Network


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

Hotspot


Dive into the research topics where Jean Tessier is active.

Publication


Featured researches published by Jean Tessier.


Clinical Cancer Research | 2012

First-in-Human, Phase I Dose-Escalation Study of the Safety, Pharmacokinetics, and Pharmacodynamics of RO5126766, a First-in-Class Dual MEK/RAF Inhibitor in Patients with Solid Tumors

Maria Martinez-Garcia; Udai Banerji; Joan Albanell; Rastilav Bahleda; Saoirse O. Dolly; Françoise Kraeber-Bodéré; Federico Rojo; E. Routier; Ernesto Guarin; Zhi-xin Xu; Ruediger Rueger; Jean Tessier; Eliezer Shochat; Steve Blotner; Valerie Meresse Naegelen; Jean-Charles Soria

Purpose: This phase I study assessed the maximum tolerated dose (MTD), dose-limiting toxicities (DLT), safety, pharmacokinetics, pharmacodynamics, and clinical activity of the first-in-class dual MEK/RAF inhibitor, RO5126766. Experimental Design: Initial dose-escalation was conducted using once daily dosing over 28 consecutive days in 4-week cycles. Further escalation was completed using 2 intermittent dosing schedules [7 days on treatment followed by 7 days off (7on/7off); 4 days on treatment followed by 3 days off (4on/3off)]. Results: Fifty-two patients received RO5126766 at doses of 0.1 to 2.7 mg once daily, 2.7 to 4.0 mg (4 on/3 off), or 2.7 to 5.0 mg (7 on/7 off). The most common DLTs were elevated creatine phosphokinase (CPK) and blurred vision. The MTD for each dosing schedule was 2.25 mg once daily, 4.0 mg (4 on/3 off), and 2.7 mg (7 on/7 off). The dose/schedule recommended for phase II (RP2D) investigation was 2.7 mg (4 on/3 off). Frequent adverse events included rash-related disorders (94.2%), elevated CPK (55.8%), and diarrhea (51.9%). Cmax occurred 1 to 2 hours after dosing and mean terminal half-life was approximately 60 hours. Pharmacodynamic changes included reduced ERK phosphorylation, an increase in apoptosis in tumor tissue, and a reduction in fluorodeoxyglucose (FDG) uptake after 15 days of dosing. Three partial responses were seen: two in BRAF-mutant melanoma tumors and one in an NRAS-mutant melanoma. Conclusion: This first-in-human study shows that oral RO5126766 has manageable toxicity, a favorable pharmacokinetic/pharmacodynamic profile, and encouraging preliminary antitumor activity in this population of heavily pretreated patients, achieving tumor shrinkage in around 40% of patients across all dose levels and all tumor types. Clin Cancer Res; 18(17); 4806–19. ©2012 AACR.


Clinical Cancer Research | 2012

Phase I Dose-Escalation Study of the Safety, Pharmacokinetics, and Pharmacodynamics of the MEK Inhibitor RO4987655 (CH4987655) in Patients with Advanced Solid Tumors

Suzanne Leijen; Mark R. Middleton; Patricia Tresca; Françoise Kraeber-Bodéré; V. Dieras; Max E. Scheulen; Avinash Gupta; Vanesa Lopez-Valverde; Zhi-xin Xu; Ruediger Rueger; Jean Tessier; Eliezer Shochat; Steve Blotner; Valerie Meresse Naegelen; Jan H. M. Schellens; Wilfried Eberhardt

Purpose: This phase I study of the mitogen-activated protein/extracellular signal–regulated kinase inhibitor RO4987655 (CH4987655) assessed its maximum tolerated dose (MTD), dose-limiting toxicities (DLT), safety, pharmacokinetic/pharmacodynamic profile, and antitumor activity in patients with advanced solid tumors. Patients and Methods: An initial dose escalation was conducted using a once-daily dosing schedule, with oral RO4987655 administered at doses of 1.0 to 2.5 mg once daily over 28 consecutive days in 4-week cycles. Doses were then escalated from 3.0 to 21.0 mg [total daily dose (TDD)] using a twice-daily dosing schedule. Results: Forty-nine patients were enrolled. DLTs were blurred vision (n = 1) and elevated creatine phosphokinase (n = 3). The MTD was 8.5 mg twice daily (TDD, 17.0 mg). Rash-related toxicity (91.8%) and gastrointestinal disorders (69.4%) were the most frequent adverse events. The pharmacokinetic profile of RO4987655 showed dose linearity and a half-life of approximately 4 hours. At the MTD, target inhibition, assessed by suppression of extracellular signal–regulated kinase phosphorylation in peripheral blood mononuclear cells, was high (mean 75%) and sustained (90% of time >IC50). Of the patients evaluable for response, clinical benefit was seen in 21.1%, including two partial responses (one confirmed and one unconfirmed). 79.4% of patients showed a reduction in fluorodeoxyglucose uptake by positron emission tomography between baseline and day 15. Conclusion: In this population of heavily pretreated patients, oral RO4987655 showed manageable toxicity, a favorable pharmacokinetics/pharmacodynamics profile, and promising preliminary antitumor activity, which has been further investigated in specific populations of patients with RAS and/or RAF mutation driven tumors. Clin Cancer Res; 18(17); 4794–805. ©2012 AACR.


Clinical Cancer Research | 2014

Phase I expansion and pharmacodynamic study of the oral MEK inhibitor RO4987655 (CH4987655) in selected patients with advanced cancer with RAS-RAF mutations.

Lisa Zimmer; Fabrice Barlesi; Maria Martinez-Garcia; V. Dieras; Jan H. M. Schellens; Jean-Philippe Spano; Mark R. Middleton; Emiliano Calvo; Luiz Paz-Ares; James Larkin; Simon Pacey; Miro Venturi; Françoise Kraeber-Bodéré; Jean Tessier; Wilfried Eberhardt; Michel Paques; Ernesto Guarin; Valerie Meresse; Jean-Charles Soria

Purpose: This phase I expansion study assessed safety, pharmacodynamic effects, and antitumor activity of RO4987655, a pure MEK inhibitor, in selected patients with advanced solid tumor. Experimental Design: We undertook a multicenter phase I two-part study (dose escalation and cohort expansion). Here, we present the part 2 expansion that included melanoma, non–small cell lung cancer (NSCLC), and colorectal cancer with oral RO4987655 administered continuously at recommended doses of 8.5 mg twice daily until progressive disease (PD). Sequential tumor sampling investigated multiple markers of pathway activation/tumor effects, including ERK phosphorylation and Ki-67 expression. BRAF and KRAS testing were implemented as selection criteria and broader tumor mutational analysis added. Results: Ninety-five patients received RO4987655, including 18 BRAF-mutant melanoma, 23 BRAF wild-type melanoma, 24 KRAS-mutant NSCLC, and 30 KRAS-mutant colorectal cancer. Most frequent adverse events were rash, acneiform dermatitis, and gastrointestinal disorders, mostly grade 1/2. Four (24%) of 17 BRAF-mutated melanoma had partial response as did four (20%) of 20 BRAF wild-type melanoma and two (11%) of 18 KRAS-mutant NSCLC. All KRAS-mutant colorectal cancer developed PD. Paired tumor biopsies demonstrated reduced ERK phosphorylation among all cohorts but significant differences among cohorts in Ki-67 modulation. Sixty-nine percent showed a decrease in fluorodeoxyglucose uptake between baseline and day 15. Detailed mutational profiling confirmed RAS/RAF screening and identified additional aberrations (NRAS/non-BRAF melanomas; PIK3CA/KRAS colorectal cancer) without therapeutic implications. Conclusions: Safety profile of RO4987655 was comparable with other MEK inhibitors. Single-agent activity was observed in all entities except colorectal cancer. Evidence of target modulation and early biologic activity was shown among all indications independent of mutational status. Clin Cancer Res; 20(16); 4251–61. ©2014 AACR.


Neuro-oncology | 2015

Phase 1 dose-escalation study of the antiplacental growth factor monoclonal antibody RO5323441 combined with bevacizumab in patients with recurrent glioblastoma

Ulrik Lassen; Olivier Chinot; Catherine McBain; Morten Mau-Sorensen; Vibeke Andrée Larsen; Maryline Barrie; Patrick Roth; Oliver Krieter; Ka Wang; Kai Habben; Jean Tessier; Angelika Lahr; Michael Weller

BACKGROUNDnWe conducted a phase 1 dose-escalation study of RO5323441, a novel antiplacental growth factor (PlGF) monoclonal antibody, to establish the recommended dose for use with bevacizumab and to investigate the pharmacokinetics, pharmacodynamics, safety/tolerability, and preliminary clinical efficacy of the combination.nnnMETHODSnTwenty-two participants with histologically confirmed glioblastoma in first relapse were treated every 2 weeks with RO5323441 (625 mg, 1250 mg, or 2500 mg) plus bevacizumab (10 mg/kg). A standard 3 + 3 dose-escalation trial design was used.nnnRESULTSnRO5323441 combined with bevacizumab was generally well tolerated, and the maximum tolerated dose was not reached. Two participants experienced dose-limiting toxicities (grade 3 meningitis associated with spinal fluid leak [1250 mg] and grade 3 cerebral infarction [2500 mg]). Common adverse events included hypertension (14 participants, 64%), headache (12 participants, 55%), dysphonia (11 participants, 50%) and fatigue (6 participants, 27%).The pharmacokinetics of RO5323441 were linear, over-the-dose range, and bevacizumab exposure was unaffected by RO5323441 coadministration. Modulation of plasmatic angiogenic proteins, with increases in VEGFA and decreases in FLT4, was observed. Dynamic contrast-enhanced/diffusion-weighted MRI revealed large decreases in vascular parameters that were maintained through the dosing period. Combination therapy achieved an overall response rate of 22.7%, including one complete response, and median progression-free and overall survival of 3.5 and 8.5 months, respectively.nnnCONCLUSIONnThe toxicity profile of RO5323441 plus bevacizumab was acceptable and manageable. The observed clinical activity of the combination does not appear to improve on that obtained with single-agent bevacizumab in patients with recurrent glioblastoma.


The Journal of Nuclear Medicine | 2012

Differences in the Biologic Activity of 2 Novel MEK Inhibitors Revealed by 18F-FDG PET: Analysis of Imaging Data from 2 Phase I Trials

Françoise Kraeber-Bodéré; Thomas Carlier; Valerie Meresse Naegelen; Eliezer Shochat; Jean Lumbroso; Carlos Trampal; James Nagarajah; Sue Chua; Florent Hugonnet; Marcel P.M. Stokkel; Fergus V. Gleeson; Jean Tessier

Two mitogen-activated protein kinase kinase (MAPK2, also known as MEK) inhibitors were assessed with 18F-FDG PET in separate phase I clinical studies, clearly illustrating the potential of metabolic imaging for dose, dosing regimen, and compound selection in early-phase trials and utility for predicting nonresponding patients. Methods: 18F-FDG PET data were collected during 2 independent, phase I, dose-escalation trials of 2 novel MEK inhibitors (RO5126766 and RO4987655). PET acquisition procedures were standardized between the 2 trials, and PET images were analyzed centrally. Imaging was performed at baseline; at cycle 1, day 15; and at cycle 3, day 1. A 10-mm-diameter region of interest was defined for up to 5 lesions, and peak standardized uptake values were determined for each lesion. The relationship between PET response and pharmacokinetic factors (dose and exposure), inhibition of extracellular-signal-regulated kinase (ERK) phosphorylation in peripheral blood mononuclear cells, and anatomic tumor response as measured by Response Evaluation Criteria in Solid Tumors was investigated for both compounds. Results: Seventy-six patients underwent PET, and 205 individual PET scans were analyzed. Strong evidence of biologic activity was seen as early as cycle 1, day 15, for both compounds. 18F-FDG PET revealed striking differences between the 2 MEK inhibitors at their recommended dose for phase II investigation. The mean amplitude of the decrease in 18F-FDG from baseline to cycle 1, day 15, was greater for patients receiving RO4987655 than for those receiving RO5126766 (47% vs. 16%, respectively; P = 0.052). Furthermore, a more pronounced relationship was seen between the change in 18F-FDG uptake and dose or exposure and phosphorylated ERK inhibition in peripheral blood mononuclear cells in patients receiving RO4987655. For both investigational drugs, PET responses tended to be greatest in patients with melanoma tumors. 18F-FDG was able to identify early nonresponding patients with a 97% negative predictive value. Conclusion: These data exemplify the role of 18F-FDG PET for guiding the selection of novel investigational drugs, choosing dose in early-phase clinical development, and predicting nonresponding patients early in treatment.


International MICCAI Workshop on Medical Computer Vision | 2013

Integrated Spatio-Temporal Segmentation of Longitudinal Brain Tumor Imaging Studies

Stefan Bauer; Jean Tessier; Oliver Krieter; Lutz-Peter Nolte; Mauricio Reyes

Consistent longitudinal segmentation of brain tumor images is a critical issue in treatment monitoring and in clinical trials. Fully automatic segmentation methods are a good candidate for reliably detecting changes of tumor volume over time. We propose an integrated 4D spatio-temporal brain tumor segmentation method, which combines supervised classification with conditional random field regularization in an energy minimization scheme. Promising results and improvements over classic 3D methods for monitoring the temporal volumetric evolution of necrotic, active and edema tumor compartments are demonstrated on a longitudinal dataset of glioma patient images from a multi-center clinical trial. Thanks to its speed and simplicity the approach is a good candidate for standard clinical use.


The Journal of Nuclear Medicine | 2013

Placental Growth Factor (PlGF)–Specific Uptake in Tumor Microenvironment of 89Zr-Labeled PlGF Antibody RO5323441

Thijs H. Oude Munnink; K. Tamas; Marjolijn N. Lub-de Hooge; Silke R. Vedelaar; Hetty Timmer-Bosscha; Annemiek M.E. Walenkamp; K. Michael Weidner; Frank Herting; Jean Tessier; Elisabeth G.E. de Vries

Placental growth factor (PlGF) is a member of the proangiogenic vascular endothelial growth factor family, which is upregulated in many tumors. RO5323441, a humanized monoclonal antibody against PlGF, showed antitumor activity in human tumor xenografts. We therefore aimed to radiolabel RO5323441 and preclinically validate this tracer to study drug tumor uptake and organ distribution by PET imaging. 89Zr-RO5323441 was tested for stability and immunoreactivity in vitro. Methods: The tumor uptake and organ distribution for 10, 50, and 500 μg of 89Zr-RO5323441 was assessed in mice bearing human PlGF–expressing hepatocellular cancer (Huh7) xenografts or human renal cell carcinoma (ACHN) xenografts without detectable human PlGF expression. The effect of pretreatment with RO5323441 (20 mg/kg) on 89Zr-RO5323441 tumor uptake was analyzed in Huh7 xenografts. 111In-IgG served as a control for nonspecific tumor uptake and organ distribution. Cy5-RO5323441 was injected to study the intratumor distribution of RO5323441 with fluorescence microscopy. Results: 89Zr-RO5323441 showed a time- and dose-dependent tumor accumulation. Uptake in Huh7 xenografts at 10 μg of 89Zr-RO5323441 was 8.2% ± 1.7% injected dose (ID)/cm3 at 144 h after injection, and in ACHN xenografts it was 5.5 ± 0.3 %ID/cm3 (P = 0.03). RO5323441 pretreatment of Huh7 xenograft–bearing mice reduced 89Zr-RO5323441 tumor uptake to the level of nonspecific 111In-IgG uptake. Cy5-RO5323441 was present in the tumors mainly in the microenvironment. Conclusion: The findings show that RO5323441 tumor uptake is PlGF-specific and time- and dose-dependent.


EJNMMI research | 2013

[18 F]FDG-PET imaging is an early non-invasive pharmacodynamic biomarker for a first-in-class dual MEK/Raf inhibitor, RO5126766 (CH5126766), in preclinical xenograft models

Tetyana Tegnebratt; Li Lu; Lucy Lee; Valerie Meresse; Jean Tessier; Nobuya Ishii; Naoki Harada; Pavel Pisa; Sharon Stone-Elander

BackgroundPositron emission tomography (PET) with [2-18u2009F]-2-fluoro-2-deoxy-D-glucose ([18u2009F]FDG-PET) was acquired at multiple time-points a) to monitor the early response to RO5126766 (CH5126766) in xenograft models b) to evaluate non-invasive small animal [18u2009F]FDG-PET imaging as a biomarker for MEK inhibitors for translation into dose-finding studies in cancer patients and c) to explore the underlying mechanism related to FDG uptake in tumors treated with RO5126766.Methods[18u2009F]FDG uptake was studied in HCT116 (K-ras), COLO205 (B-raf) mutants and COLO320DM (wild type) xenografts from day 0 to 3 of RO5126766 treatment using a microPET Focus 120 and complemented with in vitro incubations, ex-vivo phosphor imaging and immunohistochemical (IHC) analyses.ResultsIn the HCT116 (K-ras) and COLO205 (B-raf) mutant xenografts, significant decreases in [18u2009F]FDG uptake were detected in vivo on day 1 with 0.3xa0mg/kg and ex vivo on day 3 with 0.1xa0mg/kg RO5126766. [18u2009F]FDG changes correlated with decreases in tumor cells proliferation (Ki-67) and with changes in expression levels of GLUT1. No effects were observed in drug resistant COLO320DM cells. The cellular fractionation and Western blotting analyses suggested that the change of [18u2009F]FDG uptake associated with RO5126766 is due to translocation of GLUT1 from membrane to cytosol, similar to the results reported in the literature with EGFR tyrosine kinase inhibitors, which also target the MAPK pathway.ConclusionsRO5126766 inhibition resulted in a rapid time - and dose - dependent decline in [18u2009F]FDG uptake in both mutant xenografts. These results strongly resemble the clinical observations obtained with MEK/Raf inhibitors support the use of preclinical [18u2009F]FDG-PET as a translational tool for decision support in preclinical and early clinical development of MEK inhibitors.


EJNMMI research | 2014

Evaluation of efficacy of a new MEK inhibitor, RO4987655, in human tumor xenografts by [ 18 F] FDG-PET imaging combined with proteomic approaches

Tetyana Tegnebratt; Elisabeth Ruge; Sabine Bader; Nobuya Ishii; Satoshi Aida; Yasushi Yoshimura; Chia-Huey Ooi; Li Lu; Nicholas Mitsios; Valerie Meresse; Jan Mulder; Michael Pawlak; Miro Venturi; Jean Tessier; Sharon Stone-Elander

BackgroundInhibition of mitogen-activated protein kinase (MEK, also known as MAPK2, MAPKK), a key molecule of the Ras/MAPK (mitogen-activated protein kinase) pathway, has shown promising effects on B-raf-mutated and some RAS (rat sarcoma)-activated tumors in clinical trials. The objective of this study is to examine the efficacy of a novel allosteric MEK inhibitor RO4987655 in K-ras-mutated human tumor xenograft models using [18F] FDG-PET imaging and proteomics technology.Methods[18F] FDG uptake was studied in human lung carcinoma xenografts from day 0 to day 9 of RO4987655 therapy using microPET Focus 120 (CTI Concorde Microsystems, Knoxville, TN, USA). The expression levels of GLUT1 and hexokinase 1 were examined using semi-quantitative fluorescent immunohistochemistry (fIHC). The in vivo effects of RO4987655 on MAPK/PI3K pathway components were assessed by reverse phase protein arrays (RPPA).ResultsWe have observed modest metabolic decreases in tumor [18F] FDG uptake after MEK inhibition by RO4987655 as early as 2 h post-treatment. The greatest [18F] FDG decreases were found on day 1, followed by a rebound in [18F] FDG uptake on day 3 in parallel with decreasing tumor volumes. Molecular analysis of the tumors by fIHC did not reveal statistically significant correlations of GLUT1 and hexokinase 1 expressions with the [18F] FDG changes. RPPA signaling response profiling revealed not only down-regulation of pERK1/2, pMKK4, and pmTOR on day 1 after RO4987655 treatment but also significant up-regulation of pMEK1/2, pMEK2, pC-RAF, and pAKT on day 3. The up-regulation of these markers is interpreted to be indicative of a reactivation of the MAPK and activation of the compensatory PI3K pathway, which can also explain the rebound in [18F] FDG uptake following MEK inhibition with RO4987655 in the K-ras-mutated human tumor xenografts.ConclusionsWe have performed the first preclinical evaluation of a new MEK inhibitor, RO4987655, using a combination of [18F] FDG-PET imaging and molecular proteomics. These results provide support for using preclinical [18F] FDG-PET imaging in early, non-invasive monitoring of the effects of MEK and perhaps other Ras/MAPK signaling pathway inhibitors, which should facilitate a wider implementation of clinical [18F] FDG-PET to optimize their clinical use.


Clinical Cancer Research | 2018

Prediction of the optimal dosing regimen using a mathematical model of tumour uptake for immunocytokine-based cancer immunotherapy

Benjamin Ribba; Christophe Boetsch; Tapan Nayak; Hans Peter Grimm; Jehad Charo; Stefan Evers; Christian Klein; Jean Tessier; Jean Eric Charoin; Alex Phipps; Pavel Pisa; Volker Teichgräber

Purpose: Optimal dosing is critical for immunocytokine-based cancer immunotherapy to maximize efficacy and minimize toxicity. Cergutuzumab amunaleukin (CEA-IL2v) is a novel CEA-targeted immunocytokine. We set out to develop a mathematical model to predict intratumoral CEA-IL2v concentrations following various systemic dosing intensities. Experimental Design: Sequential measurements of CEA-IL2v plasma concentrations in 74 patients with solid tumors were applied in a series of differential equations to devise a model that also incorporates the peripheral concentrations of IL2 receptor–positive cell populations (i.e., CD8+, CD4+, NK, and B cells), which affect tumor bioavailability of CEA-IL2v. Imaging data from a subset of 14 patients were subsequently utilized to additionally predict antibody uptake in tumor tissues. Results: We created a pharmacokinetic/pharmacodynamic mathematical model that incorporates the expansion of IL2R-positive target cells at multiple dose levels and different schedules of CEA-IL2v. Model-based prediction of drug levels correlated with the concentration of IL2R-positive cells in the peripheral blood of patients. The pharmacokinetic model was further refined and extended by adding a model of antibody uptake, which is based on drug dose and the biological properties of the tumor. In silico predictions of our model correlated with imaging data and demonstrated that a dose-dense schedule comprising escalating doses and shortened intervals of drug administration can improve intratumoral drug uptake and overcome consumption of CEA-IL2v by the expanding population of IL2R-positive cells. Conclusions: The model presented here allows simulation of individualized treatment plans for optimal dosing and scheduling of immunocytokines for anticancer immunotherapy. Clin Cancer Res; 24(14); 3325–33. ©2018 AACR. See related commentary by Ruiz-Cerdá et al., p. 3236

Collaboration


Dive into the Jean Tessier's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge