14 results on '"Merz V"'
Search Results
2. Complete pathological response to pembrolizumab in pretreated pancreatic acinar cell carcinoma.
- Author
-
Merz V, Maines F, Marcucci S, Sartori C, Frisinghelli M, Trentin C, Kadrija D, Carbone FG, Michielan A, Gabbrielli A, Melisi D, Barbareschi M, Brolese A, and Caffo O
- Subjects
- Humans, Male, Antineoplastic Agents, Immunological therapeutic use, Middle Aged, Aged, Antibodies, Monoclonal, Humanized therapeutic use, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology, Carcinoma, Acinar Cell drug therapy, Carcinoma, Acinar Cell pathology
- Abstract
Background: Therapeutic approach used for pancreatic ductal adenocarcinoma is usually translated also for the rarer acinar counterpart, which shows a different mutational landscape nevertheless. While dMMR/MSI-H status is rare in the ductal histotype, it appears to be more prevalent in pancreatic acinar cell carcinoma (PACC)., Case Presentation: We report the case of a patient with locally advanced MSI-H PACC in whom the treatment with the anti-PD-1 pembrolizumab, administered as third line, made possible surgical resection, achieving even an exceptional pathological complete response., Conclusions: Treatment of PACC should be tailored based on the peculiar molecular features that distinguish PACC from ductal adenocarcinoma. Evaluation of potentially therapeutically targetable alterations should be mandatory in case of PACC diagnosis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. Is There Room for Liposomal Irinotecan in Biliary Tract Cancer? A Meta-analysis of Randomised Trials.
- Author
-
Merz V, Messina C, Zecchetto C, Quinzii A, Frisinghelli M, Trentin C, Salati M, Caffo O, and Melisi D
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Fluorouracil therapeutic use, Irinotecan, Leucovorin adverse effects, Liposomes therapeutic use, Biliary Tract Neoplasms drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Aims: The combination of 5-fluorouracil/leucovorin (5-FU/LV) plus oxaliplatin (FOLFOX) is widely acknowledged as the standard regimen for second-line treatment in patients with advanced biliary tract cancer. Nanoliposomal irinotecan (nal-IRI) has demonstrated its activity in patients with advanced pancreatic cancer. Recent studies have investigated the activity of nal-IRI in combination with 5-FU/LV for biliary tract cancer. However, the results have been contradictory. We conducted a meta-analysis to assess survival outcomes and response rates in randomised trials investigating the activity of nal-IRI in previously treated biliary tract cancer patients., Materials and Methods: We systematically collected potentially relevant findings from PubMed/Medline, the Cochrane library and EMBASE. Abstracts presented at major international oncological meetings were also reviewed. We extracted hazard ratios and 95% confidence intervals for progression-free survival and overall survival, as well as odds ratios and 95% confidence intervals for objective response rate. The outcomes of the accessible randomised studies evaluating the activity of nal-IRI plus 5-FU/LV were analysed., Results: The combination therapy exhibited a statistically significant decrease in the risk of progression (hazard ratio 0.70; 95% confidence interval 0.50-0.97) when compared with 5-FU/LV alone. Additionally, the dual regimen yielded longer overall survival and a higher objective response rate., Conclusion: Our meta-analysis showed that nal-IRI plus 5-FU/LV had a superior activity in comparison with 5-FU/LV. Further investigations are required to elucidate the role of nal-IRI in this setting and to identify subgroups of patients who could derive the greatest benefit from its administration., (Copyright © 2023 The Royal College of Radiologists. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Autotaxin Secretion Is a Stromal Mechanism of Adaptive Resistance to TGFβ Inhibition in Pancreatic Ductal Adenocarcinoma.
- Author
-
Pietrobono S, Sabbadini F, Bertolini M, Mangiameli D, De Vita V, Fazzini F, Lunardi G, Casalino S, Scarlato E, Merz V, Zecchetto C, Quinzii A, Di Conza G, Lahn M, and Melisi D
- Subjects
- Humans, Animals, Mice, Gemcitabine, Transforming Growth Factor beta, Signal Transduction, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology
- Abstract
The TGFβ receptor inhibitor galunisertib demonstrated efficacy in patients with pancreatic ductal adenocarcinoma (PDAC) in the randomized phase II H9H-MC-JBAJ study, which compared galunisertib plus the chemotherapeutic agent gemcitabine with gemcitabine alone. However, additional stromal paracrine signals might confer adaptive resistance that limits the efficacy of this therapeutic strategy. Here, we found that autotaxin, a secreted enzyme that promotes inflammation and fibrosis by generating lysophosphatidic acid (LPA), mediates adaptive resistance to TGFβ receptor inhibition. Blocking TGFβ signaling prompted the skewing of cancer-associated fibroblasts (CAF) toward an inflammatory (iCAF) phenotype. iCAFs were responsible for a significant secretion of autotaxin. Paracrine autotaxin increased LPA-NFκB signaling in tumor cells that triggered treatment resistance. The autotaxin inhibitor IOA-289 suppressed NFκB activation in PDAC cells and overcame resistance to galunisertib and gemcitabine. In immunocompetent orthotopic murine models, IOA-289 synergized with galunisertib in restoring sensitivity to gemcitabine. Most importantly, treatment with galunisertib significantly increased plasma levels of autotaxin in patients enrolled in the H9H-MC-JBAJ study, and median progression-free survival was significantly longer in patients without an increase of autotaxin upon treatment with galunisertib compared with those with increased autotaxin. These results establish that autotaxin secretion by CAFs is increased by TGFβ inhibition and that circulating autotaxin levels predict response to the combination treatment approach of gemcitabine plus galunisertib., Significance: TGFβ inhibition skews cancer-associated fibroblasts toward an inflammatory phenotype that secretes autotaxin to drive adaptive resistance in PDAC, revealing autotaxin as a therapeutic target and biomarker of galunisertib response., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
5. Perioperative NALIRIFOX in patients with resectable pancreatic ductal adenocarcinoma: The open-label, multicenter, phase II nITRO trial.
- Author
-
Melisi D, Zecchetto C, Merz V, Malleo G, Landoni L, Quinzii A, Casalino S, Fazzini F, Gaule M, Pesoni C, Casetti L, Esposito A, Marchegiani G, Piazzola C, D'Onofrio M, de Robertis R, Gabbrielli A, Bernardoni L, Crino SF, Pietrobono S, Luchini C, Aliberti C, Martignoni G, Milleri S, Butturini G, Scarpa A, Salvia R, and Bassi C
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Fluorouracil, Irinotecan adverse effects, Leucovorin, Neoadjuvant Therapy adverse effects, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms surgery, Pancreatic Neoplasms pathology, Adenocarcinoma pathology
- Abstract
Background: Upfront surgery followed by postoperative treatment is a commonly adopted treatment for resectable pancreatic ductal adenocarcinoma (rPDAC). However, the risk of positive surgical margins, the poor recovery that often impairs postoperative treatments, and the risk of recurrence might limit the outcome of this strategy. This study evaluated the safety and the activity of liposomal irinotecan 50 mg/m
2 + 5-fluorouracil 2400 mg/m2 + leucovorin 400 mg/m2 + oxaliplatin 60 mg/m2 (NALIRIFOX) in the perioperative treatment of patients with rPDAC., Methods: Eligible patients had a rPDAC with < 180° interface with major veins' wall. Patients received 3 cycles before and 3 cycles after resection with NALIRIFOX, days 1 and 15 of a 28-day cycle. The primary endpoint was the proportion of patients undergoing an R0 resection., Results: 107 patients began preoperative treatment. Nine patients discontinued the treatment because of related or unrelated adverse events. Disease-control rate was 92.9%. 87 patients underwent surgical exploration, 11 had intraoperative evidence of metastatic disease, and 1 died for surgical complications. R0 resection rate was 65.3%. 49 patients completed the three postoperative cycles. The most common grade ≥ 3 adverse events were diarrhea and neutropenia. Median overall survival (OS) of ITT patients was 32.3 months (95% CI 27.8-44.3). Median disease-free and OS from surgery of resected patients were 19.3 (95% CI 12.6-34.1) and 40.3 months (95% CI 29-NA), respectively., Conclusion: Perioperative NALIRIFOX was manageable and active, and deserves further investigation in randomized trials comparing it with standard upfront surgery followed by adjuvant therapy., Competing Interests: Declaration of Competing Interest DM received honoraria as an advisory board member or consultant from Servier, Incyte, iOnctura, Eli Lilly, Evotec, Baxter; received institutional support for research project from Shire, Celgene, Incyte, iOnctura, Roche. GM received honoraria as consultant from Oncosil, and institutional support for research project from Fibrogen. MDO received honoraria as an advisory board member or consultant from Bracco Diagnostics, Siemens Healthcare Diagnostic, Hitachi, Novartis. RdR received honoraria as an advisory board member or consultant from Bracco Diagnostics and Fujifilm. AS received honoraria as an advisory board member or consultant from Incyte, MSD, Amgen, GlaxoSmithkline. All other authors have declared no conflicts of interest., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
6. The role of nanoliposomal irinotecan plus fluorouracil/leucovorin in the continuum of care of patients with metastatic pancreatic ductal adenocarcinoma.
- Author
-
Procaccio L, Merz V, Fasano M, Vaccaro V, Giommoni E, Pretta A, Noventa S, Satolli MA, Giordano G, Zichi C, Pinto C, Zecchetto C, Barsotti G, De Vita F, Milella M, Antonuzzo L, Scartozzi M, Zaniboni A, Spadi R, Casalino S, Bergamo F, De Toni C, Melisi D, and Lonardi S
- Subjects
- Humans, Middle Aged, Irinotecan, Leucovorin adverse effects, Fluorouracil adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Gemcitabine, Continuity of Patient Care, Camptothecin, Pancreatic Neoplasms, Pancreatic Neoplasms pathology, Adenocarcinoma drug therapy
- Abstract
Background: The NAPOLI-I trial showed better outcome of nanoliposomal irinotecan (nal-IRI) plus 5-fluorouracil/leucovorin (5-FU/LV) compared to 5-FU/LV in patients with advanced pancreatic ductal adenocarcinoma cancer (advPDAC) progressed to gemcitabine-based therapy. This study aims to explore the real-world efficacy and safety of 5-FU/LV-nal-IRI., Methods: This is a retrospective multicenter analysis including advPDAC patients receiving 5-FU/LV-nal-IRI after failure of gemcitabine-based therapy. Survival analyses were performed using Kaplan-Meier method, univariate and multivariate analyses by Cox regression., Results: A total of 296 patients (median age 64.4 years, ECOG PS ≥1 in 56% of cases) were treated at 11 Italian institutions between 2016 and 2018. 34% of them underwent primary tumor resection, and 79% received gemcitabine-nabpaclitaxel as first line. 5-FU/LV-nal-IRI was administered as second-line in 73% of cases. Objective response and disease control rate were 12% and 41%, respectively. Treatment was well tolerated with dose reductions in 50% of patients but no one permanent discontinuation; the commonest grade ≥3 toxicities were neutropenia (14%) and diarrhea (12%). Median PFS and OS from 5-FU/LV-nal-IRI initiation was 3.2 and 7.1 months, respectively., Conclusions: These real-world data confirm the 5-FU/LV-nal-IRI efficacy and safety in advPDAC patients progressed to gemcitabine-based therapy, with outcomes comparable to NAPOLI-1, even in a less-selected population and with more modern therapeutic algorithm., (© 2023 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
7. Exceptional Clinical Response to Alectinib in Pancreatic Acinar Cell Carcinoma With a Novel ALK-KANK4 Gene Fusion.
- Author
-
Gaule M, Pesoni C, Quinzii A, Zecchetto C, Casalino S, Merz V, Contarelli S, Pietrobono S, Vissio E, Molinaro L, Manzin E, Volpatto R, Vellani G, and Melisi D
- Subjects
- Anaplastic Lymphoma Kinase genetics, Ankyrin Repeat genetics, Carcinoma, Acinar Cell genetics, Gene Fusion, Humans, Male, Middle Aged, Pancreatic Neoplasms genetics, Treatment Outcome, Carbazoles therapeutic use, Carcinoma, Acinar Cell drug therapy, Pancreatic Neoplasms drug therapy, Piperidines therapeutic use
- Abstract
Competing Interests: Valeria MerzConsulting or Advisory Role: Amgen Davide MelisiConsulting or Advisory Role: Baxter, Lilly, Evotec, Incyte, iOncturaResearch Funding: Shire, Celgene, Incyte, iOnctura, RocheNo other potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
8. Safety and activity of the TGFβ receptor I kinase inhibitor galunisertib plus the anti-PD-L1 antibody durvalumab in metastatic pancreatic cancer.
- Author
-
Melisi D, Oh DY, Hollebecque A, Calvo E, Varghese A, Borazanci E, Macarulla T, Merz V, Zecchetto C, Zhao Y, Gueorguieva I, Man M, Gandhi L, Estrem ST, Benhadji KA, Lanasa MC, Avsar E, Guba SC, and Garcia-Carbonero R
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, B7-H1 Antigen metabolism, Disease Progression, Europe, Female, Humans, Immune Checkpoint Inhibitors adverse effects, Immune Checkpoint Inhibitors pharmacokinetics, Male, Middle Aged, Neoplasm Metastasis, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Progression-Free Survival, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Pyrazoles adverse effects, Pyrazoles pharmacokinetics, Quinolines adverse effects, Quinolines pharmacokinetics, Receptor, Transforming Growth Factor-beta Type I metabolism, Republic of Korea, Signal Transduction, Time Factors, United States, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, B7-H1 Antigen antagonists & inhibitors, Immune Checkpoint Inhibitors therapeutic use, Pancreatic Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use, Quinolines therapeutic use, Receptor, Transforming Growth Factor-beta Type I antagonists & inhibitors
- Abstract
Background: We assessed the safety, efficacy, and pharmacokinetics of the transforming growth factor beta (TGFβ) receptor inhibitor galunisertib co-administered with the anti-programmed death-ligand 1 (PD-L1) antibody durvalumab in recurrent/refractory metastatic pancreatic cancer previously treated with ≤2 systemic regimens., Methods: This was a two-part, single-arm, multinational, phase Ib study. In a dose-finding phase, escalating oral doses of galunisertib were co-administered on days 1-14 with fixed-dose intravenous durvalumab 1500 mg on day 1 every 4 weeks (Q4W), followed by an expansion cohort phase., Results: The galunisertib recommended phase II dose (RP2D) when co-administered with durvalumab 1500 mg Q4W was 150 mg two times per day. No dose-limiting toxicities were recorded. Among 32 patients treated with galunisertib RP2D, 1 patient had partial response, 7 had stable disease, 15 had objective progressive disease, and 9 were not evaluable. Disease control rate was 25.0%. Median overall survival and progression-free survival were 5.72 months (95% CI: 4.01 to 8.38) and 1.87 months (95% CI: 1.58 to 3.09), respectively. Pharmacokinetic profiles for combination therapy were comparable to those published for each drug. There was no association between potential biomarkers and treatment outcomes., Conclusion: Galunisertib 150 mg two times per day co-administered with durvalumab 1500 mg Q4W was tolerable. Clinical activity was limited. Studying this combination in patients in an earlier line of treatment or selected for predictive biomarkers of TGFβ inhibition might be a more suitable approach., Trial Registration Number: ClinicalTrials.gov identifier: NCT02734160., Competing Interests: Competing interests: DM has received research funding from Celgene, Incyte, and Shire, and has a consulting role with Baxter, Eli Lilly and Company, Incyte, and Shire. D-YO has received research funding from Array, AstraZeneca, Eli Lilly and Company, and Novartis, and has a consulting/advisory role with ASLAN, AstraZeneca, Bayer, Celgene, Genentech/Roche, Halozyme, Merck Serono, Novartis, Taiho, and Zymeworks. AH has received travel and accommodation expenses from Eli Lilly and Company. EC has received research funding from AbbVie, Amcure, Amgen, AstraZeneca, BeiGene, BMS, Boehringer-Ingelheim, CytomX, Eli Lilly and Company, H3, Incyte, Kura, LOXO, Macrogenics, Menarini, Merck, Merck Serono, Merus, Millennium Pharmaceuticals, Nanobiotix Janssen, Nektar, Novartis, Pfizer, PharmaMar, Principia Bayer, PsiOxus Therapeutics, PUMA, Rigontec, Roche/Genentech, Sanofi, Tahio, Tesaro; has a consulting/advisory role with AbbVie, Amcure, AstraZeneca, Boehringer-Ingelheim, Celgene, Cerulean Pharma, EUSA, GLG, Guidepoint Global, Janssen-Cilag, Nanobiotix Janssen, Novartis, Pfizer, Pierre Pharma, PsiOxus Therapeutics, Roche/Genentech, Seattle Genetics, Servier; is employed by HM Hospitals Group and START; has shares in HM Hospitals Group, International Cancer Consultants, Oncoart Associated, and START; and is president and founder of NPO Foundation Intheos (Investigational Therapeutics in Oncological Sciences). AMV has participated in clinical trials funded by Bristol Myers Squibb, Eli Lilly and Company, GlaxoSmithKline, Silenseed, and Verastem. EB has received honoraria for consultancy from Corcept Therapeutics and Invitae, is on the speaker bureau for Ipsen, and his institution has received research funding from Biontech, Bristol-Myers Squibb, Daiichi Sankyo, Eli Lilly and Company, Helix, Mabvax, Merck, Minneamrita Therapeutics, Pharmacyclics, and Samumed. TM has received honoraria for consultancy from Baxalta, Baxter, Celgene, Genzyme, Roche, Sanofi, Shire Pharmaceuticals, Tesaro, and QED Therapeutics, and has received travel/accommodation compensation from Bayer, H3 Biomedicine, Merck, and Sanofi. VM and CZ have no conflicts of interest to disclose. YZ, IG, MM, LG, STE, and EA are employees and stock holders of Eli Lilly and Company. KAB was an employee of Eli Lilly and Company at the time this research was conducted and is a stock holder of Eli Lilly and Company and a current employee of Taiho Oncology. MCL is a current employee and stock holder of AstraZeneca. SCG was an employee of Eli Lilly and Company at the time this research was conducted and is a stock holder of Eli Lilly and Company. RG-C declares having provided scientific advice and/or received honoraria from AAA, Advanz Pharma, Amgen, Bayer, BMS, Eli Lilly and Company, HMP, Ipsen, Merck, Midatech Pharma, MSD, Novartis, PharmaMar, Pfizer, Roche, and Sanofi, and has received research support from Pfizer and BMS. Work in the unit of DM was partially supported by the Associazione Italiana per la Ricerca sul Cancro (AIRC) Investigator Grant n°23719 and 5x1000 Grant n°12182, by the Italian Ministry of Health Ricerca Finalizzata 2016 GR-2016- 02361134 grant, and by the patients associations 'Nastro Viola' and 'Voglio il Massimo' donations., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
9. Plasma IL8 Is a Biomarker for TAK1 Activation and Predicts Resistance to Nanoliposomal Irinotecan in Patients with Gemcitabine-Refractory Pancreatic Cancer.
- Author
-
Merz V, Zecchetto C, Santoro R, Simionato F, Sabbadini F, Mangiameli D, Piro G, Cavaliere A, Deiana M, Valenti MT, Bazan D, Fedele V, Lonardi S, and Melisi D
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor blood, Cell Line, Tumor, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Female, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Irinotecan pharmacology, Irinotecan therapeutic use, MAP Kinase Kinase Kinases metabolism, Male, Mice, Middle Aged, Pancreatic Neoplasms blood, Pancreatic Neoplasms genetics, Prospective Studies, Response Evaluation Criteria in Solid Tumors, Transcriptional Activation, Xenograft Model Antitumor Assays, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols pharmacology, Drug Resistance, Neoplasm genetics, Interleukin-8 blood, MAP Kinase Kinase Kinases genetics, Pancreatic Neoplasms drug therapy
- Abstract
Purpose: Pancreatic cancer is one of the most lethal solid tumors, mainly because of its intrinsic chemoresistance. We identified TAK1 as a central hub sustaining this resistance. Nanoliposomal irinotecan (nal-IRI) is a novel treatment for metastatic gemcitabine-refractory pancreatic cancer. We endeavored to identify circulating markers for TAK1 activation predicting chemoresistance in this setting., Experimental Design: In vivo activity of nal-IRI was validated in an orthotopic nude murine model expressing TAK1-specific shRNA. Plasma concentration of 20 different cytokines were measured by a multiplex xMAP/Luminex technology in patients prospectively enrolled to receive nal-IRI plus 5-fluorouracil/leucovorin (5-FU/LV). The optimal cutoff thresholds able to significantly predict patients' outcome were obtained on the basis of the maximization of the Youden's statistics., Results: Differential expression profiling revealed the gene coding for IL8 as the most significantly downregulated in shTAK1 pancreatic cancer cell lines. Mice bearing shTAK1 tumors had significantly lower plasma levels of IL8 and experienced a significant reduction in tumor growth if treated with nal-IRI, whereas those bearing TAK1-proficient tumors were resistant to this agent. In a discovery cohort of 77 patients, IL8 was the circulating factor most significantly correlated with survival (plasma levels lower vs higher than cutoff: mPFS 3.4 months vs 2.8 months; hazard ratio [HR], 2.55; 95% CI, 1.39-4.67; P = 0.0017; median overall survival 8.9 months vs 5.3 months; HR, 3.51; 95% CI, 0.84-6.68; P = 4.9e-05). These results were confirmed in a validation cohort of 50 patients., Conclusions: Our study identified IL8 as the most significant circulating factor for TAK1 pathway activation and candidates IL8 as a potential predictive biomarker of resistance to nal-IRI in gemcitabine-refractory patients with pancreatic cancer., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
10. Modulating TAK1 Expression Inhibits YAP and TAZ Oncogenic Functions in Pancreatic Cancer.
- Author
-
Santoro R, Zanotto M, Simionato F, Zecchetto C, Merz V, Cavallini C, Piro G, Sabbadini F, Boschi F, Scarpa A, and Melisi D
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Albumins administration & dosage, Albumins pharmacology, Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cell Line, Tumor, Female, Heterocyclic Compounds, 3-Ring administration & dosage, Heterocyclic Compounds, 3-Ring pharmacology, Heterografts, Humans, MAP Kinase Kinase Kinases genetics, MAP Kinase Kinase Kinases metabolism, Maleimides administration & dosage, Maleimides pharmacology, Mice, Mice, Nude, Paclitaxel administration & dosage, Paclitaxel pharmacology, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Random Allocation, Trans-Activators genetics, Trans-Activators metabolism, Transcriptional Coactivator with PDZ-Binding Motif Proteins, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing antagonists & inhibitors, MAP Kinase Kinase Kinases biosynthesis, Pancreatic Neoplasms metabolism, Trans-Activators antagonists & inhibitors
- Abstract
YAP and TAZ are central determinants of malignancy; however, their functions remain still undruggable. We identified TGFβ-activated kinase 1 (TAK1) as a central hub integrating the most relevant signals sustaining pancreatic cancer aggressiveness and chemoresistance. Glycogen synthase kinase (GSK)3 is known to stabilize TAK1, and its inhibition causes a reduction in TAK1 levels. Here, we hypothesized that TAK1 could sustain YAP/TAZ program, and thus, modulation of TAK1 expression through the inhibition of GSK3 could impair YAP/TAZ functions in pancreatic cancer.Differentially expressed transcripts between pancreatic cancer cells expressing scramble or TAK1 -specific shRNA were annotated for functional interrelatedness by ingenuity pathway analysis. TAK1 expression was modulated by using different GSK3 inhibitors, including LY2090314. In vivo activity of LY2090314 alone or in combination with nab-paclitaxel was evaluated in an orthotopic nude mouse model.Differential gene expression profiling revealed significant association of TAK1 expression with HIPPO and ubiquitination pathways. We measured a significant downregulation of YAP/TAZ and their regulated genes in shTAK1 cells. TAK1 prevented YAP/TAZ proteasomal degradation in a kinase independent manner, through a complex with TRAF6, thereby fostering their K63-ubiquitination versus K48-ubiquitination. Pharmacologic modulation of TAK1 by using GSK3 inhibitors significantly decreased YAP/TAZ levels and suppressed their target genes and oncogenic functions. In vivo , LY2090314 plus nab-paclitaxel significantly prolonged mice survival duration.Our study demonstrates a unique role for TAK1 in controlling YAP/TAZ in pancreatic cancer. LY2090314 is a novel agent that warrants further clinical development in combination with nab-paclitaxel for the treatment of pancreatic cancer., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
11. Population pharmacokinetics and exposure-overall survival analysis of the transforming growth factor-β inhibitor galunisertib in patients with pancreatic cancer.
- Author
-
Gueorguieva I, Tabernero J, Melisi D, Macarulla T, Merz V, Waterhouse TH, Miles C, Lahn MM, Cleverly A, and Benhadji KA
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Female, Half-Life, Humans, Male, Middle Aged, Pancreatic Neoplasms pathology, Pyrazoles administration & dosage, Quinolines administration & dosage, Randomized Controlled Trials as Topic, Survival Analysis, Young Adult, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols administration & dosage, CA-19-9 Antigen metabolism, Pancreatic Neoplasms drug therapy
- Abstract
Purpose: To evaluate the exposure-overall survival (OS) relationship in patients with advanced pancreatic cancer treated with galunisertib plus gemcitabine (GG) or gemcitabine plus placebo (GP)., Methods: Galunisertib 300 mg/day was given orally as intermittent dosing and gemcitabine as per label. Galunisertib exposure metrics for each patient in the GG arm (n = 99) of a phase 2 study of pancreatic cancer were calculated. Parametric survival models were used to identify influential baseline and response covariates on OS., Results: The population pharmacokinetics dataset included data from 297 patients/healthy subjects (age: 22-84 years, weight: 39-126 kg) across multiple studies, including this pancreatic cancer study. Galunisertib was rapidly absorbed with peak concentrations attained within 0.5-2 h and had an elimination half-life of 8 h. Between-subject variance on apparent clearance was estimated to be 47%. Age was the only characteristic to have a statistically significant effect on apparent clearance. A parametric Weibull survival model with treatment effect (dose) estimated a hazard ratio of 0.796, after adjusting for patient baseline factors that were significantly associated with OS. There was also a flat daily exposure-OS relationship within the observed exposure range, once all significant baseline covariates were included. Response covariates, such as reduction in CA19-9, time on treatment, and cumulative exposure over treatment cycles were also identified as significant factors for OS for patients with pancreatic cancer., Conclusions: This analysis suggests that 300 mg/day galunisertib administered as 150 mg twice daily for 14 days on/14 days off treatment is an appropriate dosing regimen for patients with pancreatic cancer.
- Published
- 2019
- Full Text
- View/download PDF
12. Outcomes of Primary Chemotherapy for Borderline Resectable and Locally Advanced Pancreatic Ductal Adenocarcinoma.
- Author
-
Maggino L, Malleo G, Marchegiani G, Viviani E, Nessi C, Ciprani D, Esposito A, Landoni L, Casetti L, Tuveri M, Paiella S, Casciani F, Sereni E, Binco A, Bonamini D, Secchettin E, Auriemma A, Merz V, Simionato F, Zecchetto C, D'Onofrio M, Melisi D, Bassi C, and Salvia R
- Subjects
- Adenocarcinoma surgery, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Female, Fluorouracil administration & dosage, Humans, Irinotecan administration & dosage, Leucovorin administration & dosage, Logistic Models, Male, Middle Aged, Oxaliplatin administration & dosage, Paclitaxel administration & dosage, Pancreatic Neoplasms surgery, Prospective Studies, Survival Analysis, Gemcitabine, Pancreatic Neoplasms, Adenocarcinoma drug therapy, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Pancreatic Neoplasms drug therapy
- Abstract
Importance: Chemotherapy is the recommended induction strategy in borderline resectable and locally advanced pancreatic ductal adenocarcinoma. However, the associated results on an intention-to-treat basis are poorly understood., Objective: To investigate pragmatically the treatment compliance, conversion to surgery, and survival outcomes of patients with borderline resectable and locally advanced pancreatic ductal adenocarcinoma undergoing primary chemotherapy., Design, Setting, and Participants: This prospective study took place in a national referral center for pancreatic diseases in Italy. Consecutive patients with borderline resectable and locally advanced pancreatic ductal adenocarcinoma were enrolled at the time of diagnosis (January 2013 through December 2015) and followed up to June 2018., Exposures: The chemotherapy regimen, assigned based on multidisciplinary evaluation, was delivered either at a hub center or at spoke centers. By convention, primary chemotherapy was considered completed after 6 months. After restaging, surgical candidates were selected based on radiologic and biochemical response. All surgeries were carried out at the hub center., Main Outcomes and Measures: Rates of receipt and completion of chemotherapy, rates of conversion to surgery, and disease-specific survival., Results: Of 680 patients, 267 (39.3%) had borderline resectable and 413 (60.7%) had locally advanced pancreatic ductal adenocarcinoma. Overall, 66 patients (9.7%) were lost to follow-up. The rate of chemotherapy receipt was 92.9% (n = 570). The chemotherapeutic regimens most commonly used included FOLFIRINOX (fluorouracil, leucovorin, oxaliplatin, and irinotecan) (260 [45.6%]) and gemcitabine plus nanoparticle albumin-bound-paclitaxel (123 [21.6%]). Nineteen patients (3.3%) receiving chemotherapy died within 6 months, mainly for disease progression. The treatment completion rate was 71.6% (408 of 570). The overall rate of resection was 15.1% (93 of 614) (borderline resectable, 60 of 249 [24.1%]; locally advanced, 33 of 365 [9%]; resection:exploration ratio, 63.3%). Independent predictors of resection were age, borderline resectable disease, chemotherapy completion, radiologic response, and biochemical response. The median survival for the whole cohort was 12.8 (95% CI, 11.7-13.9) months. Factors independently associated with survival were completion of chemotherapy, receipt of complementary radiation therapy, and resection. In patients who underwent resection, the median survival was 35.4 (95% CI, 27.0-43.7) months for initially borderline resectable and 41.8 (95% CI, 27.5-56.1) months for initially locally advanced disease. No pretreatment and posttreatment factors were associated with survival after pancreatectomy., Conclusions and Relevance: This pragmatic observational cohort study with an intention-to-treat design provides real-world evidence of outcomes associated with the most current primary chemotherapy regimens used for borderline resectable and locally advanced pancreatic ductal adenocarcinoma.
- Published
- 2019
- Full Text
- View/download PDF
13. Adipocytes sustain pancreatic cancer progression through a non-canonical WNT paracrine network inducing ROR2 nuclear shuttling.
- Author
-
Carbone C, Piro G, Gaianigo N, Ligorio F, Santoro R, Merz V, Simionato F, Zecchetto C, Falco G, Conti G, Kamga PT, Krampera M, Di Nicolantonio F, De Franceschi L, Scarpa A, Tortora G, and Melisi D
- Subjects
- Cell Line, Cell Nucleus metabolism, Epithelial-Mesenchymal Transition genetics, Humans, Mesenchymal Stem Cells, Models, Biological, Receptor Tyrosine Kinase-like Orphan Receptors genetics, Signal Transduction genetics, Wnt Proteins genetics, Adipocytes cytology, Pancreatic Neoplasms metabolism, Receptor Tyrosine Kinase-like Orphan Receptors metabolism, Wnt Proteins metabolism
- Abstract
Background: Solid epidemiological evidences connect obesity with incidence, stage and survival in pancreatic cancer. However, the underlying mechanistic basis linking adipocytes to pancreatic cancer progression remain largely elusive. We hypothesized that factors secreted by adipocytes could be responsible for epithelial-to-mesenchymal transition (EMT) induction and, in turn, a more aggressive phenotype in models of pancreatic preneoplastic lesions., Methods: We studied the role of factors secreted by two adipogenic model systems from primary human bone marrow stromal cells (hBMSCs) in an in vitro experimental cell transformation model system of human pancreatic ductal epithelial (HPDE) cell stably expressing activated KRAS (HPDE/KRAS),Results:We measured a significant induction of EMT and aggressiveness in HPDE and HPDE/KRAS cell lines when cultured with medium conditioned by fully differentiated adipocytes (ADIPO
CM ) if compared with the same cells cultured with medium conditioned by hBMSC (hBMSCCM ) from two different healthy donors. Several genes coding for soluble modulators of the non-canonical WNT signaling pathway, including FRZB, SFRP2, RSPO1, WNT5A and 5B were significantly overexpressed in fully differentiated adipocytes than in their respective in hBMSC. ADIPOCM induced the overexpression and the nuclear translocation of the Frizzled family member receptor tyrosine kinase-like orphan receptor (Ror) 2 in HPDE and HPDE/KRAS cells. Vantictumab, an anti-Frizzled monoclonal antibody, reduced ROR2 nuclear translocation and in turn the EMT and aggressiveness in HPDE and HPDE/KRAS cells., Conclusions: We demonstrated that adipocytes could induce EMT and aggressiveness in models of pancreatic preneoplastic lesions by orchestrating a complex paracrine signaling of soluble modulators of the non-canonical WNT signaling pathway that determine, in turn, the activation and nuclear translocation of ROR2. This signaling pathway could represent a novel target for pancreatic cancer chemoprevention. Most importantly, these factors could serve as novel biomarkers to select a risk population among obese subjects for screening and, thus, early diagnosis of pancreatic cancer.- Published
- 2018
- Full Text
- View/download PDF
14. Adipocytes sustain pancreatic cancer progression through a non-canonical WNT paracrine network inducing ROR2 nuclear shuttling
- Author
-
Carmine Carbone, Francesca Ligorio, F. Di Nicolantonio, Aldo Scarpa, Paul Takam Kamga, G Conti, L De Franceschi, Francesca Simionato, Davide Melisi, Geny Piro, Giampaolo Tortora, Nicola Gaianigo, Camilla Zecchetto, Valeria Merz, Mauro Krampera, Geppino Falco, Raffaela Santoro, Carbone, C, Piro, G, Gaianigo, N, Ligorio, F, Santoro, R, Merz, V, Simionato, F, Zecchetto, C, Falco, G, Conti, G, Kamga, P T, Krampera, M, Di Nicolantonio, F, De Franceschi, L, Scarpa, A, Tortora, G, and Melisi, D
- Subjects
0301 basic medicine ,EXPRESSION ,medicine.medical_specialty ,Frizzled ,Epithelial-Mesenchymal Transition ,Endocrinology, Diabetes and Metabolism ,adipocytes ,pancreatic cancer ,Medicine (miscellaneous) ,Receptor tyrosine kinase-like orphan receptor ,MICROENVIRONMENT ,Biology ,Receptor Tyrosine Kinase-like Orphan Receptors ,pancreatic cancer, epithelial-to-mesenchymal transition, adipocytes ,Models, Biological ,THERAPY ,Cell Line ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Internal medicine ,Pancreatic cancer ,medicine ,Humans ,Epithelial–mesenchymal transition ,RSPO1 ,BODY-MASS INDEX ,STEM-CELLS ,OBESITY ,RESISTANCE ,RISK ,ADENOCARCINOMA ,METAANALYSIS ,Cell Nucleus ,Nutrition and Dietetics ,Wnt signaling pathway ,Mesenchymal Stem Cells ,medicine.disease ,Pancreatic Neoplasms ,Wnt Proteins ,030104 developmental biology ,Endocrinology ,Adipogenesis ,030220 oncology & carcinogenesis ,Cancer research ,Original Article ,epithelial-to-mesenchymal transition ,Signal Transduction - Abstract
Background: Solid epidemiological evidences connect obesity with incidence, stage and survival in pancreatic cancer. However, the underlying mechanistic basis linking adipocytes to pancreatic cancer progression remain largely elusive. We hypothesized that factors secreted by adipocytes could be responsible for epithelial-to-mesenchymal transition (EMT) induction and, in turn, a more aggressive phenotype in models of pancreatic preneoplastic lesions. Methods: We studied the role of factors secreted by two adipogenic model systems from primary human bone marrow stromal cells (hBMSCs) in an in vitro experimental cell transformation model system of human pancreatic ductal epithelial (HPDE) cell stably expressing activated KRAS (HPDE/KRAS), Results: We measured a significant induction of EMT and aggressiveness in HPDE and HPDE/KRAS cell lines when cultured with medium conditioned by fully differentiated adipocytes (ADIPOCM) if compared with the same cells cultured with medium conditioned by hBMSC (hBMSCCM) from two different healthy donors. Several genes coding for soluble modulators of the non-canonical WNT signaling pathway, including FRZB, SFRP2, RSPO1, WNT5A and 5B were significantly overexpressed in fully differentiated adipocytes than in their respective in hBMSC. ADIPOCM induced the overexpression and the nuclear translocation of the Frizzled family member receptor tyrosine kinase-like orphan receptor (Ror) 2 in HPDE and HPDE/KRAS cells. Vantictumab, an anti-Frizzled monoclonal antibody, reduced ROR2 nuclear translocation and in turn the EMT and aggressiveness in HPDE and HPDE/KRAS cells. Conclusions: We demonstrated that adipocytes could induce EMT and aggressiveness in models of pancreatic preneoplastic lesions by orchestrating a complex paracrine signaling of soluble modulators of the non-canonical WNT signaling pathway that determine, in turn, the activation and nuclear translocation of ROR2. This signaling pathway could represent a novel target for pancreatic cancer chemoprevention. Most importantly, these factors could serve as novel biomarkers to select a risk population among obese subjects for screening and, thus, early diagnosis of pancreatic cancer.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.