Search Results
You are looking at 1 - 5 of 5 items for
- Author: Jaume Capdevila x
- Refine by access: All content x
Search for other papers by Jaume Capdevila in
Google Scholar
PubMed
Search for other papers by Lara Iglesias in
Google Scholar
PubMed
Search for other papers by Irene Halperin in
Google Scholar
PubMed
Search for other papers by Ángel Segura in
Google Scholar
PubMed
Search for other papers by Javier Martínez-Trufero in
Google Scholar
PubMed
Search for other papers by Maria Ángeles Vaz in
Google Scholar
PubMed
Search for other papers by Jesús Corral in
Google Scholar
PubMed
Search for other papers by Gabriel Obiols in
Google Scholar
PubMed
Search for other papers by Enrique Grande in
Google Scholar
PubMed
Search for other papers by Juan Jose Grau in
Google Scholar
PubMed
Search for other papers by Josep Tabernero in
Google Scholar
PubMed
Although thyroid cancer usually has an excellent prognosis, few therapeutic options are available in the refractory setting. Based on the recent results of phase II studies with tyrosine kinase inhibitors, we designed a retrospective analysis of patients with metastatic thyroid cancer treated with sorafenib in seven Spanish referral centers. Consecutive patients with progressive metastatic thyroid cancer (papillary, follicular, medullary, and anaplastic) not suitable for curative surgery, radioactive-iodine therapy, or radiotherapy were treated with sorafenib 400 mg twice a day. The primary end point was objective response rate (RR). Secondary end points included toxicity, median progression-free survival (mPFS), median overall survival (mOS), and correlation between tumor marker levels (thyroglobulin, calcitonin, and carcinoembryonic antigen) and efficacy. Between June 2006 and January 2010, 34 patients were included in the study. Sixteen patients presented differentiated thyroid carcinomas (DTC) of which seven (21%) were papillary, nine (26%) follicular, 15 (44%) medullary (MTC), and three (9%) were anaplastic (ATC). Eleven (32%) patients achieved partial response and 14 (41%) had stable disease beyond 6 months. Regarding histological subtype, RRs were 47% (seven of 15) for MTC, 19% (three of 16) for DTC, and 33% (one of three) for ATC. With a median follow-up of 11.5 months, mPFS were 13.5, 10.5, and 4.4 months for DTC, MTC, and ATC respectively. Tumor markers were evaluated in 22 patients, and a statistically significant association was observed between RR and decrease in tumor marker levels >50% (P=0.033). In this retrospective trial, sorafenib showed antitumor efficacy in all histological subtypes of thyroid cancer, warranting further development in this setting.
Search for other papers by Martyn E Caplin in
Google Scholar
PubMed
Search for other papers by Marianne Pavel in
Google Scholar
PubMed
Search for other papers by Jarosław B Ćwikła in
Google Scholar
PubMed
Search for other papers by Alexandria T Phan in
Google Scholar
PubMed
Search for other papers by Markus Raderer in
Google Scholar
PubMed
Search for other papers by Eva Sedláčková in
Google Scholar
PubMed
Search for other papers by Guillaume Cadiot in
Google Scholar
PubMed
Search for other papers by Edward M Wolin in
Google Scholar
PubMed
Search for other papers by Jaume Capdevila in
Google Scholar
PubMed
Search for other papers by Lucy Wall in
Google Scholar
PubMed
Search for other papers by Guido Rindi in
Google Scholar
PubMed
Search for other papers by Alison Langley in
Google Scholar
PubMed
Search for other papers by Séverine Martinez in
Google Scholar
PubMed
Search for other papers by Edda Gomez-Panzani in
Google Scholar
PubMed
Royal Free Hospital, Charité University Medicine Berlin, University of Warmia and Mazury, University of Texas MD Anderson Cancer Center, University Hospital, Department of Oncology of the First Faculty of Medicine and General Teaching Hospital, Robert‐Debré Hospital, Markey Cancer Center, Vall d'Hebron University Hospital, Western General Hospital, Università Cattolica del Sacro Cuore, Ipsen, Ipsen, Beaujon Hospital, Paris Diderot University, London, UK
Search for other papers by Philippe Ruszniewski in
Google Scholar
PubMed
Search for other papers by on behalf of the CLARINET Investigators in
Google Scholar
PubMed
In the CLARINET study, lanreotide Autogel (depot in USA) significantly prolonged progression-free survival (PFS) in patients with metastatic pancreatic/intestinal neuroendocrine tumours (NETs). We report long-term safety and additional efficacy data from the open-label extension (OLE). Patients with metastatic grade 1/2 (Ki-67 ≤10%) non-functioning NET and documented baseline tumour-progression status received lanreotide Autogel 120 mg (n=101) or placebo (n=103) for 96 weeks or until death/progressive disease (PD) in CLARINET study. Patients with stable disease (SD) at core study end (lanreotide/placebo) or PD (placebo only) continued or switched to lanreotide in the OLE. In total, 88 patients (previously: lanreotide, n=41; placebo, n=47) participated: 38% had pancreatic, 39% midgut and 23% other/unknown primary tumours. Patients continuing lanreotide reported fewer adverse events (AEs) (all and treatment-related) during OLE than core study. Placebo-to-lanreotide switch patients reported similar AE rates in OLE and core studies, except more diarrhoea was considered treatment-related in OLE (overall diarrhoea unchanged). Median lanreotide PFS (core study randomisation to PD in core/OLE; n=101) was 32.8 months (95% CI: 30.9, 68.0). A sensitivity analysis, addressing potential selection bias by assuming that patients with SD on lanreotide in the core study and not entering the OLE (n=13) had PD 24 weeks after last core assessment, found median PFS remaining consistent: 30.8 months (95% CI: 30.0, 31.3). Median time to further PD after placebo-to-lanreotide switch (n=32) was 14.0 months (10.1; not reached). This OLE study suggests long-term treatment with lanreotide Autogel 120 mg maintained favourable safety/tolerability. CLARINET OLE data also provide new evidence of lanreotide anti-tumour benefits in indolent and progressive pancreatic/intestinal NETs.
Search for other papers by James Yao in
Google Scholar
PubMed
Search for other papers by Abhishek Garg in
Google Scholar
PubMed
Search for other papers by David Chen in
Google Scholar
PubMed
Search for other papers by Jaume Capdevila in
Google Scholar
PubMed
Search for other papers by Paul Engstrom in
Google Scholar
PubMed
Search for other papers by Rodney Pommier in
Google Scholar
PubMed
Search for other papers by Eric Van Cutsem in
Google Scholar
PubMed
Search for other papers by Simron Singh in
Google Scholar
PubMed
Search for other papers by Nicola Fazio in
Google Scholar
PubMed
Search for other papers by Wei He in
Google Scholar
PubMed
Search for other papers by Markus Riester in
Google Scholar
PubMed
Search for other papers by Parul Patel in
Google Scholar
PubMed
Search for other papers by Maurizio Voi in
Google Scholar
PubMed
Search for other papers by Michael Morrissey in
Google Scholar
PubMed
Search for other papers by Marianne Pavel in
Google Scholar
PubMed
Search for other papers by Matthew Helmut Kulke in
Google Scholar
PubMed
Neuroendocrine tumors (NETs) have historically been subcategorized according to histologic features and the site of anatomic origin. Here, we characterize the genomic alterations in patients enrolled in three phase 3 clinical trials of NET of different anatomic origins and assess the potential correlation with clinical outcomes. Whole-exome and targeted panel sequencing was used to characterize 225 NET samples collected in the RADIANT series of clinical trials. Genomic profiling of NET was analyzed along with nongenomic biomarker data on the tumor grade and circulating chromogranin A (CgA) and neuron-specific enolase (NSE) levels from these patients enrolled in clinical trials. Our results highlight recurrent large-scale chromosomal alterations as a common theme among NET. Although the specific pattern of chromosomal alterations differed between tumor subtypes, the evidence for generalized chromosomal instability (CIN) was observed across all primary sites of NET. In pancreatic NET, although the P value was not significant, higher CIN suggests a trend toward longer survival (HR, 0.55, P = 0.077), whereas in the gastrointestinal NET, lower CIN was associated with longer survival (HR, 0.44, P = 0.0006). Our multivariate analyses demonstrated that when combined with other clinical data among patients with progressive advanced NETs, chromosomal level alteration adds important prognostic information. Large-scale CIN is a common feature of NET, and specific patterns of chromosomal gain and loss appeared to have independent prognostic value in NET subtypes. However, whether CIN in general has clinical significance in NET requires validation in larger patient cohort and warrants further mechanistic studies.
Search for other papers by Louis de Mestier in
Google Scholar
PubMed
Search for other papers by Angela Lamarca in
Google Scholar
PubMed
Search for other papers by Jorge Hernando in
Google Scholar
PubMed
Division of Endocrinology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
Search for other papers by Wouter Zandee in
Google Scholar
PubMed
Search for other papers by Teresa Alonso-Gordoa in
Google Scholar
PubMed
Search for other papers by Marine Perrier in
Google Scholar
PubMed
Search for other papers by Annemiek ME Walenkamp in
Google Scholar
PubMed
Search for other papers by Bipasha Chakrabarty in
Google Scholar
PubMed
Search for other papers by Stefania Landolfi in
Google Scholar
PubMed
Search for other papers by Marie-Louise F Van Velthuysen in
Google Scholar
PubMed
Search for other papers by Gursah Kats-Ugurlu in
Google Scholar
PubMed
Search for other papers by Alejandra Caminoa in
Google Scholar
PubMed
Search for other papers by Maxime Ronot in
Google Scholar
PubMed
Search for other papers by Prakash Manoharan in
Google Scholar
PubMed
Search for other papers by Alejandro Garcia-Alvarez in
Google Scholar
PubMed
Search for other papers by Tessa Brabander in
Google Scholar
PubMed
Search for other papers by María Isabel García Gómez-Muriel in
Google Scholar
PubMed
Search for other papers by Guillaume Cadiot in
Google Scholar
PubMed
Search for other papers by Anne Couvelard in
Google Scholar
PubMed
Search for other papers by Jaume Capdevila in
Google Scholar
PubMed
Search for other papers by Marianne E Pavel in
Google Scholar
PubMed
Search for other papers by Jérôme Cros in
Google Scholar
PubMed
There is no standardized treatment for grade 3 neuroendocrine tumors (G3 NETs). We aimed to describe the treatments received in patients with advanced G3 NETs and compare their efficacy. Patients with advanced digestive G3 NETs treated between 2010 and 2018 in seven expert centers were retrospectively studied. Pathological samples were centrally reviewed, and radiological data were locally reviewed. We analyzed RECIST-defined objective response (OR), tumor growth rate (TGR) and progression-free survival (PFS) obtained with first- (L1) or second-line (L2) treatments. We included 74 patients with advanced G3 NETs, mostly from the duodenal or pancreatic origin (71.6%), with median Ki-67 of 30%. The 126 treatments (L1 = 74; L2 = 52) included alkylating-based (n = 32), etoposide-platinum (n = 22) or adenocarcinoma-like (n = 20) chemotherapy, somatostatin analogs (n = 21), targeted therapies (n = 22) and liver-directed therapies (n = 7). Alkylating-based chemotherapy achieved the highest OR rate (37.9%) compared to other treatments (multivariable OR 4.22, 95% CI (1.5–12.2); P = 0.008). Adenocarcinoma-like and alkylating-based chemotherapies showed the highest reductions in 3-month TGR (P < 0.001 and P = 0.008, respectively). The longest median PFS was obtained with adenocarcinoma-like chemotherapy (16.5 months (9.0–24.0)) and targeted therapies (12.0 months (8.2–15.8)), while the shortest PFS was observed with somatostatin analogs (6.2 months (3.8–8.5)) and etoposide-platinum chemotherapy (7.2 months (5.2–9.1)). Etoposide-platinum CT achieved shorter PFS than adenocarcinoma-like (multivariable HR 3.69 (1.61–8.44), P = 0.002) and alkylating-based chemotherapies (multivariable HR 1.95 (1.01–3.78), P = 0.049). Overall, adenocarcinoma-like and alkylating-based chemotherapies may be the most effective treatments for patients with advanced G3 NETs regarding OR and PFS. Etoposide-platinum chemotherapy has poor efficacy in this setting.
Search for other papers by Marcia S Brose in
Google Scholar
PubMed
Search for other papers by Jaume Capdevila in
Google Scholar
PubMed
Search for other papers by Rossella Elisei in
Google Scholar
PubMed
Search for other papers by Lars Bastholt in
Google Scholar
PubMed
Search for other papers by Dagmar Führer-Sakel in
Google Scholar
PubMed
Department of Endocrinology, Diabetology, Nutrition and Therapeutic Education, Hôpitaux Universitaires de Genève, Geneve, Switzerland
Search for other papers by Sophie Leboulleux in
Google Scholar
PubMed
Search for other papers by Iwao Sugitani in
Google Scholar
PubMed
Search for other papers by Matthew H Taylor in
Google Scholar
PubMed
Department of Head Neck Surgery, Renji Hospital Affiliated to Jiaotong University School of Medicine, Shanghai, China
Search for other papers by Zhuoying Wang in
Google Scholar
PubMed
Search for other papers by Lori J Wirth in
Google Scholar
PubMed
Search for other papers by Francis P Worden in
Google Scholar
PubMed
Search for other papers by John Bernard in
Google Scholar
PubMed
Department of Pharmacy, University of Pisa, Pisa, Italy
Search for other papers by Paolo Caferra in
Google Scholar
PubMed
Search for other papers by Raffaella M Colzani in
Google Scholar
PubMed
Search for other papers by Shiguang Liu in
Google Scholar
PubMed
Search for other papers by Martin Schlumberger in
Google Scholar
PubMed
The VERIFY study aimed to determine the efficacy of vandetanib in patients with differentiated thyroid cancer (DTC) that is either locally advanced or metastatic and refractory to radioiodine (RAI) therapy. Specifically, VERIFY is a randomized, double-blind, multicenter phase III trial aimed to determine the efficacy and safety of vandetanib in tyrosine kinase inhibitor-naive patients with locally advanced or metastatic RAI-refractory DTC with documented progression (NCT01876784). Patients were randomized 1:1 to vandetanib or placebo. The primary endpoint was progression-free survival (PFS). Secondary endpoints included best objective response rate, overall survival (OS), safety, and tolerability. Patients continued to receive randomized treatment until disease progression or for as long as they were receiving clinical benefit unless criteria for treatment discontinuation were met. Following randomization, 117 patients received vandetanib, and 118 patients received a placebo. Median PFS was 10.0 months in the vandetanib group and 5.7 months in the placebo group (hazard ratio: 0.75; 95% CI: 0.55–1.03; P = 0.080). OS was not significantly different between treatment arms. Common Terminology Criteria for Adverse Events (CTCAE) of grade ≥3 were reported in 55.6% of patients in the vandetanib arm and 25.4% in the placebo arm. Thirty-three deaths (28.2%; one related to study treatment) occurred in the vandetanib arm compared with 16 deaths (13.6%; two related to treatment) in the placebo arm. No statistically significant improvement was observed in PFS in treatment versus placebo in patients with locally advanced or metastatic, RAI-refractory DTC. Moreover, active treatment was associated with more adverse events and more deaths than placebo, though the difference in OS was not statistically significant.