Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Antoine Hollebecque is active.

Publication


Featured researches published by Antoine Hollebecque.


European Journal of Cancer | 2016

Immune-related adverse events with immune checkpoint blockade: a comprehensive review.

Jean-Marie Michot; C. Bigenwald; Stéphane Champiat; M. Collins; F. Carbonnel; Sophie Postel-Vinay; A. Berdelou; A. Varga; R. Bahleda; Antoine Hollebecque; C. Massard; A. Fuerea; Vincent Ribrag; Anas Gazzah; Jean-Pierre Armand; N. Amellal; E. Angevin; N. Noel; C. Boutros; C. Mateus; Caroline Robert; Aurelien Marabelle; Olivier Lambotte

Cancer immunotherapy is coming of age; it has prompted a paradigm shift in oncology, in which therapeutic agents are used to target immune cells rather than cancer cells. The first generation of new immunotherapies corresponds to antagonistic antibodies that block specific immune checkpointxa0molecules cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death protein (PD-1) and its ligand PD-L1. Targeting these checkpoints in patients living with cancer had led to long-lasting tumour responses. By unbalancing the immune system, these new immunotherapies also generate dysimmune toxicities, called immune-related adverse events (IRAEs) that mainly involve the gut, skin, endocrine glands, liver, and lung but can potentially affect any tissue. In view of their undisputed clinical efficacy, anti-CTLA-4 and anti-PD-1 antibodies are entering in the routine oncological practice, and the number of patients exposed to these drugs will increase dramatically in the near future. Although steroids can be used to treat these IRAEs, the associated immunosuppression may compromise the antitumour response. Oncologists must be ready to detect and manage these new types of adverse events. This review focuses on the mechanisms of IRAE generation, putative relationship between dysimmune toxicity and antitumour efficacy, as a basis for management guidelines.


Clinical Cancer Research | 2017

Hyperprogressive Disease Is a New Pattern of Progression in Cancer Patients Treated by Anti-PD-1/PD-L1

Stéphane Champiat; Laurent Dercle; Samy Ammari; C. Massard; Antoine Hollebecque; Sophie Postel-Vinay; Nathalie Chaput; Alexander Eggermont; Aurélien Marabelle; Jean-Charles Soria; Charles Ferté

Purpose: While immune checkpoint inhibitors are disrupting the management of patients with cancer, anecdotal occurrences of rapid progression (i.e., hyperprogressive disease or HPD) under these agents have been described, suggesting potentially deleterious effects of these drugs. The prevalence, the natural history, and the predictive factors of HPD in patients with cancer treated by anti-PD-1/PD-L1 remain unknown. Experimental Design: Medical records from all patients (N = 218) prospectively treated in Gustave Roussy by anti-PD-1/PD-L1 within phase I clinical trials were analyzed. The tumor growth rate (TGR) prior (“REFERENCE”; REF) and upon (“EXPERIMENTAL”; EXP) anti-PD-1/PD-L1 therapy was compared to identify patients with accelerated tumor growth. Associations between TGR, clinicopathologic characteristics, and overall survival (OS) were computed. Results: HPD was defined as a RECIST progression at the first evaluation and as a ≥2-fold increase of the TGR between the REF and the EXP periods. Of 131 evaluable patients, 12 patients (9%) were considered as having HPD. HPD was not associated with higher tumor burden at baseline, nor with any specific tumor type. At progression, patients with HPD had a lower rate of new lesions than patients with disease progression without HPD (P < 0.05). HPD is associated with a higher age (P < 0.05) and a worse outcome (overall survival). Interestingly, REF TGR (before treatment) was inversely correlated with response to anti-PD-1/PD-L1 (P < 0.05) therapy. Conclusions: A novel aggressive pattern of hyperprogression exists in a fraction of patients treated with anti-PD-1/PD-L1. This observation raises some concerns about treating elderly patients (>65 years old) with anti-PD-1/PD-L1 monotherapy and suggests further study of this phenomenon. Clin Cancer Res; 23(8); 1920–8. ©2016 AACR. See related commentary by Sharon, p. 1879


Cancer Discovery | 2017

High-Throughput Genomics and Clinical Outcome in Hard-to-Treat Advanced Cancers: Results of the MOSCATO 01 Trial

C. Massard; Stefan Michiels; Charles Ferté; Marie-Cécile Le Deley; Ludovic Lacroix; Antoine Hollebecque; Loic Verlingue; Ecaterina Ileana; Silvia Rosellini; Samy Ammari; Maud Ngo-Camus; Rastislav Bahleda; Anas Gazzah; Andrea Varga; Sophie Postel-Vinay; Yohann Loriot; Caroline Even; Ingrid Breuskin; Nathalie Auger; Bastien Job; Thierry de Baere; Frederic Deschamps; Philippe Vielh; Jean-Yves Scoazec; Vladimir Lazar; Catherine Richon; Vincent Ribrag; Eric Deutsch; E. Angevin; Gilles Vassal

High-throughput genomic analyses may improve outcomes in patients with advanced cancers. MOSCATO 01 is a prospective clinical trial evaluating the clinical benefit of this approach. Nucleic acids were extracted from fresh-frozen tumor biopsies and analyzed by array comparative genomic hybridization, next-generation sequencing, and RNA sequencing. The primary objective was to evaluate clinical benefit as measured by the percentage of patients presenting progression-free survival (PFS) on matched therapy (PFS2) 1.3-fold longer than the PFS on prior therapy (PFS1). A total of 1,035 adult patients were included, and a biopsy was performed in 948. An actionable molecular alteration was identified in 411 of 843 patients with a molecular portrait. A total of 199 patients were treated with a targeted therapy matched to a genomic alteration. The PFS2/PFS1 ratio was >1.3 in 33% of the patients (63/193). Objective responses were observed in 22 of 194 patients (11%; 95% CI, 7%-17%), and median overall survival was 11.9 months (95% CI, 9.5-14.3 months).Significance: This study suggests that high-throughput genomics could improve outcomes in a subset of patients with hard-to-treat cancers. Although these results are encouraging, only 7% of the successfully screened patients benefited from this approach. Randomized trials are needed to validate this hypothesis and to quantify the magnitude of benefit. Expanding drug access could increase the percentage of patients who benefit. Cancer Discov; 7(6); 586-95. ©2017 AACR.See related commentary by Schram and Hyman, p. 552This article is highlighted in the In This Issue feature, p. 539.


Clinical Cancer Research | 2017

Phase I Study of GDC-0425, a Checkpoint Kinase 1 Inhibitor, in Combination with Gemcitabine in Patients with Refractory Solid Tumors

Jeffrey R. Infante; Antoine Hollebecque; Sophie Postel-Vinay; Todd Michael Bauer; Elizabeth Blackwood; Marie Evangelista; Sami Mahrus; Franklin Peale; Xuyang Lu; Srikumar Sahasranaman; Rui Zhu; Yuan Chen; Xiao Ding; Elaine Murray; Jennifer L. Schutzman; Jennifer O. Lauchle; Jean-Charles Soria; Patricia LoRusso

Purpose: Chk1 inhibition potentiates DNA-damaging chemotherapy by overriding cell-cycle arrest and genome repair. This phase I study evaluated the Chk1 inhibitor GDC-0425 given in combination with gemcitabine to patients with advanced solid tumors. Experimental Design: Patients received GDC-0425 alone for a 1-week lead-in followed by 21-day cycles of gemcitabine plus GDC-0425. Gemcitabine was initially administered at 750 mg/m2 (Arm A), then increased to 1,000 mg/m2 (Arm B), on days 1 and 8 in a 3 + 3 + 3 dose escalation to establish maximum tolerated dose (MTD). GDC-0425 was initially administered daily for three consecutive days; however, dosing was abbreviated to a single day on the basis of pharmacokinetics and tolerability. TP53 mutations were evaluated in archival tumor tissue. On-treatment tumor biopsies underwent pharmacodynamic biomarker analyses. Results: Forty patients were treated with GDC-0425. The MTD of GDC-0425 was 60 mg when administered approximately 24 hours after gemcitabine 1,000 mg/m2. Dose-limiting toxicities included thrombocytopenia (n = 5), neutropenia (n = 4), dyspnea, nausea, pyrexia, syncope, and increased alanine aminotransferase (n = 1 each). Common related adverse events were nausea (48%); anemia, neutropenia, vomiting (45% each); fatigue (43%); pyrexia (40%); and thrombocytopenia (35%). The GDC-0425 half-life was approximately 15 hours. There were two confirmed partial responses in patients with triple-negative breast cancer (TP53-mutated) and melanoma (n = 1 each) and one unconfirmed partial response in a patient with cancer of unknown primary origin. Conclusions: Chk1 inhibition with GDC-0425 in combination with gemcitabine was tolerated with manageable bone marrow suppression. The observed preliminary clinical activity warrants further investigation of this chemopotentiation strategy. Clin Cancer Res; 23(10); 2423–32. ©2016 AACR.


Annals of Oncology | 2016

Seeking the driver in tumours with apparent normal molecular profile on comparative genomic hybridization and targeted gene panel sequencing: what is the added value of whole exome sequencing?

Sophie Postel-Vinay; Y. Boursin; C. Massard; Antoine Hollebecque; Ecaterina Ileana; M. Chiron; J. Jung; J. S. Lee; Z. Balogh; Julien Adam; Philippe Vielh; E. Angevin; Ludovic Lacroix

BACKGROUNDnMolecular tumour profiling technologies have become increasingly important in the era of precision medicine, but their routine use is limited by their accessibility, cost, and tumour material availability. It is therefore crucial to assess their relative added value to optimize the sequence and combination of such technologies.nnnPATIENTS AND METHODSnWithin the MOSCATO-01 trial, we investigated the added value of whole exome sequencing (WES) in patients that did not present any molecular abnormality on array comparative genomic hybridization (aCGH) and targeted gene panel sequencing (TGPS) using cancer specific panels. The pathogenicity potential and actionability of mutations detected on WES was assessed.nnnRESULTSnAmong 420 patients enrolled between December 2011 and December 2013, 283 (67%) patients were analysed for both TGPS and aCGH. The tumour sample of 25 (8.8%) of them presented a flat (or low-dynamic) aCGH profile and no pathogenic mutation on TGPS. We selected the first eligible 10 samples-corresponding to a heterogeneous cohort of different tumour types-to perform WES. This allowed identifying eight mutations of interest in two patients: FGFR3, PDGFRB, and CREBBP missense single-nucleotide variants (SNVs) in an urothelial carcinoma; FGFR2, FBXW7, TP53, and MLH1 missense SNVs as well as an ATM frameshift mutation in a squamous cell carcinoma of the tongue. The FGFR3 alteration had been previously described as an actionable activating mutation and might have resulted in treatment by an FGFR inhibitor. CREBBP and ATM alterations might also have suggested a therapeutic orientation towards epigenetic modifiers and ataxia-telangectasia and Rad3-related inhibitors, respectively.nnnCONCLUSIONnThe therapeutic added value of performing WES on tumour samples that do not harbour any genetic abnormality on TGPS and aCGH might be limited and variable according to the histotype. Alternative techniques, including RNASeq and methylome analysis, might be more informative in selected cases.


Cancer Chemotherapy and Pharmacology | 2017

Phase I dose-escalation study of milciclib in combination with gemcitabine in patients with refractory solid tumors

Sandrine Aspeslagh; Kunwar Shailubhai; Rastilav Bahleda; Anas Gazzah; Andrea Varga; Antoine Hollebecque; C. Massard; Anna Spreafico; Michele Reni; Jean-Charles Soria

BackgroundThis phase I trial evaluated the safety and tolerability of milciclib, an inhibitor of multiple cyclin-dependent kinases and tropomycin receptor kinase A, in combination with gemcitabine in patients with refractory solid tumors.DesignSixteen patients were enrolled and treated with milciclib at three dose levels (45xa0mg/m2/day, nxa0=xa03; 60xa0mg/m2/day, nxa0=xa03; and 80xa0mg/m2/day, nxa0=xa010) with a fixed dose of gemcitabine (1000xa0mg/m2/day). Milciclib was administered orally once daily for 7xa0days on/7xa0days off in a 4-week cycle, and gemcitabine was administered intravenously on days 1, 8 and 15 in a 4-week cycle.ResultsAll 16 enrolled patients were evaluable for safety and toxicity. Dose-limiting toxicities, which occurred in only one out of nine patients treated at the maximum dose tested (milciclib 80xa0mg/m2/day and gemcitabine 1000xa0mg/m2/day), consisted of Grade 4 thrombocytopenia, Grade 3 ataxia and Grade 2 tremors in the same patient. Most frequent treatment-related AEs were neutropenia and thrombocytopenia. Among 14 evaluable patients, one NSCLC patient showed partial response and 4 patients (one each with thyroid, prostatic, pancreatic carcinoma and peritoneal mesothelioma) showed long-term disease stabilization (>6–14xa0months). Pharmacokinetics of the orally administered milciclib (~t1/2 33xa0h) was not altered by concomitant treatment with gemcitabine.ConclusionThe combination treatment was well tolerated with manageable toxicities. The recommended phase II dose was 80xa0mg/m2/day for milciclib and 1000xa0mg/m2/day for gemcitabine. This combination treatment regimen showed encouraging clinical benefit in ~36% patients, including gemcitabine refractory patients. These results support further development of combination therapies with milciclib in advanced cancer patients.


Seminars in Oncology | 2017

Angiogenesis inhibition in the second-line treatment of metastatic colorectal cancer: A systematic review and pooled analysis

Ralph Chebib; Loic Verlingue; Nathalie Cozic; Matthieu Faron; Pascal Burtin; Valérie Boige; Antoine Hollebecque; David Malka

The last two decades have seen intensive efforts devoted to the development of compounds that target angiogenesis for the treatment of metastatic colorectal cancer (mCRC). In this review, we describe supporting evidence and ongoing development of angiogenesis inhibitors in the second-line treatment of mCRC, and summarize relevant randomized trials to help therapeutic decision-making in daily practice.


European Journal of Cancer | 2017

Matching genomic molecular aberrations with molecular targeted agents: Are biliary tract cancers an ideal playground?

Loic Verlingue; Antoine Hollebecque; Valérie Boige; Michel Ducreux; David Malka; Charles Ferté

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.


European Journal of Cancer | 2018

Immunotherapy phase I trials in patients Older than 70 years with advanced solid tumours

H. Herin; Sandrine Aspeslagh; E. Castanon; V. Dyevre; Aurelien Marabelle; A. Varga; S. Postel Vinay; Jean-Marie Michot; Vincent Ribrag; Anas Gazzah; Rastislav Bahleda; O. Mir; C. Massard; Antoine Hollebecque; Capucine Baldini

BACKGROUNDnThe development of immune checkpoint blocker development brings new hope in older patients (OPs) because of clinical efficacy and low toxicity. Clinical indications are rising steadily, but very few data are available in the geriatric population where comorbidities, reduced functional reserve and immunosenescence may affect efficacy and tolerance.nnnMETHODSnAll cases of patients enrolled in immunotherapy phase I trials between January 2012 and December 2016 in the Drug Development Department (DITEP) at Gustave Roussy were retrospectively reviewed. Case-control analysis was performed in OPs (patientsxa0≥xa070 years) matched to younger patients (YPs) (patientsxa0<xa070 years) by trial and treatment dose. We compared cumulative incidence, grade and type of immune-related adverse events (IrAEs) and survival outcomes.nnnRESULTSnAmong the 46 OPs and the 174xa0YPs enrolled in 14 phase I/II trials, 10 (22%) and 23 (13%) patients experienced grade III-IV IrAEs. Cumulative incidence of grade I-II IrAEs was significantly higher in OPs than YPs (pxa0<xa00.05). No significant difference was observed between the two groups for grade III-IV IrAEs (pxa0=xa00.50). Older age was not associated with lower dose intensity of treatment (pxa0=xa00.14). No significant difference was observed between OPs and YPs in median progression-free survival (hazards ratio 1.41, 95% confidence interval [CI] [0.94-2.11] pxa0=xa00.09) or median overall survival (HR 0.92, 95% CI [0.61-1.39] pxa0=xa00.77).nnnCONCLUSIONnImmune checkpoint blockade appears to be an acceptable treatment option for OPs in the setting of phase I trials.


European Journal of Cancer | 2017

A first-in-human phase I study of SAR125844, a selective MET tyrosine kinase inhibitor, in patients with advanced solid tumours with MET amplification

E. Angevin; Gianluca Spitaleri; Jordi Rodon; Katia Fiorella Dotti; Nicolas Isambert; Stefania Salvagni; Victor Moreno; Sylvie Assadourian; Corinne Gomez; Marzia Harnois; Antoine Hollebecque; Analia Azaro; Alice Hervieu; Karim Rihawi; Filippo De Marinis

PURPOSEnDysregulated MET signalling is implicated in oncogenesis. The safety and preliminary efficacy of a highly selective MET kinase inhibitor (SAR125844) was investigated in patients with advanced solid tumours and MET dysregulation.nnnMETHODSnThis was a phase I dose-escalation (3xa0+xa03 design [50-740xa0mg/m2]) and dose-expansion study. In the dose escalation, patients had high total MET (t-MET) expression by immunohistochemistry (IHC) or MET amplification by fluorescence in situ hybridisation. In the dose expansion, patients had MET amplification (including a subset of patients with non-small cell lung cancer [NSCLC]) or phosphorylated-MET (p-MET) expression (IHC). Objectives were determination of maximum tolerated dose (MTD) of once-weekly intravenous SAR125844 based on dose-limiting toxicities; safety and pharmacokinetic profile; preliminary efficacy of SAR125844 MTD in the expansion cohort.nnnRESULTSnIn total, 72 patients were enrolled: dose escalation, Nxa0=xa033; dose expansion, Nxa0=xa039; 570xa0mg/m2 was established as the MTD. Most frequent treatment-emergent adverse events (AEs) were asthenia/fatigue (58.3%), nausea (31.9%), and abdominal pain, constipation, and dyspnea (27.8% for each); 58.3% of patients reported grade 3 AEs (19.4% were treatment related). Of the 29 evaluable patients with MET amplification treated at 570xa0mg/m2, five achieved a partial response, including four of 22 with NSCLC; 17 patients had stable disease. No response was observed in patients with high p-MET solid tumours. There was no correlation between tumour response and t-MET status or MET gene copy number.nnnCONCLUSIONnThe MTD of once-weekly SAR125844 was 570xa0mg/m2; SAR125844 was well tolerated, with significant antitumour activity in patients with MET-amplified NSCLC.nnnCLINICAL TRIAL REGISTRATION NUMBERnNCT01391533.

Collaboration


Dive into the Antoine Hollebecque's collaboration.

Top Co-Authors

Avatar

C. Massard

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anas Gazzah

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar

Vincent Ribrag

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar

E. Angevin

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea Varga

Institut Gustave Roussy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

A. Varga

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge