166 results on '"Nichetti, F."'
Search Results
52. 56P - Pro-GRP in small cell lung cancer
- Author
-
Cavalieri, S., Nichetti, F., Morelli, D., de Braud, F., Martinetti, A., Sottotetti, E., Dotti, K., Prisciandaro, M., Corti, F., and Platania, M.
- Published
- 2017
- Full Text
- View/download PDF
53. D23 - Pharmacogenomic evaluation of single nucleotide polymorphisms associated with oxaliplatin-induced peripheral neuropathy: a preliminary study
- Author
-
Nichetti, F., Infante, G., Miceli, R., Berenato, R., Caporale, M., Morano, F., Falvella, F.S., Niger, M., Bossi, I., Fucà, G., Mennitto, A., Di Bartolomeo, M., Prof de Braud, F., and Pietrantonio, F.
- Published
- 2016
- Full Text
- View/download PDF
54. A34 - Pro-gastrin releasing peptide (pro-GRP) in small cell lung cancer staging
- Author
-
Cavalieri, S., Raimondi, A., Morelli, D., Martinetti, A., de Braud, F., Agustoni, F., Dotti, K., Galli, G., Vernieri, C., Fucà, G., Peverelli, G., Prisciandaro, M., Nichetti, F., Indelicato, P., Lo Russo, G., Prinzi, N., Garassino, M., and Platania, M.
- Published
- 2016
- Full Text
- View/download PDF
55. E16 - Risk of thromboembolic events (TEE) in metastatic colorectal cancer (mCRC) patients with single nucleotide polymorphisms (SNPs) in Factor V Leiden (FVL), Prothrombin, Plasminogen Activator Inhibitor-1 (PAI-1) and Methylenetetrahydrofolate Reductase (MTHFR)
- Author
-
Falvella, S., Cremolini, C., Miceli, R., Niger, M., Berenato, R., Cheli, S., Antoniotti, C., Nichetti, F., Infante, G., Caporale, M., Martinetti, A., Marmorino, F., Sottotetti, E., Colombo, A., Bossi, I., Di Bartolomeo, M., de Braud, F., Clementi, E., Loupakis, F., and Pietrantonio, F.
- Published
- 2015
- Full Text
- View/download PDF
56. 5PSQ-097 Toxicity associated with gene polymorphisms in patients with colorectal cancer, treated with fluoropyrimidines and analogues, irinotecan and platinum coordination complexes
- Author
-
Cricchio, G Lo, Saibene, G, Ruffino, E, Laganà, G, Costanza, C Della, Pietrantonio, F, Falvella, S, Nichetti, F, Celotti, FM, and Ladisa, V
- Abstract
BackgroundGene variants, such as single nucleotide polymorphisms, have a clinical relevance in the oncological field, when they affect genes encoding enzymes involved in drug metabolism, influencing drug toxicity, treatment compliance and efficacy.PurposeThe purpose of this work is to obtain data to choose a personalised therapy based on individual gene variations, minimise adverse events (AE) and avoid the discontinuation of therapy resulting in tumour progression.Material and methodsA retrospective study was conducted on 57 males and females, age ≥18, with colorectal cancer, in therapy with five protocols using different combinations of 5-fluorouracil, irinotecan and oxaliplatin.The study evaluated the number of cases where therapy was temporarily discontinued or suspended due to AE that concerned haematological, neurological and gastrointestinal toxicity according to the CTCAE system, which provides a numerical grading scale for AE description.The prevalence of polymorphisms and association between toxicity and polymorphisms were evaluated calculating odds ratios (OR) with 95% confidence interval.The Chi-square statistical significance test was applied.Results10 polymorphisms were analysed. In order of prevalence they are:UGT1A1*28 (38.6%, n=22)GSTPi (26.32%, n=15)ABCC2rs818 (17.54%, n=10)DPYDc496A>G (15.79%, n=9)SLC31A1 (12.28%, n=7)ABCC2rs717 (10.53%, n=6)DPYDc. 1129–5923C>G (3.51%, n=2)DPYD*2Ac. 1905+1 G>A and DPYD*13 c. 1679T>G (1.75%, n=1)DPYDc. 2846A>T (0%).OR values found the association between toxicity above 2nd grade and the presence of polymorphisms. The association is:Strong positive for DPYD*2Ac. 1905+1 G>A (OR=10.68) and UGT1A1*28 (OR=7.43)Moderate positive for DPYDc. 1129–5923C>G (OR=3.58) and SLC31A1 (OR=2.13)Moderate negative for ABCC2rs818 (OR=0.33).Absent for DPYD*13 c. 1679T>G, DPYDc496A>G, ABCC2rs717 and GSTPi.ConclusionOften patients express different polymorphisms at the same time, developing a toxicity related to the total effects of all the polymorphic variants. This problem is particularly important for chemotherapeutics that are administered at very high doses, close to toxic doses, and takes on a clinical and economic relevance. The study of genes, involved in the metabolism and transport of many drugs, permits the prediction of drug toxicity and efficacy and, based on individual variations, establishing a personalised and safe therapy before the onset of the treatment.References and/or Acknowledgements1. Common Terminology Criteria for Adverse Events(CTCAE). U S department of health and human services, national institutes of health, national cancer instituteJune 14, 2010.2. Sheskin DJ. Handbook of parametric and non parametric statistical procedures2004. IIIed Boca Raton:Chapman & Hall/CRC.3. Panczyk M. Pharmacogenetics research on chemotherapy resistance in colorectal cancer over the last 20years. World J Gastroenterol2014August 7;20(29):9775–9827.No conflict of interest
- Published
- 2018
- Full Text
- View/download PDF
57. Prognostic Role of a New Index Tested in European and Korean Advanced Biliary Tract Cancer Patients: the PECS Index
- Author
-
Luca Faloppi, Hyungwoo Cho, Federico Nichetti, Francesco Leone, Giuseppe Aprile, Daniele Santini, Giulia Orsi, Mariaelena Casagrande, Mario Scartozzi, Jae Ho Jeong, Nicola Silvestris, Giovanni Brandi, Eleonora Lai, Stefania De Lorenzo, Changhoon Yoo, Filippo de Braud, Monica Niger, Elisabetta Fenocchio, Lorenzo Fornaro, Stefano Cascinu, Caterina Vivaldi, Massimo Di Maio, Giulia Rovesti, Enrico Vasile, Andrea Casadei-Gardini, Francesco Caputo, Massimo Aglietta, Andrea Palloni, Roberto Filippi, Nicoletta Pella, Rovesti, G., Leone, F., Brandi, G., Fornaro, L., Scartozzi, M., Niger, M., Yoo, C., Caputo, F., Filippi, R., Casagrande, M., Silvestris, N., Santini, D., Faloppi, L., Palloni, A., Aglietta, M., Vivaldi, C., Cho, H., Lai, E., Fenocchio, E., Nichetti, F., Pella, N., De Lorenzo, S., Di Maio, M., Vasile, E., de Braud, F., Jeong, J. H., Aprile, G., Orsi, G., Cascinu, S., Casadei-Gardini, A., Rovesti G., Leone F., Brandi G., Fornaro L., Scartozzi M., Niger M., Yoo C., Caputo F., Filippi R., Casagrande M., Silvestris N., Santini D., Faloppi L., Palloni A., Aglietta M., Vivaldi C., Cho H., Lai E., Fenocchio E., Nichetti F., Pella N., De Lorenzo S., Di Maio M., Vasile E., de Braud F., Jeong J.H., Aprile G., Orsi G., Cascinu S., and Casadei-Gardini A.
- Subjects
medicine.medical_specialty ,Index (economics) ,Multivariate analysis ,Survival ,Prognosi ,Neutrophils ,medicine.medical_treatment ,Locally advanced ,Gastroenterology ,Prognostic index ,Biliary tract cancer ,Chemotherapy ,Cholangiocarcinoma ,Gallbladder cancer ,Prognosis ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Republic of Korea ,Medicine ,Humans ,Lymphocytes ,Survival analysis ,Retrospective Studies ,Inflammation ,business.industry ,Reproducibility of Results ,medicine.disease ,Biliary Tract Neoplasms ,Oncology ,030220 oncology & carcinogenesis ,Absolute neutrophil count ,030211 gastroenterology & hepatology ,business - Abstract
Background and Aim: The aim of the present study is to evaluate a new index (PECS (PsECogSii)index) influenced by PS ECOG and systemic immune-inflammation index (SII) in unresectable locally advanced or metastatic BTC patients treated with first-line chemotherapy. Methods: This multicenter, international, study was conducted on a training cohort of 130 patients and in three European and Korean validation cohorts The PECS index was calculated as ECOG × SII index (neutrophil count × platelet count/lymphocyte count). Event-time distributions were estimated using the Kaplan–Meier method and survival curves were compared using the log-rank test. Results: In the training cohort, the median overall survival (mOS) was 13.2months, 8.7months, and 3.8months for patients with PECS-0, PECS-1, and PECS-2, respectively (PECS-0: HR=1; PECS-1: HR 1.41; PECS-2: HR 3.23) (p 
- Published
- 2021
58. Impact of baseline and on-treatment glycemia on everolimus-exemestane efficacy in patients with hormone receptor-positive advanced breast cancer (EVERMET)
- Author
-
Giovanna Catania, Fabio Puglisi, Dario Trapani, Federico Nichetti, Lorenzo Gerratana, Alessandra Fabi, Marika Cinausero, Michela Palleschi, Agnese Losurdo, Filippo Montemurro, Enrico Cortesi, Rebecca Pedersini, Andrea Rocca, Carlo Alberto Giorgi, Grazia Arpino, Gaia Griguolo, Lorenzo Castagnoli, Giancarlo Pruneri, Andrea Michelotti, Giuseppe Curigliano, Valentina Guarneri, Francesca Ligorio, Giovanni Randon, Daniele Generali, Emanuela Ferraro, Luca Moscetti, A. Fabbri, Arta Ajazi, Elisa Agostinetto, Claudia De Angelis, Amelia Vantaggiato, Carmen G. Rea, Antonio Marra, Filippo de Braud, Nicla La Verde, Debora Basile, Lucia Del Mastro, Luigi Mariani, Salvatore Lo Vullo, Mariangela Ciccarese, Anna Moretti, Simone Scagnoli, Claudio Vernieri, Ottavia Bernocchi, Chiara Molinelli, Andrea Milani, Luca Lalli, Vernieri, C., Nichetti, F., Lalli, L., Moscetti, L., Giorgi, C. A., Griguolo, G., Marra, A., Randon, G., Rea, C. G., Ligorio, F., Scagnoli, S., De Angelis, C., Molinelli, C., Fabbri, A., Ferraro, E., Trapani, D., Milani, A., Agostinetto, E., Bernocchi, O., Catania, G., Vantaggiato, A., Palleschi, M., Moretti, A., Basile, D., Cinausero, M., Ajazi, A., Castagnoli, L., Vullo, S. L., Gerratana, L., Puglisi, F., La Verde, N., Arpino, G., Rocca, A., Ciccarese, M., Pedersini, R., Fabi, A., Generali, D., Losurdo, A., Montemurro, F., Curigliano, G., Del Mastro, L., Michelotti, A., Cortesi, E., Guarneri, V., Pruneri, G., Mariani, L., De Braud, F., Vernieri, Claudio, Nichetti, Federico, Lalli, Luca, Moscetti, Luca, Giorgi, Carlo Alberto, Griguolo, Gaia, Marra, Antonio, Randon, Giovanni, Rea, Carmen G, Ligorio, Francesca, Scagnoli, Simone, De Angelis, Claudia, Molinelli, Chiara, Fabbri, Agnese, Ferraro, Emanuela, Trapani, Dario, Milani, Andrea, Agostinetto, Elisa, Bernocchi, Ottavia, Catania, Giovanna, Vantaggiato, Amelia, Palleschi, Michela, Moretti, Anna, Basile, Debora, Cinausero, Marika, Ajazi, Arta, Castagnoli, Lorenzo, Lo Vullo, Salvatore, Gerratana, Lorenzo, Puglisi, Fabio, La Verde, Nicla, Arpino, Grazia, Rocca, Andrea, Ciccarese, Mariangela, Pedersini, Rebecca, Fabi, Alessandra, Generali, Daniele, Losurdo, Agnese, Montemurro, Filippo, Curigliano, Giuseppe, Del Mastro, Lucia, Michelotti, Andrea, Cortesi, Enrico, Guarneri, Valentina, Pruneri, Giancarlo, Mariani, Luigi, and de Braud, Filippo
- Subjects
0301 basic medicine ,Oncology ,Blood Glucose ,Cancer Research ,Receptor, ErbB-2 ,medicine.medical_treatment ,metabolism everolimus ,chemistry.chemical_compound ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,ErbB-2 ,Exemestane ,Retrospective Studie ,Antineoplastic Combined Chemotherapy Protocols ,Receptors ,Hyperinsulinemia ,drug therapy breast neoplasms ,Receptors, Estrogen ,Hormone receptor ,030220 oncology & carcinogenesis ,Female ,Breast Neoplasm ,medicine.drug ,Receptor ,Human ,medicine.medical_specialty ,medicine.drug_class ,Breast Neoplasms ,genetics breast neoplasms ,03 medical and health sciences ,Diabetes mellitus ,Internal medicine ,medicine ,Humans ,Everolimus ,Retrospective Studies ,Aromatase inhibitor ,Antineoplastic Combined Chemotherapy Protocol ,blood glucose breast neoplasms ,Androstadiene ,business.industry ,Insulin ,Cancer ,medicine.disease ,Estrogen ,Androstadienes ,030104 developmental biology ,chemistry ,Phosphatidylinositol 3-Kinase ,business - Abstract
Purpose: The mTOR complex C1 (mTORC1) inhibitor everolimus in combination with the aromatase inhibitor exemestane is an effective treatment for patients with hormone receptor—positive (HR+), HER2-negative (HER2−), advanced breast cancer (HR+/HER2− aBC). However, everolimus can cause hyperglycemia and hyperinsulinemia, which could reactivate the PI3K/protein kinase B (AKT)/mTORC1 pathway and induce tumor resistance to everolimus. Experimental Design: We conducted a multicenter, retrospective, Italian study to investigate the impact of baseline and on-treatment (i.e., during first 3 months of therapy) blood glucose levels on progression-free survival (PFS) in patients with HR+/HER2− aBC treated with everolimus-exemestane. Results: We evaluated 809 patients with HR+/HER2− aBC treated with everolimus-exemestane as any line of therapy for advanced disease. When evaluated as dichotomous variables, baseline and on-treatment glycemia were not significantly associated with PFS. However, when blood glucose concentration was evaluated as a continuous variable, a multivariable model accounting for clinically relevant patient- and tumor-related variables revealed that both baseline and on-treatment glycemia are associated with PFS, and this association is largely attributable to their interaction. In particular, patients who are normoglycemic at baseline and experience on-treatment diabetes have lower PFS compared with patients who are already hyperglycemic at baseline and experience diabetes during everolimus-exemestane therapy (median PFS, 6.34 vs. 10.32 months; HR, 1.76; 95% confidence interval, 1.15–2.69; P = 0.008). Conclusions: The impact of on-treatment glycemia on the efficacy of everolimus-exemestane therapy in patients with HR+/HER2− aBC depends on baseline glycemia. This study lays the foundations for investigating novel therapeutic approaches to target the glucose/insulin axis in combination with PI3K/AKT/mTORC1 inhibitors in patients with HR+/HER2− aBC.
- Published
- 2021
59. Early changes of the standardized uptake values (SUVmax) predict the efficacy of everolimus-exemestane in patients with hormone receptor-positive metastatic breast cancer
- Author
-
Federico Nichetti, Carla Strina, Marianna Sirico, Giulia Bianchi, Maria Rosa Cappelletti, Silvia P. Corona, Valeria Cervoni, Manuela Milani, Filippo de Braud, Navid Sobhani, Giuseppina Barbieri, Daniele Generali, Martina Dester, Fabiola Giudici, Claudio Vernieri, Ottavia Bernocchi, Nicoletta Ziglioli, Sirico, M., Bernocchi, O., Sobhani, N., Giudici, F., Corona, S. P., Vernieri, C., Nichetti, F., Cappelletti, M. R., Milani, M., Strina, C., Cervoni, V., Barbieri, G., Ziglioli, N., Dester, M., Bianchi, G. V., Braud, F. D., and Generali, D.
- Subjects
Oncology ,18F-FDG PET/CT ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,∆SUV% ,lcsh:RC254-282 ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Exemestane ,Internal medicine ,medicine ,Everolimus ,Prospective cohort study ,Predictive marker ,Aromatase inhibitor ,business.industry ,F-FDG PET/CT ,Metastatic breast cancer ,Predictive biomarker ,Retrospective cohort study ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Everolimu ,chemistry ,Hormone receptor ,030220 oncology & carcinogenesis ,business ,medicine.drug - Abstract
Background: The mTORC1 inhibitor everolimus has been approved in combination with the aromatase inhibitor exemestane for the treatment of hormone receptor-positive (HR+) human epidermal growth factor receptor 2-negative (HER2&minus, ) metastatic breast cancer (HR+ mBC) progressing on prior therapy with a non-steroidal aromatase inhibitor. To date, no predictive biomarkers of tumor sensitivity/resistance for everolimus-based treatments have been identified. We hypothesized that precocious changes in the Standardized Uptake Volume (∆SUV%), as assessed by 18F-Fluorodeoxyglucosepositron-emission tomography (18F-FDG PET/CT), may be a marker of everolimus efficacy. Methods: This was a retrospective study including 31 HR+ HER2- patients treated with everolimus and exemestane in two Italian centers between 2013 and 2018. The objective of the study was to investigate ∆SUV% as a predictive marker of everolimus antitumor efficacy. 18F-FDG PET/CT scans were performed at baseline and after three months of treatment. Patients were defined as long responders (LRs) if disease progression occurred at least 10 months after treatment initiation and long survivors (LSs) if death occurred later than 36 months after starting therapy. ROC analysis was used to determine the optimal cut-off values of ∆SUV% to distinguish LRs from non-LRs and LSs from non-LSs. Progression-free survival (PFS) and overall survival (OS) were estimated by Kaplan&ndash, Meier method. Results: The SUVmax values decreased significantly from baseline to 3 months after therapy (p = 0.003). Dynamic changes of SUVmax (Delta SUV) had a higher accuracy in discriminating long-responders from non-long-responders (AUC = 0.67, Delta SUV cut-off = 28.8%) respects to its ability to identify long survivors from no-long survivors (AUC = 0.60, Delta SUV cut-off = 53.8%). Patients were divided into groups according to the Delta SUV cut-offs and survival outcomes were evaluated: patients with a decrease of ∆SUV% &ge, 28.8% had significantly better PFS (10 months-PFS: 63.2%, 95% CI: 37.9&ndash, 80.4% and 16.7%, 95% CI: 2.7&ndash, 41.3% respectively, p = 0.005). As regard as OS, patients with ∆SUV% &ge, 53.8% had longer OS when compared to patients with ∆SUV% <, 53.8% (36 month-OS: 82.5% vs. 45.9% vs. p = 0.048). Conclusion: We found two precocious ∆SUV% thresholds capable of identifying HR+ HER2-mBC patients, which would achieve long-term benefit or long-term survival during everolimus-exemestane therapy. These results warrant further validation in prospective studies and should be integrated with molecular biomarkers related to tumor metabolism and mTORC1 signaling.
- Published
- 2020
60. Thrombin generation predicts early recurrence in breast cancer patients
- Author
-
Marina Marchetti, Cinzia Giaccherini, Giovanna Masci, Cristina Verzeroli, Laura Russo, Luigi Celio, Roberta Sarmiento, Sara Gamba, Carmen J. Tartari, Erika Diani, Alfonso Vignoli, Paolo Malighetti, Daniele Spinelli, Nicole M. Kuderer, Federico Nichetti, Mauro Minelli, Carlo Tondini, Sandro Barni, Francesco Giuliani, Fausto Petrelli, Andrea D’Alessio, Giampietro Gasparini, Roberto Labianca, Armando Santoro, Filippo De Braud, Anna Falanga, Francesca Schieppati, Antonia Martinetti, Elisabetta Gennaro, Mara Ghilardi, Marchetti, M, Giaccherini, C, Masci, G, Verzeroli, C, Russo, L, Celio, L, Sarmiento, R, Gamba, S, Tartari, C, Diani, E, Vignoli, A, Malighetti, P, Spinelli, D, Kuderer, N, Nichetti, F, Tondini, C, Barni, S, Giuliani, F, Petrelli, F, D'Alessio, A, Gasparini, G, Labianca, R, Santoro, A, De Braud, F, Falanga, A, and Hypercan, I
- Subjects
Oncology ,medicine.medical_specialty ,Multivariate analysis ,medicine.medical_treatment ,Breast Neoplasms ,030204 cardiovascular system & hematology ,disease recurrence ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,risk model ,Internal medicine ,medicine ,Humans ,Derivation ,Prospective Studies ,Mastectomy ,Proportional hazards model ,business.industry ,breast cancer ,hypercoagulability ,thrombin generation ,Thrombin ,Cancer ,Hematology ,Settore ING-IND/35 - Ingegneria Economico-Gestionale ,medicine.disease ,Prognosis ,Cohort ,Neoplasm Recurrence, Local ,business ,Risk assessment - Abstract
Background: Cancer patients present with a hypercoagulable state often associated with poor disease prognosis. Objectives: This study aims to evaluate whether thrombin generation (TG), a global coagulation test, may be a useful tool to improve the identification of patients at high risk of early disease recurrence (i.e. E-DR within 2 years) after breast cancer surgery. Patients/methods: A cohort of 522 newly diagnosed patients with surgically resected high-risk breast cancer were enrolled in the ongoing prospective HYPERCAN study. TG potential was measured in plasma samples collected before starting systemic chemotherapy. Significant predictive hemostatic and clinic-pathological parameters were identified in the derivation cohort by Cox-regression analysis. A risk prognostic score for E-DR was generated in the derivation and tested in the validation cohort. Results: After a median observation period of 3.4 years, DR occurred in 51 patients, 28 of whom were E-DR. E-DR subjects presented with the highest TG values as compared to both late-DR (from 2 to 5 years) and no relapse subjects (p
- Published
- 2019
61. The Prostaglandin EP4 Antagonist Vorbipiprant Combined with PD-1 Blockade for Refractory Microsatellite-Stable Metastatic Colorectal Cancer: A Phase Ib/IIa Trial.
- Author
-
Pietrantonio F, Morano F, Niger M, Ghelardi F, Chiodoni C, Palazzo M, Nichetti F, Manca P, Cristarella E, Doldi V, Zaffaroni N, Sabella G, Brambilla N, Benincasa E, Giacovelli G, Vitalini C, Girolami F, and Rovati LC
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Adult, Microsatellite Instability, Prospective Studies, Drug Resistance, Neoplasm genetics, Neoplasm Metastasis, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Receptors, Prostaglandin E, EP4 Subtype antagonists & inhibitors, Receptors, Prostaglandin E, EP4 Subtype genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Immune Checkpoint Inhibitors therapeutic use, Immune Checkpoint Inhibitors administration & dosage, Immune Checkpoint Inhibitors adverse effects, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Abstract
Purpose: Novel combinations are required to overcome resistance to immune checkpoint inhibitors in proficient mismatch repair (pMMR) or microsatellite-stable (MSS) metastatic colorectal cancer (mCRC). We aimed to determine whether vorbipiprant, a prostaglandin E2 receptor EP4 subtype antagonist, can convert immune-resistant mCRC into a tumor responsive to anti-PD-1 inhibition., Patients and Methods: This phase Ib/IIa prospective, open-label, single-arm trial followed a 3 + 3 dose-escalation and dose-optimization design. A total of 28 patients with chemorefractory pMMR/MSS mCRC were given dose-escalated oral vorbipiprant (30, 90, or 180 mg twice daily), along with biweekly intravenous balstilimab (3 mg/kg), an anti-PD-1 antibody. The primary endpoints included safety and the disease control rate (DCR). Secondary endpoints were the overall response rate, duration of response, progression-free survival, and overall survival., Results: No dose-limiting toxicities were observed. Of the 28 patients, seven (25%) experienced serious adverse events, but only one was attributed to vorbipiprant and one to balstilimab. The trial achieved a DCR of 50% observed across the entire cohort. In the subgroup of patients with liver metastases (n = 12), the DCR was 25%. The overall response rate was 11%, with three patients showing a partial response (median duration of response, 7.4 months). The median progression-free survival was 2.6 months, and the median overall survival was 14.2 months. Translational exploratory analyses suggested that vorbipiprant may boost response to anti-PD-1 in patients with immunogenic tumors., Conclusions: The combination of vorbipiprant and a PD-1 inhibitor (balstilimab) yielded sufficient activity in refractory pMMR/MSS mCRC, which is worthy of confirmation in future clinical trials in biomarker-enriched populations., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2025
- Full Text
- View/download PDF
62. Early downmodulation of tumor glycolysis predicts response to fasting-mimicking diet in triple-negative breast cancer patients.
- Author
-
Ligorio F, Vingiani A, Torelli T, Sposetti C, Drufuca L, Iannelli F, Zanenga L, Depretto C, Folli S, Scaperrotta G, Capri G, Bianchi GV, Ferraris C, Martelli G, Maugeri I, Provenzano L, Nichetti F, Agnelli L, Lobefaro R, Fucà G, Fotia G, Mariani L, Morelli D, Ladisa V, De Santis MC, Lozza L, Trecate G, Belfiore A, Brich S, Bertolotti A, Lorenzini D, Ficchì A, Martinetti A, Sottotetti E, Arata A, Corsetto P, Sorrentino L, Rediti M, Salvadori G, Minucci S, Foiani M, Apolone G, Pagani M, Pruneri G, de Braud F, and Vernieri C
- Subjects
- Humans, Female, Middle Aged, Down-Regulation drug effects, Caloric Restriction, Adult, Diet, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms diet therapy, Triple Negative Breast Neoplasms pathology, Glycolysis, Fasting
- Abstract
In preclinical experiments, cyclic fasting-mimicking diets (FMDs) showed broad anticancer effects in combination with chemotherapy. Among different tumor types, triple-negative breast cancer (TNBC) is exquisitely sensitive to FMD. However, the antitumor activity and efficacy of cyclic FMD in TNBC patients remain unclear. Here, we show that a severely calorie-restricted, triweekly, 5-day FMD regimen results in excellent pathologic complete response (pCR) rates (primary endpoint) and long-term clinical outcomes (secondary endpoints) when combined with preoperative chemotherapy in 30 patients with early-stage TNBC enrolled in the phase 2 trial BREAKFAST. Bulk and single-cell RNA sequencing analysis revealed that highly glycolytic cancer cells, myeloid cells, and pericytes from tumors achieving pCR undergo a significant, early downmodulation of pathways related to glycolysis and pyruvate metabolism. Our findings pave the wave for conducting larger clinical trials to investigate the efficacy of cyclic FMD in early-stage TNBC patients and to validate early changes of intratumor glycolysis as a predictor of clinical benefit from nutrient restriction. This study was registered at Clinicaltrials.gov (NCT04248998)., Competing Interests: Declaration of interests The authors declare the following competing interests: G.V.B., speaker/advisory board: Novartis, Eli Lilly, Daichi/Astrazeneca, Roche, MSD, and Seagen; G.P., consulting fees: Roche, Bayer, and Astra Zeneca; C.V., consulting fees/advisory board: Novartis, Pfizer, Eli Lilly, Daiichi Sankyo, and Menarini Stemline; C.V., honoraria as speaker: Novartis, Eli Lilly, Istituto Gentili, Accademia Nazionale di Medicina, MSD, Research grant: Roche; F.d.B., consulting fees/advisory board: Roche, EMD Serono, NMS Nerviano Medical Science, Sanofi, MSD, Novartis, Incyte, BMS, Menarini, Astra Zeneca, Pierre Fabre, Mattioli 1885, MCCann Health, Taiho; F.d.B., IQVIA Speaker Bureau: BMS, Healthcare Research & Pharmacoepidemiology, Merck Group, MSD, Pfizer, Servier, Sanofi, Roche, AMGEN, Incyte, Dephaforum, Seagen, Nadirex, BMS, Ambrosetti, and Itanet; and F.d.B., research grant/funding: Novartis, F.Hoffmann-LaRoche Ltd, BMS, Ignyta Inc., Merck Sharp & Dohme Spa, Kymab, Pfizer, Tesaro, MSD, MedImmune LCC, Exelixis Inc., LOXO Oncology Inc., DAICHI SANKIO Dev. Ltd, Basilea Pharmaceutica International AG, Janssen-Cilag International NV, and Merck KGAA. C.V. and F.d.B. are co-inventors of the FMD regimen used in this study. The Italian patent (no. 102019000009954) has been released and was first deposited on June 24(th), 2019, with an extension granted under PCT WO 2020/261131 on June 24(th), 2020. The European patent (no. 207403399), Canadian patent (no. 2144217), and USA patent are pending., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
63. Is tumour sequencing effective for the identification of germline BRCA1/2 pathogenic variant carriers?
- Author
-
Azzollini J, Capone I, Duca M, Vingiani A, Piccolo A, Agnelli L, Tamborini E, Perrone F, Peissel B, Lorenzini D, Damian S, Vernieri C, Bianchi GV, Mantiero M, Ducceschi M, Polignano M, Niger M, Nichetti F, Proto C, Brambilla M, Colombo E, Stellato M, Conca E, Busico A, and Manoukian S
- Abstract
Introduction: Tumour BRCA1 /2 sequencing has progressively increased along with the expanding indications for poly(ADP-ribose) polymerase inhibitors. In our study, we investigated the feasibility and outcomes of a workflow for the identification of germline carriers based on tumour sequencing results., Methods: Between April 2020 and December 2022, BRCA1/2 tumour testing results from 2020 patients were reviewed. Analysed tumours included: 323 ovarian, 104 breast, 314 pancreas-biliary, 87 prostate, 374 gastrointestinal, 309 lung, and 509 less common histologies. Testing was performed through small (only BRCA1/2 , 16%) or comprehensive (>50 genes) next-generation sequencing panels (84%). Patients with pathogenic/likely pathogenic variants were referred for genetic counselling and germline testing., Results: Tumour BRCA1/2 pathogenic variants were identified in 145 patients (7%). The pathogenic variant frequency ranged between 23% (75/323 ovarian) and 3.5% (11/314 pancreas-biliary). The highest frequency was observed in high-grade ovarian carcinomas (27%, 64/235). By 30 June 2023, 79 out of 145 patients (54%) underwent subsequent genetic counselling and germline testing. In these patients, mostly affected with ovarian carcinoma (67%, 53/79), 48 were confirmed germline pathogenic variants (61%)., Conclusions: In our tumour-to-germline testing approach, we observed the BRCA1/2 pathogenic variant frequency reported in other large unselected ovarian cancer cohorts, thus confirming its effectiveness in identifying putative germline carriers irrespective of eligibility for germline testing. As the range of tumours subjected to genetic testing broadens, this approach is expected to also be effective in other tumour settings for enhancing the identification of carriers, reducing the burden on genetic services, and avoiding unnecessary concerns related to germline testing., Competing Interests: Declaration of conflicting interestThe author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: SD: research funding paid directly to my Institution from Roche, Incyte, Nerviano Medical Science srl, Pfizer Inc. MN: Travel expenses from AstraZeneca, speaker honorarium from Accademia della Medicina and Incyte; honoraria from Sandoz, Medpoint SRL, Incyte, AstraZeneca and Servier for editorial collaboration. Consultant honoraria from EMD Serono, Basilea Pharmaceutica, Incyte, MSD Italia, Servier, Astrazeneca and Taiho. FN: honoraria from AstraZeneca. LA: consultant honorarium from Roche Italia. All the other authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
64. Molecular Characterization and Clinical Relevance of MGMT-Silenced Pancreatic Cancer.
- Author
-
Nichetti F, Silvestri M, Agnelli L, Franza A, Pircher C, Rota S, Ambrosini P, Fotia G, Hüllein J, Randon G, Lajer P, Perrone F, Tamborini E, Leoncini G, Coppa J, Busset MDD, Pusceddu S, Milione M, Morano F, Pietrantonio F, Pruneri G, Mazzaferro V, Lipka DB, Köhler BC, Hübschmann D, Fröhling S, de Braud F, and Niger M
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Prognosis, Biomarkers, Tumor genetics, Clinical Relevance, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Pancreatic Neoplasms mortality, Tumor Suppressor Proteins genetics, DNA Modification Methylases genetics, DNA Modification Methylases metabolism, DNA Repair Enzymes genetics, DNA Methylation, Gene Silencing
- Abstract
Background: The identification of actionable molecular targets of pancreatic cancer (PAC) is key to improving patient outcomes. We hypothesized O6-methylguanine-DNA methyltransferase (MGMT) silencing may occur in a subset of PAC tumors, with unexplored clinical and molecular correlates., Experimental Design: We leveraged sequencing data from The Cancer Genome Atlas (TCGA), the Clinical Proteomic Tumor Analysis Consortium 3 (CPTAC-3), and the (Australian Pancreatic Cancer Genome Initiative) PACA-AU cohorts to characterize MGMT-silenced PAC. Genomic, transcriptomic, methylation, and clinical data were investigated, and findings were validated in silico using methylation, transcriptomic and drug sensitivity data from Cancer Cell Line Encyclopedia (CCLE) project, and in a real-world cohort of PAC patients profiled for MGMT status at Istituto Nazionale Tumori of Milan (INT)., Results: On the basis of Human Methylation 450k data, MGMT silencing was identified in ~6% of PAC cases and was enriched in tumors with non-ductal histology, with a trend toward longer overall survival. MGMT-silenced tumors were associated with a lower frequency of KRAS mutations and showed features of immune exclusion. In the INT cohort, MGMT-silencing was confirmed in ~7% of cases and prevalent in KRAS wild type tumors, with a favorable prognostic impact. In silico analysis suggested a higher sensitivity to alkylating and DNA damaging agents in MGMT-silenced PAC cell lines., Conclusions: MGMT silencing occurs in a small subgroup of PACs and is enriched in KRAS wild type cases, with a favorable prognostic impact. Our findings provide the rationale to explore combinations of alkylating with DNA damaging agents in MGMT-silenced PAC., (© 2024 The Author(s). Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
65. Lymphotoxin beta-activated LTBR/NIK/RELB axis drives proliferation in cholangiocarcinoma.
- Author
-
Xu K, Kessler A, Nichetti F, Hoffmeister-Wittmann P, Scherr AL, Nader L, Kelmendi E, Schmitt N, Schwab M, García-Beccaria M, Sobol B, Nieto OA, Isele H, Gärtner U, Vaquero-Siguero N, Volk J, Korell F, Mock A, Heide D, Ramadori P, Lenoir B, Albrecht T, Hüllein J, Jäger D, Fröhling S, Springfeld C, Jackstadt R, Heikenwälder M, Dill MT, Roessler S, Goeppert B, and Köhler BC
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, NF-kappa B metabolism, Cholangiocarcinoma pathology, Cholangiocarcinoma metabolism, Cholangiocarcinoma genetics, Cholangiocarcinoma drug therapy, Lymphotoxin beta Receptor metabolism, Lymphotoxin beta Receptor genetics, Bile Duct Neoplasms pathology, Bile Duct Neoplasms metabolism, Bile Duct Neoplasms genetics, Bile Duct Neoplasms drug therapy, Transcription Factor RelB metabolism, Transcription Factor RelB genetics, Cell Proliferation drug effects, NF-kappaB-Inducing Kinase, Signal Transduction, Lymphotoxin-beta metabolism, Lymphotoxin-beta genetics, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases genetics
- Abstract
Background and Aims: Cholangiocarcinoma (CCA) is an aggressive malignancy arising from the intrahepatic (iCCA) or extrahepatic (eCCA) bile ducts with poor prognosis and limited treatment options. Prior evidence highlighted a significant contribution of the non-canonical NF-κB signalling pathway in initiation and aggressiveness of different tumour types. Lymphotoxin-β (LTβ) stimulates the NF-κB-inducing kinase (NIK), resulting in the activation of the transcription factor RelB. However, the functional contribution of the non-canonical NF-κB signalling pathway via the LTβ/NIK/RelB axis in CCA carcinogenesis and progression has not been established., Methods: Human CCA-derived cell lines and organoids were examined to determine the expression of NF-κB pathway components upon activation or inhibition. Proliferation and cell death were analysed using real-time impedance measurement and flow cytometry. Immunoblot, qRT-PCR, RNA sequencing and in situ hybridization were employed to analyse gene and protein expression. Four in vivo models of iCCA were used to probe the activation and regulation of the non-canonical NF-κB pathway., Results: Exposure to LTα1/β2 activates the LTβ/NIK/RelB axis and promotes proliferation in CCA. Inhibition of NIK with the small molecule inhibitor B022 efficiently suppresses RelB expression in patient-derived CCA organoids and nuclear co-translocation of RelB and p52 stimulated by LTα1/β2 in CCA cell lines. In murine CCA, RelB expression is significantly increased and LTβ is the predominant ligand of the non-canonical NF-κB signalling pathway., Conclusions: Our study confirms that the non-canonical NF-κB axis LTβ/NIK/RelB drives cholangiocarcinogenesis and represents a candidate therapeutic target., (© 2024 The Author(s). Liver International published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
66. Durvalumab plus gemcitabine and cisplatin in advanced biliary tract cancer: A large real-life worldwide population.
- Author
-
Rimini M, Fornaro L, Rizzato MD, Antonuzzo L, Rossari F, Satake T, Vandeputte H, Vivaldi C, Pressiani T, Lucchetti J, Kim JW, Abidoye O, Rapposelli IG, Tamberi S, Finkelmeier F, Giordano G, Nichetti F, Chon HJ, Braconi C, Pirrone C, Castet F, Tamburini E, Yoo C, Parisi A, Diana A, Scartozzi M, Prager GW, Avallone A, Schirripa M, Kim IH, Perkhofer L, Oneda E, Verrico M, Adeva J, Chan SL, Spinelli GP, Personeni N, Garajova I, Rodriquenz MG, Leo S, Salani F, De Rosa A, Lavacchi D, Foti S, Ikeda M, Dekervel J, Niger M, Balsano R, Tonini G, Kang M, Bekaii-Saab T, Esposito L, Boccaccino A, Himmelsbach V, Landriscina M, Djaballah SA, Zanuso V, Masi G, Lonardi S, Rimassa L, and Casadei-Gardini A
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Retrospective Studies, Adult, Aged, 80 and over, Gemcitabine, Cisplatin administration & dosage, Cisplatin therapeutic use, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms pathology, Biliary Tract Neoplasms mortality, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Deoxycytidine analogs & derivatives, Deoxycytidine administration & dosage, Deoxycytidine adverse effects, Deoxycytidine therapeutic use, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal therapeutic use
- Abstract
Background: The TOPAZ-1 phase III trial showed a survival benefit with durvalumab plus gemcitabine and cisplatin in patients with advanced biliary tract cancer (BTC). To understand this combination's real-world efficacy and tolerability, we conducted a global multicenter retrospective analysis of its first-line treatment outcomes., Methods: We included patients with unresectable, locally advanced, or metastatic BTC treated with durvalumab, gemcitabine, and cisplatin at 39 sites in 11 countries (Europe, the United States, and Asia). The primary endpoint was overall survival (OS)., Results: 666 patients were enrolled. Median OS was 15.1 months and median PFS was 8.2 months. The investigator-assessed overall response rate was 32.7 %, with stable disease in 45.2 % of patients. High baseline CEA levels, ECOG PS > 0, metastatic disease, and NLR > 3 were associated with poor survival. Any grade adverse events (AEs) occurred in 92.9 % of patients (grade >2: 46.6 %). Immune-related AEs (irAEs) occurred in 20.0 % (grade >2: 2.5 %). Three deaths (0.5 %) were deemed treatment-related, none linked to immunotherapy. Common irAEs were rash (8.2 % all grades; 0.3 % grade >2), itching (10.3 % all grades; 0.2 % grade >2), and hypothyroidism (5.1 % all grades; 0.3 % grade >2). Durvalumab discontinuation rate due to AEs was 1.5 %. ESMO-recommended genes were analyzed and no outcome differences were found. A comparative analysis with a historical cohort of patients treated with chemotherapy alone confirmed the positive survival impact of durvalumab in combination with cisplatin/gemcitabine., Conclusion: This first global real-world analysis largely confirmed the TOPAZ-1 findings, supporting gemcitabine, cisplatin, and durvalumab as a first-line standard of care for patients with advanced BTC., Competing Interests: Declaration of Competing Interest LR reports consulting fees from AbbVie, AstraZeneca, Basilea, Bayer, Elevar Therapeutics, Exelixis, Genenta, Hengrui, Incyte, Ipsen, IQVIA, Jazz Pharmaceuticals, MSD, Nerviano Medical Sciences, Roche, Servier, Taiho Oncology, Zymeworks; lecture fees from AstraZeneca, Bayer, BMS, Incyte, Ipsen, Roche, Servier; travel expenses from AstraZeneca; research grants (to Institution) from Agios, AstraZeneca, BeiGene, Eisai, Exelixis, Fibrogen, Incyte, Ipsen, Lilly, MSD, Nerviano Medical Sciences, Roche, Servier, Zymeworks. ACG reports consulting fees from AstraZeneca, Bayer, BMS, Eisai, Incyte, Ipsen, IQVIA, MSD, Roche, Servier; lecture fees from AstraZeneca, Bayer, BMS, Eisai, Incyte, Ipsen, Roche, Servier; travel expenses from AstraZeneca; research grants (to Institution) from AstraZeneca, Eisai. SLC serves an advisory member for AstraZeneca, MSD, Eisai, BMS, Ipsen, and Hengrui, received research funds from MSD, Eisai, Ipsen, SIRTEX, and Zailab, and honoraria from AstraZeneca, Eisai, Roche, Ipsen, and MSD. TP received consulting fees from Bayer, Ipsen, and AstraZeneca; institutional research funding from Roche, Bayer, and AstraZeneca; travel expenses from Roche. CB received honoraria as speaker (Astrazeneca, Incyte, Servier) and consultant (Incyte, Servier, Boehringer Ingelheim, Astrazeneca), received research funds (Avacta, Medannex, Servier) and her spouse is an employee of Astrazeneca. M. Ikeda reports honoraria from AstraZeneca, Chugai Pharma, Eisai, Incyte, Lilly Japan, MSD, Novartis, Ono Pharmaceutical, Takeda, Teijin Pharma, Nihon Servier, Taiho and research funding from AstraZeneca, Bayer, Bristol-Myers Squibb, Chiome Bioscience, Chugai, Eisai, Eli Lilly Japan, Delta-Fly Pharma, Invitae, J-Pharma, Merck biopharma, Merus N.V., MSD, Novartis, Nihon Servier, Ono, Syneos Health, and Rakuten Medical. GWP: Advisories and/or Speaker fees: Servier, Bayer, Roche Amgen, Merck, MSD, BMS, Sanofi, Lilly, Astra Zeneca, Astellas, Pierre-Fabre, Incyte, Arcus, CECOG. F. F. has received travel support from Ipsen, Abbvie, Astrazeneca and speaker’s fees from AbbVie, MSD, Ipsen, Astrazeneca. LP: Advisory role: AstraZeneca, Servier, Travel expenses: AstraZeneca, Ipsen. GG: Consulting Fees: Astra Zeneca, MSD, Servier, Seagen, Bayer, Amgen, Novartis, Ipsen, BMS. Travel Expenses: Astra Zeneca, Servier, Bayer, Novartis. S.L. reports research funding (to Institution) from Amgen, Astellas, Astra Zeneca, Bayer, Bristol-Myers Squibb, Daichii Sankyo, Hutchinson, Incyte, Merck Serono, Mirati, MSD, Pfizer, Roche, Servier; personal honoraria as invited speaker from Amgen, Astra Zeneca, Bristol-Myers Squibb, Incyte, GSK, Lilly, Merck Serono, MSD, Pierre-Fabre, Roche, Servier; participation in advisory board for Amgen, Astellas, Astra Zeneca, Bayer, Bristol-Myers Squibb, Daiichi-Sankyo, GSK, Incyte, Lilly, Merck Serono, MSD, Servier, Takeda, Rottapharm. JD received consulting fees and/or speaker honoraria from Amgen, AstraZeneca, Bayer, BMS, Eisai, Need Inc., Ipsen, Lilly, MediMix, Merck, MSD, Novartis, Roche and Servier. All remaining authors have declared no conflicts of interest., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
67. Etiology-independent activation of the LTβ-LTβR-RELB axis drives aggressiveness and predicts poor prognosis in HCC.
- Author
-
Scherr AL, Nader L, Xu K, Elssner C, Ridder DA, Nichetti F, Mastel M, Fritzsche S, Kelmendi E, Schmitt N, Hoffmeister-Wittmann P, Weiler SME, Korell F, Albrecht T, Schwab M, Isele H, Kessler A, Hüllein J, Seretny A, Ye L, Urbanik T, Welte S, Leblond AL, Heilig CE, Rahbari M, Ali A, Gallage S, Lenoir B, Wilhelm N, Gärtner U, Ogrodnik SJ, Springfeld C, Tschaharganeh D, Fröhling S, Longerich T, Schulze-Bergkamen H, Jäger D, Brandl L, Schirmacher P, Straub BK, Weber A, De Toni EN, Goeppert B, Heikenwalder M, Jackstadt R, Roessler S, Breuhahn K, and Köhler BC
- Subjects
- Animals, Humans, Mice, Prognosis, Transcription Factor RelB metabolism, Transcription Factor RelB genetics, Male, Signal Transduction, Female, Lymphotoxin-beta metabolism, Lymphotoxin-beta genetics, Middle Aged, Liver Neoplasms pathology, Liver Neoplasms metabolism, Liver Neoplasms genetics, Carcinoma, Hepatocellular pathology, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular mortality, Carcinoma, Hepatocellular genetics, Lymphotoxin beta Receptor metabolism, Lymphotoxin beta Receptor genetics
- Abstract
Background and Aims: HCC is the most common primary liver tumor, with an increasing incidence worldwide. HCC is a heterogeneous malignancy and usually develops in a chronically injured liver. The NF-κB signaling network consists of a canonical and a noncanonical branch. Activation of canonical NF-κB in HCC is documented. However, a functional and clinically relevant role of noncanonical NF-κB and its downstream effectors is not established., Approach and Results: Four human HCC cohorts (total n = 1462) and 4 mouse HCC models were assessed for expression and localization of NF-κB signaling components and activating ligands. In vitro , NF-κB signaling, proliferation, and cell death were measured, proving a pro-proliferative role of v-rel avian reticuloendotheliosis viral oncogene homolog B (RELB) activated by means of NF-κB-inducing kinase. In vivo , lymphotoxin beta was identified as the predominant inducer of RELB activation. Importantly, hepatocyte-specific RELB knockout in a murine HCC model led to a lower incidence compared to controls and lower maximal tumor diameters. In silico , RELB activity and RELB-directed transcriptomics were validated on the The Cancer Genome Atlas HCC cohort using inferred protein activity and Gene Set Enrichment Analysis. In RELB-active HCC, pathways mediating proliferation were significantly activated. In contrast to v-rel avian reticuloendotheliosis viral oncogene homolog A, nuclear enrichment of noncanonical RELB expression identified patients with a poor prognosis in an etiology-independent manner. Moreover, RELB activation was associated with malignant features metastasis and recurrence., Conclusions: This study demonstrates a prognostically relevant, etiology-independent, and cross-species consistent activation of a lymphotoxin beta/LTβR/RELB axis in hepatocarcinogenesis. These observations may harbor broad implications for HCC, including possible clinical exploitation., (Copyright © 2023 American Association for the Study of Liver Diseases.)
- Published
- 2024
- Full Text
- View/download PDF
68. Correction to: A phase II/III randomized clinical trial of CisPlatin plUs Gemcitabine and Nabpaclitaxel (GAP) as pReoperative chemotherapy versus immediate resection in patIents with resecTable BiliarY tract cancers (BTC) at high risk for recurrence: PURITY study.
- Author
-
Niger M, Nichetti F, Fornaro L, Pircher C, Morano F, Palermo F, Rimassa L, Pressiani T, Berardi R, Gardini AC, Sperti E, Salvatore L, Melisi D, Bergamo F, Siena S, Mosconi S, Longarini R, Arcangeli G, Corallo S, Delliponti L, Tamberi S, Fea E, Brandi G, Rapposelli IG, Salati M, Baili P, Miceli R, Ljevar S, Cavallo I, Sottotetti E, Martinetti A, Busset MDD, Sposito C, Di Bartolomeo M, Pietrantonio F, de Braud F, and Mazzaferro V
- Published
- 2024
- Full Text
- View/download PDF
69. Prognostic and predictive impact of sex in locally advanced microsatellite instability high gastric or gastroesophageal junction cancer: An individual patient data pooled analysis of randomized clinical trials.
- Author
-
Raimondi A, Kim YW, Kang WK, Langley RE, Choi YY, Kim KM, Nankivell MG, Randon G, Kook MC, An JY, Grabsch HI, Prisciandaro M, Nichetti F, Noh SH, Sohn TS, Kim S, Wotherspoon A, Morano F, Cunningham D, Lee J, Cheong JH, Smyth EC, and Pietrantonio F
- Subjects
- Humans, Male, Female, Prognosis, Sex Factors, Esophageal Neoplasms genetics, Esophageal Neoplasms pathology, Esophageal Neoplasms mortality, Esophageal Neoplasms therapy, Esophageal Neoplasms drug therapy, Middle Aged, Aged, Chemotherapy, Adjuvant, Microsatellite Instability, Esophagogastric Junction pathology, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Stomach Neoplasms therapy, Stomach Neoplasms drug therapy, Stomach Neoplasms mortality, Randomized Controlled Trials as Topic
- Abstract
Background: Surgery plus peri-operative/adjuvant chemotherapy is the standard of care for locally advanced GC/GEJC, though with unsatisfactory results. dMMR/MSI-high tumors have better prognosis and scant benefit from chemotherapy as compared to pMMR/MSS ones. The differential outcome of therapies in terms of safety and efficacy according to sex is still debated in GC/GEJC patients., Methods: We previously performed an individual patient data pooled analysis of MAGIC, CLASSIC, ITACA-S, and ARTIST trials including GC/GEJC patients treated with surgery alone or surgery plus peri-operative/adjuvant chemotherapy to assess the value of MSI status. We performed a secondary analysis investigating the prognostic and predictive role of sex (female versus male) in the pooled analysis dataset in the overall population and patients stratified for MSI status (MSI-high versus MSS/MSI-low). Disease-free (DFS) and overall survival (OS) were calculated., Results: Patients with MSI-high tumors had improved survival as compared to MSS/MSI-low ones irrespective of sex, whereas in those with MSS/MSI-low tumors, females had numerically longer OS and DFS (5-year OS was 63.2% versus 57.6%, HR 0.842; p = 0.058, and 5-year DFS was 55.8% versus 50.8%, HR 0.850; p = 0.0504 in female versus male patients). The numerical difference for the detrimental effect of chemotherapy in MSI-high GC was higher in females than males, while the significant benefit of chemotherapy over surgery alone was confirmed in MSS/MSI-low GC irrespective of sex., Conclusions: This pooled analysis including four randomized trials highlights a relevant impact of sex in the prognosis and treatment efficacy of MSI-high and MSS/MSI-low non-metastatic GC/GEJC., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Filippo Pietrantonio reported receiving research grants from BMS, Incyte, Agenus, Amgen, Lilly and AstraZeneca, and personal fees from BMS, MSD, Amgen, Merck-Serono, Pierre-Fabre, Servier, Bayer, Takeda, Astellas, Johnson&Johnson, Rottapharm, Ipsen, AstraZeneca, GSK, Daiichi-Sankyo outside the submitted work. Alessandra Raimondi reported honoraria for advisory boards outside the submitted work from Elma Academy and Servier., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
70. Durvalumab Plus Gemcitabine and Cisplatin Versus Gemcitabine and Cisplatin in Biliary Tract Cancer: a Real-World Retrospective, Multicenter Study.
- Author
-
Rimini M, Masi G, Lonardi S, Nichetti F, Pressiani T, Lavacchi D, Jessica L, Giordano G, Scartozzi M, Tamburini E, Pastorino A, Rapposelli IG, Daniele B, Martinelli E, Garajova I, Aprile G, Schirripa M, Formica V, Salani F, Winchler C, Bergamo F, Balsano R, Gusmaroli E, Lorenzo A, Landriscina M, Pretta A, Toma I, Pirrone C, Diana A, Leone F, Brunetti O, Brandi G, Garattini SK, Satolli MA, Rossari F, Fornaro L, Niger M, Zanuso V, De Rosa A, Ratti F, Aldrighetti L, De Braud F, Foti S, Rizzato MD, Vivaldi C, Stefano C, Rimassa L, Antonuzzo L, and Casadei-Gardini A
- Subjects
- Humans, Male, Female, Retrospective Studies, Aged, Middle Aged, Adult, Aged, 80 and over, Gemcitabine, Cisplatin therapeutic use, Cisplatin pharmacology, Cisplatin administration & dosage, Deoxycytidine analogs & derivatives, Deoxycytidine therapeutic use, Deoxycytidine pharmacology, Deoxycytidine administration & dosage, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms pathology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal administration & dosage
- Abstract
Background: The TOPAZ-1 phase III trial reported a survival benefit with the anti-programmed cell death ligand 1 (anti-PD-L1) durvalumab in combination with gemcitabine and cisplatin in patients with advanced biliary tract cancer (BTC)., Objective: The present study investigated for the first time the impact on survival of adding durvalumab to cisplatin/gemcitabine compared with cisplatin/gemcitabine in a real-world setting., Patients and Methods: The analyzed population included patients with unresectable, locally advanced, or metastatic BTC treated with durvalumab in combination with cisplatin/gemcitabine or with cisplatin/gemcitabine alone. The impact of adding durvalumab to chemotherapy in terms of overall survival (OS) and progression free survival (PFS) was investigated with univariate and multivariate analysis., Results: Overall, 563 patients were included in the analysis: 213 received cisplatin/gemcitabine alone, 350 received cisplatin/gemcitabine plus durvalumab. At the univariate analysis, the addition of durvalumab was found to have an impact on survival, with a median OS of 14.8 months versus 11.2 months [hazard ratio (HR) 0.63, 95% confidence interval (CI) 0.50-0.80, p = 0.0002] in patients who received cisplatin/gemcitabine plus durvalumab compared to those who received cisplatin/gemcitabine alone. At the univariate analysis for PFS, the addition of durvalumab to cisplatin/gemcitabine demonstrated a survival impact, with a median PFS of 8.3 months and 6.0 months (HR 0.57, 95% CI 0.47-0.70, p < 0.0001) in patients who received cisplatin/gemcitabine plus durvalumab and cisplatin/gemcitabine alone, respectively. The multivariate analysis confirmed that adding durvalumab to cisplatin/gemcitabine is an independent prognostic factor for OS and PFS, with patients > 70 years old and those affected by locally advanced disease experiencing the highest survival benefit. Finally, an exploratory analysis of prognostic factors was performed in the cohort of patients who received durvalumab: neutrophil-lymphocyte ratio (NLR) and disease stage were to be independent prognostic factors in terms of OS. The interaction test highlighted NLR ≤ 3, Eastern Cooperative Oncology Group Performance Status (ECOG PS) = 0, and locally advanced disease as positive predictive factors for OS on cisplatin/gemcitabine plus durvalumab., Conclusion: In line with the results of the TOPAZ-1 trial, adding durvalumab to cisplatin/gemcitabine has been confirmed to confer a survival benefit in terms of OS and PFS in a real-world setting of patients with advanced BTC., (© 2024. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)
- Published
- 2024
- Full Text
- View/download PDF
71. A phase II/III randomized clinical trial of CisPlatin plUs Gemcitabine and Nabpaclitaxel (GAP) as pReoperative chemotherapy versus immediate resection in patIents with resecTable BiliarY Tract Cancers (BTC) at high risk for recurrence: PURITY study.
- Author
-
Niger M, Nichetti F, Fornaro L, Pircher C, Morano F, Palermo F, Rimassa L, Pressiani T, Berardi R, Gardini AC, Sperti E, Salvatore L, Melisi D, Bergamo F, Siena S, Mosconi S, Longarini R, Arcangeli G, Corallo S, Delliponti L, Tamberi S, Fea E, Brandi G, Rapposelli IG, Salati M, Baili P, Miceli R, Ljevar S, Cavallo I, Sottotetti E, Martinetti A, Busset MDD, Sposito C, Di Bartolomeo M, Pietrantonio F, de Braud F, and Mazzaferro V
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Cisplatin, Deoxycytidine, Neoadjuvant Therapy methods, Neoplasm Recurrence, Local drug therapy, Quality of Life, Retrospective Studies, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms surgery, Gemcitabine
- Abstract
Background: Biliary tract cancers (BTCs) are rare and lethal cancers, with a 5-year survival inferior to 20%(1-3). The only potential curative treatment is surgical resection. However, despite complex surgical procedures that have a remarkable risk of postoperative morbidity and mortality, the 5-year survival rate after radical surgery (R0) is 20-40% and recurrence rates are up to ~ 75%(4-6). Up to ~ 40% of patients relapse within 12 months after resection, and half of these patient will recur systemically(4-6). There is no standard of care for neoadjuvant chemotherapy (NAC) in resectable BTC, but retrospective reports suggest its potential benefit (7, 8)., Methods: PURITY is a no-profit, multicentre, randomized phase II/III trial aimed at evaluating the efficacy of the combination of gemcitabine, cisplatin and nabpaclitaxel (GAP) as neoadjuvant treatment in patients with resectable BTC at high risk for recurrence. Primary objective of this study is to evaluate the efficacy of neoadjuvant GAP followed by surgery as compared to upfront surgery, in terms of 12-month progression-free survival for the phase II part and of progression free survival (PFS) for the phase III study. Key Secondary objectives are event free survival (EFS), relapse-free survival, (RFS), overall survival (OS), R0/R1/R2 resection rate, quality of life (QoL), overall response rate (ORR), resectability. Safety analyses will include toxicity rate and perioperative morbidity and mortality rate. Exploratory studies including Next-Generation Sequencing (NGS) in archival tumor tissues and longitudinal ctDNA analysis are planned to identify potential biomarkers of primary resistance and prognosis., Discussion: Considering the poor prognosis of resected BTC experiencing early tumor recurrence and the negative prognostic impact of R1/R2 resections, PURITY study is based on the rationale that NAC may improve R0 resection rates and ultimately patients' outcomes. Furthermore, NAC should allow early eradication of microscopic distant metastases, undetectable by imaging but already present at the time of diagnosis and avoid mortality and morbidity associated with resection for patients with rapid progression or worsening general condition during neoadjuvant therapy. The randomized PURITY study will evaluate whether patients affected by BTC at high risk from recurrence benefit from a neoadjuvant therapy with GAP regimen as compared to immediate surgery., Trial Registration: PURITY is registered at ClinicalTrials.gov (NCT06037980) and EuCT(2023-503295-25-00)., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
72. Negative Hyperselection of Patients with HER2+ and RAS Wild-Type Metastatic Colorectal Cancer Receiving Dual HER2 Blockade: the PRESSING-HER2 Study.
- Author
-
Randon G, Nakamura Y, Yaeger R, Lonardi S, Cremolini C, Elez E, Nichetti F, Ghelardi F, Nasca V, Bergamo F, Conca V, Ros J, Bando H, Maddalena G, Oldani S, Prisciandaro M, Raimondi A, Schrock AB, Agnelli L, Walch H, Yoshino T, and Pietrantonio F
- Subjects
- Humans, Progression-Free Survival, Prognosis, Mutation, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Colonic Neoplasms, Rectal Neoplasms
- Abstract
Purpose: To demonstrate the negative prognostic impact of a panel of genomic alterations (PRESSING-HER2 panel) and lack of HER2 amplification by next-generation sequencing (NGS) in patients with HER2+, RAS wild-type metastatic colorectal cancer receiving dual HER2 blockade., Experimental Design: The PRESSING-HER2 panel of HER2 mutations/rearrangements and RTK/MAPK mutations/amplifications was assessed by NGS. HER2 amplification was confirmed by NGS if copy-number variation (CNV) was ≥ 6. With a case-control design, hypothesizing 30% and 5% PRESSING-HER2 positivity in resistant [progression-free survival (PFS) <4 months and no RECIST response] versus sensitive cohorts, respectively, 35 patients were needed per group., Results: PRESSING-HER2 alterations included HER2 mutations/rearrangements, EGFR amplification, and BRAF mutations and had a prevalence of 27% (9/33) and 3% (1/35) in resistant versus sensitive patients (P = 0.005) and 63% predictive accuracy. Overall, HER2 nonamplified status by NGS had 10% prevalence. Median PFS and overall survival (OS) were worse in PRESSING-HER2+ versus negative (2.2 vs. 5.3 months, P < 0.001; 5.4 vs. 14.9 months, P = 0.001) and in HER2 nonamplified versus amplified (1.6 vs. 5.2 months, P < 0.001; 7.4 vs. 12.4 months, P = 0.157). These results were confirmed in multivariable analyses [PRESSING-HER2 positivity: PFS HR = 3.06, 95% confidence interval (CI), 1.40-6.69, P = 0.005; OS HR = 2.93, 95% CI, 1.32-6.48, P = 0.007]. Combining PRESSING-HER2 and HER2 CNV increased the predictive accuracy to 75%., Conclusions: PRESSING-HER2 panel and HER2 nonamplified status by NGS warrant validation as potential predictive markers in this setting. See related commentary by Raghav et al., p. 260., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
73. NALIRIFOX, FOLFIRINOX, and Gemcitabine With Nab-Paclitaxel as First-Line Chemotherapy for Metastatic Pancreatic Cancer: A Systematic Review and Meta-Analysis.
- Author
-
Nichetti F, Rota S, Ambrosini P, Pircher C, Gusmaroli E, Droz Dit Busset M, Pusceddu S, Sposito C, Coppa J, Morano F, Pietrantonio F, Di Bartolomeo M, Mariani L, Mazzaferro V, de Braud F, and Niger M
- Subjects
- Humans, Irinotecan therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leucovorin therapeutic use, Oxaliplatin therapeutic use, Gemcitabine, Fluorouracil therapeutic use, Pancreatic Neoplasms drug therapy, Adenocarcinoma
- Abstract
Importance: The NAPOLI 3 trial showed the superiority of fluorouracil, leucovorin, liposomal irinotecan, and oxaliplatin (NALIRIFOX) over the combination of gemcitabine and nab-paclitaxel (GEM-NABP) as first-line treatment of metastatic pancreatic ductal adenocarcinoma (PDAC). Analyses comparing NALIRIFOX and GEM-NABP with fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) have not yet been reported., Objective: To derive survival, response, and toxic effects data from phase 3 clinical trials and compare NALIRIFOX, FOLFIRINOX, and GEM-NABP., Data Sources: After a systematic search of PubMed, Scopus, Embase, and American Society of Clinical Oncology and European Society for Medical Oncology meetings' libraries, Kaplan-Meier curves were extracted from phase 3 clinical trials conducted from January 1, 2011, until September 12, 2023., Study Selection: Phase 3 clinical trials that tested NALIRIFOX, FOLFIRINOX, or GEM-NABP as first-line treatment of metastatic PDAC and reported overall survival (OS) and progression-free survival (PFS) curves were selected. This study followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses of Individual Participant Data reporting guidelines., Data Extraction and Synthesis: Individual patient OS and PFS data were extracted from Kaplan-Meier plots of original trials via a graphic reconstructive algorithm. Overall response rates (ORRs) and grade 3 or higher toxic effects rates were also collected. A pooled analysis was conducted, and results were validated via a network meta-analysis., Main Outcomes and Measures: The primary end point was OS. Secondary outcomes included PFS, ORR, and toxic effects rates., Results: A total of 7 trials with data on 2581 patients were analyzed, including 383 patients treated with NALIRIFOX, 433 patients treated with FOLFIRINOX, and 1756 patients treated with GEM-NABP. Median PFS was longer in patients treated with NALIRIFOX (7.4 [95% CI, 6.1-7.7] months) or FOLFIRINOX (7.3 [95% CI, 6.5-7.9] months; [HR], 1.21 [95% CI, 0.86-1.70]; P = .28) compared with patients treated with GEM-NABP (5.7 [95% CI, 5.6-6.1] months; HR vs NALIRIFOX, 1.45 [95% CI, 1.22-1.73]; P < .001). Similarly, GEM-NABP was associated with poorer OS (10.4 [95% CI, 9.8-10.8]; months) compared with NALIRIFOX (HR, 1.18 [95% CI, 1.00-1.39]; P = .05], while no difference was observed between FOLFIRINOX (11.7 [95% CI, 10.4-13.0] months) and NALIRIFOX (11.1 [95% CI, 10.1-12.3] months; HR, 1.06 [95% CI, 0.81-1.39]; P = .65). There were no statistically significant differences in ORR among NALIRIFOX (41.8%), FOLFIRINOX (31.6%), and GEM-NABP (35.0%). NALIRIFOX was associated with lower incidence of grade 3 or higher hematological toxic effects (eg, platelet count decreased 1.6% vs 11.8% with FOLFIRINOX and 10.8% with GEM-NABP), but higher rates of severe diarrhea compared with GEM-NABP (20.3% vs 15.7%)., Conclusions and Relevance: In this systematic review and meta-analysis, NALIRIFOX and FOLFIRINOX were associated with similar PFS and OS as first-line treatment of advanced PDAC, although NALIRIFOX was associated with a different toxicity profile. Careful patient selection, financial toxic effects consideration, and direct comparison between FOLFIRINOX and NALIRIFOX are warranted.
- Published
- 2024
- Full Text
- View/download PDF
74. Peripheral blood lymphocytes predict clinical outcomes in hormone receptor-positive HER2-negative advanced breast cancer patients treated with CDK4/6 inhibitors.
- Author
-
Zattarin E, Mariani L, Menichetti A, Leporati R, Provenzano L, Ligorio F, Fucà G, Lobefaro R, Lalli L, Vingiani A, Nichetti F, Griguolo G, Sirico M, Bernocchi O, Marra A, Corti C, Zagami P, Agostinetto E, Jacobs F, Di Mauro P, Presti D, Sposetti C, Giorgi CA, Guarneri V, Pedersini R, Losurdo A, Generali D, Curigliano G, Pruneri G, de Braud F, Dieci MV, and Vernieri C
- Abstract
Background: Cyclin-Dependent Kinase 4/6 inhibitors (CDK4/6i) combined with Endocrine Therapy (ET) are the standard treatment for patients with Hormone Receptor-positive/HER2-negative advanced breast cancer (HR+/HER2- aBC)., Objectives: While CDK4/6i are known to reduce several peripheral blood cells, such as neutrophils, lymphocytes and platelets, the impact of these modulations on clinical outcomes is unknown., Design: A multicenter, retrospective-prospective Italian study., Methods: We investigated the association between baseline peripheral blood cells, or their early modifications (i.e. 2 weeks after treatment initiation), and the progression-free survival (PFS) of HR+/HER2- aBC patients treated with ETs plus CDK4/6i. Random Forest models were used to select covariates associated with patient PFS among a large list of patient- and tumor-related variables., Results: We evaluated 638 HR+/HER2- aBC patients treated with ET plus CDK4/6i at six Italian Institutions between January 2017 and May 2021. High baseline lymphocyte counts were independently associated with longer PFS [median PFS (mPFS) 20.1 versus 13.2 months in high versus low lymphocyte patients, respectively; adjusted Hazard Ratio (aHR): 0.78; 95% confidence interval (CI): 0.66-0.92; p = 0.0144]. Moreover, patients experiencing a lower early reduction of lymphocyte counts had significantly longer PFS when compared to patients undergoing higher lymphocyte decrease (mPFS 18.1 versus 14.5 months; aHR: 0.82; 95% CI: 0.73-0.93; p = 0.0037). Patients with high baseline lymphocytes and undergoing a lower reduction, or even an increase, of lymphocyte counts during CDK4/6i therapy experienced the longest PFS, while patients with lower baseline lymphocytes and undergoing a higher decrease of lymphocytes had the lowest PFS (mPFS 21.4 versus 11 months, respectively)., Conclusion: Baseline and on-treatment modifications of peripheral blood lymphocytes have independent prognostic value in HR+/HER2- aBC patients. This study supports the implementation of clinical strategies to boost antitumor immunity in patients with HR+/HER2- aBC treated with ETs plus CDK4/6i., (© The Author(s), 2023.)
- Published
- 2023
- Full Text
- View/download PDF
75. Safety and antitumor activity of metformin plus lanreotide in patients with advanced gastro-intestinal or lung neuroendocrine tumors: the phase Ib trial MetNET2.
- Author
-
Pusceddu S, Corti F, Prinzi N, Nichetti F, Ljevar S, Busico A, Cascella T, Leporati R, Oldani S, Pircher CC, Coppa J, Resi V, Milione M, Maccauro M, Miceli R, Tamborini E, Perrone F, Spreafico C, Niger M, Morano F, Pietrantonio F, Seregni E, Mariani L, Mazzaferro V, Di Liberti G, Fucà G, de Braud F, and Vernieri C
- Subjects
- Humans, Retrospective Studies, Prospective Studies, Somatostatin adverse effects, Lung pathology, Metformin pharmacology, Metformin therapeutic use, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors chemically induced, Neuroendocrine Tumors pathology, Pancreatic Neoplasms pathology, Diabetes Mellitus chemically induced, Diabetes Mellitus drug therapy
- Abstract
In retrospective studies, metformin use has been associated with better clinical outcomes in diabetic patients with advanced, well-differentiated neuroendocrine tumors (WDNETs). However, prospective evidence of metformin safety and activity is lacking. Here, we conducted the first-in-human phase Ib MetNET2 trial to investigate the safety and antitumor activity of metformin in combination with the somatostatin analog lanreotide autogel (ATG) in both diabetic and non-diabetic patients with advanced WDNETs of the gastrointestinal (GI) or thoracic tract. Enrolled patients received lanreotide ATG 120 mg plus oral metformin, up to a maximum dosage of 2550 mg/day. We enrolled 20 patients, of whom 18 (90%) and 2 (10%) had WDNETs of the GI and thoracic tract, respectively. Fourteen patients (70%) were non-diabetic. With a 5% incidence of SAEs, the study met its primary objective of demonstrating treatment safety. With a median follow-up of 39 months (95% CI 28-NE), median PFS was 24 months (95% CI 16-NE), with 12-month and 24-month PFS probability of 75% (95% CI 58-97) and 49% (95% CI 31-77), respectively. We found no statistically significant PFS differences between diabetic and non-diabetic patients. Among exploratory analyses, the presence of tumor genomic alterations in DNA damage pathways was associated with trend towards worse PFS, whereas a precocious reduction of HOMA-IR index and plasma cholesterol concentration showed a trend towards an association with better PFS. In conclusion, metformin plus lanreotide ATG is a safe and well tolerated combination treatment that is associated with promising antitumor activity in both non-diabetic and diabetic patients with WDNETs, and that warrants further investigation in larger clinical trials., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
76. Negative hyperselection of elderly patients with RAS and BRAF wild-type metastatic colorectal cancer receiving initial panitumumab plus FOLFOX or 5-FU/LV.
- Author
-
Pietrantonio F, Bergamo F, Rossini D, Ghelardi F, De Grandis MC, Germani MM, Barsotti G, Formica V, Frassineti GL, Boscolo G, Cinieri S, Di Donato S, Antonuzzo L, Antoniotti C, Ambrosini M, Piva VM, Nichetti F, Fassan M, Cremolini C, and Lonardi S
- Subjects
- Aged, Humans, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, ErbB Receptors, Fluorouracil therapeutic use, Panitumumab therapeutic use, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins B-raf genetics, Colonic Neoplasms, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Rectal Neoplasms
- Abstract
Background: Upfront anti-EGFR therapy represents the standard of care for patients with left-sided, MSS/pMMR, RAS and BRAF wild-type mCRC. Molecular 'hyperselection' may optimize EGFR inhibition by detecting additional resistance alterations., Materials and Methods: We used comprehensive genomic profiling on archival samples of elderly patients enrolled in the PANDA trial to detect: HER2 amplification/mutations; MET amplification; NTRK/ROS1/ALK/RET rearrangements; PIK3CA exon 20 mutations; PTEN alterations; AKT1 mutations; MAP2K1 mutations. We defined 'Gene Altered' (GA) patients whose tumour harboured at least one alteration, and 'Hyperselected' (HS) those without. Survival and tumour response outcomes were correlated to hyperselection status alone or combined with primary tumour sidedness or treatment arm., Results: Genomic alterations were detected in 41/147 patients (27.9%). PFS, OS and ORR were inferior in GA versus HS (median PFS: 7.6 versus 12.8 months, HR = 2.08, 95% CI: 1.43-3.03, p < 0.001; median OS: 20.0 versus 29.5 months, HR = 1.82, 95% CI:1.23-2.69, p = 0.002; ORR: 51% versus 71%; OR = 0.43, 95% CI: 0.21-0.91, p = 0.02). In the multivariable models, the impact of hyperselection on PFS and OS was confirmed. Lower ORR was observed with 5-FU/LV/panitumumab in GA (40% versus 62%), but not in HS (70% versus 72%). GA was associated with worse survival and response regardless of primary tumour sidedness, whereas in the HS subgroup, right-and left sided tumours had similar outcomes., Conclusions: Molecular hyperselection and comprehensive genomic profiling have a potential usefulness in elderly patients with RAS/BRAF wild-type, pMMR/MSS mCRC, eligible for upfront EGFR inhibition., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: F.P. received honoraria from Amgen, Bayer, Servier, Merck-Serono, MSD, BMS, Takeda, Astellas, Pierre-Fabre and received research grants for academic studies from Bristol-Myers Squibb, AstraZeneca, Agenus, Incyte, Amgen. F.B. received personal honoraria as invited speaker from Eli-Lilly, MSD, EISAI, BMS; participation in advisory board for Servier, AAA Novartis. D.R. received honoraria from Amgen, MSD and Takeda. V.F. received personal honoraria as invited speaker from Merck Serono, Pierre Fabre, Amgen, MSD, Servier. G.L.F. received personal honoraria as invited speaker from: Servier, Merck Serono; participation in advisory board for Servier, Novartis, Merck Sharp & Dohme. S.D.D received personal honoraria as invited speaker from Roche, Servier, Merck Serono, Pierre Fabre, Amgen, Merck Sharp & Dohme, iNCYTE, Bayer; participation in advisory board for Amgen, Merck Serono, Servier, Bayer. L.A. received research funding (to institution) from Novartis, Merck Serono, Bristol Myers Squibb, Astra Zeneca; personal honoraria as invited speaker from Roche, Bristol Myers Squibb, Servier, AstraZeneca, Amgen, MSD; participation in advisory board for Amgen, Merk Serono, Ely Lilly, AstraZeneca, Incite, Bristol Myers Squibb, Servier, MSD. C.A. received speaker fees from Merck; Honoraria from Merck and Nordic Pharma. M.F. received research funding (to Institution) from QED, Macrophage pharma, Diaceutics, Astellas; personal honoraria as invited speaker from Roche, Eli Lilly, Bristol Myers Squibb, MSD, Pierre Fabre, GlaxoSmithKline, Amgen, Astellas, AstraZeneca. C.A. received speaker fees from Merck; Honoraria from Merck and Nordic Pharma. M.F. received research funding (to Institution) from QED, Macrophage pharma, Diaceutics, Astellas; personal honoraria as invited speaker from Roche, Eli Lilly, Bristol Myers Squibb, MSD, Pierre Fabre, GlaxoSmithKline, Amgen, Astellas, AstraZeneca. C.C. received research funding from Merck, Bayer, Servier; Honoraria from Amgen, Bayer, Merck, MSD, Pierre Fabre, Roche and Servier; Consulting or advisory role: Amgen, Bayer, MSD, Nordic Pharma, Roche. S.L. received research funding (to Institution) from Amgen,Merck Serono, Bayer, Roche, Eli Lilly, AstraZeneca, Bristol Myers Squibb; personal honoraria as invited speaker from Roche, Eli Lilly, Bristol Myers Squibb, Servier, Merck Serono, Pierre Fabre, GlaxoSmithKline, Amgen; participation in advisory board for Amgen, Merck Serono, Eli Lilly, AstraZeneca, Incyte, Daiichi-Sankyo, Bristol Myers Squibb, Servier, Merck Sharp & Dohme, Astellas, Takeda. FG, MCDG, MMG, GBa, GBo, SC, MA, VMP and FN declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
77. Comparative Genomic Analysis and Clinical Outcomes of BRAF-mutated Advanced Biliary Tract Cancers.
- Author
-
Tang TY, Nichetti F, Kaplan B, Lonardi S, Pietrantonio F, Salvatore L, Vivaldi C, Rimassa L, de Braud F, Rizzato MD, Pavlick D, Chu R, Danner De Armas A, Sharaf R, Sokol E, Rodon Ahnert J, Ross JS, Javle M, and Niger M
- Subjects
- Humans, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins p21(ras) genetics, Mutation, Bile Ducts, Intrahepatic pathology, Genomics, Bile Duct Neoplasms genetics, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms genetics, Biliary Tract Neoplasms pathology, Cholangiocarcinoma drug therapy, Cholangiocarcinoma genetics, Cholangiocarcinoma pathology
- Abstract
Purpose: BRAF mutations are rare in biliary tract cancers (BTC), but are of interest given the recent developments in targeted therapy for BTC. We investigated the clinical outcomes in a cohort of BRAF-mutant advanced BTC treated with first-line chemotherapy. Furthermore, we investigated the genomic landscape of BRAF class I, II, and III mutations in the intrahepatic cholangiocarcinoma (iCCA) subgroup of BTC., Experimental Design: We analyzed two nonoverlapping cohorts. We examined the genomic landscape of BRAF-mutated iCCA in a "genomic cohort" [187 class I, 82 class II, 113 class III BRAF mutants and 8,026 wildtype (WT)]. We also analyzed median progression-free survival (PFS) and overall survival (OS) on first-line chemotherapy in a separate multi-institutional "clinical cohort" of patients with BTC (including iCCA and extrahepatic cholangiocarcinoma (eCCA) and gallbladder cancer; 41 class I, 32 class II+III BRAF mutants and 1,042 WT)., Results: In the entire BTC clinical cohort, the median PFS was shorter for class I [HR, 2.11 (P < 0.001)] and class II+III [HR, 1.72 (P = 0.007)] as compared with BRAF WT. OS was also shorter in class I [HR, 2.04 (P = 0.011)] and class II+III [HR, 1.86 (P = 0.002)] as compared with BRAF WT. In the iCCA subgroup, class I alterations were mutually exclusive with FGFR2, IDH1/2, ERBB2, and KRAS mutations. Class II+III mutations appear to be mutually exclusive with FGFR2 and KRAS., Conclusions: In BTC, all classes of BRAF mutations are associated with a worse prognosis. BRAF mutations occur in 5% of iCCA subgroup and may be mutually exclusive with other targetable mutations., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
78. Optimal maintenance strategy following FOLFOX plus anti-EGFR induction therapy in patients with RAS wild type metastatic colorectal cancer: An individual patient data pooled analysis of randomised clinical trials.
- Author
-
Raimondi A, Nichetti F, Stahler A, Wasan HS, Aranda E, Randon G, Kurreck A, Meade AM, Díaz-Rubio E, Niger M, Stintzing S, Palermo F, Trarbach T, Prisciandaro M, Sommerhäuser G, Fisher D, Morano F, Pietrantonio F, and Modest DP
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Cetuximab, Fluorouracil, Induction Chemotherapy, Leucovorin, Colonic Neoplasms drug therapy, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Rectal Neoplasms drug therapy
- Abstract
Background: Anti-EGFR antibodies plus doublet chemotherapy is the standard of care in RAS/BRAF wild-type metastatic colorectal cancer (mCRC). No phase-3 level of evidence is available to guide treatment de-escalation after anti-EGFR-based first-line. Several randomised clinical trials investigated de-intensification strategies with 5-fluorouracil/leucovorin (5-FU/LV) and/or anti-EGFR., Methods: We performed an individual patient data pooled analysis of Valentino, Panama, MACRO-2, COIN-B trials including RAS wild-type mCRC patients who received first-line therapy with FOLFOX plus panitumumab or cetuximab followed by pre-specified maintenance strategy. Only patients who started maintenance according to the assigned arm were included. Patients were categorised by type of maintenance (i.e. 5-FU/LV, anti-EGFR or 5-FU/LV + anti-EGFR). Progression-free survival (PFS) and overall survival (OS) were calculated from the start of maintenance; toxicity was evaluated for the maintenance treatment period., Results: A total of 518 patients were included in the pooled analysis. Overall, 123, 185 and 210 patients received maintenance with 5-FU/LV, anti-EGFR, 5-FU/LV + anti-EGFR, respectively. Median PFS was 5.6, 6.0 and 9.0 (P = 0.009) and OS was 25.7, 24.0 and 28.0 months (P = 0.134) in 5-FU/LV, anti-EGFR and 5-FU/LV + anti-EGFR arms, respectively. Monotherapy maintenance (either 5-FU/LV or anti-EGFR) was inferior to combination in terms of PFS (hazard ratios [HR] 1.26, P = 0.016) and non-significantly trending also in OS (HR 1.20, P = 0.111). An increase of overall any grade and grade ≥ 3 AEs and selected AEs was reported in combination compared to either 5-FU/LV or anti-EGFR arms., Conclusions: This pooled analysis including four randomised phase II supports the use of 5-FU/LV plus anti-EGFR as the preferred maintenance regimen. Data provide rational for a more individualised maintenance treatment approach based on tumour and patients features., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: AR: Honoraria for lectures from Elma Academy and Servier. Travel support: Amgen. HW: honoraria as a speaker and/or in an advisory role from Pierre Fabre, Merck KGaA, Incyte, Merck Sharp Dohme, Servier, Bayer, Roche Genentech and SIRTEX. EA: Honoraria for advisory role from Amgen, Bayer, Bristol Myers Squibb, Merck, Roche, Sanofi. FM: Honoraria from Servier, Lilly, Pierre-Fabre and received research grants from Incyte. FP: Honoraria from Amgen, Bayer, Servier, Merck-Serono, Lilly, MSD, BMS, Astrazeneca, Astellas, Organon, Pierre-Fabre and received research grants from Bristol-Myers Squibb, Astrazeneca, Agenus, Incyte. DPM: Honoraria for lectures and advisory boards: Merck, Amgen, Servier, Pierre Fabre, Sanofi, Lilly, BMS, MSD, AstraZeneca, G1, Cureteq, Takeda, Taiho, Onkowissen.de; GSK, Seagen, COR2ED. Travel support: Amgen, Servier. Research funding (inst): Amgen, Servier. All other authors declared no disclosures., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
79. Durvalumab plus gemcitabine and cisplatin in advanced biliary tract cancer: An early exploratory analysis of real-world data.
- Author
-
Rimini M, Fornaro L, Lonardi S, Niger M, Lavacchi D, Pressiani T, Lucchetti J, Giordano G, Pretta A, Tamburini E, Pirrone C, Rapposelli IG, Diana A, Martinelli E, Garajová I, Simionato F, Schirripa M, Formica V, Vivaldi C, Caliman E, Rizzato MD, Zanuso V, Nichetti F, Angotti L, Landriscina M, Scartozzi M, Ramundo M, Pastorino A, Daniele B, Cornara N, Persano M, Gusmaroli E, Cerantola R, Salani F, Ratti F, Aldrighetti L, Cascinu S, Rimassa L, Antonuzzo L, and Casadei-Gardini A
- Subjects
- Humans, Cisplatin therapeutic use, Antibodies, Monoclonal adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Gemcitabine, Bile Duct Neoplasms drug therapy
- Abstract
Background: The TOPAZ-1 phase III trial reported a survival benefit with the anti-programmed death cell ligand 1 (anti-PD-L1) durvalumab in combination with gemcitabine and cisplatin in patients with advanced biliary tract cancer. The present study investigated the efficacy and safety of this new standard treatment in a real-world setting., Methods: The analysed population included patients with unresectable, locally advanced or metastatic adenocarcinoma of the biliary tract treated with durvalumab in combination with gemcitabine and cisplatin at 17 Italian centres. The primary endpoint of the study was progression-free survival (PFS), whereas secondary endpoints included overall survival (OS), overall response rate (ORR) and safety. Unadjusted and adjusted hazard ratios (HRs) by baseline characteristics were calculated using the Cox proportional hazards model., Results: From February 2022 to November 2022, 145 patients were enrolled. After a median follow-up of 8.5 months (95% CI: 7.9-13.6), the median PFS was 8.9 months (95% CI: 7.4-11.7). Median OS was 12.9 months (95% CI: 10.9-12.9). The investigator-assessed confirmed ORR was 34.5%, and the disease control rate was 87.6%. Any grade adverse events (AEs) occurred in 137 patients (94.5%). Grades 3-4 AEs occurred in 51 patients (35.2%). The rate of immune-mediated AEs (imAEs) was 22.7%. Grades 3-4 imAEs occurred in 2.1% of the patients. In univariate analysis, non-viral aetiology, ECOG PS >0 and NLR ≥3 correlated with shorter PFS., Conclusion: The results reported in this first real-world analysis mostly confirmed the results achieved in the TOPAZ-1 trial in terms of PFS, ORR and safety., (© 2023 The Authors. Liver International published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
80. Molecular Tumor Board as a Clinical Tool for Converting Molecular Data Into Real-World Patient Care.
- Author
-
Vingiani A, Agnelli L, Duca M, Lorenzini D, Damian S, Proto C, Niger M, Nichetti F, Tamborini E, Perrone F, Piccolo A, Manoukian S, Azzollini J, Brambilla M, Colombo E, Lopez S, Vernieri C, Marra F, Conca E, Busico A, Capone I, Bozzi F, Angelini M, Devecchi A, Salvatori R, De Micheli V, Baggi A, Pasini S, Jommi C, Ladisa V, Apolone G, De Braud F, and Pruneri G
- Subjects
- Humans, Precision Medicine, Patient Care, Medical Oncology, High-Throughput Nucleotide Sequencing, Neoplasms
- Abstract
Purpose: The investigation of multiple molecular targets with next-generation sequencing (NGS) has entered clinical practice in oncology, yielding to a paradigm shift from the histology-centric approach to the mutational model for personalized treatment. Accordingly, most of the drugs recently approved in oncology are coupled to specific biomarkers. One potential tool for implementing the mutational model of precision oncology in daily practice is represented by the Molecular Tumor Board (MTB), a multidisciplinary team whereby molecular pathologists, biologists, bioinformaticians, geneticists, medical oncologists, and pharmacists cooperate to generate, interpret, and match molecular data with personalized treatments., Patients and Methods: Since May 2020, the institutional MTB set at Fondazione IRCCS Istituto Nazionale Tumori of Milan met weekly via teleconference to discuss molecular data and potential therapeutic options for patients with advanced/metastatic solid tumors., Results: Up to October 2021, among 1,996 patients evaluated, we identified >10,000 variants, 43.2% of which were functionally relevant (pathogenic or likely pathogenic). On the basis of functionally relevant variants, 711 patients (35.6%) were potentially eligible to targeted therapy according to European Society of Medical Oncology Scale for Clinical Actionability of Molecular Targets tiers, and 9.4% received a personalized treatment. Overall, larger NGS panels (containing >50 genes) significantly outperformed small panels (up to 50 genes) in detecting actionable gene targets across different tumor types., Conclusion: Our real-world data provide evidence that MTB is a valuable tool for matching NGS data with targeted treatments, eventually implementing precision oncology in clinical practice.
- Published
- 2023
- Full Text
- View/download PDF
81. Comprehensive transcriptomic analysis to identify biological and clinical differences in cholangiocarcinoma.
- Author
-
Silvestri M, Nghia Vu T, Nichetti F, Niger M, Di Cosimo S, De Braud F, Pruneri G, Pawitan Y, Calza S, and Cappelletti V
- Subjects
- Humans, Transcriptome, Prognosis, Bile Ducts, Intrahepatic pathology, Cholangiocarcinoma pathology, Bile Duct Neoplasms pathology
- Abstract
Background: Cholangiocarcinoma (CC) is a rare and aggressive disease with limited therapeutic options and a poor prognosis. All available public records of cohorts reporting transcriptomic data on intrahepatic cholangiocarcinoma (ICC) and extrahepatic cholangiocarcinoma (ECC) were collected with the aim to provide a comprehensive gene expression-based classification with clinical relevance., Methods: A total of 543 patients with primary tumor tissues profiled by RNAseq and microarray platforms from seven public datasets were used as a discovery set to identify distinct biological subgroups. Group predictors developed on the discovery sets were applied to a single cohort of 131 patients profiled with RNAseq for validation and assessment of clinical relevance leveraging machine learning techniques., Results: By unsupervised clustering analysis of gene expression data we identified both in the ICC and ECC discovery datasets four subgroups characterized by a distinct type of immune infiltrate and signaling pathways. We next developed class predictors using short gene list signatures and identified in an independent dataset subgroups of ICC tumors at different prognosis., Conclusions: The developed class-predictor allows identification of CC subgroups with specific biological features and clinical behavior at single-sample level. Such results represent the starting point for a complete molecular characterization of CC, including integration of genomics data to develop in clinical practice., (© 2023 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
82. Clinical Outcomes After Progression on First-Line Therapies in IDH1 Mutated Versus Wild-Type Intrahepatic Cholangiocarcinoma Patients.
- Author
-
Rimini M, Fabregat-Franco C, Persano M, Burgio V, Bergamo F, Niger M, Scartozzi M, Rapposelli IG, Aprile G, Ratti F, Pedica F, Verdaguer H, Rizzato M, Nichetti F, Lai E, Cappetta A, Macarulla T, Fassan M, De Braud F, Pretta A, Simionato F, De Cobelli F, Aldrighetti L, Fornaro L, Cascinu S, and Casadei-Gardini A
- Subjects
- Humans, Retrospective Studies, Isocitrate Dehydrogenase genetics, Prognosis, Mutation, Bile Ducts, Intrahepatic pathology, Cholangiocarcinoma drug therapy, Cholangiocarcinoma genetics, Bile Duct Neoplasms drug therapy, Bile Duct Neoplasms genetics, Bile Duct Neoplasms pathology
- Abstract
Background: Isocitrate dehydrogenase-1 (IDH1) mutations occur in a significant proportion of intrahepatic cholangiocarcinomas (iCCAs). No data are available regarding the prognostic impact of IDH1 mutations in advanced iCCA patients after progression on first-line therapies., Objective: We investigated the role of IDH1 mutation in advanced iCCA after progression on first-line therapies., Patients and Methods: After progression on first-line therapies for advanced iCCA, consecutive patients were retrospectively collected. The IDH1 status was tested at baseline. This analysis aimed to examine the association between the presence of IDH1 missense mutations and survival outcomes in patients with advanced iCCA treated with a second-line therapy., Results: The analysis included 119 patients; 56/119 (47%) were IDH1 mutated (IDH1m) and 63/119 (53%) were IDH1 wild type (IDH1 WT). At univariate analysis for overall survival (OS), the presence of IDH1 mutation was associated with a worse median OS (mOS; 8.2 vs. 14.1 months; hazard ratio [HR] 1.9, 95% confidence interval [CI] 1.2-3.0, p = 0.0047). Patients harboring IDH1 mutations showed a worse objective response rate (ORR) compared with patients without IDH1 mutation, whereas no significant differences in disease control rate (DCR) were found. Multivariate analysis confirmed IDH1 mutations as an independent negative prognostic factor for OS (HR 1.7, 95% CI 1.1-2.7, p = 0.0256). By evaluating only patients receiving FOLFOX as second-line therapy, no statistically significant differences were found in terms of both OS and PFS between IDH1m and IDH1 WT patients. In this subset of patients, those harboring an IDH1 mutation showed a worse ORR and DCR compared with those without. Finally, at univariate analysis for OS from third-line treatment, the presence of an IDH1 mutation was associated with a trend toward a worse mOS (6.0 vs. 11.9 months; HR 1.6, 95% CI 0.8-3.2, p = 0.25)., Conclusion: The present analysis constitutes the first evidence of a negative prognostic impact of IDH1 mutations in a cohort of patients treated after progression on first-line therapies in contrast to IDH1 inhibitors., (© 2023. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)
- Published
- 2023
- Full Text
- View/download PDF
83. Bcl-x L as prognostic marker and potential therapeutic target in cholangiocarcinoma.
- Author
-
Hoffmeister-Wittmann P, Mock A, Nichetti F, Korell F, Heilig CE, Scherr AL, Günther M, Albrecht T, Kelmendi E, Xu K, Nader L, Kessler A, Schmitt N, Fritzsche S, Weiler S, Sobol B, Stenzinger A, Boeck S, Westphalen CB, Schulze-Osthoff K, Trojan J, Kindler T, Weichert W, Spiekermann K, Bitzer M, Folprecht G, Illert AL, Boerries M, Klauschen F, Ochsenreither S, Siveke J, Bauer S, Glimm H, Brors B, Hüllein J, Hübschmann D, Uhrig S, Horak P, Kreutzfeldt S, Banales JM, Springfeld C, Jäger D, Schirmacher P, Roessler S, Ormanns S, Goeppert B, Fröhling S, and Köhler BC
- Subjects
- Humans, Bile Duct Neoplasms diagnosis, Bile Duct Neoplasms drug therapy, Bile Ducts, Intrahepatic, Cell Line, Tumor, Myeloid Cell Leukemia Sequence 1 Protein genetics, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Prognosis, Proto-Oncogene Proteins c-bcl-2 genetics, bcl-X Protein genetics, bcl-X Protein metabolism, Cholangiocarcinoma diagnosis, Cholangiocarcinoma drug therapy
- Abstract
Intrahepatic, perihilar, and distal cholangiocarcinoma (iCCA, pCCA, dCCA) are highly malignant tumours with increasing mortality rates due to therapy resistances. Among the mechanisms mediating resistance, overexpression of anti-apoptotic Bcl-2 proteins (Bcl-2, Bcl-x
L , Mcl-1) is particularly important. In this study, we investigated whether antiapoptotic protein patterns are prognostically relevant and potential therapeutic targets in CCA. Bcl-2 proteins were analysed in a pan-cancer cohort from the NCT/DKFZ/DKTK MASTER registry trial (n = 1140, CCA n = 72) via RNA-sequencing and transcriptome-based protein activity interference revealing high ranks of CCA for Bcl-xL and Mcl-1. Expression of Bcl-xL , Mcl-1, and Bcl-2 was assessed in human CCA tissue and cell lines compared with cholangiocytes by immunohistochemistry, immunoblotting, and quantitative-RT-PCR. Immunohistochemistry confirmed the upregulation of Bcl-xL and Mcl-1 in iCCA tissues. Cell death of CCA cell lines upon treatment with specific small molecule inhibitors of Bcl-xL (Wehi-539), of Mcl-1 (S63845), and Bcl-2 (ABT-199), either alone, in combination with each other or together with chemotherapeutics was assessed by flow cytometry. Targeting Bcl-xL induced cell death and augmented the effect of chemotherapy in CCA cells. Combined inhibition of Bcl-xL and Mcl-1 led to a synergistic increase in cell death in CCA cell lines. Correlation between Bcl-2 protein expression and survival was analysed within three independent patient cohorts from cancer centers in Germany comprising 656 CCA cases indicating a prognostic value of Bcl-xL in CCA depending on the CCA subtype. Collectively, these observations identify Bcl-xL as a key protein in cell death resistance of CCA and may pave the way for clinical application., (© 2022 The Authors. Liver International published by John Wiley & Sons Ltd.)- Published
- 2022
- Full Text
- View/download PDF
84. Molecular profile and its clinical impact of IDH1 mutated versus IDH1 wild type intrahepatic cholangiocarcinoma.
- Author
-
Rimini M, Fabregat-Franco C, Burgio V, Lonardi S, Niger M, Scartozzi M, Rapposelli IG, Aprile G, Ratti F, Pedica F, Verdaguer H, Rizzato M, Nichetti F, Lai E, Cappetta A, Macarulla T, Fassan M, De Braud F, Pretta A, Simionato F, De Cobelli F, Aldrighetti L, Fornaro L, Cascinu S, and Casadei-Gardini A
- Subjects
- Humans, Mutation, Prognosis, Bile Ducts, Intrahepatic pathology, Carrier Proteins genetics, Isocitrate Dehydrogenase genetics, Cholangiocarcinoma pathology, Bile Duct Neoplasms pathology
- Abstract
IDH1-mutated cholangiocarcinomas (CCAs) are an interesting group of neoplasia with particular behavior and therapeutic implications. The aim of the present work is to highlight the differences characterizing IDH1m and IDH1wt CCAs in terms of genomic landscape. 284 patients with iCCA treated for resectable, locally advanced or metastatic disease were selected and studied with the FOUNDATION Cdx technology. A comparative genomic analysis and survival analyses for the most relevant altered genes were performed between IDH1m and IDH1wt patients. Overall, 125 patients were IDH1m and 122 IDH1wt. IDH1m patients showed higher mutation rates compared to IDH1wt in CDKN2B and lower mutation rates in several genes including TP53, FGFR2, BRCA2, ATM, MAP3K1, NOTCH2, ZNF703, CCND1, NBN, NF1, MAP3KI3, and RAD21. At the survival analysis, IDH1m and IDH1wt patients showed no statistically differences in terms of survival outcomes, but a trend in favor of IDH1wt patients was observed. Differences in prognostic values of the most common altered genes were reported. In surgical setting, in IDH1m group the presence of CDKN2A and CDKN2B mutations negatively impact DFS, whereas the presence of CDKN2A, CDKN2B, and PBRM1 mutations negatively impact OS. In advanced setting, in the IDH1m group, the presence of KRAS/NRAS and TP53 mutations negatively impact PFS, whereas the presence of TP53 and PIK3CA mutations negatively impact OS; in the IDH1wt group, only the presence of MTAP mutation negatively impact PFS, whereas the presence of TP53 mutation negatively impact OS. We highlighted several molecular differences with distinct prognostic implications between IDH1m and IDH1wt patients., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
85. Next-generation sequencing analysis of cholangiocarcinoma identifies distinct IDH1-mutated clusters.
- Author
-
Rimini M, Loi E, Fabregat-Franco C, Burgio V, Lonardi S, Niger M, Scartozzi M, Raposelli IG, Aprile G, Ratti F, Pedica F, Verdaguer H, Rizzato M, Nichetti F, Lai E, Cappetta A, Macarulla T, Fassan M, De Braud F, Pretta A, Simionato F, De Cobelli F, Aldrighetti L, Fornaro L, Cascinu S, Zavattari P, and Casadei-Gardini A
- Subjects
- Bile Ducts, Intrahepatic metabolism, Chromatin metabolism, High-Throughput Nucleotide Sequencing, Humans, Isocitrate Dehydrogenase genetics, Mutation, Phosphatidylinositol 3-Kinases metabolism, Bile Duct Neoplasms pathology, Cholangiocarcinoma pathology
- Abstract
Background: 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., Methods: 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., Results: 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., Conclusion: We highlighted three different mutation-based clusters with prognostic significance in a cohort of IDH1m iCCAs., Competing Interests: Conflict of interest statement MN: Travel expenses from Celgene and AstraZeneca, speaker honorarium from Accademia della Medicina; honoraria from Sandoz, Medpoint SRL for editorial collaboration. Consultant honoraria from EMD Serono, Basilea Pharmaceutica, Incyte, MSD Italia and Servier. FdB: Honoraria from Roche, Pfizer, BMS, Merck, MSD, SERVIER, Sanofi, Amgen Astellas BioPharma, Incyte. Consulting or Advisory Role for Roche, Incyte, EMD Serono, BMS, Nerviano Medical Sciences, Sanofi, Novartis Italy, Menarini, research funding (institution): Novartis, Roche, Merck Serono, Pfizer, Servier, Philogen, Loxo, Tesaro, Nerviano Medical Sciences, Kymab. Research funding: BMS/Medarex, Merck KGaA, Ignyta, MedImmune, Exelis, Bayer health, Daiichi Sangyo Europe GmbH, Incyte, Basilea Pharmaceutical, Jassen Oncology. TM: (SOBI) Swedish Orpahn Biovitrum AB, Ability Pharmaceuticals SL, Aptitude Health, AstraZeneca, Basilea Pharma, Baxter, BioLineRX Ltd, Celgene, Eisai, Ellipses, Genzyme, Got It Consulting SL, Hirslanden/GITZ, Imedex, Incyte, Ipsen Bioscience, Inc, Janssen, Lilly. Marketing Farmacéutico & Investigación Clínica, S.L, MDS, Medscape, Novocure, Paraxel, PPD Development, Polaris, QED Therapeutics, Roche Farma, Sanofi-Aventis, Servier, Scilink Comunicación Científica SC, Surface Oncology, and Zymeworks., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
86. Platinum sensitivity in patients with IDH1/2 mutated vs wild-type intrahepatic cholangiocarcinoma: A propensity score-based study.
- Author
-
Niger M, Nichetti F, Casadei-Gardini A, Rizzato MD, Pircher C, Bini M, Franza A, Rimini M, Burgio V, Sposetti C, Fornaro L, Rapposelli IG, D'Amico FE, Aprile G, Vivaldi C, Frassineti GL, Milione M, Leoncini G, Cappetta A, Vasile E, Fassan M, Morano F, Perrone F, Tamborini E, Pruneri G, Lonardi S, Mazzaferro V, Pietrantonio F, Di Bartolomeo M, and de Braud F
- Subjects
- Bile Ducts, Intrahepatic pathology, Humans, Isocitrate Dehydrogenase genetics, Mutation, Propensity Score, Bile Duct Neoplasms drug therapy, Bile Duct Neoplasms genetics, Bile Duct Neoplasms pathology, Cholangiocarcinoma drug therapy, Cholangiocarcinoma genetics, Cholangiocarcinoma pathology
- 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., (© 2022 UICC.)
- Published
- 2022
- Full Text
- View/download PDF
87. Case Report: Prolonged clinical benefit with sequential trastuzumab-containing treatments in a patient with advanced extramammary Paget disease of the groin.
- Author
-
Zattarin E, Nichetti F, Ligorio F, Mazzeo L, Lobefaro R, Fucà G, Peverelli G, Vingiani A, Bianchi GV, Capri G, de Braud F, and Vernieri C
- Abstract
Extramammary Paget disease (EMPD) is a rare form of cutaneous, intraepithelial adenocarcinoma, which typically presents itself as an erythematous plaque originating from apocrine-gland rich regions, such as the vulva, the perianal region, the scrotum, the penis, or the axilla. EMPD patients typically have a good prognosis, with expected 5-year survival of 60%-92%, but it is estimated that about one-third of EMPD patients will develop lymph node or distant metastases. Treatment approaches for EMPD include locoregional therapies such as broad surgical resection, radiotherapy, or topical imiquimod, when the disease is localized, and chemotherapy and biological agents for advanced EMPD. We report the case of a 58-year-old man diagnosed with locally advanced, symptomatic HER2-overexpressing, AR-positive EMPD, who achieved long-term tumor control with a sequence of several trastuzumab-based treatments (more than 30 months with second-line carboplatin plus paclitaxel plus trastuzumab followed by trastuzumab maintenance; 9 months for third-line vinorelbine plus trastuzumab). Even if it is reported that AR expression occurs concomitantly with HER2 overexpression in more than half of the cases of EMPD, to the best of our knowledge, this is the first case report describing androgen receptor blockade therapy in combination with an anti-HER2 agent. Our patient did not benefit from androgen receptor blockade in combination with trastuzumab, thus suggesting that AR expression may simply reflect an intrinsic characteristic of the EMPD cell of origin, rather than tumor dependence upon AR signaling. Given the reported sensibility to anti-HER2 therapy, also new antibody drug conjugates targeting HER2 are worth exploring in the management of advanced EMPD., Competing Interests: FB reports an advisory role at Roche, EMD Serono, NMS Nerviano Medical Science, Sanofi, MSD, Novartis, Incyte, BMS, Menarini; speaker role for BMS, Healthcare Research and Pharmacoepidemiology, Merck Group, ACCMED, Nadirex, MSD, Pfizer, Servier, Sanofi, Roche, AMGEN, Incyte, Dephaforum; Principal Investigator for Novartis, F.Hoffmann-LaRoche Ltd, BMS, Ignyta Operating INC, Merck Sharp and Dohme Spa, Kymab, Pfizer, Tesaro, MSD, MedImmune LCC, Exelixis Inc., LOXO Oncology Incorporated, DAICHI SANKIO Dev. Limited, Basilea Pharmaceutica International AG, Janssen-Cilag International NV, Merck KGAA. CV reports an advisory role for Novartis; travel grants: Lilly, Novartis, Istituto Gentili, Roche, Pfizer; research grants: Roche. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Zattarin, Nichetti, Ligorio, Mazzeo, Lobefaro, Fucà, Peverelli, Vingiani, Bianchi, Capri, de Braud and Vernieri.)
- Published
- 2022
- Full Text
- View/download PDF
88. Gene mutational profile of BRCAness and clinical implication in predicting response to platinum-based chemotherapy in patients with intrahepatic cholangiocarcinoma.
- Author
-
Rimini M, Macarulla T, Burgio V, Lonardi S, Niger M, Scartozzi M, Rapposelli IG, Aprile G, Ratti F, Pedica F, Verdaguer H, Nappo F, Nichetti F, Lai E, Valgiusti M, Cappetta A, Febregat C, Fassan M, De Braud F, Puzzoni M, Frassineti GL, Simionato F, De Cobelli F, Aldrighetti L, Fornaro L, Cascinu S, and Casadei-Gardini A
- Subjects
- Antineoplastic Agents therapeutic use, Bile Ducts, Intrahepatic, Humans, Mutation, Prognosis, Bile Duct Neoplasms drug therapy, Bile Duct Neoplasms genetics, Cholangiocarcinoma drug therapy, Cholangiocarcinoma genetics, Genetic Profile, Organoplatinum Compounds pharmacology, Organoplatinum Compounds therapeutic use
- Abstract
Background and Aims: Biliary tract cancers are rare malignancies with a poor prognosis and scarce therapeutic strategies. The significance of BRCAness in this setting is already unknown., Method: Tissue specimens of BTC patients treated with platinum-based chemotherapy have been analyzed through the FOUNDATIONPne assay., Results: 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)., Conclusion: 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., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: DR. TERESA MACARULLA MERCADE. Dr. Teresa Macarulla reports honoraria for consultancy from (SOBI) Swedish Orpahn Biovitrum AB, Ability Pharmaceuticals SL, Advance Medical HCMS, S.A., Aptitude Health, AstraZeneca, Basilea Pharma, Baxter, BioLineRX Ltd, Celgene, Eisai, Genzyme, Got It Consulting SL, IATTGI, Imedex, Incyte, Ipsen Bioscience, Inc, Laboratorios Menarini, Lilly. Marketing Farmacéutico & Investigación Clínica, S.L, MDS, Medscape, Monte Verde SA, Novocure, Paraxel, PPD Development, QED Therapeutics, Roche Farma, Sanofi-Aventis, Servier, TRANSWORLD EDITORS, SL and Zymeworks. Dr. Teresa Macarulla declares institutional interest in form of financial support for clinical trials or contracted research: Agios, Aslan, AstraZecena, Bayer, Celgene, Genentech, Hallozyme, Immunomedics, Lilly, Merimarck, Millenim, Novartis, Pfizer, Pharmacyclics, and Roche. DR. HELENA VERDAGUER MATA declares: speaker's Bureau: Astrazeneca. Advisory Role: Merck. OTHER COAUTHORS: Declare no conflict of interest., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
89. Efficacy of mRNA anti-SARS-CoV-2 vaccination and dynamics of humoral immune response in patients with solid tumors: results from the institutional registry of an Italian tertiary cancer center.
- Author
-
Fucà G, Lecchi M, Ciniselli CM, Ottini A, Spagnoletti A, Mazzeo L, Morelli D, Frati P, Stroscia M, Ebrahem E, Sottotetti E, Galli G, D'Elia MG, Lobefaro R, Ducceschi M, Di Guardo L, Bhoori S, Provenzano S, Platania M, Niger M, Colombo E, Nichetti F, Duca M, Rivoltini L, Mortarini R, Baili P, Apolone G, de Braud F, Verderio P, and Damian S
- Abstract
Background: Systemic immunosuppression characterizing cancer patients represents a concern regarding the efficacy of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination, and real-world evidence is needed to define the efficacy and the dynamics of humoral immune response to mRNA-based anti-SARS-CoV-2 vaccines., Methods: We conducted an observational study that included patients with solid tumors who were candidates for mRNA anti-SARS-CoV-2 vaccination at the Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy. The primary objective was to monitor the immunologic response to the mRNA anti-SARS-CoV-2 vaccination in terms of anti-spike antibody levels. All the patients received two doses of the mRNA-1273 vaccine or the BNT162b2 vaccine. Healthcare workers served as a control group of healthy subjects., Results: Among the 243 patients included in the present analysis, 208 (85.60%) and 238 (97.94%) resulted seroconverted after the first and the second dose of vaccine, respectively. Only five patients (2.06%) had a negative titer after the second dose. No significant differences in the rate of seroconversion after two vaccine doses were observed in patients as compared with the control group of healthy subjects. Age and anticancer treatment class had an independent impact on the antibody titer after the second dose of vaccination. In a subgroup of 171 patients with available data about the third timepoint, patients receiving immunotherapy with immune checkpoint inhibitors seem to have a higher peak of antibodies soon after the second dose (3 weeks after), but a more pronounced decrease at a late timepoint (3 months after)., Conclusions: The systemic immunosuppression characterizing cancer patients did not seem to dramatically affect the humoral response to anti-SARS-CoV-2 mRNA vaccines in our population of patients with solid tumors. Further investigation is needed to dissect the interplay between immunotherapy and longitudinal dynamics of humoral response to mRNA vaccines, as well as to analyze the cellular response to mRNA vaccines in cancer patients., Competing Interests: Conflict of interest statement: The authors declare that there is no conflict of interest., (© The Author(s), 2022.)
- Published
- 2022
- Full Text
- View/download PDF
90. MGMT inactivation as a new biomarker in patients with advanced biliary tract cancers.
- Author
-
Niger M, Nichetti F, Casadei-Gardini A, Morano F, Pircher C, Tamborini E, Perrone F, Canale M, Lipka DB, Vingiani A, Agnelli L, Dobberkau A, Hüllein J, Korell F, Heilig CE, Pusceddu S, Corti F, Droz M, Ulivi P, Prisciandaro M, Antista M, Bini M, Cattaneo L, Milione M, Glimm H, Köhler BC, Pruneri G, Hübschmann D, Fröhling S, Mazzaferro V, Pietrantonio F, Di Bartolomeo M, and de Braud F
- Subjects
- Biomarkers, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Humans, O(6)-Methylguanine-DNA Methyltransferase genetics, O(6)-Methylguanine-DNA Methyltransferase metabolism, Precision Medicine, Tumor Suppressor Proteins genetics, Bile Duct Neoplasms, Biliary Tract Neoplasms genetics
- Abstract
Biliary tract cancers (BTCs) have poor prognosis and limited therapeutic options. The impact of O
6 -methylguanine-DNA methyltransferase (MGMT) inactivation in advanced BTC patients is not established. We investigated the prevalence, prognostic, and predictive impact of MGMT inactivation in two multicenter cohorts. MGMT inactivation was assessed through PCR and immunohistochemistry (IHC) in an Italian cohort; the results were then externally validated using RNA sequencing (RNA-seq) data from the BTC subcohort of the Molecularly Aided Stratification for Tumor Eradication Research (MASTER) precision oncology program of the National Center for Tumor Diseases Heidelberg and the German Cancer Consortium. Among 164 Italian cases, 18% presented MGMT promoter hypermethylation (> 14%) and 73% had negative MGMT protein expression. Both were associated with worse overall survival (OS; HR 2.31; P < 0.001 and HR 1.99, P = 0.012, respectively). In the MASTER cohort, patients with lower MGMT mRNA expression showed significantly poorer OS (median OS [mOS] 20.4 vs 31.7 months, unadjusted HR 1.89; P = 0.043). Our results suggest that MGMT inactivation is a frequent epigenetic alteration in BTC, with a significant prognostic impact, and provide the rationale to explore DNA-damaging agents in MGMT-inactivated BTCs., (© 2022 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2022
- Full Text
- View/download PDF
91. Prognostic Role of a New Index Tested in European and Korean Advanced Biliary Tract Cancer Patients: the PECS Index.
- Author
-
Rovesti G, Leone F, Brandi G, Fornaro L, Scartozzi M, Niger M, Yoo C, Caputo F, Filippi R, Casagrande M, Silvestris N, Santini D, Faloppi L, Palloni A, Aglietta M, Vivaldi C, Cho H, Lai E, Fenocchio E, Nichetti F, Pella N, De Lorenzo S, Di Maio M, Vasile E, de Braud F, Jeong JH, Aprile G, Orsi G, Cascinu S, and Casadei-Gardini A
- Subjects
- Humans, Inflammation, Neutrophils pathology, Prognosis, Reproducibility of Results, Republic of Korea epidemiology, Retrospective Studies, Biliary Tract Neoplasms diagnosis, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms pathology, Lymphocytes pathology
- Abstract
Background and Aim: The aim of the present study is to evaluate a new index (PECS (PsECogSii)index) influenced by PS ECOG and systemic immune-inflammation index (SII) in unresectable locally advanced or metastatic BTC patients treated with first-line chemotherapy., Methods: This multicenter, international, study was conducted on a training cohort of 130 patients and in three European and Korean validation cohorts The PECS index was calculated as ECOG × SII index (neutrophil count × platelet count/lymphocyte count). Event-time distributions were estimated using the Kaplan-Meier method and survival curves were compared using the log-rank test., Results: In the training cohort, the median overall survival (mOS) was 13.2 months, 8.7 months, and 3.8 months for patients with PECS-0, PECS-1, and PECS-2, respectively (PECS-0: HR = 1; PECS-1: HR 1.41; PECS-2: HR 3.23) (p < 0.0001). In the first validation cohort, the mOS was 12.8 months, 10.1 months, and 5.3 months for patients with PECS-0, PECS-1, and PECS-2, respectively (PECS-0: HR = 1; PECS-1: HR 1.29; PECS-2: HR 2.40) (p < 0.0001). In the second validation cohort, the mOS was 21.2 months, 10.2 months, and 3.0 months for patients with PECS-0, PECS-1, and PECS-2, respectively (PECS-0: HR = 1; PECS-1: HR 2.25; PECS-2: HR 9.00) (p < 0.0001). In the third validation cohort, the median OS was 15.5 months, 7.5 months, and 3.7 months for patients with PECS-0, PECS-1, and PECS-2, respectively (PECS-0: ref HR = 1; PECS-1: HR 2.14; PECS-2: HR 5.00) (p < 0.0001). Multivariate analysis in all cohorts confirmed the PECS index as an independent prognostic factor for OS., Conclusions: The easy assessment, low cost, and reproducibility make PECS index a promising tool to assess the prognosis of BTC patients in future clinical practice., (© 2021. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
92. Isocitrate Dehydrogenase Mutations in Cholangiocarcinoma: Still a Long Road Ahead.
- Author
-
Nichetti F and Niger M
- Subjects
- Bile Duct Neoplasms genetics, Cholangiocarcinoma genetics, Humans, Bile Duct Neoplasms enzymology, Cholangiocarcinoma enzymology, Isocitrate Dehydrogenase genetics, Mutation
- Abstract
Competing Interests: Monica NigerConsulting or Advisory Role: Incyte, Basilea Pharmaceutical, EMD Serono, MSD/AstraZenecaTravel, Accommodations, Expenses: CelgeneNo other potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
93. Acquired Resistance Mechanisms to PD-L1 Blockade in a Patient With Microsatellite Instability-High Extrahepatic Cholangiocarcinoma.
- Author
-
Niger M, Nichetti F, Dell'Angelo F, Cappelletti V, Pircher C, Vismara M, Cotsoglou C, Bhoori S, Vingiani A, Di Bartolomeo M, Pietrantonio F, de Braud F, Pruneri G, Daidone MG, and Mazzaferro V
- Subjects
- B7-H1 Antigen genetics, Bile Ducts, Intrahepatic pathology, Humans, Microsatellite Instability, Bile Duct Neoplasms drug therapy, Cholangiocarcinoma drug therapy
- Abstract
Competing Interests: Monica NigerConsulting or Advisory Role: Incyte, Basilea Pharmaceutica, EMD Serono, MSD/AstraZenecaTravel, Accommodations, Expenses: Celgene Sherrie BhooriHonoraria: Eisai, Ipsen, Boston ScientificConsulting or Advisory Role: Eisai Maria Di BartolomeoHonoraria: Lilly, MSD Oncology, ServierConsulting or Advisory Role: Lilly, MSD OncologyResearch Funding: LillyTravel, Accommodations, Expenses: Roche, Sanofi Filippo PietrantonioHonoraria: Servier, Bayer, AstraZeneca/MedImmune, Lilly, Sanofi, MSD Oncology, AmgenConsulting or Advisory Role: Amgen, Servier, MSD Oncology, MerckResearch Funding: Bristol Myers Squibb (Inst), AstraZeneca (Inst) Filippo de BraudHonoraria: Roche, Pfizer, BMS, Merck, MSD, Servier, Sanofi, Amgen Astellas BioPharma, IncyteConsulting or Advisory Role: Roche, Incyte, EMD SERONO, Bristol Myers Squibb, Nerviano Medical Sciences, Sanofi, Novartis Italy, NMS Medical Science, MenariniResearch Funding: Novartis (Inst), Roche (Inst), Merck Serono (Inst), Pfizer (Inst), Servier (Inst), Philogen (Inst), Loxo (Inst), Tesaro (Inst), Nerviano Medical Sciences (Inst), Kymab (Inst), Bristol Myers Squibb/Medarex, Merck KGaA, Ignyta, MedImmune, Exelixis, Bayer Health, Daiichi Sankyo Europe GmbH, Incyte, Basilea Pharmaceutical, Janssen Oncology Giancarlo PruneriHonoraria: Novartis, Roche, Genomic HealthConsulting or Advisory Role: ADS BiotecResearch Funding: Roche, Roche Molecular DiagnosticsNo other potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
94. Transforming Growth Factor-β Activated Kinase 1 (Tak1) Is Activated in Hepatocellular Carcinoma, Mediates Tumor Progression, and Predicts Unfavorable Outcome.
- Author
-
Ridder DA, Urbansky LL, Witzel HR, Schindeldecker M, Weinmann A, Berndt K, Gerber TS, Köhler BC, Nichetti F, Ludt A, Gehrke N, Schattenberg JM, Heinrich S, Roth W, and Straub BK
- Abstract
Although knowledge on inflammatory signaling pathways driving cancer initiation and progression has been increasing, molecular mechanisms in hepatocarcinogenesis are still far from being completely understood. Hepatocyte-specific deletion of the MAPKKK Tak1 in mice recapitulates important steps of hepatocellular carcinoma (HCC) development, including the occurrence of cell death, steatohepatitis, dysplastic nodules, and HCCs. However, overactivation of Tak1 in mice upon deletion of its deubiquitinase Cyld also results in steatohepatitis and HCC development. To investigate Tak1 and Cyld in human HCCs, we created a tissue microarray to analyze their expression by immunohistochemistry in a large and well-characterized cohort of 871 HCCs of 561 patients. In the human liver and HCC, Tak1 is predominantly present as its isoform Tak1A and predominantly localizes to cell nuclei. Tak1 is upregulated in diethylnitrosamine-induced mouse HCCs as well as in human HCCs independent of etiology and is further induced in distant metastases. A high nuclear Tak1 expression is associated with short survival and vascular invasion. When we overexpressed Tak1A in Huh7 cells, we observed increased tumor cell migration, whereas overexpression of full-length Tak1 had no significant effect. A combined score of low Cyld and high Tak1 expression was an independent prognostic marker in a multivariate Cox regression model.
- Published
- 2022
- Full Text
- View/download PDF
95. Prolonged benefit from palbociclib plus letrozole in heavily pretreated advanced male breast cancer: case report.
- Author
-
Zattarin E, Ligorio F, Nichetti F, Bianchi G, Capri G, and de Braud F
- Subjects
- Adult, Breast Neoplasms, Male metabolism, Breast Neoplasms, Male pathology, Humans, Letrozole administration & dosage, Male, Piperazines administration & dosage, Prognosis, Pyridines administration & dosage, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms, Male drug therapy, Salvage Therapy
- 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
- 2021
- Full Text
- View/download PDF
96. Risk assessment of thromboembolic events in hospitalized cancer patients.
- Author
-
Nichetti F, Ligorio F, Montelatici G, Porcu L, Zattarin E, Provenzano L, Franza A, Lalli L, de Braud F, and Platania M
- Subjects
- Aged, Female, Humans, Inpatients statistics & numerical data, Male, Middle Aged, Neoplasms epidemiology, Neoplasms complications, Thromboembolism epidemiology
- Abstract
Hospitalized cancer patients are at increased risk for Thromboembolic Events (TEs). As untailored thromboprophylaxis is associated with hemorrhagic complications, the definition of a risk-assessment model (RAM) in this population is needed. INDICATE was a prospective observational study enrolling hospitalized cancer patients, with the primary objective of assessing the Negative Predictive Value (NPV) for TEs during hospitalization and within 45 days from discharge of low-grade Khorana Score (KS = 0). Secondary objectives were to assess KS Positive Predictive Value (PPV), the impact of TEs on survival and the development of a new RAM. Assuming 7% of TEs in KS = 0 patients as unsatisfactory percentage and 3% of as satisfactory, 149 patients were needed to detect the favorable NPV with one-sided α = 0.10 and power = 0.80. Stepwise logistic regression was adopted to identify variables included in a new RAM. Among 535 enrolled patients, 153 (28.6%) had a KS = 0. The primary study objective was met: 29 (5.4%) TEs were diagnosed, with 7 (4.6%) cases in the KS = 0 group (NPV = 95.4%, 95% CI 90.8-98.1%; one-sided p = 0.084). However, the PPV was low (5.7%, 95% CI 1.9-12.8%); a new RAM based on albumin (OR 0.34, p = 0.003), log(LDH) (OR 1.89, p = 0.023) and presence of vascular compression (OR 5.32, p < 0.001) was developed and internally validated. Also, TEs were associated with poorer OS (median, 5.7 vs 24.8 months, p < 0.001). INDICATE showed that the KS has a good NPV but poor PPV for TEs in hospitalized cancer patients. A new RAM was developed, and deserves further assessment in external cohorts., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
97. Impact of Baseline and On-Treatment Glycemia on Everolimus-Exemestane Efficacy in Patients with Hormone Receptor-Positive Advanced Breast Cancer (EVERMET).
- Author
-
Vernieri C, Nichetti F, Lalli L, Moscetti L, Giorgi CA, Griguolo G, Marra A, Randon G, Rea CG, Ligorio F, Scagnoli S, De Angelis C, Molinelli C, Fabbri A, Ferraro E, Trapani D, Milani A, Agostinetto E, Bernocchi O, Catania G, Vantaggiato A, Palleschi M, Moretti A, Basile D, Cinausero M, Ajazi A, Castagnoli L, Lo Vullo S, Gerratana L, Puglisi F, La Verde N, Arpino G, Rocca A, Ciccarese M, Pedersini R, Fabi A, Generali D, Losurdo A, Montemurro F, Curigliano G, Del Mastro L, Michelotti A, Cortesi E, Guarneri V, Pruneri G, Mariani L, and de Braud F
- Subjects
- Androstadienes, Antineoplastic Combined Chemotherapy Protocols adverse effects, Blood Glucose, Female, Humans, Phosphatidylinositol 3-Kinases, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism, Retrospective Studies, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms metabolism, Everolimus
- Abstract
Purpose: The mTOR complex C1 (mTORC1) inhibitor everolimus in combination with the aromatase inhibitor exemestane is an effective treatment for patients with hormone receptor-positive (HR
+ ), HER2-negative (HER2- ), advanced breast cancer (HR+ /HER2- aBC). However, everolimus can cause hyperglycemia and hyperinsulinemia, which could reactivate the PI3K/protein kinase B (AKT)/mTORC1 pathway and induce tumor resistance to everolimus., Experimental Design: We conducted a multicenter, retrospective, Italian study to investigate the impact of baseline and on-treatment (i.e., during first 3 months of therapy) blood glucose levels on progression-free survival (PFS) in patients with HR+ /HER2- aBC treated with everolimus-exemestane., Results: We evaluated 809 patients with HR+ /HER2- aBC treated with everolimus-exemestane as any line of therapy for advanced disease. When evaluated as dichotomous variables, baseline and on-treatment glycemia were not significantly associated with PFS. However, when blood glucose concentration was evaluated as a continuous variable, a multivariable model accounting for clinically relevant patient- and tumor-related variables revealed that both baseline and on-treatment glycemia are associated with PFS, and this association is largely attributable to their interaction. In particular, patients who are normoglycemic at baseline and experience on-treatment diabetes have lower PFS compared with patients who are already hyperglycemic at baseline and experience diabetes during everolimus-exemestane therapy (median PFS, 6.34 vs. 10.32 months; HR, 1.76; 95% confidence interval, 1.15-2.69; P = 0.008)., Conclusions: The impact of on-treatment glycemia on the efficacy of everolimus-exemestane therapy in patients with HR+ /HER2- aBC depends on baseline glycemia. This study lays the foundations for investigating novel therapeutic approaches to target the glucose/insulin axis in combination with PI3K/AKT/mTORC1 inhibitors in patients with HR+ /HER2- aBC., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
98. Poziotinib for EGFR and HER2 exon 20 insertion mutation in advanced NSCLC: Results from the expanded access program.
- Author
-
Prelaj A, Bottiglieri A, Proto C, Lo Russo G, Signorelli D, Ferrara R, Galli G, De Toma A, Viscardi G, Brambilla M, Lobefaro R, Nichetti F, Manglaviti S, Occhipinti M, Labianca A, Ganzinelli M, Gallucci R, Zilembo N, Greco GF, Torri V, de Braud F, and Garassino MC
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung secondary, Compassionate Use Trials, Disease Progression, ErbB Receptors genetics, Exons, Female, Humans, Lung Neoplasms genetics, Lung Neoplasms mortality, Lung Neoplasms pathology, Male, Middle Aged, Progression-Free Survival, Protein Kinase Inhibitors adverse effects, Quinazolines adverse effects, Time Factors, Antineoplastic Agents administration & dosage, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Mutagenesis, Insertional, Protein Kinase Inhibitors administration & dosage, Quinazolines administration & dosage, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 genetics
- Abstract
Background: The treatment of metastatic non-small-cell lung cancer (mNSCLC) patients with EGFR/HER2 exon 20 insertion mutation (i-mut) remains an unmet clinical need. Poziotinib, a new generation tyrosine kinase inhibitor, is currently under investigation as a potential targeted therapy. This compassionate study of its use aims to describe the activity/toxicity of poziotinib in mNSCLC with EGFR/HER2-exon-20-i-mut., Patients and Methods: NSCLC patients who were treated either with EGFR or HER2 exon 20-i-mut within an expanded access program were included in this study. Poziotinib (16 mg or less) was administrated orally quaque die (QD). The primary end-point was the overall response rate (ORR) assessed by central review using RECIST v1.1, and secondary end-points were median progression free survival (PFS), disease control rate (DCR), median overall survival (OS) and toxicity., Results: The median age of all the 30 patients was 58 years (25-80 years), most of them were females (73%); ECOG 0-1 (83%), EGFR i-mut (73%) and pre-treated (83%). 73% started with poziotinib at a dose of 16 mg. At data cut-off, 22 of 33 patients (73%) experienced a progress in the disease and 12 of 30 (40%) died. Median PFS was 5.6 months (95% CI: 3.6-6.7 months) and the mOS 9.5 months (95% CI: 5.3 - not-reached months). The ORR was 30% (EGFR/HER2: 23/50%) and DCR 80%. G3 AEs were reported in 66% of the patients and were found with skin rash (50%), diarrhoea (17.6%), mucositis (7%) and paronychia (3%). G5, possibly associated with pneumonitis might also have occurred., Conclusions: Poziotinib exhibited effects in mNSCLC patients with EGFR/HER2-exon 20-i-mut. The toxicity rate was high leading to frequent dose interruption and reduction, thereby reducing mPFS in patients with good ORR/DCR. ZENITH20 trial is now being used to evaluate the low dose and new scheduled dose (e.g. bis in die (BID))., Competing Interests: Conflict of interest statement M.C.G. declares personal financial interests with the following organizations: AstraZeneca, MSD International GmbH, BMS, Boehringer Ingelheim Italia S.p.A, Celgene, Eli Lilly, Ignyta, Incyte, Inivata, MedImmune, Novartis, Pfizer, Roche, Takeda. F.dB. declares Consultant Advisory Board for Ignyta, BMS, Daiichi Sankyo, Pfizer, Octimet Oncology, Incyte, Teofarma, Pierre Fabre, Roche, EMD Serono, Sanofi, NMS Nerviano Medical Science, Pharm Research Associated (U.K) Ltd; as a Speaker BMS, Roche, MSD, Ignyta, Bayer, ACCMED, Dephaforum S.r.l., Nadirex, Merck, Biotechspert Ltd, PriME Oncology, Pfizer, Servier, Celgene, Tesaro, Loxo Oncology Inc., Sanofi, Healthcare Research & Pharmacoepidemiology, as P.I for Novartis, Roche, BMS, Celgene, Incyte, NMS, Merck KGAA, Kymab, Pfizer, Tesaro, MSD. A.P. declares personal fees from Roche, AstraZeneca and BMS outside the submitted work. C.P. declares personal fees from BMS and MSD, outside the submitted work. G.LR. declares personal fees from BMS, MSD and Astra Zeneca outside the submitted work. D.S. declares personal fees from AstraZeneca, Boehringer Ingelheim and BMS, outside the submitted work. The other authors report no conflict of interest., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
99. Antitumor activity and efficacy of shorter versus longer duration of anthracycline-taxane neoadjuvant chemotherapy in stage II-III HER2-negative breast cancer: a 10-year, retrospective analysis.
- Author
-
Lobefaro R, Zattarin E, Nichetti F, Prisciandaro M, Ligorio F, Brambilla M, Sepe P, Corti F, Peverelli G, Ottini A, Beninato T, Mazzeo L, Rea CG, Mariani G, de Braud F, Bianchi GV, Vernieri C, and Capri G
- Abstract
Background: Neoadjuvant anthracycline-taxane-based chemotherapy (ChT) is a standard of care treatment option for stage II-III breast cancer (BC) patients. However, the optimal duration of neoadjuvant ChT has been poorly investigated so far., Material and Methods: We retrospectively retrieved clinical data of patients with stage II-III human epidermal growth factor receptor 2-negative (HER2-) BC who were treated between October 2007 and January 2018 with neoadjuvant AT (doxorubicin-paclitaxel) for three cycles followed by CMF (cyclophosphamide-methotrexate-5-fluorouracil) for three cycles (cohort A) or with four AT cycles followed by four CMF cycles (cohort B). The aim of our study was to investigate the impact of neoadjuvant ChT duration (cohort A versus cohort B) on pathological complete response (pCR) rates, disease-free survival (DFS) and overall survival (OS)., Results: Of 209 HER2- BC patients included, 62 had triple-negative breast cancer (TNBC) and 147 had hormone receptor-positive (HR+) BC. Median age was 48 years (range 30-74 years). A total of 111 patients belonged to cohort A and 98 patients belonged to cohort B. pCR was detected in 29 (13.9%) patients, 25 (40.3%) of whom had TNBC and four (2.7%) had HR+ HER2- BC. Patients achieving pCR had significantly longer DFS and OS, with statistical significance reached only in patients with TNBC. We found no differences between cohort A and cohort B in terms of pCR rates (15.3% versus 12.2%; p = 0.55), DFS ( p = 0.49) or OS ( p = 0.94). The incidence of grade 3/4 adverse events was similar in cohort A versus cohort B as well (22.5% versus 19.4%; p = 0.54)., Conclusion: Shorter duration of neoadjuvant anthracycline-taxane ChT was not associated with worse clinical outcomes in patients with stage II-III BC. Prospective studies are needed to evaluate whether the duration of neoadjuvant anthracycline-taxane-based ChT can be reduced in specific patient subgroups without negatively affecting clinical outcomes., Competing Interests: Conflict of interest: The authors declare that there is no conflict of interest., (© The Author(s), 2020.)
- Published
- 2020
- Full Text
- View/download PDF
100. Pembrolizumab plus axitinib: another step ahead in advanced renal cell carcinoma.
- Author
-
Procopio G, Nichetti F, and Verzoni E
- Subjects
- Antibodies, Monoclonal, Humanized, Axitinib, Follow-Up Studies, Humans, Sunitinib, Carcinoma, Renal Cell, Kidney Neoplasms
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.