69 results on '"Ducreux M"'
Search Results
2. Oral ADAGRASIB associated with cetuximab unblocks obstructed colorectal cancer.
- Author
-
Mercier L, Delaye M, Fuerea A, Ducreux M, and Boilève A
- Subjects
- Humans, Male, Administration, Oral, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Aged, Female, Intestinal Obstruction etiology, Middle Aged, Cetuximab administration & dosage, Cetuximab adverse effects, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology
- Abstract
Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: DUCREUX Michel: Merck Serono, MSD, AMGEN, Roche, Bayer, Ipsen, Pfizer, Servier, Pierre Fabre, HalioDx, Lilly, Sanofi, BMS. BOILEVE Alice: Merck Serono, Opsen, Servier. FUEREA Alina, DELAYE Matthieu and MERCIER Lea: no COI.
- Published
- 2024
- Full Text
- View/download PDF
3. Precision medicine for KRAS wild-type pancreatic adenocarcinomas.
- Author
-
Ben-Ammar I, Rousseau A, Nicolle R, Tarabay A, Boige V, Valery M, Pudlarz T, Malka D, Gelli M, Fernandez-De-Sevilla E, Fuerea A, Tanguy ML, Rouleau E, Barbe R, Mathieu JRR, Jaulin F, Smolenschi C, Hollebecque A, Ducreux M, and Boileve A
- Subjects
- Female, Humans, Male, Middle Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Mutation, Precision Medicine, Proto-Oncogene Proteins p21(ras) genetics, Retrospective Studies, Aged, Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics
- Abstract
Background: KRAS mutation is the most common molecular alteration in pancreatic adenocarcinoma (PDAC), and around 10% of patients harbor KRAS wild-type tumors (KRAS
WT )., Methods: A retrospective chart review of clinical/molecular data was performed including all PDAC patients with a determined KRAS status (tumor molecular profiling on tissue or liquid biopsy)., Results: 342 patients were included with 54 KRASWT PDAC (16%) compared to 288 patients with KRASm PDAC. Median age was 61 years [IQR:54.0;67.0] and 164 pts (48%) were female. At diagnosis, KRASWT patients (63%) were more frequently diagnosed at a non-metastatic stage compared to KRASm patients (41%) (p = 0.003). Regarding metastatic sites, liver was less frequent in KRASWT (39%, p < 0.0001). Median overall survival (mOS) from initial diagnosis was significantly higher in the KRASWT group compared to KRASm (50.8 months, CI95% [32.0-NR] vs 21.1 months, CI95% [18.9-23.4] (p < 0.004 after adjustment on age, ECOG and stage at diagnosis). In first-line systemic treatment, (mostly FOLFIRINOX) progression-free survival (PFS) was also higher in KRASWT . Based on ESCAT classification, a putative actionable alteration (ESCAT I-III) was identified in 19 (36%) KRASWT pts and 46 (16%) KRASm patients (p < 0.0001) with more alterations in FGFR2, BRAF(V600E), NRTK and more MSI tumors. KRASWT harbored also fewer alterations in TP53, CDKN2A, and SMAD4. 12 KRASWT patients received a molecularly-matched treatment with clinical benefit and improved outcomes compared to KRASm patients., Conclusions: KRASWT patients display distinct disease characteristics and outcomes with prolonged overall survival. KRASWT patients also harbor more actionable molecular alterations, leading to higher survival rates after receiving molecularly matched treatments., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Antoine Hollebecque: Amgen, AstraZeneca, Debiopharm, Eli Lilly and Company, Incyte Corporation, QED Therapeutics. David Malka: Roche, Amgen, Bayer, Sanofi, Merck Serono, Servier, Sanofi, Pierre Fabre, Viatris, Bristol Myers Squibb, MSD Oncology, LEO Pharma, Incyte, AstraZeneca, Taiho Oncology, Pfizer. Fanny Jaulin: ORAKL. Valérie Boige: Amgen, AstraZeneca, Bayer Schering Pharma, Ipsen, Merck Serono, MSD Oncology, Roche/Genentech. Michel Ducreux: Merck Serono, MSD, AMGEN, Roche, Bayer, Ipsen, Pfizer, Servier, Pierre Fabre, HalioDx, Lilly, Sanofi, BMS. Alice Boilève: Merck Serono, Ipsen. Other authors report no conflict of interest., (Copyright © 2023 Elsevier Ltd. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
4. Hepatic arterial oxaliplatin plus intravenous 5-fluorouracil and cetuximab for first-line treatment of colorectal liver metastases: A multicenter phase II trial.
- Author
-
Malka D, Verret B, Faron M, Guimbaud R, Caramella C, Edeline J, Galais MP, Bengrine-Lefevre L, Smith D, Dupont-Bierre E, De Baere T, Goéré D, Dartigues P, Lacroix L, Boige V, Gelli M, Pignon JP, and Ducreux M
- Subjects
- Humans, Oxaliplatin, Cetuximab, Infusions, Intra-Arterial, Fluorouracil, Leucovorin, Antineoplastic Combined Chemotherapy Protocols adverse effects, Treatment Outcome, Colorectal Neoplasms pathology, Liver Neoplasms secondary
- Abstract
Background: The efficacy and tolerability of hepatic arterial infusion (HAI) oxaliplatin plus systemic 5-fluorouracil and cetuximab as frontline treatment in patients with colorectal liver metastases (CRLM) are unknown., Methods: In this multicenter, single-arm phase II study, patients with CRLM not amenable to curative-intent resection or requiring complex/major liver resection, and no prior chemotherapy for metastatic disease, received HAI oxaliplatin and intravenous 5-fluorouracil, leucovorin and cetuximab, every two weeks until disease progression, limiting toxicity or at least 3 months after complete response or curative-intent resection/ablation. The primary endpoint was overall response rate (ORR)., Results: 35 patients, mostly with bilateral (89%), multiple CRLM (>4, 86%; >10, 46%) were enrolled in eight centers. The ORR was 88% (95% CI, 71%-96%) among evaluable patients (n = 32), and 95% (95% CI 70-100%) among the 22 wild-type RAS/BRAF evaluable patients. After a median follow-up of 8.8 years (95% CI, 8.7-not reached), median progression-free survival was 17.9 months (95% CI, 15-23) and median overall survival (OS) was 46.3 months (95% CI, 40.0-not reached). 23 of the 35 patients (66%), including 22 (79%) of the 25 patients with wild-type RAS tumor, underwent curative-intent surgical resection and/or ablation of CRLM. HAI catheter remained patent in 86% of patients, allowing for a median of eight oxaliplatin infusions (range, 1-19). Treatment toxicity was manageable, without toxic death., Conclusion: HAI oxaliplatin plus systemic 5-fluorouracil and cetuximab appears highly effective in the frontline treatment of patients with unresectable CRLM and should be investigated further., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: DM: Consulting/advisory role: AbbVie, Amgen, AstraZeneca, Bayer, Bionest Partners, BMS, Incyte, Merck Serono, MSD, Pierre Fabre, Roche, Sanofi, Simon-Kutcher & Partners, Servier, Taiho. Honoraria for lectures: Amgen, AstraZeneca, Bayer, BMS, Foundation Medicine, Incyte, Leo Pharma, Merck Serono, MSD, Pierre Fabre, Roche, Sanofi, Servier, Veracyte, Viatris. Personal fees for a writing engagement: Medscape. Travel expenses for medical congresses: Amgen, Bayer, BMS, Merck Serono, MSD, Pierre Fabre, Roche, Sanofi, Servier, Viatris. BV: none. MF: honoraria for lectures: Novartis. Travel expenses for medical congresses: Ipsen. RG: Consulting/advisory role: Pierre Fabre, BMS, Servier, MSD. Honoraria for lectures: MSD, Servier, Ipsen, BMS, Viatris. Travel expenses for medical congresses: MSD, Ipsen, Roche, Pierre Fabre, Advanced Accelerator Applications (AAA). CC: none. JE: Consulting/advisory role: MSD, Eisai, BMS, AstraZeneca, Bayer, Roche, Ipsen, Basilea, Merck Serono, Incyte, Servier, Beigene. Travel expenses for medical congresses: Amgen. Research funding (institutional): BMS, Beigene. MPG: honoraria: Servier, BMS. Travel expenses for medical congresses: AAA, Bayer, Servier, Amgen. LBG: honoraria: AstraZeneca, Servier, KSK, MSD, Bayer, Eisai. DS: travel expenses for medical congresses: Servier, Pierre Fabre. EDB: none. TDB: Consulting/advisory role: Terumo, Boston Scientific. Research funding (institutional): Quantum, Terumo. DG: honoraria for lectures: Merck Serono, Amgen, Sanofi, Servier. PD: Consulting/advisory role: MSD. Honoraria for lectures: AstraZeneca, MSD. Travel expenses for medical congresses: MSD. LL: none. VB: Consulting/advisory role: Merck Serono, Ipsen, Bayer, Eisai, BMS, Roche, AstraZeneca. Honoraria for lectures: Merck Serono, Novartis, Roche, Bayer, MSD, Amgen, Ipsen, AstraZeneca. Travel expenses for medical congresses: Amgen, Merck Serono, Sanofi Genzyme, Bayer, Roche, MSD, Ipsen, AstraZeneca. Research funding (institutional): Merck Serono. MG: none. JPP: none. MD: honoraria for advisory boards or as a presenter in symposium: Merck Serono, MSD, Amgen, Roche, Bayer, Ipsen, Pfizer, Servier, Pierre Fabre, Lilly, Sanofi, Servier, Daiichi Sankyo., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. Molecular profiling and target actionability for precision medicine in neuroendocrine neoplasms: real-world data.
- Author
-
Boilève A, Faron M, Fodil-Cherif S, Bayle A, Lamartina L, Planchard D, Tselikas L, Kanaan C, Scoazec JY, Ducreux M, Italiano A, Baudin E, and Hadoux J
- Subjects
- Humans, Prognosis, Retrospective Studies, Precision Medicine, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors genetics, Carcinoma, Neuroendocrine drug therapy, Carcinoma, Neuroendocrine genetics, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics
- Abstract
Background: Key molecular alterations (MA) of neuroendocrine neoplasm (NEN) of various grade/primaries have been described but the applicability of molecular profiling (MP) for precision medicine in NEN remains to be demonstrated., Methods: We conducted a retrospective study of all patients with metastatic NEN who had MP on tumour tissue at Gustave Roussy. The primary objective was to assess the clinical applicability of MP by evaluating the growth modulator index (GMI) as the primary end-point., Results: MPs were obtained in 114 out of 156 eligible patients, including 12% NET-G1, 42% NET-G2, 13% NET-G3 and 35% neuroendocrine carcinoma (NEC). Primary sites were lung/thymus (40%), pancreas (19%), gastro-intestinal (16%), head&neck (10%), unknown (10%) and others (10%) with synchronous metastases in 61% of the patients. Most frequent MA were: MEN1 (25%), PTEN (13%), TP53 (11%) and TSC2 (9%), in neuroendocrine tumour (NET), and TP53 (50%) and RB1 (18%) in NEC. ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT) classification of these MA were: I(5%), III(20%), IV(23%), X(27%); a putative actionable MA was identified in 48% patients. Median TMB was 5.7 Mut/Mb, with 3 TMB > 10 and 1 MSI NET. No MA was found in 26% patients. Molecularly matched treatment was administered to 19 patients (4 NEC, 15 NET): immunotherapy (n = 3), tipifarnib (n = 1), NOTCHi (n = 1), EGFRi (n = 2), HER2i (n = 1) and everolimus (n = 11). Overall, 67% of patients had a clinical benefit defined as a GMI over 1.3 with a 78% disease control rate., Conclusion: We report 48% of NEN with a putative actionable MA of which 35% received molecularly matched treatment, with a clinical benefit in 67% of the cases., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: EB received honorarium for board participation and grants from HRA. TSL: consultant: GE, Terumo, quantum surgical (none related to this paper) AI: Research grant (AstraZeneca, Bayer, BMS, Merck, MSD, Pharmamar) DP: reports receiving consulting, advisory role, or lectures from AstraZeneca, Bristol-Myers Squibb, Boehringer Ingelheim, Celgene, Daiichi Sankyo, Eli Lilly, Merck, Novartis, Pfizer, prIME Oncology, Peer CME, Roche, Janssen, and AbbVie; reports receiving honoraria from AstraZeneca, Bristol-Myers Squibb, Boehringer Ingelheim, Celgene, Eli Lilly, Merck, Novartis, Pfizer, prIME Oncology, Peer CME, Roche, Janssen, and AbbVie; clinical trial research as principal or co-investigator (institutional financial interests) for AstraZeneca, Bristol-Myers Squibb, Boehringer Ingelheim, Eli Lilly, Merck, Novartis, Pfizer, Roche, Medimmune, Sanofi-Aventis, Taiho Pharma, Novocure, Daiichi Sankyo, Janssen, and AbbVie; and travel, accommodations, and expenses from AstraZeneca, Roche, Novartis, and Pfizer. ARB: Personal: Advisory board: Sanofi, Janssen. As part of the Drug Development Department (DITEP) = Principal/sub-Investigator of Clinical Trials for Abbvie, Adaptimmune, Adlai Nortye USA Inc, Aduro Biotech, Agios Pharmaceuticals, Amgen, Astex Pharmaceuticals, Astra Zeneca Ab, Aveo, Basilea Pharmaceutica International Ltd, Bayer Healthcare Ag, Bbb Technologies Bv, Beigene, BicycleTx Ltd, Blueprint Medicines, Boehringer Ingelheim, Boston Pharmaceuticals, Bristol Myers Squibb, Ca, Casi Pharmaceuticals, Inc, Celgene Corporation, Cellcentric, Chugai Pharmaceutical Co, Cullinan-Apollo, Curevarc, Daiichi Sankyo, Debiopharm, Eisai, Eisai Limited, Eli Lilly, Exelixis, Faron Pharmaceuticals Ltd, Forma Tharapeutics, Gamamabs, Genentech, Glaxosmithkline, H3 Biomedicine, Hoffmann La Roche Ag, Imcheck Therapeutics, Incyte Corporation, Innate Pharma, Institut De Recherche Pierre Fabre, Iris Servier, Iteos Belgium SA, Janssen Cilag, Janssen Research Foundation, Janssen R&D LLC, Kura Oncology, Kyowa Kirin Pharm. Dev, Lilly France, Loxo Oncology, Medimmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Molecular Partners Ag, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology Nv, Oncoethix, Oncopeptides, Orion Pharma, Genomics, Ose Pharma, Pfizer, Pharma Mar, Pierre Fabre Medicament, Relay Therapeutics, Inc, Roche, Sanofi Aventis, Seattle Genetics, Sotio A.S, Syros Pharmaceuticals, Taiho Pharma, Tesaro, Transgene S.A, Turning Point Therapeutics, XencorResearch Grants from Astrazeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Pfizer, Roche, Sanofi Non-financial support (drug supplied) from Astrazeneca, BMS, Boringher Ingelheim, GSK, Medimmune, Merck, NH TherAGuiX, Pfizer, Roche MD: declares conflicts of interest with Merck Serono, MSD, AMGEN, Roche, Bayer, Ipsen, Pfizer, Servier, Pierre Fabre, HalioDx, Lilly, Sanofi, BMS. JH: declares grant/research support from Novartis and consultant for IPSEN, Lilly, Roche, Pharma Mar and Eisai Other authors report no conflict of interest., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
6. Clinical and exploratory biomarker findings from the MODUL trial (Cohorts 1, 3 and 4) of biomarker-driven maintenance therapy for metastatic colorectal cancer.
- Author
-
Ducreux M, Tabernero J, Grothey A, Arnold D, O'Dwyer PJ, Gilberg F, Abbas A, Thakur MD, Prizant H, Irahara N, Tahiri A, Schmoll HJ, Van Cutsem E, and de Gramont A
- Subjects
- Humans, Bevacizumab, Cetuximab, Proto-Oncogene Proteins B-raf genetics, Vemurafenib therapeutic use, Fluorouracil, Biomarkers, Antineoplastic Combined Chemotherapy Protocols adverse effects, Leucovorin, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Colonic Neoplasms drug therapy, Rectal Neoplasms drug therapy
- Abstract
Purpose: MODUL is an adaptable, signal-seeking trial of biomarker-driven maintenance therapy following first-line induction treatment in patients with metastatic colorectal cancer (mCRC). We report findings from Cohorts 1 (BRAF
mut ), 3 (human epidermal growth factor 2 [HER2]+) and 4 (HER2‒/high microsatellite instability, HER2‒/microsatellite stable [MSS]/BRAFwt or HER2‒/MSS/BRAFmut /RASmut )., Methods: Patients with unresectable, previously untreated mCRC without disease progression following standard induction treatment (5-fluorouracil/leucovorin [5-FU/LV] plus oxaliplatin plus bevacizumab) were randomly assigned to control (fluoropyrimidine plus bevacizumab) or cohort-specific experimental maintenance therapy (Cohort 1: vemurafenib plus cetuximab plus 5-FU/LV; Cohort 3: capecitabine plus trastuzumab plus pertuzumab; Cohort 4: cobimetinib plus atezolizumab). The primary efficacy end-point was progression-free survival (PFS)., Results: Cohorts 1, 3 and 4 did not reach target sample size because of early study closure. In Cohort 1 (n = 60), PFS did not differ between treatment arms (hazard ratio, 0.95; 95% confidence intervals 0.50-1.82; P = 0.872). However, Cohort 1 exploratory biomarker data showed preferential selection for mitogen-activated protein kinase (MAPK) pathway mutations (mainly KRAS, NRAS, MAP2K1 or BRAF) in the experimental arm but not the control arm. In Cohort 3 (n = 5), PFS ranged from 3.6 to 14.7 months versus 4.0 to 5.4 months in the experimental and control arms, respectively. In Cohort 4 (n = 99), PFS was shorter in the experimental arm (hazard ratio, 1.44; 95% confidence intervals 0.90-2.29; P = 0.128)., Conclusions: Vemurafenib plus cetuximab plus 5-FU/LV warrants further investigation as first-line maintenance treatment for BRAFmut mCRC. MAPK-pathway emergent genomic alterations may offer novel therapeutic opportunities in BRAFmut mCRC. Cobimetinib plus atezolizumab had an unfavourable benefit:risk ratio in HER2‒/MSS/BRAFwt mCRC. New strategies are required to increase the susceptibility of MSS mCRC to immunotherapy., Trial Registration: ClinicalTrials.gov: NCT02291289., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
7. FGFR2 fusion in metastatic pancreatic ductal adenocarcinoma: Is there hope?
- Author
-
Helal C, Valéry M, Ducreux M, Hollebecque A, and Smolenschi C
- Subjects
- Humans, Receptor, Fibroblast Growth Factor, Type 2 genetics, Pancreatic Neoplasms, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal pathology
- Abstract
Competing Interests: Conflict of interest statement The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
- Published
- 2022
- Full Text
- View/download PDF
8. Individual patient data meta-analysis of neoadjuvant chemotherapy followed by surgery versus upfront surgery for carcinoma of the oesophagus or the gastro-oesophageal junction.
- Author
-
Faron M, Cheugoua-Zanetsie AM, Thirion P, Nankivell M, Winter K, Cunningham D, Van der Gaast A, Law S, Langley R, de Vathaire F, Valmasoni M, Mauer M, Roth J, Gebski V, Burmeister BH, Paoletti X, van Sandick J, Fu J, Ducreux M, Blanchard P, Tierney J, Pignon JP, and Michiels S
- Subjects
- Adenocarcinoma mortality, Adenocarcinoma pathology, Carcinoma, Squamous Cell mortality, Carcinoma, Squamous Cell pathology, Chemotherapy, Adjuvant statistics & numerical data, Disease-Free Survival, Esophageal Neoplasms mortality, Esophageal Neoplasms pathology, Esophagectomy statistics & numerical data, Esophagogastric Junction pathology, Esophagogastric Junction surgery, Humans, Neoplasm Recurrence, Local prevention & control, Postoperative Complications etiology, Adenocarcinoma therapy, Carcinoma, Squamous Cell therapy, Esophageal Neoplasms therapy, Esophagectomy adverse effects, Neoadjuvant Therapy statistics & numerical data, Neoplasm Recurrence, Local epidemiology
- Abstract
Introduction: Which neoadjuvant treatment for locally advanced thoracic oesophagus (TE) or gastro-oesophageal junction carcinoma is best remains an open question. Randomised controlled trials variously accrued patients with adenocarcinoma and squamous cell carcinoma, making strong conclusions hard to obtain. The primary objective of this individual participant data meta-analysis was to investigate the effect of neoadjuvant chemotherapy on overall survival (OS)., Patients and Methods: Eligible trials should have closed to accrual before 2016 and compared neoadjuvant chemotherapy and surgery (CS) to surgery alone. All relevant published and unpublished trials were identified via searches of electronic databases, conference proceedings and clinical trial registers. The main end-point was OS. Investigators were contacted to obtain the individual patient data, which was recorded, harmonised and checked. A random-effects Cox model, stratified by trial, was used for meta-analysis and subgroup analyses were preplanned., Results: 16 trials were identified as eligible. Individual patient data were obtained from 12 trial and 2478 patients. CS was associated with an improved OS versus surgery, hazard ratio (HR) = 0.83 [0.72-0.96], p < 0.0001, translating to an absolute benefit of 5.7% at 5-years from 16.8% to 22.5%. Treatment effects did not vary substantially between adenocarcinoma (HR = 0.73 [0.62-0.87]) and squamous cell carcinoma (HR = 0.91 [0.76-1.08], interaction p = 0.26). A somewhat more pronounced effect was observed in gastro-oesophageal junction (HR = 0.68 [0.50-0.93]) versus TE (HR = 0.87 [0.75-1.00], interaction p = 0.07). CS was also associated with a greater disease-free survival (HR = 0.74 [0.64-0.85], p < 0.001)., Conclusions: Neoadjuvant chemotherapy conferred a better OS than surgery alone and should be considered in all anatomical location and histological subtypes., Competing Interests: Conflict of interest statement No authors had conflict of interest related to the current work., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
9. Impact of COVID-19 on healthcare organisation and cancer outcomes.
- Author
-
Bardet A, Fraslin AM, Marghadi J, Borget I, Faron M, Honoré C, Delaloge S, Albiges L, Planchard D, Ducreux M, Hadoux J, Colomba E, Robert C, Bouhir S, Massard C, Micol JB, Ter-Minassian L, Michiels S, Auperin A, Barlesi F, and Bonastre J
- Subjects
- COVID-19 mortality, COVID-19 virology, Computer Simulation, Delivery of Health Care organization & administration, Hospital Administration, Hospitals, Humans, Neoplasms pathology, Pandemics, Proportional Hazards Models, SARS-CoV-2 isolation & purification, COVID-19 epidemiology, Neoplasms mortality, Neoplasms therapy
- Abstract
Background: Changes in the management of patients with cancer and delays in treatment delivery during the COVID-19 pandemic may impact the use of hospital resources and cancer mortality., Patients and Methods: Patient flows, patient pathways and use of hospital resources during the pandemic were simulated using a discrete event simulation model and patient-level data from a large French comprehensive cancer centre's discharge database, considering two scenarios of delays: massive return of patients from November 2020 (early-return) or March 2021 (late-return). Expected additional cancer deaths at 5 years and mortality rate were estimated using individual hazard ratios based on literature., Results: The number of patients requiring hospital care during the simulation period was 13,000. In both scenarios, 6-8% of patients were estimated to present a delay of >2 months. The overall additional cancer deaths at 5 years were estimated at 88 in early-return and 145 in late-return scenario, with increased additional deaths estimated for sarcomas, gynaecological, liver, head and neck, breast cancer and acute leukaemia. This represents a relative additional cancer mortality rate at 5 years of 4.4 and 6.8% for patients expected in year 2020, 0.5 and 1.3% in 2021 and 0.5 and 0.5% in 2022 for each scenario, respectively., Conclusions: Pandemic-related diagnostic and treatment delays in patients with cancer are expected to impact patient survival. In the perspective of recurrent pandemics or alternative events requiring an intensive use of limited hospital resources, patients should be informed not to postpone care, and medical resources for patients with cancer should be sanctuarised., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: No author reported potential conflict of interest in link with the submitted work. A.B. reports personal fees from Roche SAS. I.B. reports personal fees from Roche, Novartis, CSL Behring and Takeda. M.F. reports fees from HRA Pharma (participation to board) and travel grants from Ipsen, Pfizer and Novartis. S.D. reports grants and non-financial support from Pfizer, grants from Novartis, grants and non-financial support from AstraZeneca, grants and non-financial support from Roche Genentech, grants from Lilly, grants from Puma, grants from Myriad, grants from Orion, grants from Amgen, grants from Sanofi, grants from Genomic Health, grants from GE, grants from Servier, grants from MSD, grants from BMS, grants from Pierre Fabre, grants from Seagen and grants from Exact Sciences. L.A. reports a consulting/advisory role for Novartis (Institution), Amgen (Institution), Bristol Myers Squibb (Institution), Ipsen (Institution), Roche (Institution), Pfizer (Institution), Astellas Pharma (Institution), Merck (Institution), MSD (Institution), AstraZeneca (Institution), Exelixis (Institution), Corvus Pharmaceuticals (Institution) and Peloton Therapeutics (Institution) and research Funding from Bristol Myers Squibb (Institution). D.P. reports a consulting, advisory role or lectures for AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Celgene, Daiichi Sankyo, Eli Lilly, Merck, Novartis, Pfizer, prIME Oncology, Peer CME, Roche and Samsung; honoraria from AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Celgene, Eli Lilly, Merck, Novartis, Pfizer, prIME Oncology, Peer CME, Roche and Samsung; institutional financial interests (clinical trials research as the principal or co-investigator) from AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Eli Lilly, Merck, Novartis, Pfizer, Roche, MedImmune, Sanofi-Aventis, Taiho Pharma, Novocure and Daiichi Sankyo and travel and accommodation expenses from AstraZeneca, Roche, Novartis, prIME Oncology and Pfizer. M.D. reports honoraria for advisory boards or as a presenter in symposium from Merck Serono, MSD, AMGEN, Roche, Bayer, Ipsen, Pfizer, Servier, Pierre Fabre, HalioDx, Lilly and Sanofi. E.C. reports consulting advisory board activity for BMS, Ipsen, Sanofi, Pfizer, GSK and Merck. C.R. reports a consultant/advisory role for Bristol Myers Squibb, MSD, Roche, Novartis, Amgen, Sanofi and Pierre Fabre. C.M. reports consultant/advisory fees from Amgen, Astellas, AstraZeneca, Bayer, BeiGene, BMS, Celgene, Debiopharm, Genentech, Ipsen, Janssen, Lilly, MedImmune, MSD, Novartis, Pfizer, Roche, Sanofi and Orion and as acted as a principal/sub-investigator of clinical trials for AbbVie, Aduro, Agios, Amgen, Argen-x, Astex, AstraZeneca, Aveo pharmaceuticals, Bayer, BeiGene, Blueprint, BMS, Boehringer Ingelheim, Celgene, Chugai, Clovis, Daiichi Sankyo, Debiopharm, Eisai, Eos, Exelixis, Forma, GamaMabs, Genentech, Gortec, GSK, H3 Biomedecine, Incyte, Innate Pharma, Janssen, Kura Oncology, Kyowa, Lilly, Loxo, Lysarc, Lytix Biopharma, MedImmune, Menarini, Merus, MSD, Nanobiotix, Nektar Therapeutics, Novartis, Octimet, Oncoethix, Oncopeptides AB, Orion, Pfizer, PharmaMar, Pierre Fabre, Roche, Sanofi, Servier, Sierra Oncology, Taiho, Takeda, Tesaro and Xencor. J.B.M. reports personal fees from AbbVie, Jazz PHARMA and Astellas. S.M. reports personal fees from statistical advice to IDDI, Janssen Cilag and Amaris and from data and safety monitoring membership of clinical trials (Hexal, Steba, IQVIA, Roche, Sensorion, Biophytis, Servier and Yuhan). A.A. reports grant support, paid to her institution, from F. Hoffmann-La Roche. F.B. reports personal financial interests with AstraZeneca, Bayer, Bristol Myers Squibb, Boehringer Ingelheim, Eli Lilly Oncology, F. Hoffmann–La Roche Ltd, Novartis, Merck, Mirati, MSD, Pierre Fabre, Pfizer, Seattle Genetics and Takeda; institutional financial interests with AbbVie, ACEA, Amgen, AstraZeneca, Bayer, Bristol Myers Squibb, Boehringer Ingelheim, Eisai, Eli Lilly Oncology, F. Hoffmann–La Roche Ltd, Genentech, Ipsen, Ignyta, Innate Pharma, Loxo, Novartis, MedImmune, Merck, MSD, Pierre Fabre, Pfizer, Sanofi-Aventis and Takeda and non-financial interests (as a principal investigator) for AstraZeneca, BMS, Merck, Pierre Fabre and F. Hoffmann-La Roche, Ltd–sponsored trials (or ISR). J.B. reports honoraria from Bristol Myers Squibb; a consulting or advisory role for Bristol Myers Squibb, MSD, PharmaMar (Inst), Bristol Myers Squibb (Inst) and Merck Serono (Inst) and travel and accommodation expenses from Bristol Myers Squibb. Other authors report no conflict of interest. Y. Takahashi declares no conflict of interest. Her editorial work for the present article has been funded by Inserm., (Copyright © 2021 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
10. Predictive factors of severe early treatment-related toxicity in patients receiving first-line treatment for metastatic colorectal cancer: Pooled analysis of 2190 patients enrolled in Fédération Francophone de Cancérologie Digestive (FFCD) trials.
- Author
-
Breton C, Aparicio T, Le Malicot K, Ducreux M, Lecomte T, Bachet JB, Taieb J, Legoux JL, De Gramont A, Bennouna J, Bouché O, Boussari O, Manfredi S, and Gornet JM
- Subjects
- Aged, Colorectal Neoplasms pathology, Female, Humans, Male, Middle Aged, Neoplasm Metastasis, Prognosis, Prospective Studies, Treatment Outcome, Colorectal Neoplasms complications
- Abstract
Aim: Few studies have explored the association between baseline characteristics and the occurrence of early toxicities in patients treated with first-line chemotherapy for metastatic colorectal cancer (mCRC)., Patients and Methods: Individual patient data of 2190 patients enrolled in 10 prospective FFCD (Fédération Francophone de Cancérologie Digestive) trials were analysed. Severe early toxicity was defined as the occurrence of grade ≥III toxicity within 3 months after initiation of chemotherapy (ET3)., Results: Patients received monotherapy based on 5-FU (n = 1068), a cytotoxic doublet (n = 395) or tritherapy with a cytotoxic doublet plus anti-VEGF agent or a cytotoxic triplet (n = 727). The patients received 5-FU (100%), Irinotecan (39.6%), Oxaliplatin (13.4%), Bevacizumab (29.6%) or Aflibercept (1.8%). ET3 occurred in 244 patients (22.8%) with monotherapy, 248 patients (62.8%) with doublet and 392 patients (53.9%) with tritherapy. The most frequent ET3s were related to biological abnormalities and/or gastrointestinal, general and vascular disorders. The prognostic factors for the occurrence of an ET3 in multivariate analysis were a performance status of 2 rather than 0-1 (OR 2.57; 95% CI [1.16, 5.73]; p = 0.02), tritherapy versus monotherapy (OR 2.31; 95% CI [0.84, 6.33]; p = 0.02), alkaline phosphatase > 300 UI/l (OR 3.07; 95% CI [1.79, 5.27]; p < 0.001) and non-resected primary tumour versus resection (OR 1.59; 95% CI [1.06, 2.39]; p = 0.02). Median overall survival in patients without ET3 was significantly longer than that in patients with ET3 (HR 0.87; 95% CI [0.80-0.96]; p = 0.004)., Conclusion: ET3 is frequent whatever the treatment regimen and is associated with certain baseline characteristics. The clinical impact of ET3 on prognosis in mCRC warrants further investigation., Competing Interests: Conflict of interest statement CB, KLM, ADG and OB have no conflict of interest to declare. TA has received personal fees from AstraZeneca, Amgen, Servier, Bioven, Sanofi and Roche, and non-financial support from Bayer, Roche and Ipsen. MD has received personal fees from Amgen, AstraZeneca, Bayer, Merck Serono, Pierre Fabre, Roche, Servier, Lilly, Merck and Ipsen, and non-financial support from Roche, Merck Serono and Bayer. TL and TA have received personal fees from Amgen, Merck Serono, Servier and Sanofi, and non-financial support from Amgen. JBB has received personal fees from Amgen, AstraZeneca, Bayer, Merck Serono, Pierre Fabre, Roche, Sanofi, Servier and Shire, and non-financial support from Amgen, Merck Serono and Roche. JT has received personal fees from Amgen, Roche, Merck Serono, Pierre Fabre, MSD, Sanofi, Servier and Shire, and non-financial support from Amgen, Merck Serono and Roche. JLL has received personal fees from Novartis and Pfizer, and non-financial support from Servier, Novartis and Keocyt. JB has received personal fees from Amgen, AstraZeneca, Bayer, Merck Serono, Pierre Fabre, Roche, Sanofi, Servier and Shire, and non-financial support from Amgen, Merck Serono and Roche. OB has received personal fees or non-financial support from Amgen, Roche, Merck, Pierre Fabre, Servier, MSD, Bayer and Sanofi. SM has received non-financial support from Amgen, Roche, Servier, Astra Zeneca, Ipsen pharma, Novartis pharma, Bayer, MSD and Sirtex medical Europe. JMG has received personal fees from Amgen, Janssen Cilag, Sanofi, Takeda and Tillots Pharma)., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
11. Prognostic factors of metastatic neuroendocrine carcinoma under first-line treatment with platinum etoposide with a focus on NEC score and Rb expression: Results from the multicentre RBNEC study of the Groupe d'Etude des Tumeurs Endocrines (GTE) and the ENDOCAN-RENATEN network.
- Author
-
Hadoux J, Kanaan C, Durand A, Hescot S, Hautefeuille V, Cadiot G, Tauveron I, Laboureau S, Do Cao C, Walter T, Petorin C, Blanchet O, Jannin A, Gu C, Faron M, Leteurtre E, Rousselet MC, Zakeyh JJ, Marchal A, Chatelain D, Beaulaton C, Hervieu V, Ducreux M, Scoazec JY, and Baudin E
- Subjects
- Adult, Age Factors, Aged, Aged, 80 and over, Biomarkers, Tumor metabolism, Carboplatin administration & dosage, Carcinoma, Neuroendocrine diagnosis, Carcinoma, Neuroendocrine drug therapy, Carcinoma, Neuroendocrine pathology, Cisplatin administration & dosage, Drug Administration Schedule, Female, Humans, Immunohistochemistry, Male, Middle Aged, Prognosis, Progression-Free Survival, Retinoblastoma Binding Proteins metabolism, Retrospective Studies, Risk Assessment, Ubiquitin-Protein Ligases metabolism, Young Adult, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Biomarkers, Tumor analysis, Carcinoma, Neuroendocrine mortality, Etoposide administration & dosage, Retinoblastoma Binding Proteins analysis, Ubiquitin-Protein Ligases analysis
- Abstract
Introduction and Aim: Neuroendocrine carcinomas (NECs) are aggressive malignant diseases. Platinum-etoposide (PE) combination is the standard first-line treatment, whatever the primary location. The NEC score and also retinoblastoma protein (Rb) status have been suggested to be predictive/prognostic factors in NEC. The primary objective of our multicentric retrospective study was to evaluate the prognostic relevance of the NEC score and Rb status, assessed by immunohistochemistry in PE-treated patients with metastatic NEC., Methods: Seven centres participated. The inclusion criteria were NEC, whatever the primary site, metastatic stage, first-line treatment with PE and tissue samples available. Rb status was determined centrally., Results: We report multicentric data from 185 metastatic patients (37% women, median age 63). There were 108 small-cell NECs (SCNECs, 58.4%), 50 large-cell NECs (LCNECs, 27%) and 27 not otherwise specified NECs (nosNECs, 14.6%). The primary sites were the thorax (37%), gastroenteropancreatic sites (38%), unknown (15%) and other (9%). The mean Ki-67 index was 76% (range 20-100). Rb status was interpretable in 122 cases. Rb expression was lost in 74% of the cases: 84% of SCNEC vs. 60% and 63% of LCNEC and nosNEC, respectively (p = 0.016). Objective response was seen in 70% of SCNEC, 45% of LCNEC and 48% of nosNEC (p < 0.001) and in 62% of Rb-negative tumours vs. 46% of Rb-positive tumours (p = 0.3). There was no difference in median progression-free survival or overall survival (OS) as per Rb status. Age, NEC score and response to chemotherapy were the main factors associated with OS in our cohort., Conclusion: In our series, Rb status had no prognostic impact in PE-treated metastatic patients with NEC, whereas age, NEC score and response to chemotherapy were the main factors associated with OS., Competing Interests: Conflict of interest statement The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
12. Severe acute respiratory syndrome coronavirus 2 vaccination for patients with solid cancer: Review and point of view of a French oncology intergroup (GCO, TNCD, UNICANCER).
- Author
-
Tougeron D, Hentzien M, Seitz-Polski B, Bani-Sadr F, Bourhis J, Ducreux M, Gaujoux S, Gorphe P, Guiu B, Hoang-Xuan K, Huguet F, Lecomte T, Lièvre A, Louvet C, Maggiori L, Mansi L, Mariani P, Michel P, Servettaz A, Thariat J, Westeel V, Aparicio T, Blay JY, and Bouché O
- Subjects
- Humans, SARS-CoV-2, COVID-19 prevention & control, COVID-19 Vaccines therapeutic use, Neoplasms complications
- Abstract
The impacts of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic on cancer care are multiple, entailing a high risk of death from coronavirus disease 2019 (COVID-19) in patients with cancer treated by chemotherapy. SARS-CoV-2 vaccines represent an opportunity to decrease the rate of severe COVID-19 cases in patients with cancer and also to restore normal cancer care. Patients with cancer to be targeted for vaccination are difficult to define owing to the limited contribution of these patients in the phase III trials testing the different vaccines. It seems appropriate to vaccinate not only patients with cancer with ongoing treatment or with a treatment having been completed less than 3 years ago but also household and close contacts. High-risk patients with cancer who are candidates for priority access to vaccination are those treated by chemotherapy. The very high-priority population includes patients with curative treatment and palliative first- or second-line chemotherapy, as well as patients requiring surgery or radiotherapy involving a large volume of lung, lymph node and/or haematopoietic tissue. When possible, vaccination should be carried out before cancer treatment begins. SARS-CoV-2 vaccination can be performed during chemotherapy while avoiding periods of neutropenia and lymphopenia. For organisational reasons, vaccination should be performed in cancer care centres with messenger RNA vaccines (or non-replicating adenoviral vaccines in non-immunocompromised patients). Considering the current state of knowledge, the benefit-risk ratio strongly favours SARS-CoV-2 vaccination of all patients with cancer. To obtain more data concerning the safety and effectiveness of vaccines, it is necessary to implement cohorts of vaccinated patients with cancer., Competing Interests: Conflict of interest statement None declared., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
13. Hepatic arterial infusion of chemotherapy as an option in a multimodal treatment of metastatic squamous cell carcinoma of the anus.
- Author
-
Dall'Armellina F, Perret A, Smolenschi C, Hollebecque A, Malka D, Boige V, and Ducreux M
- Subjects
- Aged, Female, Humans, Male, Neoplasm Metastasis, Anal Canal pathology, Anus Neoplasms drug therapy, Carcinoma, Squamous Cell drug therapy, Combined Modality Therapy methods, Hepatic Artery drug effects, Infusions, Intra-Arterial methods
- Published
- 2021
- Full Text
- View/download PDF
14. Immune scores in colorectal cancer: Where are we?
- Author
-
Malka D, Lièvre A, André T, Taïeb J, Ducreux M, and Bibeau F
- Subjects
- CD8-Positive T-Lymphocytes immunology, Colorectal Neoplasms pathology, Humans, Immunologic Memory immunology, Lymphocytes, Tumor-Infiltrating immunology, Prognosis, Colorectal Neoplasms immunology, Immunity immunology
- Abstract
There is growing evidence that the immune system may prevent the occurrence, growth and metastatic diffusion of colorectal cancer (CRC). The role played by the adaptive immune response at the tumour site is critical in the balance between tumour invasion and defence against cancer. Recent data have shown that the evaluation of this immune response may help to define the prognosis and possibly the treatment of localised CRC as well as metastatic CRC. Tumour infiltrates with T cells (CD3+), cytotoxic T cells (CD8+) and memory T cells (CD45RO+) are the immune parameters most consistently and strongly associated with good clinical outcome in CRC. Several scoring systems have been developed, including the Immunoscore®, based on the immunohistochemical determination with a digital image analysis system of the density of CD3+ and CD8+ lymphocytes in the centre and the invasive margin of the tumour. This review will focus on the different immunoscoring systems developed in CRC, their performance, their limitations and their potential for improving patients' care in the future., Competing Interests: Conflict of interest statement David Malka: Honoraria and non-financial support from Amgen, Bayer, Ipsen, Incyte, MSD, Merck Serono, Roche, Sanofi, Servier; honoraria from BMS, Leo Pharma, Shire, HalioDx and Agios. Astrid Lièvre: Personal fees from HalioDX, AAA, Amgen, Bayer, BMS, Celgene, Incyte, Ipsen, Lilly, Merck, Novartis, Pierre Fabre, Roche, Sandoz, Sanofi, Servier; non-financial support from AAA, Amgen, Bayer, Incyte, Ipsen, Merck, Novartis, Pierre Fabre, Pfizer, Roche, Sandoz, Servier, Integragen; grants from Bayer, Lilly, Merck, Novartis Thierry André: Consulting/advisory role and/or honoraria from Amgen, Bristol-Myers Squibb, Chugai, Clovis, Gristone Oncology, GlaxoSmithKline, Haliodx, MSD Oncology, Pierre Fabre, Roche/Ventana, Sanofi, and Servier; travel, accommodations, and expenses from Roche/Genentech, MSD Oncology, and Bristol-Myers Squibb. Julien Taïeb: Consultancy or speaker roles for Amgen, Roche, Merck, Celgene, Lilly, Sanofi, Sirtex, Pierre Fabre, Merck Sharp & Dohme and Servier Michel Ducreux: Personal fees from Servier, MSD, Novartis, Ipsen, Lilly, Amgen, Bayer, Roche, Merck Serono and Shire, grants from Roche and Merck Serono, other from Roche, Amgen and Bayer; his spouse is the Head of the Oncology Business Unit of Sandoz Affiliate in France. Frédéric Bibeau: Personal fees from HalioDX, Amgen, Astra-Zeneca, Merck, Bayer, GSK, BMS and MSD and non-financial support from BMS and MSD., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
15. Hepatic arterial infusion of oxaliplatin plus systemic chemotherapy and targeted therapy for unresectable colorectal liver metastases.
- Author
-
Boilève A, De Cuyper A, Larive A, Mahjoubi L, Najdawi M, Tazdait M, Gelli M, Tselikas L, Smolenschi C, Malka D, Pignon JP, Ducreux M, and Boige V
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Bevacizumab administration & dosage, Camptothecin administration & dosage, Camptothecin analogs & derivatives, ErbB Receptors antagonists & inhibitors, Female, Fluorouracil administration & dosage, Hepatic Artery, Humans, Infusions, Intra-Arterial, Leucovorin administration & dosage, Liver Neoplasms mortality, Male, Middle Aged, Oxaliplatin adverse effects, Colorectal Neoplasms pathology, Liver Neoplasms drug therapy, Liver Neoplasms secondary, Oxaliplatin administration & dosage
- Abstract
Background: Hepatic arterial infusion (HAI) combined with systemic chemotherapy has shown promising results in patients with unresectable colorectal liver metastases (CRLM), even after failure to systemic therapy. Addition of systemic targeted therapies has been investigated with controversial results regarding tolerance, especially with HAI-floruxidine when combined with systemic bevacizumab. Our study aimed to analyse feasibility, safety and efficacy of HAI-oxaliplatin plus systemic chemotherapy and targeted therapies., Methods: Between 2005 and 2016, single-centre consecutive patients with unresectable CRLM who received at least one cycle of HAI-oxaliplatin plus systemic chemotherapy and targeted therapies (cetuximab/panitumumab or bevacizumab) were analysed., Results: A total of 89 patients (median age 55 years (range, 26-76 years) who previously received a median number of one systemic chemotherapy regimen (range, 0-5) including oxaliplatin in 78% of cases were included. Median number of HAI-oxaliplatin cycles was 9 (range, 1-28) combined with systemic chemotherapy and targeted therapies (LV5FU2 [63%], FOLFIRI [36%]) plus anti-EGFR (30%), or bevacizumab (70%). Grade 3/4 toxicities included neutropenia (40%), HAI-related abdominal pain (43%) and neurotoxicity (12%). The intent-to-treat objective response rate was 42%, and 45% had stable disease, allowing complete CRLM resection/ablation in 27% of patients. After a median follow-up of 72 months, median overall and progression-free survival was 20 and 9 months, respectively., Conclusion: Addition of targeted therapy to systemic chemotherapy combined with HAI-oxaliplatin is feasible, safe and shows promising activity, even after systemic chemotherapy failure., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
16. Chemotherapy beyond immune checkpoint inhibitors in patients with metastatic colorectal cancer.
- Author
-
Martin-Romano P, Ammari S, El-Dakdoukti Y, Baldini C, Varga A, Vuagnat P, Angevin E, Bahleda R, Gazzah A, Champiat S, Michot JM, Postel-Vinay S, Marabelle A, Soria JC, Boige V, Malka D, Ducreux M, Massard C, and Hollebecque A
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Immune Checkpoint Inhibitors pharmacology, Male, Middle Aged, Retrospective Studies, Colorectal Neoplasms drug therapy, Immune Checkpoint Inhibitors therapeutic use
- Abstract
Background: Immune checkpoint inhibitors (ICIs) alone or in combination with chemotherapy (CT) are the current standard of therapy in several cancer types. Patients (pts) with lung cancer display higher response rates to CT when given after ICIs. Although ICIs have failed to demonstrate antitumour activity in microsatellite stable (MSS) metastatic colorectal cancer (mCRC), little is known about CT effect after ICIs. We aimed to assess whether sequential ICIs followed by CT may be an alternative therapeutic approach in a population of pts with mCRC., Material and Methods: We retrospectively assessed CT after ICI (CAICI) failure in pts with mCRC. The ICI regimen consisted of anti-PD(L)1 alone or in combination. The primary end-point was objective response rate. Progression-free survival (PFS) and overall survival (OS) were secondary end-points., Results: Between 2014 and 2018, 29 pts with mCRC received CAICI (MSS tumours, 27 pts [86%]). The median number of previous lines was 4 (range, 2-7). Regimens included TAS-102 (n = 14), FOLFIRI (irinotecan, leucovorin, and fluorouracil; n = 6) or FOLFOX (oxaliplatin, leucovorin, and fluorouracil; n = 4), regorafenib (n = 3) and carboplatin (1 pt with BRCA mutation). Partial response and stable disease were observed in 4 (19%) and 9 (43%) pts, respectively (disease control rate, 62%). The median PFS and OS were 3.8 months (95% confidence interval [CI] = 1.5-5.4) and 8.0 months (95% CI = 4.2-14.0), respectively., Conclusion: ICIs administered before CT might enhance cytotoxic effects even in pts with immunorefractory MSS mCRC. The results of this small cohort need to be validated in independent prospective cohorts. The role of ICIs as modifiers of both tumour cells and microenvironment in mCRC deserves further research., Competing Interests: Conflict of interest statement All authors report being a Principal/sub-Investigator of Clinical Trials for Abbvie, Adaptimmune, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Arno Therapeutics, Astex Pharmaceuticals, AstraZeneca Ab, Aveo, Basilea Pharmaceutica International Ltd, Bayer Healthcare Ag, Bbb Technologies Bv, Beigene, Blueprint Medicines, Boehringer Ingelheim, Boston Pharmaceuticals, Bristol Myers Squibb, Ca, Celgene Corporation, Chugai Pharmaceutical Co, Clovis Oncology, Cullinan-Apollo, Daiichi Sankyo, Debiopharm, Eisai, Eisai Limited, Eli Lilly, Exelixis, Forma Tharapeutics, Gamamabs, Genentech, Glaxosmithkline, H3 Biomedicine, Hoffmann La Roche Ag, Imcheck Therapeutics, Innate Pharma, Institut De Recherche Pierre Fabre, Iris Servier, Janssen Cilag, Janssen Research Foundation, Kyowa Kirin Pharm. Dev, Lilly France, Loxo Oncology, Lytix Biopharma As, Medimmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Millennium Pharmaceuticals, Molecular Partners Ag, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology Nv, Oncoethix, Oncopeptides, Orion Pharma, Ose Pharma, Pfizer, Pharma Mar, Pierre Fabre, Medicament, Roche, Sanofi Aventis, Sotio A.S, Syros Pharmaceuticals, Taiho Pharma, Tesaro and Xencor; report receiving research grants from Astrazeneca, BMS, Boehringer Ingelheim, Janssen Cilag, Merck, Novartis, Pfizer, Roche and Sanofi and report receiving non-financial support (drug supplied) from Astrazeneca, Bayer, BMS, Boringher Ingelheim, Medimmune, Merck, NH TherAGuiX, Pfizer and Roche. C.M. reports receiving honoraria from Astellas, AstraZeneca, Bayer, BeiGene, BMS, Celgene, Debiopharm, Genentech, Ipsen, Janssen, Lilly, MedImmune, MSD, Novartis, Pfizer, Roche, Sanofi and Orion. S.C. reports receiving honoraria from AstraZeneca, BMS, Janssen, MSD, Novartis and Roche. A. H. has received honoraria from Amgen, Spectrum Pharmaceuticals, Lilly. J.C.S. reports receiving consultancy fees from AstraZeneca, Astex, Clovis, GSK, GamaMabs, Lilly, MSD, Mission Therapeutics, Merus, Pfizer, PharmaMar, Pierre Fabre, Roche-Genentech, Sanofi, Servier, Symphogen and Takeda over the past 3 years; reports serving as a full-time employee for AstraZeneca between September 2017–December 2019 and reports being a shareholder of Gritstone. A.M. reports receiving honoraria from GSK, AstraZeneca, Oncovir, Merck Serono, eTheRNA, Lytix pharma, Kyowa Kirin Pharma, Bayer, Novartis, BMS, Symphogen, Genmab, Amgen, Biothera, Nektar, Pfizer, Seattle Genetics, Flexus Bio, Roche/Genentech, OSE immunotherapeutics, Transgene, Gritstone, Merck (MSD), Cerenis, Innate pharma, Protagen, Partner Therapeutics and Servier. S.P.V. reports receiving honoraria from Merck KGaA and AstraZeneca. V. B. reports receiving honoraria from Bayer, Merck Serono, MSD, Roche, Sanofi and Ipsen. D.M. reports receiving honoraria from Amgen, Bayer, Merck Serono, MSD, Roche, Sanofi, HalioDx and Servier., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
17. Palliation of dysphagia in metastatic oesogastric cancers: An international multidisciplinary position.
- Author
-
Levy A, Wagner AD, Chargari C, Moehler M, Verheij M, Durand-Labrunie J, Kissel M, Chirat E, Burtin P, Ducreux M, Boige V, Nilsson M, Boku N, Chau I, and Deutsch E
- Subjects
- Brachytherapy, Consensus, Deglutition Disorders etiology, Deglutition Disorders physiopathology, Esophageal Neoplasms complications, Esophageal Neoplasms pathology, Humans, Neoplasm Metastasis, Nutritional Support, Quality of Life, Recovery of Function, Self Expandable Metallic Stents, Treatment Outcome, Antineoplastic Agents therapeutic use, Deglutition, Deglutition Disorders therapy, Esophageal Neoplasms drug therapy, Palliative Care
- Abstract
Malignant dysphagia is the most common symptom in advanced oesogastric cancers patients. Relief of dysphagia allows quality of life improvement, nutritional replenishment and potentially improves prognosis. Chemotherapy alone is effective and should be prioritised in patients with metastatic disease a good performance status, and its impact on dysphagia should be determined before further interventions are planned. Regarding local treatments, the insertion of a covered self-expandable metallic stent is the most commonly used alternative, as it allows for the rapid relief of severe dysphagia. Although several randomised trials have highlighted the role of oesophageal brachytherapy, this technique is often not easily accessible. Contemporary trials are ongoing to better define the role of external radiation therapy. While awaiting these results, external radiation therapy can be considered as a second-best option for patients with a life-expectancy > 3 months. It is important to offer nutritional support and to integrate quality of life measures in the palliative management of dysphagia. This multidisciplinary international position paper aims to propose a decision-making process and highlight randomised trials for the management of malignant dysphagia in metastatic oesogastric cancer patients., Competing Interests: Conflict of interest statement Following authors declared financial support, but not related to this work: Other authors did not declare conflict of interest., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
18. 5-Fluorouracil-induced hyperammonaemic encephalopathy: A French national survey.
- Author
-
Boilève A, Thomas L, Lillo-Le Louët A, Gaboriau L, Chouchana L, Ducreux M, Malka D, Boige V, Hollebecque A, Hillaire-Buys D, and Jozwiak M
- Subjects
- Aged, Ammonia blood, Antimetabolites, Antineoplastic administration & dosage, Brain Diseases blood, Brain Diseases chemically induced, Brain Diseases therapy, Citric Acid Cycle drug effects, Dose-Response Relationship, Drug, Female, Fluorouracil administration & dosage, France epidemiology, Humans, Hyperammonemia blood, Hyperammonemia chemically induced, Hyperammonemia therapy, Incidence, Male, Middle Aged, Pharmacovigilance, Retrospective Studies, Treatment Outcome, Urea metabolism, Antimetabolites, Antineoplastic adverse effects, Brain Diseases epidemiology, Fluorouracil adverse effects, Hyperammonemia epidemiology, Neoplasms drug therapy
- Abstract
Background: 5-Fluorouracil (5-FU)-induced hyperammonaemic encephalopathy is a rare but serious 5-FU adverse drug reaction (ADR). Given the growing number of cancers treated with 5-FU and the paucity of data regarding this ADR, we performed a retrospective national survey to better characterise 5-FU-induced hyperammonaemic encephalopathy., Patients and Methods: Since inception of the French pharmacovigilance database, we identified all patients who experienced 5-FU-induced hyperammonaemic encephalopathy. Variables regarding demographics, characteristics, management and outcome of patients were collected., Results: From 1986 to 2018, 30 patients were included. 5-FU-induced hyperammonaemic encephalopathy started 2 [1-4] days after 5-FU infusion onset. Most common neurological disorders were consciousness impairment, seizures and confusion. hyperammonaemia tended to be higher in patients with the lowest Glasgow score and admitted in intensive care unit (ICU) compared to non-ICU patients (250 [133-522] versus 139 [68-220] μmol/L respectively, p = NS). Dihydropyrimidine dehydrogenase deficiency was found in 27% of tested patients (n = 3/11). Encephalopathy-induced mortality was 17%, 57% of patients were admitted in ICU and 70% had a complete neurological recovery within 5 [2-10] days. A 5-FU rechallenge was considered in 14 (67%) patients with neurological recovery and a relapse was observed in 57% of them. No 5-FU-induced hyperammonaemic encephalopathy relapse was observed as long as 5-FU rechallenge was performed with decreased 5-FU dosage., Conclusion: We report the largest cohort of 5-FU-induced hyperammonaemic encephalopathy cases so far. This ADR should be suspected and ammonaemia measured in all patients experiencing neurological disorders after 5-FU administration. In patients with complete neurological recovery, a 5-FU rechallenge could be cautiously considered., Competing Interests: Conflict of interest statement The authors have declared no conflicts of interest., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
19. Safety and effectiveness of regorafenib in patients with metastatic colorectal cancer in routine clinical practice in the prospective, observational CORRELATE study.
- Author
-
Ducreux M, Petersen LN, Öhler L, Bergamo F, Metges JP, de Groot JW, Wang JY, García Paredes B, Dochy E, Fiala-Buskies S, Cervantes A, O'Connor JM, and Falcone A
- Subjects
- Adult, Aged, Aged, 80 and over, Bone Neoplasms genetics, Bone Neoplasms secondary, Carcinoma secondary, Colorectal Neoplasms genetics, Fatigue chemically induced, Female, GTP Phosphohydrolases genetics, Hand-Foot Syndrome etiology, Humans, Hypertension chemically induced, Liver Neoplasms genetics, Liver Neoplasms secondary, Lung Neoplasms genetics, Lung Neoplasms secondary, Lymph Nodes pathology, Male, Membrane Proteins genetics, Middle Aged, Neoplasm Metastasis, Peritoneal Neoplasms genetics, Peritoneal Neoplasms secondary, Progression-Free Survival, Prospective Studies, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins p21(ras) genetics, Young Adult, Bone Neoplasms drug therapy, Carcinoma drug therapy, Colorectal Neoplasms pathology, Liver Neoplasms drug therapy, Lung Neoplasms drug therapy, Peritoneal Neoplasms drug therapy, Phenylurea Compounds therapeutic use, Pyridines therapeutic use
- Abstract
Background: Regorafenib prolonged overall survival (OS) versus placebo in patients with treatment-refractory metastatic colorectal cancer (mCRC) in phase III trials. We conducted an observational study of regorafenib for patients with mCRC in real-world clinical practice., Methods: The international, prospective, CORRELATE study recruited patients with mCRC previously treated with approved therapies, for whom the decision to treat with regorafenib was made by the treating physician according to the local health authority approved label. The primary objective was safety, assessed by treatment-emergent adverse events (TEAEs; National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.03)., Results: A total of 1037 patients were treated. The median age was 65 years (range: 24-93); 87% of patients had Eastern Cooperative Oncology Group performance status 0-1, 56% of patients had KRAS, 7% had NRAS and 4% had BRAF mutations. The initial regorafenib dose was 160 mg/day in 57% of patients. The most common grade III or IV drug-related TEAEs were fatigue (9%), hand-foot skin reaction (7%) and hypertension (6%). Drug-related grade V (fatal) TEAEs occurred in 1% of patients. Dose reductions for drug-related TEAEs occurred in 24% of patients. Median OS was 7.7 months (95% confidence interval [CI]: 7.2-8.3), and median progression-free survival (PFS) was 2.9 months (95% CI: 2.8-3.0)., Conclusions: In this real-world, observational study of patients with mCRC, the regorafenib toxicity profile was similar to that reported in phase III trials. The starting dose for almost half of patients was less than the approved 160-mg dose, and the median OS and PFS were in the range observed in phase III trials., Trial Registration: NCT02042144., (Copyright © 2019 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
20. Vision loss after chemotherapy: an irinotecan-induced retinopathy.
- Author
-
Boilève A, Jozwiak M, Malka D, Boige V, Le Roy F, Paques M, and Ducreux M
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colonic Neoplasms drug therapy, Female, Humans, Irinotecan therapeutic use, Middle Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Irinotecan adverse effects, Retinal Diseases chemically induced, Vision Disorders chemically induced
- Published
- 2019
- Full Text
- View/download PDF
21. The 4th St. Gallen EORTC Gastrointestinal Cancer Conference: Controversial issues in the multimodal primary treatment of gastric, junctional and oesophageal adenocarcinoma.
- Author
-
Lutz MP, Zalcberg JR, Ducreux M, Adenis A, Allum W, Aust D, Carneiro F, Grabsch HI, Laurent-Puig P, Lordick F, Möhler M, Mönig S, Obermannova R, Piessen G, Riddell A, Röcken C, Roviello F, Schneider PM, Seewald S, Smyth E, van Cutsem E, Verheij M, Wagner AD, and Otto F
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Combined Modality Therapy methods, Humans, Neoadjuvant Therapy methods, Adenocarcinoma therapy, Esophageal Neoplasms therapy, Esophagogastric Junction drug effects, Stomach Neoplasms therapy
- Abstract
Multimodal primary treatment of localised adenocarcinoma of the stomach, the oesophagus and the oesophagogastric junction (AEG) was reviewed by a multidisciplinary expert panel in a moderated consensus session. Here, we report the key points of the discussion and the resulting recommendations. The exact definition of the tumour location and extent by white light endoscopy in conjunction with computed tomography scans is the backbone for any treatment decision. Their value is limited with respect to the infiltration depth, lymph node involvement and peritoneal involvement. Additional endoscopic ultrasound was recommended mainly for tumours of the lower oesophagogastric junction (i.e. AEG type II and III according to Siewert) and in early cancers before endoscopic resection. Laparoscopy to diagnose peritoneal involvement was thought to be necessary before the start of neoadjuvant treatment in all gastric cancers and in AEG type II and III. In general, perioperative multimodal treatment was suggested for all locally advanced oesophageal tumours and for gastric cancers with a clinical stage above T1N0. There was consensus that the combination of fluorouracil, folinic acid, oxaliplatin and docetaxel is now a new standard chemotherapy (CTx) regimen for fit patients. In contrast, the optimal choice of perioperative CTx versus neoadjuvant radiochemotherapy (neoRCTx), especially for AEG, was identified as an open question. Expert treatment recommendations depend on the tumour location, biology, the risk of incomplete (R1) resection, response to treatment, local or systemic recurrence risks, the predicted perioperative morbidity and patients' comorbidities. In summary, any treatment decision requires an interdisciplinary discussion in a comprehensive multidisciplinary setting., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
22. Screening and surveillance in hereditary gastrointestinal cancers: Recommendations from the European Society of Digestive Oncology (ESDO) expert discussion at the 20th European Society for Medical Oncology (ESMO)/World Congress on Gastrointestinal Cancer, Barcelona, June 2018.
- Author
-
Vangala DB, Cauchin E, Balmaña J, Wyrwicz L, van Cutsem E, Güller U, Castells A, Carneiro F, Hammel P, Ducreux M, van Laethem JL, Matysiak-Budnik T, and Schmiegel W
- Subjects
- Age of Onset, Chemoprevention, Colonoscopy, DNA Mutational Analysis, Early Detection of Cancer methods, Gastrointestinal Neoplasms diagnosis, Gastrointestinal Neoplasms epidemiology, Gastrointestinal Neoplasms prevention & control, Genetic Counseling, Genetic Testing standards, Germ-Line Mutation, Humans, Molecular Diagnostic Techniques standards, Neoplastic Syndromes, Hereditary diagnosis, Oncogenes, Organ Specificity, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms epidemiology, Pancreatic Neoplasms prevention & control, Risk Assessment, Sequence Analysis, DNA methods, Sequence Analysis, DNA standards, Early Detection of Cancer standards, Gastrointestinal Neoplasms genetics, Neoplastic Syndromes, Hereditary epidemiology, Pancreatic Neoplasms genetics
- Abstract
Patients with hereditary gastrointestinal (GI) cancers represent a substantial fraction of the overall affected population. Although awareness for hereditary GI cancer syndromes is on the rise, identification of patients and measures of surveillance are often unclear in everyday clinical routine. Therefore, the European Society of Digestive Oncology expert discussion 2018 at the World Congress on Gastrointestinal Cancer focussed on screening and surveillance of hereditary colorectal, gastric and pancreatic cancers. An international panel of experts and opinion leaders developed the here presented recommendations based on published evidence and on profound clinical expertise to facilitate clinical routine in identification and caretaking of patients with familial GI cancers., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
23. Peritoneal and extraperitoneal relapse after previous curative treatment of peritoneal metastases from colorectal cancer: What survival can we expect?
- Author
-
Gelli M, Huguenin JFL, de Baere T, Benhaim L, Mariani A, Boige V, Malka D, Sourouille I, Ducreux M, Elias D, and Goéré D
- Subjects
- Adolescent, Adult, Aged, Colorectal Neoplasms mortality, Databases, Factual, Disease-Free Survival, Female, Humans, Male, Middle Aged, Peritoneal Neoplasms mortality, Recurrence, Retreatment, Retrospective Studies, Risk Factors, Time Factors, Treatment Outcome, Young Adult, Colorectal Neoplasms pathology, Cytoreduction Surgical Procedures adverse effects, Cytoreduction Surgical Procedures mortality, Hyperthermia, Induced adverse effects, Hyperthermia, Induced mortality, Peritoneal Neoplasms secondary, Peritoneal Neoplasms therapy
- Abstract
Introduction: Over the last 20 years, complete cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) dramatically increased the survival of patients with colorectal peritoneal metastases (CRPM). However, despite better knowledge of the disease, around 70% of patients relapse after CRS with HIPEC. This study was designed to analyse the pattern of recurrence and the outcomes of different treatment modalities., Methods: Patients relapsing after CRS plus HIPEC for CRPM were selected from a prospective database. The impact of iterative curative-intent treatments was analysed using Kaplan-Meier estimates and multivariate Cox regression models., Results: Between April 1993 and December 2014, 190 of 274 (69%) patients previously treated by CRS plus HIPEC developed relapse, as an isolated peritoneal recurrence (31%), isolated distant recurrence (35%), or multisite recurrence (34%). The curative-intent treatment rate was 48% for isolated peritoneal recurrences, 49% for isolated distant recurrences and 22% for multisite recurrences (p = 0.002). From the diagnosis of relapse, 3- and 5-year overall survival were 77% and 46% after curative-intent treatment and 14% and 4.7% after non-curative treatment, with median survival of 59.7 and 18.3 months (log-rank p < 0.0001), respectively. Regression analysis identified the initial extent of CRPM (hazard ratio [HR]: 2.25; p < 0.0001), iterative curative-intent treatment (HR: 0.22; p < 0.0001) and disease-free interval (HR: 1.77; p = 0.01) as independent predictors of prolonged survival., Conclusions: Iterative curative-intent treatment can be performed in up to 40% of patients with relapse after CRS and HIPEC for CRPM, and is associated with prolonged survival in selected patients., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
24. Overweight is associated to a better prognosis in metastatic colorectal cancer: A pooled analysis of FFCD trials.
- Author
-
Aparicio T, Ducreux M, Faroux R, Barbier E, Manfredi S, Lecomte T, Etienne PL, Bedenne L, Bennouna J, Phelip JM, François E, Michel P, Legoux JL, Gasmi M, Breysacher G, Rougier P, De Gramont A, Lepage C, Bouché O, and Seitz JF
- Subjects
- Aged, Angiogenesis Inhibitors administration & dosage, Clinical Trials as Topic, Colorectal Neoplasms pathology, Colorectal Neoplasms physiopathology, Female, Humans, Kaplan-Meier Estimate, Male, Neoplasm Metastasis, Outcome Assessment, Health Care methods, Outcome Assessment, Health Care statistics & numerical data, Prognosis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Body Mass Index, Colorectal Neoplasms drug therapy, Overweight physiopathology
- Abstract
Background: Previous studies showed that high and low body mass index (BMI) was associated with worse prognosis in early-stage colorectal cancer (CRC), and low BMI was associated with worse prognosis in metastatic CRC (mCRC). We aimed to assess efficacy outcomes according to BMI., Patients and Methods: A pooled analysis of individual data from 2085 patients enrolled in eight FFCD first-line mCRC trials from 1991 to 2013 was performed. Comparisons were made according to the BMI cut-off: Obese (BMI ≥30), overweight patients (BMI ≥ 25), normal BMI patients (BMI: 18.5-24) and thin patients (BMI <18.5). Interaction tests were performed between BMI effect and sex, age and the addition of antiangiogenics to chemotherapy., Results: The rate of BMI ≥25 patients was 41.5%, ranging from 37.6% (1991-1999 period) to 41.5% (2000-2006 period) and 44.8% (2007-2013 period). Comparison of overweight patients versus normal BMI range patients revealed a significant improvement of median overall survival (OS) (18.5 versus 16.3 months, HR = 0.88 [0.80-0.98] p = 0.02) and objective response rate (ORR) (42% versus 36% OR = 1.23 [1.01-1.50] p = 0.04) but a comparable median progression-free survival (PFS) (7.8 versus 7.2 months, HR = 0.96 [0.87-1.05] p = 0.35). Subgroup analyses revealed that overweight was significantly associated with better OS in men. OS and PFS were significantly shorter in thin patients., Conclusion: Overweight patients had a prolonged OS compared with normal weight patients with mCRC. The association of overweight with better OS was only observed in men. The pejorative prognosis of BMI <18.5 was confirmed., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
25. Corrigendum to "Precision medicine for patients with advanced biliary tract cancers: An effective strategy within the prospective MOSCATO-01 trial" [Eur J Cancer 87 (2017) 122-130].
- Author
-
Verlingue L, Malka D, Allorant A, Massard C, Ferté C, Lacroix L, Rouleau E, Auger N, Ngo M, Nicotra C, De Baere T, Tselikas L, Ba B, Michiels S, Scoazec JY, Boige V, Ducreux M, Soria JC, and Hollebecque A
- Published
- 2018
- Full Text
- View/download PDF
26. Circulating oncometabolite D-2-hydroxyglutarate enantiomer is a surrogate marker of isocitrate dehydrogenase-mutated intrahepatic cholangiocarcinomas.
- Author
-
Delahousse J, Verlingue L, Broutin S, Legoupil C, Touat M, Doucet L, Ammari S, Lacroix L, Ducreux M, Scoazec JY, Malka D, Paci A, and Hollebecque A
- Subjects
- Adult, Aged, Bile Duct Neoplasms blood, Bile Duct Neoplasms genetics, Cholangiocarcinoma blood, Cholangiocarcinoma genetics, Female, Humans, Isocitrate Dehydrogenase genetics, Isomerism, Male, Middle Aged, Mutation, Bile Duct Neoplasms diagnosis, Biomarkers, Tumor blood, Cholangiocarcinoma diagnosis, Glutarates blood
- Abstract
Therapeutic resources are limited for advanced biliary tract cancers and prognosis remains poor. Somatic mutations in isocitrate dehydrogenase (IDH)1/2 gene are found in 5-36% of patients with intrahepatic cholangiocarcinoma (ICC). The mutant forms of IDH1/2 catalyse the non-reversible accumulation of 2-hydroxyglutarate (2HG). Increasing numbers of indirect or direct-targeted therapies are developed to IDH1/2 mutations and could be assisted by a routinely feasible, rapid and inexpensive serum 2HG measurement by liquid chromatography coupled to tandem mass spectrometry. By comparing eight patients with an IDH1/2-mutated ICC to nine patients with wild-type IDH1/2 ICC, we found significantly higher levels of 2HG in patients with IDH1/2 mutations versus the wild-type group (median, 10.9 vs. 0.8 μmol/L, p = 0.0037). D and L-2HG enantiomer levels significantly differed between the two groups with a higher level of D-2HG (p < 0.0001) in patients with IDH1/2 mutations. Accordingly, the D/L ratio was markedly higher in the patients with IDH1/2 mutations compared with the wild-type group (38.0 vs. 0.9 μmol/L, p < 0.0001). D-2HG measurement ensured 100% sensitivity and specificity at a cut-off of 0.6 μmol/L. D-2HG levels were correlated with tumour burden and tumour response to treatment with IDH-targeted therapies or indirect therapies. D-2HG serum level measurement by liquid chromatography coupled to tandem mass spectrometry is a sensitive, specific, precise (a coefficient of variation <10% and an accuracy >95%), fast (9 min run per sample) and inexpensive surrogate marker of IDH1/2 somatic mutation in ICC. Systematic measurement in patients with ICC may facilitate access to, and monitoring of, IDH-driven therapies., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. Precision medicine for patients with advanced biliary tract cancers: An effective strategy within the prospective MOSCATO-01 trial.
- Author
-
Verlingue L, Malka D, Allorant A, Massard C, Ferté C, Lacroix L, Rouleau E, Auger N, Ngo M, Nicotra C, De Baere T, Tselikas L, Ba B, Michiels S, Scoazec JY, Boige V, Ducreux M, Soria JC, and Hollebecque A
- Subjects
- Adult, Aged, Biliary Tract Neoplasms mortality, Biliary Tract Neoplasms pathology, Biopsy, Disease-Free Survival, Female, Genetic Predisposition to Disease, Humans, Male, Middle Aged, Phenotype, Predictive Value of Tests, Prospective Studies, Risk Factors, Time Factors, Treatment Outcome, Antineoplastic Agents therapeutic use, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms genetics, Biomarkers, Tumor genetics, High-Throughput Nucleotide Sequencing, Molecular Diagnostic Techniques, Precision Medicine
- Abstract
Background: Recommended treatments of patients with advanced biliary tract cancer (aBTC) are limited to one chemotherapy doublet. Nevertheless, efficacy of treatment personalisation for aBTCs is supported by accumulating evidences but remains to be evaluated., Patients and Methods: Patients with aBTCs included in the prospective clinical trial MOSCATO-01 were treated by at least one previous systemic treatment, had an ECOG performance status of 0-1, and at least one tumour site accessible to biopsy. Multiple high-throughput molecular analysis was performed on biopsies to drive the administration of molecular targeted agents (MTAs)., Results: From November 2011 to March 2016, 43 patients (4%) of the 1035 adult patients included in MOSCATO-01 had aBTCs with a majority of intrahepatic localisation (67%). Successful biopsy procedures and DNA extractions led to molecular portraits for 34 patients (79%). Orientation to an appropriate early clinical trial or accessible MTA(s) was possible for 23 of these patients (68%), and 18 (53%) have received matched MTA(s). Among them, the overall response rate was 33% and the disease control rate was 88%. A PFS ≥6 months was observed in 37% and the PFS ratio was >1.3 for 50% of the patients. These patients had a lower risk for death as compared to the 20 patients not orientated to a matched MTA (HR, 0.29; 95% CI, 0.11-0.76; p = 0.008)., Conclusions: Within the MOSCATO-01 trial, patients with aBTCs had the highest rate of orientation to matched MTAs and derived a clear clinical benefit. A broader evaluation of these findings may improve future treatments strategies for aBTCs., Trial Registration: NCT01566019., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
28. Clinical relevance of molecular diagnostics in gastrointestinal (GI) cancer: European Society of Digestive Oncology (ESDO) expert discussion and recommendations from the 17th European Society for Medical Oncology (ESMO)/World Congress on Gastrointestinal Cancer, Barcelona.
- Author
-
Baraniskin A, Van Laethem JL, Wyrwicz L, Guller U, Wasan HS, Matysiak-Budnik T, Gruenberger T, Ducreux M, Carneiro F, Van Cutsem E, Seufferlein T, and Schmiegel W
- Subjects
- Consensus, Gastrointestinal Neoplasms pathology, Gastrointestinal Neoplasms therapy, Genetic Predisposition to Disease, Humans, Interdisciplinary Communication, Phenotype, Predictive Value of Tests, Prognosis, Biomarkers, Tumor genetics, Gastrointestinal Neoplasms genetics, Molecular Diagnostic Techniques standards, Precision Medicine standards
- Abstract
Background and Scope: In the epoch of precision medicine and personalised oncology, which aims to deliver the right treatment to the right patient, molecular genetic biomarkers are a topic of growing interest. The aim of this expert discussion and position paper is to review the current status of various molecular tests for gastrointestinal (GI) cancers and especially considering their significance for the clinical routine use., Methodology: Opinion leaders and experts from diverse nationalities selected on scientific merit were asked to answer to a prepared set of questions about the current status of molecular diagnostics in different GI cancers. All answers were then discussed during a plenary session and reported here in providing a well-balanced reflection of both clinical expertise and updated evidence-based medicine., Results: Preselected molecular genetic biomarkers that are described and disputed in the current medical literature in different GI cancers were debated, and recommendations for clinical routine practice were made whenever possible. Furthermore, the preanalytical variations were commented and proposals for quality controls of biospecimens were made., Conclusion: The current article summarises the recommendations of the expert committee regarding prognostic and predictive molecular genetic biomarkers in different entities of GI cancers. The briefly and comprehensively formulated guidelines should assist clinicians in the process of decision making in daily clinical practice., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
29. Matching genomic molecular aberrations with molecular targeted agents: Are biliary tract cancers an ideal playground?
- Author
-
Verlingue L, Hollebecque A, Boige V, Ducreux M, Malka D, and Ferté C
- Subjects
- Biliary Tract Neoplasms genetics, Clinical Trials as Topic, Epigenomics, Humans, Antineoplastic Agents therapeutic use, Biliary Tract Neoplasms drug therapy, Immunotherapy methods, Molecular Targeted Therapy methods, Mutation
- Abstract
Biliary tract cancers (BTCs) are a heterogeneous group of tumours with geographical discrepancies in terms of incidence and risk factors. However, a convergent genomic and epigenetic mutational landscape emerges from the genome-wide screens of BTCs in South East Asia, Latin America and in the Western World. Specificities are observed for some alterations and anatomical subtypes: frequent fibroblast growth factor receptor 2 (FGFR2) and isocitrate dehydrogenase 1/2 (IDH1/2) alterations are specific to intrahepatic cholangiocarcinomas (ICCs), whereas frequent ERBB2 oncogene alterations are specific to extrahepatic cholangiocarcinomas (ECCs) and gallbladder carcinomas (GBCs). Until now, the outcome of patients with BTCs treated by molecular targeted agents (MTAs) alone or in combination with conventional chemotherapy in non-biology driven trials remains poor and does not exceed the outcome of patients treated with chemotherapy alone. Encouraging reports of biology-driven therapeutic approaches should accelerate the clinical development of MTAs in BTCs. Additionally, frequent epigenetic aberrations such as IDH1/2 mutations and switch/sucrose non-fermenting (SWI/SNF) complex dysfunctions suggest that epidrugs must also be considered. In this review, we expose the rationale and feasibility to biologically drive the treatment of BTC patients., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. Dynamic evaluation of circulating tumour cells in patients with advanced gastric and oesogastric junction adenocarcinoma: Prognostic value and early assessment of therapeutic effects.
- Author
-
Pernot S, Badoual C, Terme M, Castan F, Cazes A, Bouche O, Bennouna J, Francois E, Ghiringhelli F, De La Fouchardiere C, Samalin E, Bachet JB, Borg C, Ducreux M, Marcheteau E, Stanbury T, Gourgou S, Malka D, and Taieb J
- Subjects
- Antibodies, Monoclonal, Humanized administration & dosage, Cell Count, Disease Progression, Disease-Free Survival, Esophageal Neoplasms drug therapy, Female, Fluorouracil administration & dosage, Humans, Kaplan-Meier Estimate, Leucovorin administration & dosage, Male, Organoplatinum Compounds administration & dosage, Oxaliplatin, Prognosis, Stomach Neoplasms drug therapy, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Esophageal Neoplasms pathology, Esophagogastric Junction pathology, Neoplastic Cells, Circulating pathology, Stomach Neoplasms pathology
- Abstract
Background: The identification of dynamic biomarkers in advanced gastric and oesogastric junction adenocarcinoma (GOA) could help to tailor strategies for each patient. Enumeration of circulating tumour cells (CTCs) is approved by the US Food and Drug Administration in breast, colon and prostate cancer but is not in advanced GOA. Our study aims to establish the optimal threshold and the clinical significance of CTC count in advanced GOA before and during treatment., Methods: One hundred six patients with untreated advanced GOA were included in the ancillary study of the PRODIGE 17-ACCORD 20 trial. CTCs were detected in the peripheral blood using the CellSearch system on day 0 (D0) and day 28 (D28). The prognostic value of CTCs at D0 and D28 was analysed by testing several thresholds., Results: At baseline, median CTC count was 1 (range, 0-415). While CTCs ≥1, 2 or 3 at D0 were all significantly associated with worse overall survival (OS) and progression-free survival (PFS), CTCs ≥2 were the optimal threshold, on D0 or D28. CTCs ≥2 at D28 were also predictive of disease control. Taking into account both D0 and D28 CTC count defined 3 groups (low/low, high/low and low-high/high) with significantly different PFS (p = 0.0002) and OS (p = 0.003)., Conclusion: Quantification of CTCs at baseline and during treatment may be a useful prognostic tool in advanced GOA, as it is associated with worse PFS and OS. A threshold ≥2 CTCs seems to have the best discriminant value. Change in CTC count between baseline and D28 could help to tailor treatment to each individual patient., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
31. 3rd St. Gallen EORTC Gastrointestinal Cancer Conference: Consensus recommendations on controversial issues in the primary treatment of pancreatic cancer.
- Author
-
Lutz MP, Zalcberg JR, Ducreux M, Aust D, Bruno MJ, Büchler MW, Delpero JR, Gloor B, Glynne-Jones R, Hartwig W, Huguet F, Laurent-Puig P, Lordick F, Maisonneuve P, Mayerle J, Martignoni M, Neoptolemos J, Rhim AD, Schmied BM, Seufferlein T, Werner J, van Laethem JL, and Otto F
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chemotherapy, Adjuvant methods, Cholangiography methods, Consensus, Humans, Neoadjuvant Therapy methods, Neoplasm Staging, Pancreatic Neoplasms pathology, Stents, Pancreatic Neoplasms therapy
- Abstract
The primary treatment of pancreatic cancer was the topic of the 3rd St. Gallen Conference 2016. A multidisciplinary panel reviewed the current evidence and discussed controversial issues in a moderated consensus session. Here we report on the key expert recommendations. It was generally accepted that radical surgical resection followed by adjuvant chemotherapy offers the only evidence-based treatment with a chance for cure. Initial staging should classify localised tumours as resectable or unresectable (i.e. locally advanced pancreatic cancer) although there remains a large grey-zone of potentially resectable disease between these two categories which has recently been named as borderline resectable, a concept which was generally accepted by the panel members. However, the definition of these borderline-resectable (BR) tumours varies between classifications due to their focus on either (i) technical hurdles (e.g. the feasibility of vascular resection) or (ii) oncological outcome (e.g. predicting the risk of a R1 resection and/or occult metastases). The resulting expert discussion focussed on imaging standards as well as the value of pretherapeutic laparoscopy. Indications for biliary drainage were seen especially before neoadjuvant therapy. Following standard resection, the panel unanimously voted for the use of adjuvant chemotherapy after R0 resection and considered it as a reasonable standard of care after R1 resection, even though the optimal pathologic evaluation and the definition of R0/R1 was the issue of an ongoing debate. The general concept of BR tumours was considered as a good basis to select patients for preoperative therapy, albeit its current impact on the therapeutic strategy was far less clear. Main focus of the conference was to discuss the limits of surgical resection and to identify ways to standardise procedures and to improve curative outcome, including adjuvant and perioperative treatment., (Copyright © 2017 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
32. Antitumour activity of somatostatin analogues in sporadic, progressive, metastatic pulmonary carcinoids.
- Author
-
Sullivan I, Le Teuff G, Guigay J, Caramella C, Berdelou A, Leboulleux S, Déandréis D, Hadoux J, Ducreux M, Duvillard P, Adam J, Scoazec JY, Baudin E, and Planchard D
- Subjects
- Adult, Aged, Carcinoid Tumor mortality, Cell Proliferation drug effects, Disease Progression, Disease-Free Survival, Female, Humans, Lung Neoplasms mortality, Male, Middle Aged, Neoplasm Metastasis, Retrospective Studies, Somatostatin therapeutic use, Tomography, X-Ray Computed, Treatment Outcome, Antineoplastic Agents therapeutic use, Carcinoid Tumor drug therapy, Lung Neoplasms drug therapy, Octreotide therapeutic use, Peptides, Cyclic therapeutic use, Somatostatin analogs & derivatives
- Abstract
Purpose: Antiproliferative activity of somatostatin analogues (SSAs) has been demonstrated in digestive neuroendocrine tumours but few data have been published on pulmonary carcinoids (PC). The aim of this retrospective study was to report the antitumour activity of SSAs in patients with progressive, metastatic PC., Methods: Patients with PC and treated with SSA monotherapy were reviewed. Disease was classified according to the tumour slope prior to SSA initiation as rapidly progressive (at least 20% increase in the sum of the longest diameter of target lesions or the appearance of one or more new lesions within 6 months) or slowly progressive (if progression occurred over 6 months). Survival outcomes were progression-free survival (PFS) and overall survival (OS). We additionally examined the overall response rate and safety. Prognostic factors associated with PFS and OS were sought. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using the Cox model., Results: Among 67 patients reviewed, 61 were included in the study. Forty-one (67%) of them exhibited slowly progressive disease prior to SSAs, 41 (67%) had atypical carcinoids and 29 (48%) had functioning tumours. Forty-six (76%) patients had received SSAs as first-line therapy. The best overall response was stable disease in 47 (77%) patients. The median duration of SSAs was 13.7 months. With a median follow-up of 5.8 years, median PFS and OS were 17.4 (95% CI: 8.7-26.0) and 58.4 (95% CI: 44.2-102.7) months, respectively. Functioning tumours and slowly progressive disease were significantly associated with longer PFS: HR = 0.48 ([95% CI: 0.24-0.95], p = 0.03) and HR = 7.43 ([95% CI: 3.02-18.25], p < 0.0001), respectively. Only functioning tumours remained significantly associated with OS: HR = 0.33 ([95% CI: 0.14-0.79], p = 0.01). Treatment had been discontinued in two patients due to side-effects., Conclusions: Median PFS observed in our study is encouraging for PC patients. Patients with functioning tumours and slowly progressive disease treated with SSAs have better prognosis., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
33. The role of image-guided therapy in the management of colorectal cancer metastatic disease.
- Author
-
de Baere T, Tselikas L, Yevich S, Boige V, Deschamps F, Ducreux M, Goere D, Nguyen F, and Malka D
- Subjects
- Bone Neoplasms secondary, Brachytherapy methods, Chemoembolization, Therapeutic methods, Chemotherapy, Cancer, Regional Perfusion methods, Forecasting, Humans, Liver Neoplasms secondary, Lung Neoplasms secondary, Radiotherapy, Image-Guided, Randomized Controlled Trials as Topic, Surgery, Computer-Assisted methods, Bone Neoplasms therapy, Catheter Ablation methods, Colorectal Neoplasms, Liver Neoplasms therapy, Lung Neoplasms therapy
- Abstract
The European Society for Medical Oncology (ESMO) have stressed that the option for treating oligometastatic disease is a strategy of local ablative therapy, the goal of which is to improve disease control. The spectrum of the local ablative therapy toolbox described by the ESMO includes surgical R0 resection, percutaneous ablation and intra-arterial therapies, the choice of treatment being left to the multidisciplinary team. Interventional therapy involving image-guided treatment offers the possibility of less invasive treatments for colorectal cancer metastases in the liver, lung and bone by preserving from toxicity distant healthy organs or even parts of the diseased organs. Oligometastases can be targeted by image-guided puncture for percutaneous ablation by delivering locally, through inserted probes, heat (radiofrequency, microwaves), extreme cold (cryoablation) or electric pulses (electroporation). Radiofrequency (RFA) is the mainstay of percutaneous ablation and provides local control rates of around 90% when metastases are small (<3 cm), located away from hilum and large vessels, and perfectly visible under imaging guidance. The lung provides a specific environment with excellent visibility of the target tumour, and insulation of the tumour by the healthy lung improves thermal delivery. RFA of colorectal lung metastases provides a 5-year overall survival of 56.0%, with a 91.6% control rate for metastases with a diameter <3 cm. These results are comparable to results of surgical series. Non-resectable, non-ablatable liver metastases can be targeted through their preferential arterial vascularisation with hepatic arterial infusion chemotherapy (HAIC) or selective internal radiation therapy (SIRT) with radioactive microspheres. HAIC with oxaliplatin has demonstrated an impressive response rate when patients who have previously failed intravenous oxaliplatin are rechallenged. The response rate in first-line therapy is around 90%, with conversion to surgery in roughly 40% of patients. SIRT has recently demonstrated a benefit for progression-free survival in the liver when used as first-line treatment in combination with systemic therapy., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
34. Bevacizumab versus anti-epidermal growth factor receptor in first-line metastatic colorectal cancer. A meta-analysis: The last building block?
- Author
-
Ducreux M and Pignon JP
- Subjects
- Colorectal Neoplasms pathology, ErbB Receptors antagonists & inhibitors, ErbB Receptors immunology, Humans, Neoadjuvant Therapy, Neoplasm Metastasis, Panitumumab, Antibodies, Monoclonal therapeutic use, Bevacizumab therapeutic use, Cetuximab therapeutic use, Colorectal Neoplasms drug therapy, Meta-Analysis as Topic
- Published
- 2016
- Full Text
- View/download PDF
35. Early tumour response as a survival predictor in previously- treated patients receiving triplet hepatic artery infusion and intravenous cetuximab for unresectable liver metastases from wild-type KRAS colorectal cancer.
- Author
-
Bouchahda M, Boige V, Smith D, Karaboué A, Ducreux M, Hebbar M, Lepère C, Focan C, Guimbaud R, Innominato P, Awad S, Carvalho C, Tumolo S, Truant S, De Baere T, Castaing D, Rougier P, Morère JF, Taieb J, Adam R, and Lévi F
- Subjects
- Adenocarcinoma secondary, Adult, Aged, Camptothecin administration & dosage, Camptothecin analogs & derivatives, Cetuximab administration & dosage, Colorectal Neoplasms pathology, Fatigue chemically induced, Female, Fluorouracil administration & dosage, Hepatic Artery, Humans, Infusions, Intra-Arterial, Infusions, Intravenous, Irinotecan, Liver Neoplasms secondary, Logistic Models, Male, Middle Aged, Multivariate Analysis, Organoplatinum Compounds administration & dosage, Oxaliplatin, Proportional Hazards Models, Retrospective Studies, Survival Rate, Time Factors, Treatment Outcome, Adenocarcinoma drug therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, Liver Neoplasms drug therapy
- Abstract
Background: Early tumour shrinkage has been associated with improved survival in patients receiving cetuximab-based systemic chemotherapy for liver metastases from colorectal cancer (LM-CRC). We tested this hypothesis for previously treated LM-CRC patients receiving cetuximab (500 mg/m
2 ) and triplet hepatic artery infusion (HAI) within European trial OPTILIV., Methods: Irinotecan (180 mg/m2 ), 5-fluorouracil (2800 mg/m2 ) and oxaliplatin (85 mg/m2 ) were given as chronomodulated or conventional delivery. Patients were retrospectively categorised as early responders (complete or partial RECIST response after three courses) or non-early responders (late or no response). Prognostic factors were determined using multivariate logistic or Cox regression models., Results: Response was assessed in 57 of 64 registered patients (89%), who had previously received one to three prior systemic chemotherapy protocols. An early response occurred at 6 weeks in 16 patients (28%; 9 men, 7 women), aged 33-76 years, with a median of 12 liver metastases (LMs) (2-50), involving five segments (1-8). Ten patients had a late response, and 31 patients had no response. Grade 3-4 fatigue selectively occurred in the non-early responders (0% versus 26%; p = 0.024). Early tumour response was jointly predicted by chronomodulation-odds ratio (OR): 6.0 (1.2-29.8; p = 0.029)-and LM diameter ≤57 mm-OR: 5.3 (1.1-25.0; p = 0.033). Early tumour response predicted for both R0-R1 liver resection-OR: 11.8 (1.4-100.2; p = 0.024) and overall survival-hazard ratio: 0.39 (0.17-0.88; p = 0.023) in multivariate analyses., Conclusions: Early tumour response on triplet HAI and systemic cetuximab predicted for complete macroscopic liver resection and prolonged survival for LM-CRC patients within a multicenter conversion-to-resection medicosurgical strategy. Confirmation is warranted for early response on HAI to guide decision making. Protocol numbers: EUDRACT 2007-004632-24 NCT00852228., (Copyright © 2016 Elsevier Ltd. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
36. Corrigendum to "2nd St. Gallen EORTC Gastrointestinal Cancer Conference: Consensus recommendations on controversial issues in the primary treatment of rectal cancer" [Eur J Cancer 63 (August 2016) 11-24].
- Author
-
Lutz MP, Zalcberg JR, Glynne-Jones R, Ruers T, Ducreux M, Arnold D, Aust D, Brown G, Bujko K, Cunningham C, Evrard S, Folprecht G, Gerard JP, Habr-Gama A, Haustermans K, Holm T, Kuhlmann KF, Lordick F, Mentha G, Moehler M, Nagtegaal ID, Pigazzi A, Pucciarelli S, Roth A, Rutten H, Schmoll HJ, Sorbye H, Van Cutsem E, Weitz J, and Otto F
- Published
- 2016
- Full Text
- View/download PDF
37. Acute neurovascular events in cancer patients receiving anti-vascular endothelial growth factor agents: Clinical experience in Paris University Hospitals.
- Author
-
Tlemsani C, Mir O, Psimaras D, Vano YA, Ducreux M, Escudier B, Rousseau B, Loirat D, Ceccaldi B, André T, Goldwasser F, and Ricard D
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Angiogenesis Inhibitors adverse effects, Brain Ischemia chemically induced, Cerebral Hemorrhage chemically induced, Female, Hospitalization statistics & numerical data, Hospitals, University statistics & numerical data, Humans, Male, Middle Aged, Paris, Prospective Studies, Risk Factors, Young Adult, Ischemic Attack, Transient chemically induced, Neoplasms drug therapy, Posterior Leukoencephalopathy Syndrome chemically induced, Stroke chemically induced, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
Background: Despite the increasing and broadening use of agents targeting the vascular endothelial growth factor (VEGF) pathway, little is known on their acute neurovascular toxicities., Methods: This retrospective, multi-centre study examined the characteristics of patients with solid tumours who experienced an ischaemic or haemorrhagic stroke, a transient ischaemic accident (TIA) or a posterior reversible encephalopathy syndrome (PRES) while under anti-VEGF and until 8 weeks after termination of treatment and evaluated their management in our institutions from 2004 to 2014. Patients with newly diagnosed or progressive cerebral metastases at the time of the acute neurovascular event were excluded., Results: Thirty-four patients (55.9% men) were identified, and experienced either ischaemic stroke (n = 18), PRES (n = 9), TIA (n = 6) or haemorrhagic stroke (n = 1). At initiation of anti-VEGF agents, 64.7% of patients had previous cardiovascular risk factors, and 52.9% had hypertension. Eight patients (23.5%) had received cerebral radiotherapy, five of which concomitantly to anti-VEGF treatment. Six (17%) patients died in the 8 weeks following the acute neurovascular event, and only 55.9% recovered their initial neurological status. Overall, 1-year and 2-year survival rates after the acute neurovascular event were 67.9% and 50%, respectively. When anti-VEGF agents were reintroduced (n = 6), severe vascular toxicity recurred in two patients., Conclusions: Neurovascular events under VEGF treatments are potentially severe, and the management of comorbid conditions has to be improved. A prospective collection of data and standardised management of such events is therefore being structured in our institutions., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
38. Second St. Gallen European Organisation for Research and Treatment of Cancer Gastrointestinal Cancer Conference: consensus recommendations on controversial issues in the primary treatment of rectal cancer.
- Author
-
Lutz MP, Zalcberg JR, Glynne-Jones R, Ruers T, Ducreux M, Arnold D, Aust D, Brown G, Bujko K, Cunningham C, Evrard S, Folprecht G, Gerard JP, Habr-Gama A, Haustermans K, Holm T, Kuhlmann KF, Lordick F, Mentha G, Moehler M, Nagtegaal ID, Pigazzi A, Pucciarelli S, Roth A, Rutten H, Schmoll HJ, Sorbye H, Van Cutsem E, Weitz J, and Otto F
- Subjects
- Antineoplastic Agents therapeutic use, Chemoradiotherapy methods, Combined Modality Therapy, Diagnostic Imaging methods, Europe, Humans, Neoadjuvant Therapy methods, Neoplasm Staging methods, Rectal Neoplasms diagnosis, Risk Assessment methods, Rectal Neoplasms therapy
- Abstract
Primary treatment of rectal cancer was the focus of the second St. Gallen European Organisation for Research and Treatment of Cancer (EORTC) Gastrointestinal Cancer Conference. In the context of the conference, a multidisciplinary international expert panel discussed and voted on controversial issues which could not be easily answered using published evidence. Main topics included optimal pretherapeutic imaging, indication and type of neoadjuvant treatment, and the treatment strategies in advanced tumours. Here we report the key recommendations and summarise the related evidence. The treatment strategy for localised rectal cancer varies from local excision in early tumours to neoadjuvant radiochemotherapy (RCT) in combination with extended surgery in locally advanced disease. Optimal pretherapeutic staging is a key to any treatment decision. The panel recommended magnetic resonance imaging (MRI) or MRI + endoscopic ultrasonography (EUS) as mandatory staging modalities, except for early T1 cancers with an option for local excision, where EUS in addition to MRI was considered to be most important because of its superior near-field resolution. Primary surgery with total mesorectal excision was recommended by most panellists for some early tumours with limited risk of recurrence (i.e. cT1-2 or cT3a N0 with clear mesorectal fascia on MRI and clearly above the levator muscles), whereas all other stages were considered for multimodal treatment. The consensus panel recommended long-course RCT over short-course radiotherapy for most clinical situations where neoadjuvant treatment is indicated, with the exception of T3a/b N0 tumours where short-course radiotherapy or even no neoadjuvant therapy were regarded to be an option. In patients with potentially resectable tumours and synchronous liver metastases, most panel members did not see an indication to start with classical fluoropyrimidine-based RCT but rather favoured preoperative short-course radiotherapy with systemic combination chemotherapy or alternatively a liver-first resection approach in resectable metastases, which both allow optimal systemic therapy for the metastatic disease. In general, proper patient selection and discussion in an experienced multidisciplinary team was considered as crucial component of care., (Copyright © 2016 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
39. Immunotherapy in gastrointestinal cancer: Recent results, current studies and future perspectives.
- Author
-
Moehler M, Delic M, Goepfert K, Aust D, Grabsch HI, Halama N, Heinrich B, Julie C, Lordick F, Lutz MP, Mauer M, Alsina Maqueda M, Schild H, Schimanski CC, Wagner AD, Roth A, and Ducreux M
- Subjects
- Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, Biomarkers, Tumor metabolism, Cell Cycle Checkpoints drug effects, Epidemiologic Methods, Forecasting, Genetic Markers physiology, Humans, Immunotherapy methods, Molecular Targeted Therapy methods, Molecular Targeted Therapy trends, Oncolytic Virotherapy methods, Treatment Outcome, Gastrointestinal Neoplasms therapy, Immunotherapy trends
- Abstract
The new therapeutic approach of using immune checkpoint inhibitors as anticancer agents is a landmark innovation. Early studies suggest that immune checkpoint inhibition might also be effective in patients with gastrointestinal cancer. To improve the efficacy of immunotherapy, different strategies are currently under evaluation. This review summarises the discussion during the European Organisation for Research and Treatment of Cancer Gastrointestinal Tract Cancer Translational Research Meeting in Mainz in November 2014 and provides an update on the most recent results of immune therapy in gastrointestinal cancers. Knowledge of potential relationships between tumour cells and their microenvironment including the immune system will be essential in gastrointestinal malignancies. In this context, the density of T cell infiltration within colorectal cancer metastases has been associated with response to chemotherapy, and a high expression of programmed cell death ligand 1 (PD-L1) in advanced gastric cancer has been related with poor prognosis. Effective targets might include neo-antigens encoded from genes carrying tumour-specific somatic mutations. Tailored immunotherapy based on such mutations could enable the effective targeting of an individual patient's tumour with vaccines produced on demand. Other strategies considering checkpoint inhibitors have shown efficacy by targeting cytotoxic T-lymphocyte-associated protein 4 and PD-1 or PD-L1. DNA mismatch repair-deficient tumours appear to be potentially the best candidates for these therapies. Finally, the combination of oncolytic viruses with immunotherapy might boost antitumour activity as well. Further evaluation of these promising immunological therapeutic approaches will require large prospective clinical studies., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
40. Current standards and new innovative approaches for treatment of pancreatic cancer.
- Author
-
Conroy T, Bachet JB, Ayav A, Huguet F, Lambert A, Caramella C, Maréchal R, Van Laethem JL, and Ducreux M
- Subjects
- Adenocarcinoma pathology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chemotherapy, Adjuvant methods, Diagnostic Imaging methods, Humans, Molecular Targeted Therapy methods, Neoadjuvant Therapy methods, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local therapy, Neoplasm Staging, Palliative Care methods, Pancreatic Neoplasms pathology, Precision Medicine methods, Reference Standards, Adenocarcinoma therapy, Pancreatic Neoplasms therapy, Therapies, Investigational methods
- Abstract
Pancreatic adenocarcinoma remains a devastating disease with a 5-year survival rate not exceeding 6%. Treatment of this disease remains a major challenge. This article reviews the state-of-the-art in the management of this disease and the new innovative approaches that may help to accelerate progress in treating its victims. After careful pre-therapeutic evaluation, only 15-20% of patients diagnosed with a pancreatic cancer (PC) are eligible for upfront radical surgery. After R0 or R1 resection in such patients, evidence suggests a significantly positive impact on survival of adjuvant chemotherapy comprising 6 months of gemcitabine or fluorouracil/folinic acid. Delayed adjuvant chemoradiation is considered as an option in cases of positive margins. Borderline resectable pancreatic cancer (BRPC) is defined as a tumour involving the mesenteric vasculature to a limited extend. Resection of these tumours is technically feasible, yet runs the high risk of a R1 resection. Neoadjuvant treatment probably offers the best chance of achieving successful R0 resection and long-term survival, but the best treatment options should be determined in prospective randomised studies. Gemcitabine has for 15 years been the only validated therapy for advanced PC. Following decades of negative phase III studies, increasing evidence now suggests that further significant improvements to overall survival can be achieved via either Folfirinox or gemcitabine + nab-paclitaxel regimens. Progress in systemic therapy may improve the chances of resection in borderline resectable pancreatic cancer (BRPC) or locally advanced PC. This requires first enhancing knowledge of the genetic events driving carcinogenesis, which may then be translated into clinical studies., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
41. Evaluating iatrogenic prescribing: development of an oncology-focused trigger tool.
- Author
-
Hébert G, Netzer F, Ferrua M, Ducreux M, Lemare F, and Minvielle E
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Female, Guideline Adherence statistics & numerical data, Humans, Incidence, Infant, Infant, Newborn, Length of Stay statistics & numerical data, Male, Middle Aged, Retrospective Studies, Young Adult, Antineoplastic Agents adverse effects, Drug Monitoring methods, Drug Prescriptions statistics & numerical data, Drug-Related Side Effects and Adverse Reactions diagnosis, Drug-Related Side Effects and Adverse Reactions epidemiology, Iatrogenic Disease epidemiology
- Abstract
Background: Drug-related iatrogenic effects are common in oncology because chemotherapy is toxic. The evaluation of the application of the guidelines may be a way to understand the occurrence of adverse drug-related event (ADE). There is no specific method for identifying ADEs and measuring harm to patients in oncology., Objective: Our objective was to develop and test an Oncology Trigger Tool (OTT) for ADEs and to describe ADE characteristics and incidence., Methods: A clinical advisory panel identified situations at high risk of ADE occurrence and built 22 triggers with, in each case, an analysis flowchart to confirm or refute occurrence. The OTT was used to review 288 random admissions (Oct. 2010-Sept. 2011) and measure ADE incidence and severity (CTCAE 4.03 - Common Terminology Criteria for Adverse Events). Tool feasibility (time required), inter-rater (IR) reproducibility and positive predictive value (PPV) were measured., Results: Overall, 884 triggers were detected and 122 ADEs, with 42.4 ADEs/100 admissions or 46.0 ADEs/1000 patient-days, and a 31.1% rate of severe ADEs. The most common ADEs were hyperglycaemia (14.5%), unplanned drug-related admission within 30 days (13.7%) and opiate-induced constipation (12.1%). Unplanned drug-related admission was the most serious (82.4% incidence of severe harm). Mean time for OTT implementation was 21.8 min; IR reproducibility was high (κ=0.965 (trigger); κ=0.935 (ADE); κ=0.853 (harm)); PPV 22-trigger version was 20.7%., Conclusions: ADE analysis flowcharts coupled with standardised grading of harm considerably reduced IR variability, thus providing a robust oncology-focused trigger tool for use in ADE audits and hospital comparisons. The involvement of a clinical advisory panel in tool development should help drive changes for improving practice. Further research on the OTT is warranted., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
42. Systemic treatment of advanced hepatocellular carcinoma: from disillusions to new horizons.
- Author
-
Hollebecque A, Malka D, Ferté C, Ducreux M, and Boige V
- Subjects
- Angiogenesis Inhibitors therapeutic use, Carcinoma, Hepatocellular pathology, Chemotherapy, Adjuvant, Disease Progression, Humans, Liver Neoplasms pathology, Neoadjuvant Therapy, Niacinamide analogs & derivatives, Niacinamide therapeutic use, Phenylurea Compounds therapeutic use, Sorafenib, Treatment Failure, Antineoplastic Agents therapeutic use, Carcinoma, Hepatocellular drug therapy, Drugs, Investigational therapeutic use, Liver Neoplasms drug therapy
- Abstract
Hepatocellular carcinoma (HCC) is an aggressive malignancy, which accounts for a third of all cancer deaths globally each year. The management of patients with HCC is complex, as both the tumour stage and any underlying liver disease must be considered conjointly. Since the approval of sorafenib in advanced HCC, several phase III clinical trials have failed to demonstrate any superiority over sorafenib in the frontline setting, and no agent has been shown to impact outcomes after sorafenib failure. This review will focus on the range of experimental therapeutics for patients with advanced HCC and highlight the successes and failures of these treatments as well as areas for future development. Specifics such as dose limiting toxicity and safety profile in patients with liver dysfunction related to the underlying chronic liver disease should be considered when developing therapies in HCC. Finally, robust validated and reproducible surrogate end-points as well as predictive biomarkers should be defined in future randomised trials., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
43. Is primary tumour resection associated with survival improvement in patients with colorectal cancer and unresectable synchronous metastases? A pooled analysis of individual data from four randomised trials.
- Author
-
Faron M, Pignon JP, Malka D, Bourredjem A, Douillard JY, Adenis A, Elias D, Bouché O, and Ducreux M
- Subjects
- Aged, Carcinoembryonic Antigen blood, Colorectal Neoplasms pathology, Combined Modality Therapy, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Multivariate Analysis, Neoplasm Metastasis, Outcome Assessment, Health Care standards, Prognosis, Proportional Hazards Models, Randomized Controlled Trials as Topic, Colorectal Neoplasms drug therapy, Colorectal Neoplasms surgery, Outcome Assessment, Health Care methods
- Abstract
Objective: To evaluate the impact on survival of primary tumour resection in patients with unresectable synchronous metastases from colorectal cancer (CRC)., Summary Background Data: Primary tumour resection in this setting remains controversial., Patients and Methods: We retrieved individual data of 1155 patients with metastatic CRC included in four first-line chemotherapy trials: Fédération Francophone de Cancérologie Digestive (FFCD)-9601, FFCD-2000-05, Actions Concertées dans les cancers COloRectaux et Digestifs (ACCORD)-13, and ML-16987. Patients with unresectable synchronous metastases were eligible for this study. We used univariate and multivariate analyses (Cox models stratified on the trial) to assess the impact of primary tumour resection and other potential prognostic variables on overall survival (OS) (the primary endpoint)., Results: Amongst the 1155 patients, 810 patients met the inclusion criteria and 59% (n = 478) underwent resection of their primary tumour, prior to trial entry (resection group). Compared to patients in the non-resection group (n =3 32 [41%]), those in the resection group were more likely to have a colonic primary, lower baseline carcinoembryonic antigen (CEA) and alkaline phosphatase levels, and normal white-blood-cell count (p < 0.001 each). Primary tumour resection was independently associated to better OS in multivariate analysis (hazard ratio (HR), 0.63 [0.53-0.75]; p < 0.001, with a more favourable impact of resection on OS in case of rectal primary and low CEA level. Primary tumour resection was also independently associated to a better progression-free survival in multivariate analysis (HR, 0.82 [0.70-0.95]; p < 0.001)., Conclusion: Primary tumour resection was independently associated to a better OS in patients with CRC and unresectable synchronous metastases., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
44. Bevacizumab plus capecitabine in patients with progressive advanced well-differentiated neuroendocrine tumors of the gastro-intestinal (GI-NETs) tract (BETTER trial)--a phase II non-randomised trial.
- Author
-
Mitry E, Walter T, Baudin E, Kurtz JE, Ruszniewski P, Dominguez-Tinajero S, Bengrine-Lefevre L, Cadiot G, Dromain C, Farace F, Rougier P, and Ducreux M
- Subjects
- Administration, Oral, Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Bevacizumab, Capecitabine, Deoxycytidine administration & dosage, Deoxycytidine adverse effects, Deoxycytidine analogs & derivatives, Disease-Free Survival, Female, Fluorouracil administration & dosage, Fluorouracil adverse effects, Fluorouracil analogs & derivatives, Gastrointestinal Neoplasms mortality, Humans, Infusions, Intravenous, Male, Middle Aged, Neuroendocrine Tumors mortality, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Gastrointestinal Neoplasms drug therapy, Neuroendocrine Tumors drug therapy
- Abstract
Aim of the Study: Gastro-intestinal neuroendocrine tumours (GI-NETs) are chemotherapy-resistant tumours. Bevacizumab, an inhibitor of vascular endothelial growth factor (VEGF), has shown promising results in several phase II trials of gastro-entero-pancreatic-NETs. We assessed bevacizumab combined with capecitabine, specifically in GI-NET patients., Patients and Methods: BEvacizumab in The Treament of neuroEndocrine tumoRs (BETTER) was a multicentre, open-label, non-randomised, two-group phase II trial. Here we present the group of patients with progressive, metastatic, well-differentiated GI-NETs. Patients Eastern Cooperative Oncology Group-performance status (ECOG-PS)⩽2, Ki-67 proliferation rate <15% and no prior systemic chemotherapy were treated with bevacizumab (7.5 mg/kg/q3w) and capecitabine (1000 mg/m2 twice daily, orally d1-14, resumed on d22) for 6-24 months. The primary end-point was progression-free survival (PFS); secondary end-points included overall survival (OS), response rate, safety and quality of life., Results: Of the 49 patients included, 53% were men, median age was 60 years (41-82), primary tumour site was ileal in 82% patients and Ki-67 was <15% in 48 patients and not available for one patient. After a maximum of 24 month follow-up per patient, the median PFS by investigator assessment was 23.4 months [95% confidence interval (CI): 13.2; not reached] and the overall disease control rate was 88% (18% partial response, 70% stable disease). The 2-year survival rate was 85%. Median OS was not reached. The most frequent grade 3-4 adverse events were hypertension (31%), diarrhoea (14%) and hand-foot syndrome (10%)., Conclusion: The combination of bevacizumab and capecitabine showed clinical activity and a manageable safety profile in the treatment of GI-NETs that warrant confirmation in a randomised phase III trial., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
45. Bevacizumab combined with 5-FU/streptozocin in patients with progressive metastatic well-differentiated pancreatic endocrine tumours (BETTER trial)--a phase II non-randomised trial.
- Author
-
Ducreux M, Dahan L, Smith D, O'Toole D, Lepère C, Dromain C, Vilgrain V, Baudin E, Lombard-Bohas C, Scoazec JY, Seitz JF, Bitoun L, Koné S, and Mitry E
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Bevacizumab, Disease-Free Survival, Drug Administration Schedule, Female, Fluorouracil administration & dosage, Fluorouracil adverse effects, Humans, Male, Middle Aged, Neoplasm Grading, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Quality of Life, Streptozocin administration & dosage, Streptozocin adverse effects, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Pancreatic Neoplasms drug therapy
- Abstract
Aim of the Study: Neuroendocrine tumours are highly vascular neoplasms known to overexpress vascular endothelial growth factor (VEGF) and its receptor. Bevacizumab, an inhibitor of VEGF, was assessed in combination with chemotherapy in pancreatic neuroendocrine tumour (P-NET)., Patients and Methods: BETTER was a multicentre, open-label, non-randomised, two-group phase II trial. Patients with progressive metastatic, well-differentiated P-NET received a minimum of 6 month treatment of bevacizumab at 7.5 mg/kg IV on d1 q3w with 5-FU at 400 mg/m2/day and streptozocin at 500 mg/m2/day IV from d1 to d5 every 42 days. The primary end-point was progression-free survival (PFS); secondary end-points were overall survival (OS), overall response rate, safety and quality of life., Results: A total of 34 patients were included. Median age was 55 years, 65% of patients were men, 97% had an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 and 97% had a Ki-67 proliferative index of <15%. After a maximum of 24 month follow-up per patient, the median PFS assessed by investigators was 23.7 months [95% confidence interval (CI): 13.1; not reached], 19 (56%) patients had a partial response and 15 (44%) had stable disease as best response. OS rate at 24 months was 88%. The most frequently reported grade 3-4 adverse events were hypertension (21% patients), abdominal pain (12%) and thromboembolic events (9%)., Conclusion: Bevacizumab with 5-FU/streptozocin in the treatment of pancreatic NETs seems to be feasible with a PFS of 23.7 months, which deserves further attention. No unexpected toxicity was observed., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
46. Guidelines for time-to-event end-point definitions in trials for pancreatic cancer. Results of the DATECAN initiative (Definition for the Assessment of Time-to-event End-points in CANcer trials).
- Author
-
Bonnetain F, Bonsing B, Conroy T, Dousseau A, Glimelius B, Haustermans K, Lacaine F, Van Laethem JL, Aparicio T, Aust D, Bassi C, Berger V, Chamorey E, Chibaudel B, Dahan L, De Gramont A, Delpero JR, Dervenis C, Ducreux M, Gal J, Gerber E, Ghaneh P, Hammel P, Hendlisz A, Jooste V, Labianca R, Latouche A, Lutz M, Macarulla T, Malka D, Mauer M, Mitry E, Neoptolemos J, Pessaux P, Sauvanet A, Tabernero J, Taieb J, van Tienhoven G, Gourgou-Bourgade S, Bellera C, Mathoulin-Pélissier S, and Collette L
- Subjects
- Consensus, Delphi Technique, Disease-Free Survival, Endpoint Determination, Humans, Pancreatic Neoplasms mortality, Pancreatic Neoplasms therapy, Randomized Controlled Trials as Topic
- Abstract
Background: Using potential surrogate end-points for overall survival (OS) such as Disease-Free- (DFS) or Progression-Free Survival (PFS) is increasingly common in randomised controlled trials (RCTs). However, end-points are too often imprecisely defined which largely contributes to a lack of homogeneity across trials, hampering comparison between them. The aim of the DATECAN (Definition for the Assessment of Time-to-event End-points in CANcer trials)-Pancreas project is to provide guidelines for standardised definition of time-to-event end-points in RCTs for pancreatic cancer., Methods: Time-to-event end-points currently used were identified from a literature review of pancreatic RCT trials (2006-2009). Academic research groups were contacted for participation in order to select clinicians and methodologists to participate in the pilot and scoring groups (>30 experts). A consensus was built after 2 rounds of the modified Delphi formal consensus approach with the Rand scoring methodology (range: 1-9)., Results: For pancreatic cancer, 14 time to event end-points and 25 distinct event types applied to two settings (detectable disease and/or no detectable disease) were considered relevant and included in the questionnaire sent to 52 selected experts. Thirty experts answered both scoring rounds. A total of 204 events distributed over the 14 end-points were scored. After the first round, consensus was reached for 25 items; after the second consensus was reached for 156 items; and after the face-to-face meeting for 203 items., Conclusion: The formal consensus approach reached the elaboration of guidelines for standardised definitions of time-to-event end-points allowing cross-comparison of RCTs in pancreatic cancer., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
47. Role of hyperthermic intraoperative peritoneal chemotherapy in the management of peritoneal metastases.
- Author
-
Elias D, Goéré D, Dumont F, Honoré C, Dartigues P, Stoclin A, Malka D, Boige V, and Ducreux M
- Subjects
- Combined Modality Therapy, Female, Humans, Neuroendocrine Tumors pathology, Ovarian Neoplasms drug therapy, Ovarian Neoplasms pathology, Ovarian Neoplasms surgery, Peritoneal Neoplasms secondary, Stomach Neoplasms drug therapy, Stomach Neoplasms pathology, Stomach Neoplasms surgery, Survival Analysis, Treatment Outcome, Hyperthermia, Induced, Intraoperative Care methods, Peritoneal Neoplasms drug therapy, Peritoneal Neoplasms surgery
- Abstract
The peritoneal cavity must be oncologically considered as an organ in its own right and peritoneal metastases (PM) must be treated with the same curative intent (and the same results) as liver metastases. The package combining complete cytoreductive surgery (CCRS) (treating the visible disease) plus hyperthermic intraoperative peritoneal chemotherapy (HIPEC) (treating the remaining non-visible disease) achieves cure in many patients. Twenty years of publication allow us to assemble sufficient background information and data to point out the good and poor indications for CCRS+HIPEC. HIPEC is the standard of care for the treatment of peritoneal pseudomyxomas and peritoneal mesotheliomas and also, recently for the treatment of colorectal PM with limited peritoneal extension. HIPEC is in the evaluation phase for gastric PM and ovarian PM after initially disappointing results, but it is highly probable that it will be useful in particular settings. PM from neuroendocrine tumours are in the same situation. HIPEC is not currently indicated for the treatment of PM from sarcomas, from GIST, and for small round-cell desmoplastic tumours, given the poor results obtained. HIPEC can be useful, on a case-by-case basis, to treat rare tumours complicated by isolated peritoneal diffusion (e.g. Frantz's tumours). HIPEC can be used in the prophylactic setting to prevent PM in patients with a high risk of developing PM, and the first results of the 'second-look' approach are promising. Finally, CCRS+HIPEC appear to be indispensable tools in the oncologist's armentarium., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
48. Value of cardiophrenic angle lymph node for the diagnosis of colorectal peritoneal carcinomatosis.
- Author
-
Caramella C, Pottier E, Borget I, Malka D, Goéré D, Boige V, Honoré C, Dartigues P, Dumont F, Ducreux M, Elias D, and Dromain C
- Subjects
- Adult, Aged, Aged, 80 and over, Diaphragm, Female, Humans, Logistic Models, Lymphatic Metastasis, Male, Middle Aged, Multivariate Analysis, Pericardium, Peritoneum diagnostic imaging, Prognosis, Retrospective Studies, Sensitivity and Specificity, Tomography, X-Ray Computed, Young Adult, Colorectal Neoplasms pathology, Lymph Nodes pathology, Peritoneal Neoplasms diagnosis, Peritoneal Neoplasms secondary
- Abstract
Background: Cardiophrenic angle lymph nodes (CPALN) have been reported in patients with abdominopelvic malignancies. We aimed to assess whether the presence of CPALN is associated with peritoneal carcinomatosis (PC) in colorectal cancer., Patients and Methods: Between 2007 and 2011, 550 patients with colorectal cancer, including 165 (30%) with PC, had undergone surgery with complete peritoneal exploration. We retrospectively reviewed preoperative CT scans for the presence of CPALN and assessed its association with confirmed PC by univariate and multivariate analyses., Results: CPALN were present in 123 (75%) patients with PC, but absent in 263 (68%) patients without PC (Se: 0.72; Sp: 0.68; PPV: 0.49; NPV: 0.85; [OR], 3.3; p<0.001). PC was the only factor independently associated with CPALN in the multivariate analysis. CPALN was not correlated with the presence of liver metastases. 99 of the 165 patients with PC (62%) had visible signs of PC on CT scan. Among the remaining 66 patients, CPALN were the only potential sign of PC in 41 (62%), (Se 0.62, Sp 0.68, PPV 0.24, and NPV 0.92)., Conclusion: The detection of CPALN on CT may be of valuable help for the diagnosis of PC in patients with CRC., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
49. Improving outcomes in colorectal cancer: where do we go from here?
- Author
-
Van Cutsem E, Borràs JM, Castells A, Ciardiello F, Ducreux M, Haq A, Schmoll HJ, and Tabernero J
- Subjects
- Biomarkers, Tumor analysis, Colorectal Neoplasms economics, Colorectal Neoplasms pathology, Cost-Benefit Analysis, Humans, Survival Analysis, Treatment Outcome, Colorectal Neoplasms diagnosis, Colorectal Neoplasms therapy
- Abstract
Colorectal cancer (CRC) places a considerable burden on individuals and society in Europe, being the second most common cause of cancer-related death in the region. While earlier diagnosis and advances in treatment have considerably improved survival in recent years, further progress is needed. One of the greatest challenges associated with the treatment of CRC is the fact that current therapies for advanced disease are not curative, necessitating treatment for many years and placing a significant healthcare burden on society. To reduce the burden of CRC, care delivery must be more efficient and cost-effective. In particular, development of adequate screening programmes is needed, along with chemo-preventative strategies and newer, more active therapies. Further challenges include the lack of optimal selection of patients for adjuvant therapy, identification of the most appropriate target populations for current treatments and the optimum sequence for new molecular targeted agents. This article outlines current developments and unmet needs in CRC, and provides a detailed vision for improvements in the management of the disease. Implementation of some of these strategies will go some way to improving outcomes for patients with CRC., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
50. The KRAS mutation detection within the initial management of patients with metastatic colorectal cancer: a status report in France in 2011.
- Author
-
Lièvre A, Artru P, Guiu M, Laurent-Puig P, Merlin JL, Sabourin JC, Viguier J, Bastie A, Seronde A, and Ducreux M
- Subjects
- Adult, Aged, Antineoplastic Agents therapeutic use, Colonic Neoplasms drug therapy, DNA Mutational Analysis statistics & numerical data, ErbB Receptors antagonists & inhibitors, Female, France, Genetic Testing statistics & numerical data, Genotype, Humans, Male, Middle Aged, Neoplasm Metastasis, Practice Patterns, Physicians' statistics & numerical data, Rectal Neoplasms drug therapy, Referral and Consultation statistics & numerical data, Retrospective Studies, Time Factors, Colonic Neoplasms genetics, Genes, ras genetics, Mutation genetics, Rectal Neoplasms genetics
- Abstract
Background: The detection of KRAS mutations is mandatory to initiate an anti-epidermal growth factor receptor (EGFR) antibody in the treatment of metastatic colorectal carcinoma (mCRC)., Patients and Methods: This observational retrospective study was performed in 160 French centres during a 2-week period in 2011. Its main objective was to evaluate the rate of KRAS testing in patients with mCRC having initiated their first-line therapy. Secondary objectives included time of process, techniques used and reasons for non-prescription., Results: Five hundred and thirty eight mCRC patients (67.1 ± 11.3 years, synchronous metastases: 69.9%) were enrolled in the study. KRAS testing was prescribed in 81.1% of patients, in a median of 15 days after the diagnosis of metastases, and of 15 days prior to the initiation of the first-line metastatic chemotherapy. KRAS status was available for 87% of patients, after 23.6 ± 28.2 days, but after the choice of the first-line therapy in 56.6% of patients. Heterogeneity of reception time was noteworthy within regions (8.3 ± 7 days to 38.8 ± 101 days). KRAS testing was not prescribed mainly due to the planned non-prescription of an anti-EGFR antibody., Conclusion: This study confirmed that KRAS testing is definitely part of the management of most of mCRC patients, despite discrepancies observed in the rate of prescription and the time of results., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.