene (IFN metagene) expression. In 97 chemo‐naive HGBCs from the METABRIC dataset, the dichotomous ECM3/IFN (dECIF) variable identified a group of high‐risk patients (ECM3+/IFN− vs other; hazard ratio = 3.2, 95% confidence interval: 1.5–6.7). Notably, ECM3+/IFN− tumors showed low tumor‐infiltrating lymphocytes, high levels of CD33‐positive cells, absence of PD‐1 expression, or low expression of PD‐L1, as suggested by immune profiles and immune‐histochemical analysis on an independent cohort of 131 HGBCs. To make our results transferable to clinical use, we refined the dECIF biomarker using reduced ECM3 and IFN signatures; notably, the prognostic value of this reduced dECIF was comparable to that of the original dECIF. After validation in a new BC cohort, reduced dECIF was translated into a robust qPCR classifier for real‐world clinical use., This study investigates the combination of two relevant molecular signatures of the tumor microenvironment (TME) in identifying aggressive high‐grade breast cancers (HGBCs). The novel molecular biomarker described here highlighted patients with worst HGBC prognosis and a peculiar TME. Finally, to improve the potential of the identified biomarker in the clinic, we reduced the two original signatures and technically validated the biomarker's expression through a qPCR‐based assay.
- Published
- 2021
22. Recent Advances in the Management of Typical and Atypical Lung Carcinoids
- Author
-
Michele Prisciandaro, Carlo Spreafico, Massimo Milione, Claudia Proto, Francesca Corti, Alessandra Raimondi, Jorgelina Coppa, Giovanni Leuzzi, Maria Di Bartolomeo, Sara Pusceddu, Martina Torchio, Tommaso Cascella, Natalie Prinzi, Filippo de Braud, Roberta Elisa Rossi, Giuseppe Lo Russo, Teresa Beninato, and Elena Colombo
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Poor prognosis ,Lung Neoplasms ,Carcinoid Tumor ,Neuroendocrine tumors ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Epidemiology ,Humans ,Medicine ,Lung cancer ,Patient Care Team ,Lung ,business.industry ,Incidence ,Poorly differentiated ,medicine.disease ,Neuroendocrine Tumors ,Locoregional disease ,030104 developmental biology ,medicine.anatomical_structure ,Current management ,030220 oncology & carcinogenesis ,business - Abstract
Neuroendocrine neoplasms of the lung represent about 20% to 30% of all neuroendocrine tumors. On the basis of clinical and pathologic characteristics, 2 different categories of tumors may be defined: poorly differentiated neuroendocrine neoplasms, characterized by a high rate of recurrences and poor prognosis, and well-differentiated neuroendocrine neoplasms (typical carcinoids and atypical carcinoids), which generally display an indolent course. Lung carcinoids represent only 1% to 5% of all lung malignancies, but their incidence has significantly increased over the past 30 years. Surgery is the reference standard of treatment for lung carcinoids with locoregional disease. For advanced or unresectable lung carcinoids, several therapeutic options are available, but the choice should be shared within a multidisciplinary team to ensure optimal therapeutic outcomes. We describe the current management of these rare neoplasms.
- Published
- 2021
- Full Text
- View/download PDF
23. Corrigendum to ‘Gene mutational profile of BRCAness and clinical implication in predicting response to platinum-based chemotherapy in patients with intrahepatic cholangiocarcinoma’ [Euro J Cancer 171 (2022) 232–241]
- Author
-
Margherita Rimini, Teresa Macarulla, Valentina Burgio, Sara Lonardi, Monica Niger, Mario Scartozzi, Ilario G. Rapposelli, Giuseppe Aprile, Francesca Ratti, Federica Pedica, Helena Verdaguer, Floriana Nappo, Federico Nichetti, Eleonora Lai, Martina Valgiusti, Alessandro Cappetta, Carles Fabregat-Franco, Matteo Fassan, Filippo De Braud, Marco Puzzoni, Giovanni L. Frassineti, Francesca Simionato, Francesco De Cobelli, Luca Aldrighetti, Lorenzo Fornaro, Stefano Cascinu, and Andrea Casadei-Gardini
- Subjects
Cancer Research ,Oncology - Published
- 2022
- Full Text
- View/download PDF
24. Efficacy and Safety of First-line Carboplatin-paclitaxel and Carboplatin-gemcitabine in Patients With Advanced Triple-negative Breast Cancer: A Monocentric, Retrospective Comparison
- Author
-
Riccardo Lobefaro, Luigi Mariani, Giorgia Peverelli, Francesca Ligorio, Giovanni Fucà, Alessandro Rametta, Emma Zattarin, Rita Leporati, Daniele Presti, Beatrice Cantarelli, Catherine Depretto, Andrea Vingiani, Siranoush Manoukian, Gianfranco Scaperrotta, Giulia V. Bianchi, Giuseppe Capri, Giancarlo Pruneri, Filippo de Braud, and Claudio Vernieri
- Subjects
Cancer Research ,Oncology - Abstract
Platinum-based chemotherapy is widely used in patients with advanced triple-negative breast cancer (TNBC). However, the most effective platinum-based combination in the first-line treatment setting remains unclear.We evaluated the efficacy of first-line carboplatin-paclitaxel (CP) or carboplatin-gemcitabine (CG) combinations in advanced TNBC patients treated between April 2007 and April 2021. CP and CG were compared in terms of progression-free survival (PFS), overall survival (OS), and incidence of adverse events (AEs). Multivariable Cox Models were used to adjust the efficacy of CP versus CG for clinically relevant covariates.Of 88 consecutive advanced TNBC patients receiving first-line carboplatin-based doublets, 56 (63.6%) received CP and 32 (36.4%) CG. After adjusting for clinically relevant variables, patients receiving CG had significantly better PFS when compared to CP-treated patients (HR: 0.49 (95% CI, 0.27-0.87), P value 0.014). Of note, CG was associated with better PFS only among patients previously treated with taxanes in the (neo)adjuvant setting (HR: 0.39; 95% CI, 0.21-0.75), but not in patients not exposed to taxanes (HR: 1.20; 95% CI, 0.37-3.88). CG was also independently associated with better OS when compared to CP (HR: 0.31 (95% CI: 0.15-0.64), P value 0.002). Overall, grade 3-4 AEs were more common in patients treated with CG than in patients treated with CP (68.8% vs. 21.4%, P value .009).CG and CP are effective and well tolerated first-line platinum doublets in advanced TNBC patients. CG could be more effective than CP in patients previous exposed to taxanes despite worse toxicity profile.
- Published
- 2022
25. Thrombin Generation and D-Dimer for Prediction of Disease Progression and Mortality in Patients with Metastatic Gastrointestinal Cancer
- Author
-
Cinzia Giaccherini, Cristina Verzeroli, Laura Russo, Sara Gamba, Carmen Julia Tartari, Silvia Bolognini, Francesca Schieppati, Chiara Ticozzi, Roberta Sarmiento, Luigi Celio, Giovanna Masci, Carlo Tondini, Fausto Petrelli, Francesco Giuliani, Andrea D’Alessio, Filippo De Braud, Armando Santoro, Roberto Labianca, Giampietro Gasparini, Marina Marchetti, and Anna Falanga
- Subjects
Cancer Research ,Oncology ,gastrointestinal cancer ,hypercoagulability ,prognosis ,survival ,D-dimer ,thrombin generation - Abstract
Background: the tight and reciprocal interaction between cancer and hemostasis has stimulated investigations on the possible role of hemostatic biomarkers in predicting specific cancer outcomes, such as disease progression (DP) and overall survival (OS). In a prospective cohort of newly diagnosed metastatic gastrointestinal (GI) cancer patients from the HYPERCAN study, we aimed to assess whether the hemostatic biomarker levels measured before starting any anticancer therapy may specifically predict for 6-months DP (6m-DP) and for 1-year OS (1y OS). Methods: plasma samples were collected and tested for thrombin generation (TG) as global hemostatic assay, and for D-dimer, fibrinogen, and prothrombin fragment 1 + 2 as hypercoagulation biomarkers. DP and mortality were monitored during follow-up. Results: A prospective cohort of 462 colorectal and 164 gastric cancer patients was available for analysis. After 6 months, DP occurred in 148 patients, providing a cumulative incidence of 24.8% (21.4–28.4). D-dimer and TG endogenous thrombin potential (ETP) were identified as independent risk factors for 6m-DP by multivariate Fine–Gray proportional hazard regression model corrected for age, cancer site, and >1 metastatic site. After 1 year, we observed an OS of 75.7% (71.9–79.0). Multivariate Cox regression analysis corrected for age, site of cancer, and performance status identified D-dimer and ETP as independent risk factors for 1y OS. Patients with one or both hemostatic parameters above the dichotomizing threshold were at higher risk for both 6m-DP and 1-year mortality. Conclusion.: in newly diagnosed metastatic GI cancer patients, pretreatment ETP and D-dimer appear promising candidate biomarkers for predicting 6m-DP and 1y OS. In this setting, for the first time, the role of TG as a prognostic biomarker emerges in a large prospective cohort.
- Published
- 2022
26. Abstract PS11-28: Efficacy and safety of entrectinib in NTRK fusion-positive (NTRK-fp) breast cancer
- Author
-
Janice Lu, Todd M. Bauer, Minal A. Barve, S. Osborne, Filippo de Braud, Tira Tan, Christophe Le Tourneau, B. Simmons, Alexander I. Spira, Samuel McCallum, Collin M. Blakely, Saiama N. Waqar, and David S. Hong
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Nausea ,Population ,Cancer ,Entrectinib ,medicine.disease ,Dysgeusia ,Clinical trial ,Breast cancer ,Internal medicine ,medicine ,medicine.symptom ,education ,business ,Adverse effect - Abstract
Neurotrophic tyrosine receptor kinase genes (NTRK1/2/3) act as oncogenic drivers across a range of tumors. NTRK fusions occur at low frequency (90% of secretory breast carcinomas. Furthermore, up to 30% of patients (pts) with breast cancer will develop central nervous system (CNS) metastases (mets). Entrectinib is a potent, oral TRKA/B/C, ROS1 and ALK inhibitor, specifically selected for its CNS penetration properties. Entrectinib was evaluated in 3 global phase 1/2 clinical trials (ALKA-372-001 [EudraCT 2012-000148-88], STARTRK-1 [NCT02097810], and STARTRK-2 [NCT02568267]), where it demonstrated strong and durable systemic and intracranial efficacy in pts with NTRK-fp solid tumors, including those with breast cancer. We present updated data from this integrated analysis (data cut-off: 31 October 2018) focusing on pts with breast cancer. The entrectinib trials were conducted at >150 sites in 15 countries, and enrolled pts with locally advanced/metastatic NTRK-fp tumors (with or without baseline CNS mets) confirmed by nucleic acid-based methods. Tumor assessments were performed at the end of cycle 1 (4 weeks) and every 8 weeks thereafter, and evaluated by blinded independent central review (BICR) using RECIST v1.1. Primary endpoints were objective response rate (ORR) and duration of response (DoR) by BICR. Secondary endpoints included progression-free survival (PFS), overall survival (OS), and safety. At clinical cut-off, the overall efficacy-evaluable population included 74 adults from the 3 trials, with 12 different NTRK-fp tumor types and >25 histopathologies. In these pts, ORR by BICR was 63.5% (95% CI 51.5-74.4), including five complete responses (CR) and 42 partial responses (PR). Median (95% CI) DoR, PFS and OS were 12.9 (9.3-not estimable [NE]), 11.2 (8.0-15.7) and 23.9 (16.0-NE) months, respectively. The efficacy-evaluable NTRK-fp breast cancer cohort included 6 pts with a median age of 63 (range 36-67) years; most had an Eastern Cooperative Oncology Group performance status of 0 (3/6; 50%) or 1 (1/6; 17%). Breast cancer tumors were classified as secretory (4/6; 67%; all NTRK3-fp) or non-secretory (2/6; 33%; all NTRK1-fp). Pts had received 0 (3/6; 50%), 1 (1/6; 17%) or ≥4 (2/6; 33%) lines of prior therapy for metastatic disease. At data cut off, the median survival follow-up was 17.4 (range 1.7-23.9) months. ORR was 100% (2 CR, 2 PR; 95% CI 39.8-100.0) in pts with secretory and 50% (1 PR, 1 missing/unevaluable; 95% CI 1.3-98.7) in pts with non-secretory histology. Median (95% CI) DoR, PFS, and OS were 12.9 (4.2-NE), 10.1 (5.1-NE), and 23.9 (5.1-23.9) months, respectively. At baseline, 2 pts had CNS mets per investigator assessment; 1 of these pts had missing response data. CNS mets were confirmed by BICR in the other pt (non-secretory); this pt had received whole brain radiotherapy 2-6 months before starting entrectinib treatment, and had systemic PR and intracranial non-CR/non-progressive disease (non-measurable CNS lesion). The overall integrated safety-evaluable population comprised 504 pts with any gene fusion who received ≥1 dose of entrectinib. Most treatment-related adverse events (TRAEs) were Grade ≤3 (96.1%); the most frequently reported TRAEs were dysgeusia (39.7%) and fatigue (31.5%). Seven breast cancer pts were evaluated for safety, of whom six (85.7%) reported TRAEs; all were Grade ≤3. The most frequently reported TRAEs (each occurring in 3/7 pts; 42.9%) were nausea, anemia, and increased alanine or aspartate aminotransferase. Dose reductions and interruptions due to TRAEs were each reported in 3/7 pts (42.9%); no discontinuations or deaths due to TRAEs were recorded. In this updated integrated analysis, entrectinib induced objective responses in all pts with NTRK-fp breast cancer who had data available, and was generally well tolerated with no discontinuations due to TRAEs. Citation Format: Janice Lu, Collin M. Blakely, Christophe Le Tourneau, Saiama N. Waqar, Todd M. Bauer, Filippo de Braud, David S. Hong, Alexander Spira, Tira Tan, Samuel McCallum, Stuart Osborne, Brian Simmons, Minal Barve. Efficacy and safety of entrectinib in NTRK fusion-positive (NTRK-fp) breast cancer [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS11-28.
- Published
- 2021
- Full Text
- View/download PDF
27. Impact of early tumor shrinkage and depth of response on the outcomes of panitumumab-based maintenance in patients with RAS wild-type metastatic colorectal cancer
- Author
-
Lorenza Rimassa, Sara Lonardi, Filippo Pagani, Federica Morano, Filippo de Braud, Giovanni Randon, Gianluca Tomasello, Giuseppina Calareso, Francesca Bergamo, Alberto Zaniboni, Michele Prisciandaro, Marta Vaiani, Salvatore Corallo, Maria Di Bartolomeo, Paolo Manca, Carlotta Antoniotti, Valeria Smiroldo, Maria Antista, Chiara Cremolini, Marco Tampellini, Matteo Clavarezza, Francesca Greco, Filippo Pietrantonio, Federica Palermo, Roberto Murialdo, Alessandra Raimondi, and Patrizia Racca
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Treatment duration ,Maintenance Chemotherapy ,03 medical and health sciences ,0302 clinical medicine ,FOLFOX ,Maintenance therapy ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Molecular hyper-selection ,medicine ,Humans ,Panitumumab ,In patient ,Neoplasm Metastasis ,RAS wild-type ,Aged ,Metastatic colorectal cancer ,business.industry ,Tumor shrinkage ,Wild type ,Prognosis ,medicine.disease ,Bevacizumab ,Oxaliplatin ,Survival Rate ,Depth of response ,Early tumor shrinkage ,030104 developmental biology ,030220 oncology & carcinogenesis ,ras Proteins ,Female ,Fluorouracil ,Colorectal Neoplasms ,business ,Follow-Up Studies ,medicine.drug - Abstract
In patients with metastatic colorectal cancer (mCRC) receiving highly active first-line combination treatments, early tumor shrinkage (ETS) and depth of response (DoR) are associated with survival, but their influence on outcomes during maintenance therapy is unknown. The Valentino study showed inferior PFS in 229 RAS wild-type mCRC patients randomized to panitumumab plus FOLFOX followed by maintenance with panitumumab vs. panitumumab + 5-FU/LV.After blinded independent central review of ETS (≥20% reduction of the sum of target lesions) and DoR in patients enrolled in Valentino, the prognostic and predictive role of such parameters was investigated, along with their combination with PRESSING panel (uncommon genomic alterations associated with anti-EGFRs resistance beyond RAS and BRAF).One hundred and ninety-six patients were included (ETS in 132 [67.3%], median DoR: 44.1%). Both ETS and DoR ≥34% were associated with longer mPFS (p = 0.010 and p 0.001) and mOS (p = 0.006 and p 0.001). The PFS benefit of 5-FU/LV added to panitumumab maintenance, reported in the study, was independent from ETS and DoR status (interaction tests NS for both PFS and OS). However, outcomes were extremely poor in patients who received single-agent panitumumab and had no-ETS (mPFS and mOS: 7.7 and 18.7 months) or DoR 34% (mPFS and mOS: 6.5 and 18 months). Combining PRESSING panel ('molecular hyperselection') and response dynamics allowed to stratify both PFS (p 0.001 and p 0.001 for ETS and DoR, respectively) and OS (p 0.001 and p = 0.017 for ETS and DoR, respectively).ETS and DoR allow on-treatment anticipation of outcomes following an anti-EGFR-based strategy planning de-escalation, and poor radiological response may guide enrolment in crossover strategy trials. As in vivo markers of drug sensitivity, ETS and DoR may be integrated with several patient- and tumor-related factors to wisely drive decision-making on upfront treatment duration and intensity.
- Published
- 2021
- Full Text
- View/download PDF
28. Prolonged benefit from palbociclib plus letrozole in heavily pretreated advanced male breast cancer: case report
- Author
-
Giulia Bianchi, Emma Zattarin, Francesca Ligorio, Federico Nichetti, Filippo de Braud, and Giuseppe Capri
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pyridines ,Receptor, ErbB-2 ,Palbociclib ,Piperazines ,Breast Neoplasms, Male ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,030212 general & internal medicine ,Salvage Therapy ,business.industry ,Letrozole ,General Medicine ,Prognosis ,medicine.disease ,Clinical trial ,Receptors, Estrogen ,030220 oncology & carcinogenesis ,Male breast cancer ,Receptors, Progesterone ,business ,medicine.drug - Abstract
Introduction: Breast cancer in men is less common than in women and treatment recommendations are often derived from clinical trials exclusively involving women. Data on efficacy of CDK 4/6 inhibitors, which are the mainstay of treatment for hormone receptor–positive/HER2-negative advanced breast cancer, are lacking in male patients. Case report: We present a clinical case of prolonged benefit from palbociclib in combination with letrozole and LHRH analogue in a man who had previously been treated with six lines of endocrine therapies and chemotherapy regimens but was still in excellent clinical condition. Conclusions: This clinical case demonstrates that male breast cancer stands out as an endocrine-sensitive disease, which could potentially benefit from CDK 4/6 inhibitors in combination with endocrine agents even in very heavily pretreated settings of disease, underscoring both the importance of an accurate selection of patients for later treatment lines, taking into account disease history and previous treatment responses, and the peculiarity of breast cancer in men, which deserves dedicated clinical trials to tailor future recommendations.
- Published
- 2020
- Full Text
- View/download PDF
29. Phase I Study of Rucaparib in Combination with Bevacizumab in Ovarian Cancer Patients: Maximum Tolerated Dose and Pharmacokinetic Profile
- Author
-
Francesco Raspagliesi, Maurizio D'Incalci, Cristina Matteo, Sara Cresta, Tommaso Ceruti, Giuseppa Maltese, Ilaria Sabatucci, Serena Giolitto, Massimo Zucchetti, Domenica Lorusso, Cristina Sonetto, Dominique Ronzulli, Filippo de Braud, and Valentina Sinno
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Indoles ,Bevacizumab ,Maximum Tolerated Dose ,medicine.medical_treatment ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Pharmacology (medical) ,Original Research Article ,Stage (cooking) ,Adverse effect ,Rucaparib ,Aged ,Ovarian Neoplasms ,Chemotherapy ,business.industry ,Middle Aged ,medicine.disease ,Clinical trial ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Female ,Ovarian cancer ,business ,medicine.drug - Abstract
Background Targeted agents, such as antiangiogenic drugs (e.g., bevacizumab) and poly(ADP-ribose) polymerase inhibitors (e.g., rucaparib), have been shown to improve outcomes in patients with newly diagnosed or recurrent ovarian cancer. Evidence suggests that combinations of these two classes of targeted agents may result in synergistic antitumor activity. Objective The phase I portion of MITO 25 was designed to determine the maximum tolerated dose, pharmacokinetics, and the safety profile of rucaparib when administered in combination with bevacizumab as maintenance treatment for patients with high-grade epithelial ovarian, fallopian tube, or primary peritoneal cancer. Methods This was a single-arm, phase I dose-escalation study. Cohorts of three patients were recruited to receive increasing rucaparib doses of 400 mg, 500 mg, or 600 mg twice daily for 28 days. Bevacizumab 15 mg/kg was administered at day 1 every 21 days. Results We enrolled nine patients. Two patients in the rucaparib 600-mg group had four grade 3 treatment-emergent adverse events: increased in alanine aminotransferase and aspartate aminotransferase levels, depression, and hallucinations. These were deemed to be dose-limiting toxicities related to rucaparib. Because these dose-limiting toxicities occurred in the 600-mg group and affected more than one in three patients, the maximum tolerated dose for rucaparib was considered 500 mg twice daily when combined with bevacizumab 15 mg/kg at day 1 every 21 days. There were no new safety concerns from using the combination. No substantial difference in pharmacokinetic parameters was found between the cohorts or in the pharmacokinetic profiles of rucaparib administered alone or with bevacizumab with respect to historical controls. Conclusions The maximum tolerated dose of rucaparib is 500 mg twice daily when co-administered with bevacizumab. The plasma concentration–time profiles of rucaparib in combination with bevacizumab suggest no pharmacokinetic interactions between the drugs. The randomized phase II portion of MITO 25 will further investigate rucaparib maintenance treatment with or without bevacizumab in patients with newly diagnosed stage III–IV ovarian cancer who responded to carboplatin-paclitaxel chemotherapy with or without bevacizumab. Trial Registration ClinicalTrials.gov identifier NCT03462212; registered March 2018.
- Published
- 2020
30. Validation of the Colon Life nomogram in patients with refractory metastatic colorectal cancer enrolled in the RECOURSE trial
- Author
-
Francesca Corti, Giovanni Fucà, Filippo Pagani, Alessandra Raimondi, Rosalba Miceli, Filippo de Braud, Giovanni Randon, Paolo Manca, Chiara Cremolini, Michele Prisciandaro, and Filippo Pietrantonio
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colon ,Colorectal cancer ,Trifluridine ,Colon Life nomogram ,metastatic colorectal cancer ,prognosis ,randomized clinical trial ,Trifluridine/tipiracil ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,Refractory ,law ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Overall survival ,Humans ,Multicenter Studies as Topic ,In patient ,030212 general & internal medicine ,Peritoneal Neoplasms ,Aged ,Randomized Controlled Trials as Topic ,Retrospective Studies ,business.industry ,General Medicine ,Nomogram ,Prognosis ,medicine.disease ,Survival Rate ,Nomograms ,Clinical Trials, Phase III as Topic ,030220 oncology & carcinogenesis ,Female ,Colorectal Neoplasms ,business ,Follow-Up Studies ,medicine.drug - Abstract
Background: The RECOURSE trial (Study of TAS-102 in Patients With Metastatic Colorectal Cancer Refractory to Standard Chemotherapies) demonstrated an overall survival (OS) benefit of trifluridine/tipiracil (FTD/TPI) vs placebo in refractory metastatic colorectal cancer (mCRC). Given the limited benefit of later line treatments, we developed the Colon Life nomogram to assess the 12-week death probability in the refractory setting. Methods: This post hoc analysis of RECOURSE included patients with available data to calculate the nomogram score: Eastern Cooperative Oncology Group Performance Status, primary tumor resection, lactate dehydrogenase, and peritoneal metastases. The nomogram calibration was assessed by calibration plots and C-index. The nomogram prognostic and predictive ability was assessed by Cox model analyses and the nomogram score predictive value was explored according to the cutoff identified at maximum value of the Youden index in time-dependent receiver operating characteristic curve analysis. Results: Overall, 251 trial patients were evaluable: 90 in the placebo arm and 161 in the FTD/TPI arm. The calibration was optimal in the placebo arm (C-index 0.807) and suboptimal in the FTD/TPI arm (0.657). The cutoff of the nomogram score of 23 showed the best discriminative ability for 12-week OS (hazard ratio 3.46, 95% confidence interval 2.17–5.51 for scores 40 vs 15) and had maximum value of the Youden index (0.381). Median OS and 3-month PFS were 9.0 vs 7.5 months and 39.3% vs 5.2%, respectively, for FTD/TPI vs placebo in the low-risk group (score Conclusion: The Colon Life nomogram is an accurate tool for estimating life expectancy in refractory mCRC. The benefit of FTD/TPI was independent of the predicted risk of early death.
- Published
- 2020
- Full Text
- View/download PDF
31. SMO mutations confer poor prognosis in malignant pleural mesothelioma
- Author
-
Laura Botta, Giulia Galli, Annalisa Trama, Marcello Tiseo, Claudia Proto, Ugo Pastorino, Giulia Pasello, Roberto Ferrara, Adele Busico, Martina Imbimbo, Arsela Prelaj, Diego Signorelli, Gemma Gatta, Giuseppe Lo Russo, Monica Ganzinelli, Elena Tamborini, Alessandro De Toma, Alessandra Fabbri, Marina Chiara Garassino, and Filippo de Braud
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Gene mutation ,medicine.disease_cause ,Hedgehog pathway ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Malignant pleural mesothelioma (MPM) ,Lung cancer ,Hedgehog ,BAP1 ,Mutation ,business.industry ,SMO ,Immunotherapy ,Prognosis ,medicine.disease ,030104 developmental biology ,Gene mutations ,PTCH1 ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Original Article ,business - Abstract
BACKGROUND: Malignant pleural mesothelioma (MPM) is an aggressive tumor but approximately 12% of patients survive more than 3 years. The biological differences underlying better outcomes are not known. Several targeted agents and immunotherapy have been ineffective. Hedgehog (Hh) is one emerging pathway. We compared the biological profiles of patients with different survival, investigating the most frequently altered genes, including the Hh pathway. METHODS: We analyzed 56 MPM. A 36-month overall survival (OS) cut-off divided patients into 32 normo (NS) and 24 long (LS) survivors. We used next generation sequencing to test 21 genes, immunohistochemistry to evaluate SMO expression. Mutation differences between NS and LS and their associations with clinical features were analysed by Fisher’s test, OS with the Kaplan-Meier method and its association with mutations by univariate and multivariate Cox proportional hazard models. RESULTS: Clinical features were similar in both groups. Eighteen out of 56 patients (32%) were wild-type for the genes analysed. At least five had mutations in BAP1, NF2, TP53, SMO and PTCH1 with no significant differences between the groups except for SMO. SMO, a member of the Hh pathway, was mutated only in NS (15.6%) and only SMO mutations were significantly associated with poor prognosis at univariate (HR =4.36, 95% CI: 2.32–8.18, P
- Published
- 2020
- Full Text
- View/download PDF
32. Dabrafenib plus trametinib in patients with BRAFV600E-mutated biliary tract cancer (ROAR): a phase 2, open-label, single-arm, multicentre basket trial
- Author
-
Angelica Fasolo, Antoine Italiano, Kert Viele, Gerald W. Prager, Richard Greil, Vivek Subbiah, Milind Javle, Patrick Y. Wen, Eduard Gasal, Giuseppe Curigliano, Jan H.M. Schellens, Aislyn Boran, Palanichamy Ilankumaran, Alexander Stein, Ulrik Lassen, Paul Burgess, Elena Elez, Filippo de Braud, and Zev A. Wainberg
- Subjects
0301 basic medicine ,Trametinib ,education.field_of_study ,medicine.medical_specialty ,Biliary tract neoplasm ,business.industry ,Population ,Dabrafenib ,Interim analysis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Clinical endpoint ,Progression-free survival ,education ,business ,medicine.drug - Abstract
Summary Background Effective treatments for patients with cholangiocarcinoma after progression on gemcitabine-based chemotherapy are urgently needed. Mutations in the BRAF gene have been found in 5% of biliary tract tumours. The combination of dabrafenib and trametinib has shown activity in several BRAFV600E-mutated cancers. We aimed to assess the activity and safety of dabrafenib and trametinib combination therapy in patients with BRAFV600E-mutated biliary tract cancer. Methods This study is part of an ongoing, phase 2, open-label, single-arm, multicentre, Rare Oncology Agnostic Research (ROAR) basket trial in patients with BRAFV600E-mutated rare cancers. Patients were eligible for the biliary tract cancer cohort if they were aged 18 years or older, had BRAFV600E-mutated, unresectable, metastatic, locally advanced, or recurrent biliary tract cancer, an Eastern Cooperative Oncology Group performance status of 0–2, and had received previous systemic treatment. All patients were treated with oral dabrafenib 150 mg twice daily and oral trametinib 2 mg once daily until disease progression or intolerance of treatment. The primary endpoint was the overall response rate, which was determined by Response Evaluation Criteria in Solid Tumors version 1.1 in the intention-to-treat evaluable population, which comprised all enrolled patients regardless of receiving treatment who were evaluable (ie, had progression, began a new anticancer treatment, withdrew consent, died, had stable disease for 6 weeks or longer, or had two or more post-baseline assessments). The ROAR trial is registered with ClinicalTrials.gov , NCT02034110 . These results are based on an interim analysis; the study is active but not recruiting. Findings Between March 12, 2014, and July 18, 2018, 43 patients with BRAFV600E-mutated biliary tract cancer were enrolled to the study and were evaluable. Median follow-up was 10 months (IQR 6–15). An investigator-assessed overall response was achieved by 22 (51%, 95% CI 36–67) of 43 patients. An independent reviewer-assessed overall response was achieved by 20 (47%, 95% CI 31–62) of 43 patients. The most common grade 3 or worse adverse event was increased γ-glutamyltransferase in five (12%) patients. 17 (40%) patients had serious adverse events and nine (21%) had treatment-related serious adverse events, the most frequent of which was pyrexia (eight [19%]). No treatment-related deaths were reported. Interpretation Dabrafenib plus trametinib combination treatment showed promising activity in patients with BRAFV600E-mutated biliary tract cancer, with a manageable safety profile. Routine testing for BRAFV600E mutations should be considered in patients with biliary tract cancer. Funding GlaxoSmithKline and Novartis.
- Published
- 2020
- Full Text
- View/download PDF
33. Ramucirumab and durvalumab for previously treated, advanced non–small-cell lung cancer, gastric/gastro-oesophageal junction adenocarcinoma, or hepatocellular carcinoma: An open-label, phase Ia/b study (JVDJ)
- Author
-
Laetitia Dahan, Victor Moreno, Yung-Jue Bang, Laura W. Goff, Zev A. Wainberg, Filippo de Braud, Naomi Laing, Gu Mi, Talia Golan, Chia-Chi Lin, Joana M Oliveira, Heather Wasserstrom, Ravit Geva, Siqing Fu, Keunchil Park, and Martin Reck
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Durvalumab ,Esophageal Neoplasms ,Antineoplastic Agents ,Adenocarcinoma ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,Ramucirumab ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Lung cancer ,Adverse effect ,Aged ,Aged, 80 and over ,Hepatitis ,business.industry ,Liver Neoplasms ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,digestive system diseases ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Cohort ,Female ,business - Abstract
Background Emerging evidence supports combining immune checkpoint inhibitors (ICIs) with conventional or targeted therapies to enhance ICI antitumour activity and broaden the spectrum of patients who respond to ICIs. Here, we present the safety and preliminary efficacy of ramucirumab, an anti-VEGFR2 IgG1, plus durvalumab, an anti–PD-L1 IgG1, in previously treated patients with advanced non–small-cell lung cancer (NSCLC), gastric/gastro-oesophageal junction adenocarcinoma (gastric/GEJ), or hepatocellular carcinoma (HCC). Patients and methods A 25-centre, phase Ia/b single-arm, non-randomised, multi-cohort study was undertaken in patients with advanced/metastatic disease, Eastern Cooperative Oncology Group performance status, 0–1, progression on prior therapy, no prior ramucirumab or immunotherapy and any PD-L1 status. Patients received ramucirumab (10 mg/kg) plus durvalumab (1125 mg) intravenously Q3W (NSCLC), or ramucirumab (8 mg/kg) plus durvalumab (750 mg) Q2W (gastric/GEJ, HCC). Results Phase Ia treatment was found safe for phase Ib expansion; final enrolment was NSCLC (n = 28), gastric/GEJ (n = 29), HCC (n = 28). Grade ≥3 treatment–related adverse events occurred in 32.1%, 37.9% and 42.9% of patients, respectively. The most common were fatigue (35.7%), hypertension (34.5%) and diarrhoea (28.6%), respectively. Two patients died owing to an adverse event; one was treatment-related (hepatitis acute, HCC cohort). Objective response rate was 11% for NSCLC and HCC and 21% for gastric/GEJ. Median progression-free survival and overall survival were, respectively, 2.7 and 11 months in NSCLC; 2.6 and 12.4 months in gastric/GEJ; 4.4 and 10.7 months in HCC, with more prolonged survival in patients with high PD-L1 expression. Conclusion Ramucirumab/durvalumab exhibited manageable safety. The combination showed antitumour activity in all cohorts, particularly in patients with high PD-L1 expression.
- Published
- 2020
- Full Text
- View/download PDF
34. The emerging role of PARP inhibitors in prostate cancer
- Author
-
Alessia Mennitto, Giuseppe Procopio, Pierangela Sepe, Giuseppe Tonini, Melanie Claps, Marco Stellato, Elena Verzoni, Emma Zattarin, Riccardo Valdagni, Daniele Santini, Filippo de Braud, and Valentina Guadalupi
- Subjects
Male ,0301 basic medicine ,DNA Repair ,Veliparib ,Poly ADP ribose polymerase ,Antineoplastic Agents ,Poly(ADP-ribose) Polymerase Inhibitors ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Drug Development ,Animals ,Humans ,Medicine ,Talazoparib ,Pharmacology (medical) ,Rucaparib ,Gene ,business.industry ,medicine.disease ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,business ,Homologous recombination ,human activities - Abstract
In prostate cancer , there has recently been an emerging interest in mutations in genes belonging to the homologous recombination repair (HRR) pathway and in the inhibition of poly (ADP-ribose) polymerase (PARP) proteins.Mutations in the HRR genes, including BRCA1, BRCA2, and Ataxia-Telangiesctasia mutated (ATM), have been reported in prostate cancer, with different incidence in the localized and advanced settings. The PARP enzyme complex is involved in repair of DNA damage and its inhibition causes the accumulation of DNA mutations in HRR deficient cells. Several PARP inhibitors (PARPi) are under development, such as olaparib, talazoparib, niraparib, rucaparib, and veliparib. In metastatic castration resistant prostate cancer (mCRPC), olaparib has been the most studied and its clinical efficacy has been validated in a phase III clinical trial. Rucaparib and niraparib have also shown promising results in the preliminary analyzes of two phase II trials, while talazoparib is currently under development.PARPi have become part of the treatment of mCRPC. Early results of combination therapy with PARPi and new hormonal therapy are promising and are supported by a strong biological rationale. Current results need to be validated in randomized phase III-controlled trials in order to translate the use of PARPi into real world practice.
- Published
- 2020
- Full Text
- View/download PDF
35. Prognostic impact of immune-microenvironment in colorectal liver metastases resected after triplets plus a biologic agent: A pooled analysis of five prospective trials
- Author
-
Marco Maria Germani, Alessandra Boccaccino, Giuseppe Aprile, Chiara Cremolini, Salvatore Corallo, Filippo Pietrantonio, Daniele Rossini, Umberto Cillo, Giovanni Centonze, Massimo Milione, Alfredo Falcone, Roberto Moretto, Sara Lonardi, Antonino Belfiore, Michele Prisciandaro, Gabriella Fontanini, Filippo de Braud, Lucio Urbani, Fotios Loupakis, Laura Cattaneo, Federica Morano, Luca Morelli, Silvia Brich, Matteo Fassan, and Federica Marmorino
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Time Factors ,Organoplatinum Compounds ,Colorectal cancer ,Leucovorin ,Colorectal liver metastasis ,Immunological parameters ,Triplet plus targeted agent ,Tumour microenvironment ,Cetuximab ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Microenvironment ,Clinical Trials as Topic ,FOLFOXIRI ,Liver Neoplasms ,Middle Aged ,Neoadjuvant Therapy ,Bevacizumab ,Oxaliplatin ,Treatment Outcome ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Female ,Fluorouracil ,Colorectal Neoplasms ,medicine.drug ,medicine.medical_specialty ,Irinotecan ,Capecitabine ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Internal medicine ,medicine ,Hepatectomy ,Humans ,Aged ,Retrospective Studies ,business.industry ,medicine.disease ,Clinical trial ,030104 developmental biology ,Camptothecin ,business - Abstract
Immune-contexture of tumour microenvironment (TME) influences prognosis of colorectal cancer (CRC) patients and can be altered by cytotoxic and targeted agents. Limited data are available regarding the immune-TME of CRC after treatment.An extensive immunohistochemistry evaluation of immunological parameters on tumour cells and TME of colorectal liver metastases from 106 patients who underwent secondary resection, after receiving triplets FOLFOXIRI (5-fluorouracil, oxaliplatin and irinotecan) or COI (capecitabine, oxaliplatin and irinotecan) plus bevacizumab (N = 59) or cetuximab (N = 47) in five first-line no-profit clinical trials was performed.No substantial differences were reported in immunological parameters according to administered targeted agent, RAS/BRAF mutational status and histopathological or Response Evaluation Criteria in Solid Tumours response. Stromal expression of Cyclooxygenase-2 (COX-2) (p = 0.002), Human leukocyte antigen (HLA) (p = 0.003) and Programmed cell death protein 1 (PD1) (p = 0.002) were independent prognostic factors for longer relapse-free survival (RFS) at multivariate analysis with a positive trend for post-resection overall survival (OS). Patients whose metastases expressed stromal COX-2, HLA and PD1 (inflamed-score positive) reported longer RFS (25.5 versus 9.8 months; p 0.001) and post-resection OS (64.3 versus 37.7 months; p = 0.003) as compared with others. In addition, patients with higher expression of CD4 and CD8 T-cells in tumour core and invasive margin (CD4/CD8-score) showed a better post-resection OS (not-reached versus 41.6 months; p = 0.032). A combined score of inflamed-score and CD4/CD8-score (combo-score) showed a clear prognostic role.The present study emphasises the role of immune-TME as independent predictor of survival in patients resected after triplets plus biologic. Inflamed-, CD4/C8- and combo-scores should be confirmed as prognostic factors in further studies.
- Published
- 2020
- Full Text
- View/download PDF
36. One size does not fit all for pancreatic cancers: A review on rare histologies and therapeutic approaches
- Author
-
Melissa Anna Teresa Monica, Sara Pusceddu, Sara Manglaviti, Federico Nichetti, Filippo de Braud, Vincenzo Mazzaferro, Natalie Prinzi, Maria Di Bartolomeo, Marta Brambilla, Martina Torchio, Monica Niger, Jorgelina Coppa, Maria Antista, Laura Cattaneo, Francesca Corti, and Michele Prisciandaro
- Subjects
Oncology ,Pancreatoblastoma ,medicine.medical_specialty ,Pancreatic acinar cell cancer ,business.industry ,Gastroenterology ,Review ,medicine.disease ,Pancreatic adenosquamous cancer ,Pseudopapillary pancreatic cancer ,03 medical and health sciences ,0302 clinical medicine ,Rare pancreatic cancers ,Undifferentiated pancreatic cancer ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,030211 gastroenterology & hepatology ,business - Abstract
Exocrine pancreatic neoplasms represent up to 95% of pancreatic cancers (PCs) and are widely recognized among the most lethal solid cancers, with a very poor 5-year survival rate of 5%-10%. The remaining < 5% of PCs are neuroendocrine tumors that are usually characterized by a better prognosis, with a median overall survival of 3.6 years. The most common type of PC is pancreatic ductal adenocarcinoma (PDAC), which accounts for roughly 85% of all exocrine PCs. However up to 10% of exocrine PCs have rare histotypes, which are still poorly understood. These subtypes can be distinguished from PDAC in terms of pathology, imaging, clinical presentation and prognosis. Additionally, due to their rarity, any knowledge regarding these specific histotypes is mostly based on case reports and a small series of retrospective analyses. Therefore, treatment strategies are generally deduced from those used for PDAC, even if these patients are often excluded or not clearly represented in clinical trials for PDAC. For these reasons, it is essential to collect as much information as possible on the management of PC, as assimilating it with PDAC may lead to the potential mistreatment of these patients. Here, we report the most significant literature regarding the epidemiology, typical presentation, possible treatment strategies, and prognosis of the most relevant histotypes among rare PCs.
- Published
- 2020
37. Ceritinib plus Nivolumab in Patients with Advanced ALK-Rearranged Non–Small Cell Lung Cancer: Results of an Open-Label, Multicenter, Phase 1B Study
- Author
-
Luojun Victor Wang, Yvonne Y. Lau, Thomas John, Giovanni Selvaggi, Filippo de Braud, Jeffrey W. Scott, Enriqueta Felip, Vanessa Giannone, O. Alejandro Balbin, Michela Maur, Daniel Shao-Weng Tan, Pilar Cazorla, Martijn P. Lolkema, Herbert H. Loong, Johan Vansteenkiste, Alice T. Shaw, Jason Baum, Geoffrey Liu, Medical Oncology, Institut Català de la Salut, [Felip E] Vall d'Hebron Hospital Universitari, Barcelona, Spain. Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. [de Braud FG] University of Milan, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy. [Maur M] AOU Policlinico of Modena, University of Modena and Reggio Emilia, Modena, Italy. [Loong HH] The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China. [Shaw AT] Massachusetts General Hospital, Boston, Massachusetts. [Vansteenkiste JF] University Hospital KU Leuven, Leuven, Belgium, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Ceritinib ,NSCLC ,Gastroenterology ,Quimioteràpia combinada ,03 medical and health sciences ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Therapeutics::Therapeutics::Drug Therapy::Antineoplastic Protocols::Therapeutics::Drug Therapy::Antineoplastic Combined Chemotherapy Protocols [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,PD-1 ,Maculopapular rash ,medicine ,Humans ,Anaplastic Lymphoma Kinase ,Other subheadings::/therapeutic use [Other subheadings] ,Sulfones ,Pulmons - Càncer - Quimioteràpia ,Lung cancer ,Adverse effect ,nivolumab ,Chemotherapy ,Otros calificadores::/uso terapéutico [Otros calificadores] ,business.industry ,terapéutica::terapéutica::farmacoterapia::protocolos antineoplásicos::terapéutica::farmacoterapia::protocolos de quimioterapia antineoplásica combinada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Neoplasms::Neoplasms by Site::Thoracic Neoplasms::Respiratory Tract Neoplasms::Lung Neoplasms::Bronchial Neoplasms::Carcinoma, Bronchogenic::Carcinoma, Non-Small-Cell Lung [DISEASES] ,neoplasias::neoplasias por localización::neoplasias torácicas::neoplasias del tracto respiratorio::neoplasias pulmonares::neoplasias de los bronquios::carcinoma broncogénico::carcinoma de pulmón de células no pequeñas [ENFERMEDADES] ,medicine.disease ,Rash ,Nivolumab ,Pyrimidines ,030104 developmental biology ,ALK ,Oncology ,030220 oncology & carcinogenesis ,Cohort ,medicine.symptom ,business ,medicine.drug - Abstract
INTRODUCTION: Induction of programmed death ligand 1 (PD-L1) expression due to constitutive oncogenic signaling has been reported in NSCLC models harboring echinoderm microtubule associated protein like 4 gene (EML4)-ALK receptor tyrosine kinase gene (ALK) rearrangements. We assessed the safety and activity of ceritinib plus nivolumab in these patients. METHODS: In this open-label, phase 1B, multicenter, dose escalation and expansion study, previously treated (with ALK receptor tyrosine kinase [ALK] inhibitor [ALKI]/chemotherapy) or treatment-naive patients with stage IIIB or IV ALK-rearranged NSCLC received nivolumab, 3 mg/kg intravenously every 2 weeks, plus ceritinib, 450 mg/300 mg daily, with a low-fat meal. RESULTS: In total, 36 patients were treated (a 450-mg cohort [n=14] and a 300-mg cohort [n=22]). In the 450-mg cohort, four patients experienced dose-limiting toxicities. In the 300-mg cohort, two patients experienced dose-limiting toxicities. Among ALKI-naive patients, the overall response rate (ORR) was 83% (95% confidence interval [CI]: 35.9-99.6) in the 450-mg cohort and 60% (95% CI: 26.2-87.8) in the 300-mg cohort. Among ALKI-pretreated patients, the ORR was 50% (95% CI: 15.7-84.3) in the 450-mg cohort and 25% (95% CI: 5.5-57.2) in the 300-mg cohort. The ORR point estimate was observed to be greater in patients who were positive for PD-L1 than in those who were negative for PD-L1, with overlapping CIs (e.g., at a cutoff ≥1% PD-L1, 64% of patients [95% CI: 35.1-87.2] had confirmed responses as compared with those with negative PD-L1 staining (31% [95% CI: 11.0-58.7]). The most frequently reported grade 3 or 4 adverse events were increased alanine aminotransferase level (25%), increased gamma-glutamyl transferase level (22%), increased amylase level (14%), increased lipase level (11%), and maculopapular rash (11%). The incidence of all-grade rash (grouped term) was 64% in both cohorts; grade 3 rash was reported in 29% and 14% of patients in the 450-mg and 300-mg cohorts, respectively; no grade 4 rash was reported. CONCLUSION: Ceritinib plus nivolumab has activity; ORR appears to correlate with PD-L1 at baseline. Toxicity, especially rash, is more common than with either single agent. ispartof: Journal of Thoracic Oncology vol:15 issue:3 pages:392-403 ispartof: location:United States status: published
- Published
- 2020
- Full Text
- View/download PDF
38. Metastatic pheochromocytomas and paragangliomas: where are we?
- Author
-
Natalie Prinzi, Francesca Corti, Martina Torchio, Monica Niger, Maria Antista, Filippo Pagani, Teresa Beninato, Iolanda Pulice, Roberta Elisa Rossi, Jorgelina Coppa, Tommaso Cascella, Luca Giacomelli, Maria Di Bartolomeo, Massimo Milione, Filippo de Braud, and Sara Pusceddu
- Subjects
Paraganglioma ,Cancer Research ,Oncology ,Peripheral Nervous System Neoplasms ,Adrenal Gland Neoplasms ,Humans ,Neoplasms, Second Primary ,General Medicine ,Pheochromocytoma - Abstract
Pheochromocytomas and paragangliomas (PPGLs) can metastasize in approximately 15–20% of cases. This review discusses the available evidence on the biology and treatment of metastatic PPGLs. Chemotherapy is the first-line treatment option for this evolving and symptomatic disease. In patients with high MIBG uptake and positive PETGa-68, radiometabolic treatment may be considered. The efficacy of sunitinib has been shown in observational studies, and pembrolizumab has been evaluated in phase II clinical studies, while other agents investigated in this setting are anti-angiogenic drugs cabozantinib, dovitinib, axitinib and lenvatinib. As these agents' efficacy and safety data, alone or in combination, are scant and based on few treated patients, enrollment in clinical trials is mandatory. Future therapeutic options may be represented by DNA repair system inhibitors (such as olaparib), HIF2 inhibitors and immunotherapy.
- Published
- 2022
39. Updated Integrated Analysis of the Efficacy and Safety of Entrectinib in Patients With NTRK Fusion-Positive Solid Tumors
- Author
-
George D. Demetri, Filippo De Braud, Alexander Drilon, Salvatore Siena, Manish R. Patel, Byoung Chul Cho, Stephen V. Liu, Myung-Ju Ahn, Chao-Hua Chiu, Jessica J. Lin, Koichi Goto, Jeeyun Lee, Lyudmila Bazhenova, Thomas John, Marwan Fakih, Sant P. Chawla, Rafal Dziadziuszko, Takashi Seto, Sebastian Heinzmann, Bethany Pitcher, David Chen, Timothy R. Wilson, and Christian Rolfo
- Subjects
Adult ,Cancer Research ,Lung Neoplasms ,Indazoles ,Carcinoma ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,Protein-Tyrosine Kinases ,Brain Disorders ,Oncology ,Clinical Research ,Carcinoma, Non-Small-Cell Lung ,Proto-Oncogene Proteins ,6.1 Pharmaceuticals ,Benzamides ,Humans ,Oncology & Carcinogenesis ,Non-Small-Cell Lung ,Child ,Cancer - Abstract
Purpose: Entrectinib potently inhibits tropomyosin receptor kinases (TRKAs)/B/C and ROS1, and previously induced deep [objective response rate (ORR) 57.4%] and durable [median duration of response (DoR) 10.4 months] responses in adults with NTRK fusion-positive solid tumors from three phase I/II trials. This article expands prior reports with additional patients and longer follow-up. Patients and Methods: Patients with locally advanced/metastatic NTRK fusion-positive solid tumors and ≥12 months' follow-up were included. Primary endpoints were ORR and DoR by blinded independent central review (BICR); secondary endpoints included progression-free survival (PFS), intracranial efficacy, and safety. The safety-evaluable populations included all patients who had received ≥1 entrectinib dose. Results: At clinical cut-off (August 31, 2020), the efficacy-evaluable population comprised 121 adults with 14 tumor types and ≥30 histologies. Median follow-up was 25.8 months; 61.2% of patients had a complete (n = 19) or partial response (n = 55). Median DoR was 20.0 months [95% confidence interval (CI), 13.0–38.2]; median PFS was 13.8 months (95% CI, 10.1–19.9). In 11 patients with BICR-assessed measurable central nervous system (CNS) disease, intracranial ORR was 63.6% (95% CI, 30.8–89.1) and median intracranial DoR was 22.1 (95% CI, 7.4–not estimable) months. The safety profile of entrectinib in adults and pediatric patients was aligned with previous reports. Most treatment-related adverse events (TRAEs) were grade 1/2 and manageable/reversible with dose modifications. TRAE-related discontinuations occurred in 8.3% of patients. Conclusions: With additional clinical experience, entrectinib continues to demonstrate durable systemic and intracranial responses and can address the unmet need of a CNS-active treatment in patients with NTRK fusion-positive solid tumors.
- Published
- 2022
40. Exceptional tumour responses to fasting-mimicking diet combined with standard anticancer therapies: A sub-analysis of the NCT03340935 trial
- Author
-
Francesca Ligorio, Giovanni Fucà, Leonardo Provenzano, Riccardo Lobefaro, Lucrezia Zanenga, Andrea Vingiani, Antonino Belfiore, Alice Lorenzoni, Alessandra Alessi, Giancarlo Pruneri, Filippo de Braud, and Claudio Vernieri
- Subjects
Cancer Research ,Oncology ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Fasting - Abstract
Cyclic fasting or calorie-restricted, low-carbohydrate, low-protein diets, collectively referred to as fasting-mimicking diets (FMDs), demonstrated additive or synergistic antitumour effects when combined with chemotherapy, targeted therapies, or immunotherapy in several preclinical in vivo models, including murine models of breast cancer, lung cancer, and colorectal cancer. However, no data on the antitumour efficacy of cyclic FMD in patients with cancer have been published so far. Here, we aim at reporting on patients with advanced cancer achieving complete and long-lasting tumour remissions with cyclic FMD in combination with standard anticancer therapies in the context of the phase Ib NCT03340935 trial.The NCT03340935 trial enrolled 101 patients with different tumour types, and it showed that a severely calorie-restricted FMD regimen is safe and feasible in patients with cancer receiving concomitant standard-of-care antineoplastic therapies. In addition, cyclic FMD resulted in positive metabolic and immunologic modifications, thus recapitulating the biological effects that in preclinical models were found to mediate the antitumour effects of fasting/FMD.Of the 101 patients enrolled in the NCT03340935 trial, we identified five patients with advanced, poor prognosis solid neoplasms (n = 1: extensive stage small cell lung cancer; n = 1: metastatic pancreatic adenocarcinoma; n = 1: metastatic colorectal cancer; n = 2: metastatic triple-negative breast cancer), who achieved complete and long-lasting tumour responses when treated with a combination of cyclic FMD and standard systemic treatments in the context of the NCT03340935 trial.These excellent responses prompt the initiation of clinical trials to investigate cyclic FMD in combination with standard antitumour therapies in specific clinical contexts.
- Published
- 2022
41. Abstract 5506: Circulating immature neutrophils early detect hyperprogressive disease upon first-line PD-1/PD-L1 inhibitors in non-small cell lung cancer patients selecting best candidates for platinum-based chemotherapy and PD-1/PD-L1 inhibitors combinations
- Author
-
Roberto Ferrara, Giuseppe Lo Russo, Chiara Maura Ciniselli, Annamaria Piva, Barbara Bassani, Elena Jachetti, Giuseppina Calareso, Valeria Duroni, Settimio Di Gregorio, Claudia Proto, Arsela Prelaj, Alessandro De Toma, Mario Occhipinti, Marta Brambilla, Sara Manglaviti, Laura Mazzeo, Arturo Rinaldi, Teresa Beninato, Monica Ganzinelli, Filippo De Braud, Marina Chiara Garassino, Paolo Verderio, Mario Paolo Colombo, and Sabina Sangaletti
- Subjects
Cancer Research ,Oncology - Abstract
Background: Hyperprogressive disease (HPD) has been described in ≃14-25% of pretreated non-small cell lung cancer (NSCLC) patients upon single-agent (SA) PD-1/PD-L1 inhibitors (ICI) and has not been reported upon platinum-based chemotherapy (PCT) and ICI combinations. So far, no predictive biomarkers are available for HPD early detection. Methods: NSCLC patients treated with 1st line SA-ICI or PCT-ICI were assessed for HPD and circulating neutrophils. HPD was defined as delta tumor growth rate (TGR) >50% and/or TGR ratio ≥2. Circulating low density neutrophils (LDNs) were assessed by flow cytometry on peripheral blood mononuclear cells (PMBCs). LDNs were defined as CD66b+CD15+ cells among CD11b+ PBMCs and immature subtypes as CD10− LDNs. The LDNs predictive role was assessed by penalized model-based tests. Results: 144 NSCLC patients were included: 75 treated with SA-ICI, 69 with PCT-ICI. In the SA-ICI cohort, HPD occurred in 8 (11%) patients, while progressive disease (PD) and response or stable disease (PR/SD) occurred in 33 (44%) and 34 (45%) of patients respectively. Immature circulating CD10− LDNs were significantly higher in baseline blood samples of HPD patients [median (ME): 39.3, interquartile range (IQR): 28.7] compared to PD [ME: 7.4, IQR: 14.9, p Conclusions: Baseline circulating immature neutrophils characterize HPD upon 1st line SA-ICI and a 30.5% cut-off of immature neutrophils could select NSCLC patients to be addressed to PCT-ICI combinations. Citation Format: Roberto Ferrara, Giuseppe Lo Russo, Chiara Maura Ciniselli, Annamaria Piva, Barbara Bassani, Elena Jachetti, Giuseppina Calareso, Valeria Duroni, Settimio Di Gregorio, Claudia Proto, Arsela Prelaj, Alessandro De Toma, Mario Occhipinti, Marta Brambilla, Sara Manglaviti, Laura Mazzeo, Arturo Rinaldi, Teresa Beninato, Monica Ganzinelli, Filippo De Braud, Marina Chiara Garassino, Paolo Verderio, Mario Paolo Colombo, Sabina Sangaletti. Circulating immature neutrophils early detect hyperprogressive disease upon first-line PD-1/PD-L1 inhibitors in non-small cell lung cancer patients selecting best candidates for platinum-based chemotherapy and PD-1/PD-L1 inhibitors combinations. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5506.
- Published
- 2023
- Full Text
- View/download PDF
42. Abstract HER2-02: HER2-02 HER2-Low Status is Associated with Worse Clinical Outcomes in Hormone Receptor-Positive, HER2-Negative Advanced Breast Cancer Patients Treated With First-Line Cyclin-Dependent Kinase 4/6 Inhibitors Plus Endocrine Therapy
- Author
-
Emma Zattarin, Caterina Sposetti, Rita Leporati, Luigi Mariani, Alice Menichetti, Chiara Corti, Chiara Benvenuti, Giovanni Fucà, Riccardo Lobefaro, Francesca Ligorio, Daniele Presti, Leonardo Provenzano, Andrea Vingiani, Gaia Griguolo, Marianna Sirico, Ottavia Bernocchi, Antonio Marra, Paola Zagami, Elisa Agostinetto, Flavia Jacobs, Pierluigi Di Mauro, Andrea Esposito, Carlo Alberto Giorgi, Luca Lalli, Laura Boldrini, Pier Paolo Maria Berton Giachetti, Ambra Carnevale Schianca, Valentina Guarneri, Rebecca Pedersini, Agnese Losurdo, Alberto Zambelli, Daniele Giulio Generali, Giuseppe Curigliano, Giancarlo Pruneri, Filippo de Braud, Maria Vittoria Dieci, and Claudio Vernieri
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) in combination with endocrine therapy (ET) are the standard first-line treatment for patients with hormone receptor-positive, human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer (HR+/HER2- aBC). HER2-low BC, which is defined by an IHC score for HER2 of 1+ or 2+ with negative ISH assay, accounts for more than half of all HR+/HER2- aBC cases, and it is associated with remarkable clinical benefit from the novel anti-HER2 antibody drug conjugate (ADC) trastuzumab-deruxtecan. Evidence on the prognostic impact of HER2-low status is controversial in both limited-stage and advanced BC. Here, we sought to investigate the possible prognostic relevance of HER2-low status in a population of aBC patients treated with CDK4/6i plus ET. Methods: We conducted a retrospective-prospective study in six Italian Cancer Centers to investigate the impact of HER2 status (low vs. 0) on the progression-free survival (PFS) and overall survival (OS) of consecutive HR+/HER2- aBC patients treated with CDK4/6i plus ET (aromatase inhibitors or fulvestrant) as a first-line therapy. In the main study analysis, we considered HER2 status in the last tumor assessment (i.e., primary tumor, or, when available, a metastatic lesion). We also performed a subgroup analysis including only patients with HER2 status evaluation in a metastatic lesion collected before CDK4/6i plus ET therapy initiation. The association between HER2 status (low vs. 0) and PFS or OS was evaluated using log-rank test and Cox regression modeling. Results: We evaluated 767 consecutive HR+/HER2- aBC patients treated with CDK4/6i plus ET between January 2017 and January 2022. Of these, 436 patients (56.8%) received CDK4/6i plus ET as a first-line therapy, and they were included in this analysis. Median age was 63 years (range 27-87), and 362 patients (83.0%) were postmenopausal. The majority of patients were treated with palbociclib (68.3%), while 91 (20.9%) and 47 (10.8%) patients received ribociclib and abemaciclib, respectively. Regarding HER2 status, 269 (62.9%) patients had HER2-low tumors, while 159 (37.1%) patients had HER2-0 neoplasms. HER2-low status was associated with significantly lower PFS when compared to HER2-0 status [median PFS (mPFS) 23.6 vs. 32.3 months, respectively; p=0.014]. HER2-low status was also associated with significantly worse OS (mOS 48.7 vs 58.3 months, respectively; p=0.025). These results were confirmed in multivariable models adjusting the impact of HER2 status for clinically-relevant covariates, namely estrogen receptor status, Ki-67, age, number of metastatic sites, presence of liver metastases, disease free interval, ECOG Performance Status. In this analysis, HER2-low status, compared with HER2-0 status, was independently associated with worse PFS [adjusted Hazard Ratio (aHR): 1.62; 95% confidence interval (CI): 1.17-2.24; p< 0.01] and OS (aHR: 1.74; 95% CI: 1.09-2.76; p=0.019). Subgroup analysis conducted in the subset of 256 patients with available metastatic tumor samples collected before CDK4/6i plus ET initiation confirmed that HER2-low status (n=157), when compared to HER2-0 status (n=99), was independently associated with worse PFS (mPFS 24.5 vs 35.2 months, p=0.01; aHR 2.07; 95% CI: 1.28-3.34, p< 0.01) and worse OS (mPFS 48.7 vs 72.3 months, p=0.027; aHR 3.12; 95% CI 1.44-6.77, p< 0.01). Conclusions: This multicenter Italian study revealed that HER2-low status has independent, negative prognostic value in patients with HR+/HER2- aBC treated with CDK4/6i plus ET in the first-line setting. Our results suggest that HER2-low status might be associated with different clinical benefit from standard anticancer therapies in specific clinical settings. The definition of treatment algorithms also taking into account HER2 status is a clinical priority in patients with HR+/HER2- aBC. Citation Format: Emma Zattarin, Caterina Sposetti, Rita Leporati, Luigi Mariani, Alice Menichetti, Chiara Corti, Chiara Benvenuti, Giovanni Fucà, Riccardo Lobefaro, Francesca Ligorio, Daniele Presti, Leonardo Provenzano, Andrea Vingiani, Gaia Griguolo, Marianna Sirico, Ottavia Bernocchi, Antonio Marra, Paola Zagami, Elisa Agostinetto, Flavia Jacobs, Pierluigi Di Mauro, Andrea Esposito, Carlo Alberto Giorgi, Luca Lalli, Laura Boldrini, Pier Paolo Maria Berton Giachetti, Ambra Carnevale Schianca, Valentina Guarneri, Rebecca Pedersini, Agnese Losurdo, Alberto Zambelli, Daniele Giulio Generali, Giuseppe Curigliano, Giancarlo Pruneri, Filippo de Braud, Maria Vittoria Dieci, Claudio Vernieri. HER2-02 HER2-Low Status is Associated with Worse Clinical Outcomes in Hormone Receptor-Positive, HER2-Negative Advanced Breast Cancer Patients Treated With First-Line Cyclin-Dependent Kinase 4/6 Inhibitors Plus Endocrine Therapy [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr HER2-02.
- Published
- 2023
- Full Text
- View/download PDF
43. Cabozantinib beyond progression improves survival in advanced renal cell carcinoma patients: the CABEYOND study (Meet-URO 21)
- Author
-
Giuseppe Procopio, Filippo de Braud, Pierangela Sepe, Emma Zattarin, Giandomenico Roviello, Giuseppe Di Lorenzo, Arianna Ottini, Sebastiano Buti, Ugo De Giorgi, Sandro Pignata, Carlo Messina, Davide Bimbatti, Marco Stellato, Melanie Claps, Valentina Guadalupi, Daniele Santini, Consuelo Buttigliero, Massimo Di Maio, Elena Verzoni, Mariella Sorarù, Alessia Mennitto, Claudia Mucciarini, and Chiara Casadei
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Cabozantinib ,kidney cancer treatment ,metastatic renal cell carcinoma ,post-progression survival ,treatment beyond progression ,Pyridines ,chemistry.chemical_compound ,Renal cell carcinoma ,Internal medicine ,Clinical endpoint ,Humans ,Medicine ,Anilides ,Pharmacology (medical) ,Carcinoma, Renal Cell ,Retrospective Studies ,Prior treatment ,Response rate (survey) ,business.industry ,Carcinoma ,Disease progression ,Renal Cell ,medicine.disease ,Kidney Neoplasms ,chemistry ,Tolerability ,Multicenter study ,Female ,business - Abstract
BACKGROUND Cabozantinib improves survival in metastatic renal cell carcinoma (mRCC) after prior antiangiogenics. The best treatment at disease progression (PD) is unknown. Being also a AXL/MET inhibitor, involved in acquired resistance, we hypothesized a prolonged tumor growth control in patients continuing cabozantinib despite PD. RESEARCH DESIGN AND METHODS This retrospective multicenter study enrolled patients receiving cabozantinib after the first line between 2014 and 2020. We compared patients maintaining cabozantinib after first PD due to clinical benefit and good tolerability with those who changed therapy. The postprogression survival (PPS) of both was our primary endpoint. RESULTS We analyzed 89 patients: 45 received cabozantinib beyond PD and 44 switched therapy. 40.4%, 31.5%, and 28.1% of patients received 1, 2, or >2 prior treatment, respectively. 84.3% were intermediate-poor International Metastatic Renal Cell Carcinoma Database risk. Patients continuing cabozantinib showed a higher response rate to cabozantinib before PD (46.7% vs 25%, p = 0.03) and were more heavily pretreated. Continuing cabozantinib showed a significantly longer PPS compared with switching therapy (median PPS 16.9 vs 13.2 months, HR 0.66, 95%CI 0.48-0.92, p = 0.011). CONCLUSIONS We observed longer PPS in patients continuing cabozantinib beyond PD, suggesting that this could be an effective option.
- Published
- 2022
44. Platinum sensitivity in patients with IDH1/2 mutated versus wild-type intrahepatic cholangiocarcinoma: a propensity score-based study
- Author
-
Monica Niger, Federico Nichetti, Andrea Casadei‐Gardini, Mario Domenico Rizzato, Chiara Pircher, Marta Bini, Andrea Franza, Margherita Rimini, Valentina Burgio, Caterina Sposetti, Lorenzo Fornaro, Ilario Giovanni Rapposelli, Francesco Enrico D'Amico, Giuseppe Aprile, Caterina Vivaldi, Giovanni Luca Frassineti, Massimo Milione, Giuseppe Leoncini, Alessandro Cappetta, Enrico Vasile, Matteo Fassan, Federica Morano, Federica Perrone, Elena Tamborini, Giancarlo Pruneri, Sara Lonardi, Vincenzo Mazzaferro, Filippo Pietrantonio, Maria Di Bartolomeo, and Filippo de Braud
- Subjects
Intrahepatic ,Cancer Research ,Settore MED/06 - Oncologia Medica ,DNA repair ,Isocitrate Dehydrogenase ,Cholangiocarcinoma ,IDH1 ,IDH2 ,cholangiocarcinoma ,homologous recombination deficiency ,Bile Ducts, Intrahepatic ,Humans ,Mutation ,Propensity Score ,Bile Duct Neoplasms ,Oncology ,Bile Ducts ,Homologous Recombination Deficiency - Abstract
Isocitrate dehydrogenase (IDH)1/2 mutations are the most frequent druggable alterations in intrahepatic cholangiocarcinoma (iCCA), reported in ~20% of cases. Preclinical evidence indicates that these mutations are associated with homologous recombination deficiency (HRD), which could be exploited as a target for platinum chemotherapy (ChT) and PARP inhibitors. However, the role of IDH1/2 mutations as surrogate biomarkers for platinum efficacy is unknown. We conducted a multicenter, propensity score-matched analysis to investigate the impact of IDH1/2 mutations on progression-free survival (PFS), overall response rate (ORR) and disease control rate (DCR) in patients with iCCA treated with platinum-based ChT. An exploratory comparison of complex HRD estimates between IDH1/2 mutated and wild-type tumors from TCGA was also performed. A total of 120 cases were matched in a 1:1 ratio (60 IDH1/2 mutant and 60 wild-type). No differences were observed for platinum-based PFS (7.7 vs 7.3 months, P = .970), DCR (66.1% vs 74.1%, P = .361) and ORR (27.8% vs 25.0%, P = .741). IDH1/2 mutations showed mutual exclusivity with genomic alterations in ATM, BRCA2, MST1R, NF1, FGFR2 and CDKN2A/B losses, respectively, with no clear survival and response differences. Among TCGA tumors, IDH1/2 mutated CCA did not show higher HRD compared to wild-type cases. IDH1/2 mutations are not associated with increased sensitivity to platinum-based ChT in iCCA patients. Deeper genomic sequencing is needed to elucidate the HRD phenotype in IDH1/2 mutant iCCA and exploit its therapeutic vulnerabilities.
- Published
- 2022
45. Prognostic role of neutrophil-to-lymphocyte ratio and EPSILoN score in advanced non-small-cell lung cancer patients treated with first-line chemo-immunotherapy
- Author
-
Emma Zattarin, Sara Manglaviti, Giulia Apollonio, Teresa Beninato, Laura Mazzeo, Giacomo Massa, Achille Bottiglieri, Edoardogregorio Galli, Alessandro De Toma, Mario Occhipinti, Marta Brambilla, Roberto Ferrara, Monica Ganzinelli, Claudia Proto, Marina Chiara Garassino, Filippo de Braud, Giuseppe Lo Russo, and Arsela Prelaj
- Subjects
Cancer Research ,Lung Neoplasms ,Neutrophils ,Carcinoma ,General Medicine ,first-line setting ,Prognosis ,prognostic score ,EPSILoN score ,Oncology ,neutrophil-to-lymphocyte ratio ,advanced non-small-cell lung cancer ,Carcinoma, Non-Small-Cell Lung ,Humans ,chemo-immunotherapy ,Immunotherapy ,Lymphocytes ,Non-Small-Cell Lung ,Biomarkers ,Retrospective Studies - Abstract
Patients affected by inoperable lung cancer, due to great extension or to the presence of metastases, are currently treated with intravenous drugs that act on immune system activation alone or in combination with chemotherapy as first-line treatment. The characteristics of these patients (both their medical history and their blood exams) need to be studied to find out if some of them can help clinicians to predict if they will benefit from the combination of immunotherapy with chemotherapy. The authors collected the data of patients with advanced lung cancer treated in their hospital and found out that a value calculated from their blood exams, collected before the start of treatment and a combination of values named EPSILoN score (which considers patients' clinical condition, their history of tobacco smoking, the presence of metastases in the liver and two blood exam parameters, namely the neutrophil-to-lymphocyte ratio and LDH level) can predict how their disease will evolve during first-line treatment with chemotherapy in combination with immunotherapy.
- Published
- 2022
46. Biomarker Landscape in Neuroendocrine Tumors With High-Grade Features: Current Knowledge and Future Perspective
- Author
-
Michele Prisciandaro, Maria Antista, Alessandra Raimondi, Francesca Corti, Federica Morano, Giovanni Centonze, Giovanna Sabella, Alessandro Mangogna, Giovanni Randon, Filippo Pagani, Natalie Prinzi, Monica Niger, Salvatore Corallo, Erica Castiglioni di Caronno, Marco Massafra, Maria Di Bartolomeo, Filippo de Braud, Massimo Milione, Sara Pusceddu, Prisciandaro, Michele, Antista, Maria, Raimondi, Alessandra, Corti, Francesca, Morano, Federica, Centonze, Giovanni, Sabella, Giovanna, Mangogna, Alessandro, Randon, Giovanni, Pagani, Filippo, Prinzi, Natalie, Niger, Monica, Corallo, Salvatore, Castiglioni di Caronno, Erica, Massafra, Marco, Di Bartolomeo, Maria, de Braud, Filippo, Milione, Massimo, and Pusceddu, Sara
- Subjects
PD-L1 ,Cancer Research ,neuroendocrine carcinoma (NEC) ,Oncology ,next-generation sequencing (NGS) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,high microsatellite instability (MSI-H) ,neuroendocrine tumors ,RC254-282 ,neuroendocrine tumor - Abstract
Neuroendocrine tumors (NETs) are classified based on morphology and are graded based on their proliferation rate as either well-differentiated low-grade (G1) to intermediate (G2–G3) or poorly differentiated high-grade neuroendocrine carcinomas (NEC G3). Recently, in gastroenteropancreatic (GEP) NETs, a new subgroup of well-differentiated high-grade tumors (NET G3) has been divided from NEC by WHO due to its different clinical–pathologic features. Although several mutational analyses have been performed, a molecular classification of NET is an unmet need in particular for G3, which tends to be more aggressive and have less benefit to the available therapies. Specifically, new possible prognostic and, above all, predictive factors are highly awaited, giving the basis for new treatments. Alteration of KRAS, TP53, and RB1 is mainly reported, but also druggable alterations, including BRAF and high microsatellite instability (MSI-H), have been documented in subsets of patients. In addition, PD-L1 demonstrated to be highly expressed in G3 NETs, probably becoming a new biomarker for G3 neuroendocrine neoplasm (NEN) discrimination and a predictive one for immunotherapy response. In this review, we describe the current knowledge available on a high-grade NET molecular landscape with a specific focus on those harboring potentially therapeutic targets in the advanced setting.
- Published
- 2022
- Full Text
- View/download PDF
47. Management of BRCA Tumour Testing in an Integrated Molecular Tumour Board Multidisciplinary Model
- Author
-
Jacopo Azzollini, Andrea Vingiani, Luca Agnelli, Elena Tamborini, Federica Perrone, Elena Conca, Iolanda Capone, Adele Busico, Bernard Peissel, Erica Rosina, Monika Ducceschi, Mara Mantiero, Salvatore Lopez, Francesco Raspagliesi, Monica Niger, Matteo Duca, Silvia Damian, Claudia Proto, Filippo de Braud, Giancarlo Pruneri, and Siranoush Manoukian
- Subjects
Cancer Research ,ovarian cancer ,Oncology ,somatic variants ,Settore MED/06 - Oncologia Medica ,homologous recombination ,BRCA1 ,BRCA2 ,genetic counselling ,HBOC (hereditary breast and ovarian cancer) ,Settore MED/08 - Anatomia Patologica - Abstract
Tumour testing of the BRCA1/2 genes is routinely performed in patients with different cancer histological subtypes. To accurately identify patients with tumour-detected germline pathogenic variants (PVs) is a relevant issue currently under investigation. This study aims at evaluating the performance of the tumour-to-germline diagnostic flowchart model defined at our Institutional Molecular Tumour Board (MTB). Results from tumour BRCA sequencing of 641 consecutive unselected cancer patients were discussed during weekly MTB meetings with the early involvement of clinical geneticists for appropriate referral to genetic counselling. The overall tumour detection rate of BRCA1/2 PVs was 8.7% (56/641), ranging from 24.4% (31/127) in high-grade ovarian cancer to 3.9% (12/304) in tumours not associated with germline BRCA1/2 PVs. Thirty-seven patients with PVs (66%) were evaluated by a clinical geneticist, and in 24 of them (64.9%), germline testing confirmed the presence of the PV in blood. Nine of these patients (37.5%) were not eligible for germline testing according to the criteria in use at our institution. Cascade testing was subsequently performed on 18 relatives. The tumour-to-germline diagnostic pipeline, developed in the framework of our institutional MTB, compared with guideline-based germline testing following genetic counselling, proved to be effective in identifying a higher number of germline BRCA PVs carriers.
- Published
- 2022
48. Immune-Checkpoint Inhibitors in Advanced Non-Small Cell Lung Cancer With Uncommon Histology
- Author
-
Emma Zattarin, Giuseppe Viscardi, Elisa Sottotetti, A. Prelaj, Marta Brambilla, Giulia Galli, Riccardo Lobefaro, Monica Ganzinelli, Claudia Proto, Roberto Ferrara, Rosa Maria Di Mauro, Sara Manglaviti, Diego Signorelli, Marta Bini, Mario Occhipinti, Giuseppe Lo Russo, Giulia Apollonio, Giacomo Massa, Teresa Beninato, Filippo de Braud, Alessandra Fabbri, A. Bottiglieri, M.C. Garassino, Alessandro De Toma, and Benedetta Trevisan
- Subjects
Pulmonary and Respiratory Medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Adenosquamous carcinoma ,medicine.medical_treatment ,Population ,Uncommon NSCLC ,Kaplan-Meier Estimate ,Gastroenterology ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,medicine ,80 and over ,Humans ,ICIs ,education ,Sarcomatoid carcinoma ,Lung cancer ,Non-Small-Cell Lung ,Immune Checkpoint Inhibitors ,Aged ,Proportional Hazards Models ,Retrospective Studies ,Aged, 80 and over ,education.field_of_study ,business.industry ,Not Otherwise Specified ,Carcinoma ,Histology ,Immunotherapy ,Middle Aged ,medicine.disease ,Survival Analysis ,immunotherapy ,lung cancer ,rare histology ,uncommon NSCLC ,Rare histology ,Oncology ,Adenocarcinoma ,Female ,business - Abstract
Immune-checkpoint inhibitors (ICIs) have significantly improved outcome of advanced non-small cell lung cancer (aNSCLC) patients. However, their efficacy remains uncertain in uncommon histologies (UH).Data from ICI treated aNSCLC patients (April,2013-January,2021) in one Institution were retrospectively collected. Univariate and multivariate survival analyses were estimated by Kaplan-Meier and Cox proportional hazards regression model, respectively. Objective response rate (ORR) and disease control rate (DCR) were assessed.Of 375 patients, 79 (21.1%) had UH: 19 (24.1%) sarcomatoid carcinoma, 15 (19.0%) mucinous adenocarcinoma, 10 (12.6%) enteric adenocarcinoma, 8 (10.1%) adenocarcinoma not otherwise specified, 7 (8.9%) large-cell neuroendocrine carcinoma, 6 (7.6%) mixed histology non-adenosquamous, 5 (6.3%) adenosquamous carcinoma, 9 (11.4%) other UH. In UH group, programmed death-ligand 1 (PD-L1)1%, 1-49%, ≥50% and unknown expression were reported in 27.8%, 22.8%, 31.7% and 17.7% patients respectively and ICI was the second/further-line in the majority of patients. After a median follow-up of 35.64 months (m), median progression-free survival (mPFS) was 2.5 m in UH [95% CI 2.2-2.9 m] versus (vs.) 2.7 m in CH [95% CI 2.3-3.2 m, P-value = .584]; median overall survival (mOS) was 8.8 m [95% CI 4.9-12.6 m] vs. 9.7 m [95% CI 8.0-11.3 m, P-value = .653]. At multivariate analyses only ECOG PS was a confirmed prognostic factor in UH. ORR and DCR were 25.3% and 40.5% in UH vs. 21.6% and 49.5% in CH [P-value = .493 and .155 respectively].No significant differences were detected between UH and CH groups. Prospective trials are needed to understand ICIs role in UH population.
- Published
- 2022
49. The pan-immune-inflammation value is associated with clinical outcomes in patients with advanced TNBC treated with first-line, platinum-based chemotherapy: an institutional retrospective analysis
- Author
-
Leonardo Provenzano, Riccardo Lobefaro, Francesca Ligorio, Emma Zattarin, Luca Zambelli, Caterina Sposetti, Daniele Presti, Giulia Montelatici, Angela Ficchì, Antonia Martinetti, Alessio Arata, Marta Del Vecchio, Claudia Lauria Pantano, Barbara Formisano, Giulia Valeria Bianchi, Giuseppe Capri, Filippo de Braud, Claudio Vernieri, and Giovanni Fucà
- Subjects
Oncology - Abstract
Background: Advanced triple-negative breast cancer (aTNBC) has a poor prognosis; thus, there is a need to identify novel biomarkers to guide future research and improve clinical outcomes. Objectives: We tested the prognostic ability of an emerging, complete blood count (CBC)-based inflammatory biomarker, the pan-immune-inflammation value (PIV), in patients with aTNBC treated with first-line, platinum-based chemotherapy. Design: This was a retrospective, monocentric, observational study. Methods: We included consecutive aTNBC patients treated with platinum-based, first-line chemotherapy at our Institution, and for whom baseline (C1) CBC data were available. We collected CBC data early on-treatment, when available. PIV was calculated as: (neutrophil count × platelet count × monocyte count)/lymphocyte count. Patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer (aBC) were included in a control, non-TNBC cohort. Results: A total of 78 aTNBC patients were included. When evaluated as a continuous variable, PIV-C1 was associated with worse overall survival (OS; p Conclusion: PIV has a promising prognostic discrimination ability in aTNBC patients treated with first-line, platinum-based chemotherapy. Both baseline and early on-treatment PIV are associated with clinical outcomes and may be exploited for creating PIV-based risk classifiers if further validated.
- Published
- 2023
- Full Text
- View/download PDF
50. EGFR Amplification in Metastatic Colorectal Cancer
- Author
-
Jeeyun Lee, Rona Yaeger, Elena Elez, Henry Walch, Chiara Cremolini, Marco Maria Germani, Henry Wood, Annunziata Gloghini, Paolo Manca, Alberto Bardelli, Jaclyn F. Hechtman, Daniele Rossini, Margherita Ratti, Filippo Pagani, J. Seligmann, Filippo Pietrantonio, Benedetta Mussolin, Giovanni Fucà, Margherita Ambrosini, Francesc Salvà, Filippo de Braud, Federica Morano, Giovanni Randon, Massimo Milione, Gouri Nanjangud, and Susan D Richman
- Subjects
Oncology ,Proto-Oncogene Proteins B-raf ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Colorectal cancer ,Monoclonal antibody ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,030304 developmental biology ,0303 health sciences ,business.industry ,Hazard ratio ,Articles ,medicine.disease ,Primary tumor ,Confidence interval ,digestive system diseases ,Clinical trial ,ErbB Receptors ,030220 oncology & carcinogenesis ,Cohort ,Colonic Neoplasms ,business ,Colorectal Neoplasms ,Cohort study - Abstract
Background EGFR amplification occurs in about 1% of metastatic colorectal cancers (mCRCs) but is not routinely tested as a prognostic or predictive biomarker for patients treated with anti-EGFR monoclonal antibodies. Herein, we aimed to characterize the clinical and molecular landscape of EGFR-amplified mCRC. Methods In this multinational cohort study, we compared clinical data of 62 patients with EGFR-amplified vs 1459 EGFR nonamplified mCRC, as well as comprehensive genomic data of 35 EGFR-amplified vs 439 EGFR nonamplified RAS/BRAF wild-type and microsatellite stable (MSS) tumor samples. All statistical tests were 2-sided. Results EGFR amplification was statistically significantly associated with left primary tumor sidedness and RAS/BRAF wild-type status. All EGFR-amplified tumors were MSS and HER2 nonamplified. Overall, EGFR-amplified samples had higher median fraction of genome altered compared with EGFR-nonamplified, RAS/BRAF wild-type MSS cohort. Patients with EGFR-amplified tumors reported longer overall survival (OS) (median OS = 71.3 months, 95% confidence interval [CI] = 50.7 to not available [NA]) vs EGFR-nonamplified ones (24.0 months; 95% CI = 22.8 to 25.6; hazard ratio [HR] = 0.30, 95% CI = 0.20 to 0.44; P < .001; adjusted HR = 0.46, 95% CI = 0.30 to 0.69; P < .001). In the subgroup of patients with RAS/BRAF wild-type mCRC exposed to anti-EGFR-based therapy, EGFR amplification was again associated with better OS (median OS = 54.0 months, 95% CI = 35.2 to NA, vs 29.1 months, 95% CI = 27.0 to 31.9, respectively; HR = 0.46, 95% CI = 0.28 to 0.76; P = .002). Conclusion Patients with EGFR-amplified mCRC represent a biologically defined subgroup and merit dedicated clinical trials with novel and more potent EGFR-targeting strategies beyond single-agent monoclonal antibodies.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.