Network


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

Hotspot


Dive into the research topics where Olivier Rixe is active.

Publication


Featured researches published by Olivier Rixe.


The Journal of Clinical Pharmacology | 2013

Axitinib in Metastatic Renal Cell Carcinoma: Results of a Pharmacokinetic and Pharmacodynamic Analysis

Brian I. Rini; May Garrett; Bill Poland; Janice P. Dutcher; Olivier Rixe; George Wilding; Walter M. Stadler; Yazdi K. Pithavala; Sinil Kim; Jamal Tarazi; Robert J. Motzer

Axitinib is a potent and selective inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, approved for second‐line therapy for advanced renal cell carcinoma (RCC). Axitinib population pharmacokinetic and pharmacokinetic/pharmacodynamic relationships were evaluated. Using nonlinear mixed effects modeling with pooled data from 383 healthy volunteers, 181 patients with metastatic RCC, and 26 patients with other solid tumors in 17 trials, the disposition of axitinib was best described by a 2‐compartment model with first‐order absorption and a lag time, with estimated mean systemic clearance (CL) of 14.6u2009L/h and central volume of distribution (Vc) of 47.3u2009L. Of 12 covariates tested, age over 60 years and Japanese ethnicity were associated with decreased CL, whereas Vc increased with body weight. However, the magnitude of predicted changes in exposure based on these covariates does not warrant dose adjustments. Multivariate Cox proportional hazard regression and logistic regression analyses showed that higher exposure and diastolic blood pressure were independently associated with longer progression‐free and overall survivals and higher probability of partial response in metastatic RCC patients. These findings support axitinib dose titration to increase plasma exposure in patients who tolerate axitinib, and also demonstrate diastolic blood pressure as a potential marker of efficacy.


Journal for ImmunoTherapy of Cancer | 2014

The indoleamine 2,3-dioxygenase pathway controls complement-dependent enhancement of chemo-radiation therapy against murine glioblastoma

Minghui Li; Aaron Bolduc; Nasrul Hoda; Denise N. Gamble; Sarah-Bianca Dolisca; Anna Bolduc; Kelly Hoang; Claire N. Ashley; David McCall; Amyn M. Rojiani; Bernard L. Maria; Olivier Rixe; Tobey J. MacDonald; Peter S. Heeger; Andrew L. Mellor; David H. Munn; Theodore S. Johnson

BackgroundIndoleamine 2,3-dioxygenase (IDO) is an enzyme with immune-suppressive properties that is commonly exploited by tumors to evade immune destruction. Anti-tumor T cell responses can be initiated in solid tumors, but are immediately suppressed by compensatory upregulation of immunological checkpoints, including IDO. In addition to these known effects on the adaptive immune system, we previously showed widespread, T cell-dependent complement deposition during allogeneic fetal rejection upon maternal treatment with IDO-blockade. We hypothesized that IDO protects glioblastoma from the full effects of chemo-radiation therapy by preventing vascular activation and complement-dependent tumor destruction.MethodsTo test this hypothesis, we utilized a syngeneic orthotopic glioblastoma model in which GL261 glioblastoma tumor cells were stereotactically implanted into the right frontal lobes of syngeneic mice. These mice were treated with IDO-blocking drugs in combination with chemotherapy and radiation therapy.ResultsPharmacologic inhibition of IDO synergized with chemo-radiation therapy to prolong survival in mice bearing intracranial glioblastoma tumors. We now show that pharmacologic or genetic inhibition of IDO allowed chemo-radiation to trigger widespread complement deposition at sites of tumor growth. Chemotherapy treatment alone resulted in collections of perivascular leukocytes within tumors, but no complement deposition. Adding IDO-blockade led to upregulation of VCAM-1 on vascular endothelium within the tumor microenvironment, and further adding radiation in the presence of IDO-blockade led to widespread deposition of complement. Mice genetically deficient in complement component C3 lost all of the synergistic effects of IDO-blockade on chemo-radiation-induced survival.ConclusionsTogether these findings identify a novel mechanistic link between IDO and complement, and implicate complement as a major downstream effector mechanism for the beneficial effect of IDO-blockade after chemo-radiation therapy. We speculate that this represents a fundamental pathway by which the tumor regulates intratumoral vascular activation and protects itself from immune-mediated tumor destruction.


Cancer Gene Therapy | 2000

Inhibition of glioma cells in vitro and in vivo using a recombinant adenoviral vector containing an astrocyte-specific promoter

Didier Vandier; Olivier Rixe; Francois Besnard; Min Kim; Toshiki Rikiyama; Merrill E. Goldsmith; Michael Brenner; Alain Gouyette; Kenneth H. Cowan

Gene therapy using the herpes simplex virus thymidine kinase (HSV-TK) gene in combination with the drug ganciclovir (GCV) is a promising approach for the treatment of cancer-inducing gliomas, a tumor with a poor prognosis. In an attempt to limit the toxic effects on normal tissues, we constructed a recombinant adenoviral vector, Adgfa2TK, in which the HSV-TK gene is driven by the promoter for the gene encoding glial fibrillary acidic protein, an intermediate filament protein expressed primarily in astrocytes. Infection by Adgfa2TK of a glial cell line (C6) and a non-glial cell line (MDA-MB-231) revealed markedly increased expression of HSV-TK in glial cells as determined by Western blot. In comparison, high HSV-TK protein levels were produced in both cell lines after infection with a control virus, AdCMVTK, in which the constitutive cytomegalovirus viral promoter was used to direct HSV-TK expression. Infection of two glial cell lines (C6, U251) and two non-glial cell lines (HepG2, MDA-MB-231) with Adgfa2TK followed by GCV treatment revealed high toxicity in glial cell lines (50% growth inhibitory concentration: <2 μg/mL of GCV) with little or no toxicity (50% growth inhibitory concentration: >75 μg/mL) in the non-glial cell lines. In vivo, injection of Adgfa2TK into C6 tumors grown in nude mice followed by intraperitoneal GCV treatment significantly repressed tumor growth compared with the controls. Adgfa2TK may be useful for directing expression of the HSV-TK gene to gliomas.


Gastroenterologie Clinique Et Biologique | 2007

Place de l’imagerie par Tomographie par Émission de Positons pour les tumeurs stromales gastro-intestinales

Jean-Louis Alberini; Malik Al Nakib; Myriam Wartski; Eric Gontier; Frédérique Cvitkovic; Olivier Rixe; Philippe Rougier; Alain-Paul Pecking

Resume La TDM abdominopelvienne est consideree comme la methode d’imagerie de reference pour le bilan d’extension et le suivi therapeutique des tumeurs stromales gastro-intestinales. L’interet d’un examen corps entier de TEP au FDG dans le bilan d’extension apparait limite en raison de la rarete des lesions extra-abdominales et de sa sensibilite inferieure a celle de la TDM. Toutefois, la realisation d’une TEP lors du bilan pretherapeutique peut etre indiquee s’il est prevu d’evaluer precocement l’efficacite d’un traitement par imatinib, car elle permet d’observer une reponse des le 8e jour de traitement, beaucoup plus precocement que la TDM. La diminution de l’activite metabolique tumorale est rapide, importante et plus facile a quantifier que des modifications de taille ou de densite analysees en TDM. La TEP peut etre aussi utile en cas de problemes d’interpretation de la TDM tels que des images equivoques suspectes de metastases, un doute sur une reponse au traitement ou au contraire une progression en TDM, notamment lorsqu’il existe une discordance avec les donnees cliniques. La TEP et la TDM peuvent donc etre complementaires et la combinaison de ces 2 modalites avec les appareils de TEP/TDM a montre son interet pour les tumeurs stromales gastro-intestinales. La TEP pourrait etre proposee comme methode d’imagerie d’evaluation de l’efficacite therapeutique dans toute etude prospective utilisant l’imatinib ou de nouvelles molecules. L’indication de cet examen chez les malades porteurs de tumeurs stromales gastro-intestinales doit etre portee lors d’une approche pluridisciplinaire oncologique.


Cancer Chemotherapy and Pharmacology | 2014

Phase I study of cabazitaxel plus cisplatin in patients with advanced solid tumors: study to evaluate the impact of cytochrome P450 3A inhibitors (aprepitant, ketoconazole) or inducers (rifampin) on the pharmacokinetics of cabazitaxel

John Sarantopoulos; Alain C. Mita; James L. Wade; John C. Morris; Olivier Rixe; Monica M. Mita; Jean François Dedieu; Claudine Wack; Laurent Kassalow; A. Craig Lockhart

PurposeCabazitaxel is primarily metabolized by CYP3A. This study evaluated the impact of moderate/strong CYP3A inhibitors [aprepitant (Study Part 2); ketoconazole (Study Part 3)] or strong CYP3A inducers [rifampin (Study Part 4)] on the pharmacokinetics of cabazitaxel.MethodsAdult patients received IV cabazitaxel/cisplatin 15/75xa0mg/m2 on Day 1 of 3-week cycles (5/75xa0mg/m2 in Cycles 1 and 2 of Part 3 to allow a safety margin to the cabazitaxel MTD). Patients received repeated oral doses of aprepitant, ketoconazole or rifampin before/during Cycle 2. Cabazitaxel clearance was the primary endpoint; clearance and area under the plasma concentration–time curve (AUC) were normalized to body surface area and dose, respectively.ResultsThe PK population included 13 (Part 2), 23 (Part 3) and 21 patients (Part 4). Repeated aprepitant administration did not affect cabazitaxel clearance [geometric mean ratio (GMR) 0.98; 90xa0% confidence interval (CI) 0.80–1.19]. Repeated ketoconazole administration resulted in 20xa0% decrease in cabazitaxel clearance (GMR 0.80; 90xa0% CI 0.55–1.15), associated with 25xa0% increase in AUC (GMR 1.25; 90xa0% CI 0.86–1.81). Repeated rifampin administration resulted in 21xa0% increase in cabazitaxel clearance (GMR 1.21; 90xa0% CI 0.95–1.53), associated with 17xa0% decrease in AUC (GMR 0.83; 90xa0% CI 0.65–1.05). The GMR of AUC0–24 with rifampin administration was 1.09 (90xa0% CI 0.9–1.33), suggesting that rifampin had a low impact during the initial phases of cabazitaxel elimination. Safety findings were consistent with previous results.ConclusionsCabazitaxel pharmacokinetics are modified by drugs strongly affecting CYP3A. Co-administration of cabazitaxel with strong CYP3A inhibitors or inducers should be avoided.


Expert Opinion on Drug Discovery | 2015

Axitinib: from preclinical development to future clinical perspectives in renal cell carcinoma

Yousef Zakharia; Kais Zakharia; Olivier Rixe

Introduction: Based on extensive preclinical data and abundant evidence for clinical activity, vascular endothelial growth factor receptor (VEGFR) inhibitors are currently standard of care for metastatic renal cell carcinoma (mRCC). Axitinib is one of the most selective molecules in the class of anti-angiogenic agents, which confers an optimal profile between its safety and anti-cancer activity spectrum. Area covered: In this review, the authors discuss the different stages that lead to the approval of axitinib in the clinic as well as the current perspectives for its clinical use with other promising therapies in mRCC such as immune checkpoint inhibitors and vaccines. Expert opinion: In 2015, axitinib has emerged as one of the major agents used in mRCC. Based on robust preclinical data, this highly specific VEGFR inhibitor continues to be evaluated in different indications, including the adjuvant setting but also sequential administration with other molecularly targeted agents or combinations with immune therapies.


Targeted Oncology | 2016

Effect of Renal Impairment on the Pharmacokinetics and Safety of Axitinib.

Ying Chen; Brian I. Rini; Robert J. Motzer; Janice P. Dutcher; Olivier Rixe; George Wilding; Walter M. Stadler; Jamal Tarazi; May Garrett; Yazdi K. Pithavala

AbstractBackgroundAxitinib, an inhibitor of vascular endothelial growth factor (VEGF) receptors, is approved as second-line treatment for advanced renal cell carcinoma (RCC). Agents targeting the VEGF pathway may induce renal toxicities, which may be influenced by pre-existing renal dysfunction.ObjectiveThe objective was to characterize axitinib pharmacokinetics and safety in patients with renal impairment.Patients and MethodsEffect of renal function (baseline creatinine clearance [CrCL]) on axitinib clearance was evaluated in a population pharmacokinetic model in 207 patients with advanced solid tumors who received a standard axitinib starting dose, and in 383 healthy volunteers. Axitinib safety according to baseline CrCL was assessed in previously treated patients with RCC (nu2009=u2009350) who received axitinib in the phase 3 AXIS study.ResultsMedian axitinib clearance was 14.0, 10.7, 12.3, 7.81, and 12.6xa0L/h, respectively, in individuals with normal renal function (≥90xa0ml/min; nu2009=u2009381), mild renal impairment (60–89xa0ml/min; nu2009=u2009139), moderate renal impairment (30–59xa0ml/min; nu2009=u200964), severe renal impairment (15–29xa0ml/min; nu2009=u20095), and end-stage renal disease (<15xa0ml/min; nu2009=u20091). The population pharmacokinetic model adequately predicted axitinib clearance in individuals with severe renal impairment or end-stage renal disease. Grade ≥3 adverse events (AEs) were reported in 63xa0% of patients with normal renal function or mild impairment, 77xa0% with moderate impairment, and 50xa0% with severe impairment; study discontinuations due to AEs were 10xa0%, 11xa0%, and 0xa0%, respectively.ConclusionsAxitinib pharmacokinetics and safety were similar regardless of baseline renal function; no starting-dose adjustment is needed for patients with pre-existing mild to severe renal impairment.n


Clinical Cancer Research | 2015

High-Dose Sirolimus And Immune Selective Pentostatin Plus Cyclophosphamide Conditioning Yields Stable Mixed Chimerism and Insufficient Graft-Versus-Tumor Responses

Miriam E. Mossoba; David Halverson; Roger Kurlander; Bazetta Blacklock Schuver; Ashley Carpenter; Brenna Hansen; Seth M. Steinberg; Syed Abbas Ali; Nishant Tageja; Frances T. Hakim; Juan Gea-Banacloche; Claude Sportes; Nancy M. Hardy; Dennis D. Hickstein; Steven Z. Pavletic; Hanh Khuu; Marianna Sabatini; David F. Stroncek; Bruce L. Levine; Carl H. June; Jacopo Mariotti; Olivier Rixe; Antonio Tito Fojo; Michael R. Bishop; Ronald E. Gress; Daniel H. Fowler

Purpose: We hypothesized that lymphoid-selective host conditioning and subsequent adoptive transfer of sirolimus-resistant allogeneic T cells (T-Rapa), when combined with high-dose sirolimus drug therapy in vivo, would safely achieve antitumor effects while avoiding GVHD. Experimental Design: Patients (n = 10) with metastatic renal cell carcinoma (RCC) were accrued because this disease is relatively refractory to high-dose conditioning yet may respond to high-dose sirolimus. A 21-day outpatient regimen of weekly pentostatin (P; 4 mg/m2/dose) combined with daily, dose-adjusted cyclophosphamide (C; ≤200 mg/d) was designed to deplete and suppress host T cells. After PC conditioning, patients received matched sibling, T-cell–replete peripheral blood stem cell allografts, and high-dose sirolimus (serum trough target, 20–30 ng/mL). To augment graft-versus-tumor (GVT) effects, multiple T-Rapa donor lymphocyte infusions (DLI) were administered (days 0, 14, and 45 posttransplant), and sirolimus was discontinued early (day 60 posttransplant). Results: PC conditioning depleted host T cells without neutropenia or infection and facilitated donor engraftment (10 of 10 cases). High-dose sirolimus therapy inhibited multiple T-Rapa DLI, as evidenced by stable mixed donor/host chimerism. No antitumor responses were detected by RECIST criteria and no significant classical acute GVHD was observed. Conclusions: Immune-selective PC conditioning represents a new approach to safely achieve alloengraftment without neutropenia. However, allogeneic T cells generated ex vivo in sirolimus are not resistant to the tolerance-inducing effects of in vivo sirolimus drug therapy, thereby cautioning against use of this intervention in patients with refractory cancer. Clin Cancer Res; 21(19); 4312–20. ©2015 AACR.


Targeted Oncology | 2014

Significant anti-tumor effect of bevacizumab in treatment of pineal gland glioblastoma multiforme.

Joshua Mansour; Braxton Fields; Samuel D. Macomson; Olivier Rixe

Glioblastoma multiforme (GBM) is the most aggressive subtype of malignant gliomas. Current standard treatment for GBM involves a combination of cytoreduction through surgical resection, followed by radiation with concomitant and adjuvant chemotherapy (temozolomide). The role of bevacizumab in the treatment of GBM continues to be a topic of ongoing research and debate. Despite aggressive treatment, these tumors remain undoubtedly fatal, especially in the elderly. Furthermore, tumors present in the pineal gland are extremely rare, accounting for only 0.1–0.4xa0% of all adult brain tumors, with this location adding to the complexity of treatment. We present a case of GBM, at the rare location of pineal gland, in an elderly patient who was refractory to initial standard of care treatment with radiation and concomitant and adjuvant temozolomide, but who developed a significant response to anti-angiogenic therapy using bevacizumab.


Anti-Cancer Drugs | 2015

Phase I dose-escalation study of cabazitaxel administered in combination with gemcitabine in patients with metastatic or unresectable advanced solid malignancies

Olivier Rixe; Igor Puzanov; Patricia LoRusso; Roger B. Cohen; John C. Morris; Olugbenga Olowokure; Jian Y. Yin; Séverine Doroumian; Liji Shen; Anthony J. Olszanski

Taxane–gemcitabine combinations have demonstrated antitumor activity. This phase I study (NCT01001221) aimed to determine the maximum tolerated dose (MTD) and dose-limiting toxicities (DLTs) of cabazitaxel plus gemcitabine and to assess the preliminary efficacy of this combination. The patients included had metastatic or unresectable solid tumors and had exhausted standard treatment. Cohorts of three to six patients received cabazitaxel (15–20u2009mg/m2) before (part 1a) or after (part 1b) gemcitabine (700–1000u2009mg/m2) on Day 1 and gemcitabine alone on Day 8. Prophylactic growth factors were not allowed in cycle 1. In part 1a (n=12), five patients received 20u2009mg/m2 cabazitaxel plus 1000u2009mg/m2 gemcitabine (20/1000), five received 15/900, two received 15/700. In part 1b, all six patients received the lowest dose (700/15). At all doses, two or more patients experienced a DLT, regardless of administration sequence, including febrile neutropenia (n=4), grade 4 neutropenia (n=2), grade 4 thrombocytopenia (n=2), and grade 3 aspartate transaminase increase (n=1). The MTD was not established as all cohorts exceeded the MTD by definition. All patients experienced an adverse event; the most frequent all-grade nonhematologic events were fatigue (66.7%), decreased appetite (50.0%), and diarrhea (44.4%). The most frequent grade 3–4 hematologic abnormalities were neutropenia (83.3%), leukopenia (77.8%), and lymphopenia (72.2%). Toxicity was sequence-independent but appeared worse with gemcitabine followed by cabazitaxel. Durable partial responses were observed in three patients (prostate cancer, appendiceal cancer, and melanoma). The unacceptable DLTs with cabazitaxel plus gemcitabine, at doses reduced more than 25% from single-agent doses, preclude further investigation.

Collaboration


Dive into the Olivier Rixe's collaboration.

Top Co-Authors

Avatar

John C. Morris

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

David H. Munn

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Didier Vandier

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

George Wilding

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Janice P. Dutcher

National Foundation for Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge