Network


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

Hotspot


Dive into the research topics where Alain Thibault is active.

Publication


Featured researches published by Alain Thibault.


Journal of Clinical Oncology | 2004

Phase II Study of R115777, a Farnesyl Transferase Inhibitor, in Myelodysplastic Syndrome

Razelle Kurzrock; Maher Albitar; Jorge Cortes; Elihu H. Estey; Stefan Faderl; Guillermo Garcia-Manero; Deborah A. Thomas; Francis J. Giles; Mary Ellen Ryback; Alain Thibault; P. De Porre; H. Kantarjian

PURPOSE To perform a phase II study of the farnesyl transferase inhibitor R115777 (Zarnestra; Johnson and Johnson Pharmaceutical Research and Development, Raritan, NJ) in patients with myelodysplastic syndrome (MDS), using doses recommended in a phase I study in relapsed/refractory leukemia. PATIENTS AND METHODS Patients with MDS were treated with R115777 at doses of 600 mg orally (PO) bid in cycles of 4 weeks of therapy followed by a 2-week rest period. Dose reduction rules for toxicity were applied. RESULTS Twenty-seven of the 28 patients treated were assessable. Three patients responded (complete remission, n = 2; partial remission, n = 1). Responders included two patients with refractory anemia with excess blasts and one patient with refractory anemia with excess blasts in transformation. Two of the responders had a diploid karyotype and one had multiple cytogenetic abnormalities including monosomy 5 and 7. The starting dose of 600 mg PO bid resulted in side effects (myelosuppression, fatigue, neurotoxicity, rash, or leg pain) necessitating dose reduction (n = 4) or discontinuation of therapy (n = 7) in 11 (41%) of 27 patients during the induction period (12 weeks). Lower doses of 300 mg PO bid were well tolerated. All responses occurred in patients who had been reduced to this dose level during the initial two cycles. CONCLUSION This study suggests that R115777 has modest activity in MDS patients, but that, in this patient population, 4 weeks of daily doses of 600 mg PO bid is not tolerated. Further exploration of the optimal dose/schedule and correlation with biologic end points are warranted.


Journal of Neurochemistry | 2002

Lipid Metabolism as a Target for Brain Cancer Therapy: Synergistic Activity of Lovastatin and Sodium Phenylacetate Against Human Glioma Cells

Premakala Prasanna; Alain Thibault; Lei Liu; Dvorit Samid

Abstract: Malignant gliomas, the most common form of primary brain tumors, are highly dependent on the mevalonate (MVA) pathway for the synthesis of lipid moieties critical to cell replication. Human glioblastoma cells were found to be uniquely vulnerable to growth arrest by lovastatin, a competitive inhibitor of the enzyme regulating MVA synthesis, 3‐hydroxy‐3‐methylglutaryl coenzyme A reductase. The sodium salt of phenylacetic acid (NaPA), an inhibitor of MVA‐pyrophosphate decarboxylase, the enzyme that controls MVA use, acted synergistically with lovastatin to suppress malignant growth. When used at pharmacologically attainable concentrations, the two compounds induced profound cytostasis and loss of malignant properties such as invasiveness and expression of the transforming growth factor‐β2 gene, coding for a potent immunosuppressive cytokine. Supplementation with exogenous ubiquinone, an end product of the MVA pathway, failed to rescue the cells, suggesting that decreased synthesis of intermediary products are responsible for the antitumor effects observed. In addition to blocking the MVA pathway, lovastatin alone and in combination with NaPA increased the expression of the peroxisome proliferator‐activated receptor, a transcription factor implicated in the control of lipid metabolism, cell growth, and differentiation. Our results indicate that targeting lipid metabolism with lovastatin, used alone or in combination with the aromatic fatty acid NaPA, may offer a novel approach to the treatment of malignant gliomas.


Cancer | 1995

Antitumor activity of suramin in hormone-refractory prostate cancer controlling for hydrocortisone treatment and flutamide withdrawal as potentially confounding variables†

Nancy A. Dawson; Michael R. Cooper; William D. Figg; Donna Headlee; Alain Thibault; Raymond C. Bergan; Seth M. Steinberg; Edward A. Sausville; Charles E. Myers; Oliver Sartor

Background. A prospective Phase II clinical trial was conducted to assess the clinical activity of a pharmacokinetically guided suramin regimen in patients who had documented progression of metastatic prostate cancer after hydrocortisone plus antecedent or simultaneous withdrawal of flutamide.


mAbs | 2014

ARGX-110, a highly potent antibody targeting CD70, eliminates tumors via both enhanced ADCC and immune checkpoint blockade

Karen Silence; Torsten Dreier; Mahan Moshir; Peter Ulrichts; Sofie Me Gabriels; Michael A. Saunders; Harald Wajant; Peter Brouckaert; Leander Huyghe; Tim Van Hauwermeiren; Alain Thibault; Hans de Haard

Overexpression of CD70 has been documented in a variety of solid and hematological tumors, where it is thought to play a role in tumor proliferation and evasion of immune surveillance. Here, we describe ARGX-110, a defucosylated IgG1 monoclonal antibody (mAb) that selectively targets and neutralizes CD70, the ligand of CD27. ARGX-110 was generated by immunization of outbred llamas. The antibody was germlined to 95% human identity, and its anti-tumor efficacy was tested in several in vitro assays. ARGX-110 binds CD70 with picomolar affinity. In depletion studies, ARGX-110 lyses tumor cells with greater efficacy than its fucosylated version. In addition, ARGX-110 demonstrates strong complement-dependent cytotoxicity and antibody-dependent cellular phagocytosis activity. ARGX-110 inhibits signaling of CD27, which results in blocking of the activation and proliferation of Tregs. In a Raji xenograft model, administration of the fucosylated version of ARGX-110 resulted in a prolonged survival at doses of 0.1 mg/kg and above. The pharmacokinetics of ARGX-110 was tested in cynomolgus monkeys; the calculated half-life is 12 days. In conclusion, ARGX-110 is a potent blocking mAb with a dual mode of action against both CD70-bearing tumor cells and CD70-dependent Tregs. This antibody is now in a Phase 1 study in patients with advanced malignancies expressing CD70 (NCT01813539).


The Prostate | 1996

The differentiating agent phenylacetate increases prostate-specific antigen production by prostate cancer cells

Ron M. Walls; Alain Thibault; Lei Liu; Chris Wood; James M. Kozlowski; William D. Figg; Maureen Sampson; Ronald J. Elin; Dvorit Samid

The prostatic‐specific antigen (PSA) is the tumor marker most widely relied upon for the monitoring of patients with prostate cancer. Recently, declines in the serum concentrations of PSA have been advocated as a surrogate marker of tumor response in clinical trials of investigational antitumor agents. We examined the hypothesis that this postulate may not apply to the evaluation of drugs such as phenylacetate, a differentiating agent endowed with mechanisms of action different from those of classic cytotoxic chemotherapy. Using human prostatic carcinoma LNCaP cells as a model, we show that phenylacetate induces PSA production despite inhibition of tumor cell proliferation. Incubation of LNCaP cultures with cytostatic doses of phenylacetate (3–10 mM) resulted in a three‐ to fourfoLd increase in PSA secretion per cell. This appears to result from upregulation of PSA gene expression, as indicated by elevated PSA mRNA steady‐state levels in treated cells. The increase in PSA production per cell was confirmed in rats bearing subcutaneous LNCaP tumor implants that were treated systemically with phenylacetate. Further comparative studies indicate that upregulation of PSA is common to various differentiation inducers, including all, trans‐retinoic acid, 1,25‐dihydroxyvitamin D3, and butyrate but is not induced by other antitumor agents of clinical interest such as suramin. We conclude that declines in PSA may be treatment specific and that the exclusive use of this criterion as a marker of disease response may mislead the proper evaluation of differentiating agents in prostate cancer patients.


Cancer Research | 2015

Depleting MET-Expressing Tumor Cells by ADCC Provides a Therapeutic Advantage over Inhibiting HGF/MET Signaling

Anna Hultberg; Virginia Morello; Leander Huyghe; Natalie De Jonge; Christophe Blanchetot; Val erie Hanssens; Gitte De Boeck; Karen Silence; Els Festjens; Raimond Heukers; Benjamin Roux; Fabienne Lamballe; Christophe Ginestier; Emmanuelle Charafe-Jauffret; Flavio Maina; Peter Brouckaert; Michael A. Saunders; Alain Thibault; Torsten Dreier; Hans de Haard; Paolo Michieli

Hepatocyte growth factor (HGF) and its receptor MET represent validated targets for cancer therapy. However, HGF/MET inhibitors being explored as cancer therapeutics exhibit cytostatic activity rather than cytotoxic activity, which would be more desired. In this study, we engineered an antagonistic anti-MET antibody that, in addition to blocking HGF/MET signaling, also kills MET-overexpressing cancer cells by antibody-dependent cellular cytotoxicity (ADCC). As a control reagent, we engineered the same antibody in an ADCC-inactive form that is similarly capable of blocking HGF/MET activity, but in the absence of any effector function. In comparing these two antibodies in multiple mouse models of cancer, including HGF-dependent and -independent tumor xenografts, we determined that the ADCC-enhanced antibody was more efficacious than the ADCC-inactive antibody. In orthotopic mammary carcinoma models, ADCC enhancement was crucial to deplete circulating tumor cells and to suppress metastases. Prompted by these results, we optimized the ADCC-enhanced molecule for clinical development, generating an antibody (ARGX-111) with improved pharmacologic properties. ARGX-111 competed with HGF for MET binding, inhibiting ligand-dependent MET activity, downregulated cell surface expression of MET, curbing HGF-independent MET activity, and engaged natural killer cells to kill MET-expressing cancer cells, displaying MET-specific cytotoxic activity. ADCC assays confirmed the cytotoxic effects of ARGX-111 in multiple human cancer cell lines and patient-derived primary tumor specimens, including MET-expressing cancer stem-like cells. Together, our results show how ADCC provides a therapeutic advantage over conventional HGF/MET signaling blockade and generates proof-of-concept for ARGX-111 clinical testing in MET-positive oncologic malignancies.


Anti-Cancer Drugs | 1994

In vitro antitumor effect of hydroxyurea on hormone-refractory prostate cancer cells and its potentiation by phenylbutyrate.

William D. Figg; Ronald G. Walls; Michael R. Cooper; Alain Thibault; Oliver Sartor; Natalie A. McCall; Charles E. Myers; Dvorit Samid

Previous clinical trials have suggested that hydroxyurea may possess some activity against prostate cancer. The in vitro antiproliferative activity of hydroxyurea was evaluated in three hormone-refractory prostate cancer cell lines, PC-3, DU-145 and PC-3M. Fifty-percent inhibition of growth in all three cell lines required prolonged (120 h) exposure to hydroxyurea at a concentration of approximately 100 microM. Using pharmacokinetic data obtained during the course of a clinical trial of hydroxyurea, we simulated a dosing regimen that would sustain plasma drug concentrations above 100 microM for 120 h (1 g loading dose, followed by 500 mg every 6 h for 5 days in a 70 kg man). Since this dosing regimen is likely to generate an unacceptable degree of myelosuppression, in vitro combination studies were conducted with hydroxyurea and phenylbutyrate, a new differentiating agent with no myelosuppressive effects. These studies resulted in a reduction of the hydroxyurea concentration necessary for 50% growth inhibition (50 microM of hydroxyurea plus 0.5 mM of phenylbutyrate). A regimen designed to achieve that hydroxyurea concentration (400 mg loading dose, followed by 200 mg every 6 h for 5 days) should be clinically achievable. Based on these results, this combination deserves further evaluation in patients with stage D prostate cancer.


Clinical Cancer Research | 2017

Phase I dose-escalation study of the anti-CD70 antibody ARGX-110 in advanced malignancies

Philippe Aftimos; Christian Rolfo; Sylvie Rottey; Fritz Offner; D. Bron; Marie Maerevoet; Jean-Charles Soria; Mahan Moshir; Torsten Dreier; Luc Van Rompaey; Jean-Marie Michot; Karen Silence; Anna Hultberg; Domenica Gandini; Hans de Haard; Vincent Ribrag; Marc Peeters; Alain Thibault; Nicolas Leupin; Ahmad Awada

Purpose: The purpose of this study was to evaluate safety, pharmacokinetics, pharmacodynamics, and preliminary antitumor efficacy of ARGX-110, a glyco-engineered monoclonal antibody, targeting CD70, in patients with CD70 expressing advanced malignancies. Experimental Design: Dose escalation with a sequential 3+3 design was performed in five steps at the 0.1, 1, 2, 5, and 10 mg/kg dose levels (N = 26). ARGX-110 was administered intravenously every 3 weeks until progression or intolerable toxicity. Dose-limiting toxicity was evaluated in the 21 days following the first ARGX-110 administration (Cycle 1). Samples for pharmacokinetics and pharmacodynamics were collected. Results: Dose-limiting toxicity was not observed and the maximum tolerated dose was not reached. ARGX-110 was generally well tolerated, with no dose-related increase in treatment-emergent adverse events (TEAE). The most common TEAE were fatigue and drug related infusion-related reactions (IRR). Of the 20 SAEs reported, five events, all IRRs, were considered related to ARGX-110. ARGX-110 demonstrates dose proportionality over the dose range 1 to 10 mg/kg, but not at 0.1 mg/kg and a terminal half-life of 10 to 13 days. The best overall response was stable disease (14/26) in all 26 evaluable patients with various malignancies and the mean duration of treatment was 15 weeks. No dose–response related antitumor activity was observed, but biomarker readouts provided signs of biological activity, particularly in patients with hematologic malignancies. Conclusions: This dose-escalation phase I trial provides evidence of good tolerability of ARGX-110, pharmacokinetics, and preliminary antitumor activity at all dose levels in generally heavily pretreated patients with advanced CD70-positive malignancies. Clin Cancer Res; 23(21); 6411–20. ©2017 AACR.


Urologic Oncology-seminars and Original Investigations | 1997

Meeting summary: Workshop conference on endocrine therapy of advanced prostate cancer, airlie house, November 3–4, 1996

Richard J. Santen; Alain Thibault; Charles E. Myers; Leland W. K. Chung

Investigators from a range of disciplines met at the Airlie House Conference Center in Warrenton, Virginia, on November 3 and 4, 1996, to discuss the biologic principles, prior data, and potential new strategies for use of endocrine therapy in advanced prostate cancer. A general goal of this workshop was to consider whether androgen deprivation of prostate cancer cells may be associated with development of dependence on other hormonal mechanisms for cellular proliferation. The initial session reviewed recent data regarding mutations of the androgen receptor that allow it to initiate transcription in response to estrogens, progestins, and antiandrogens. Evolving concepts of receptor processing and competitive interactions among receptors were presented. The next session addressed the role of estrogens on prostate cellular function and carcinogenesis. Themes included the interaction of androgens with estrogens in mediating differentiated function as well as cell proliferation. The concept that estrogens may mediate the process of carcinogenesis was reviewed. The process of programmed cell death in regulating the rate of tumor growth and the mechanistic role of bcl-2 in androgen ablative therapy were discussed. The following session reviewed data obtained over the past two decades relating to secondary hormonal therapies in prostate cancer. Presentations focused on the use of aromatase inhibitors and antiestrogens, and on withdrawal responses to antiandrogens. The final session considered the difficulties in evaluating objective responses in patients with advanced prostate cancer. A series of endpoints for potential use were identified. A clear consensus emerged that better assessment tools for evaluating pain relief and other quality of life parameters be used for trials of endocrine therapy at this stage of the disease. Side effects and morbidity from experimental therapies should be a major concern in designing new trials. The new biologic data regarding receptor mutations and tumor cell evolution do not provide compelling evidence for initiating large multicenter trials but point to the need for small phase II studies to examine potential efficacy of additional endocrine therapies. Positive results would then lead to larger multicenter trials with incorporation of more highly developed evaluative tools of assessment.


Cancer Research | 2016

Abstract P2-05-17: ARGX-111 depletes MET-expressing circulating tumor cells via enhanced ADCC, resulting in inhibition of metastasis

Virginia Morello; Anna Hultberg; N. De Jonge; Leander Huyghe; Valérie Hanssens; Peter Brouckaert; Michael A. Saunders; Torsten Dreier; Alain Thibault; Christian Rolfo; Philippe Aftimos; Ahmad Awada; Paolo Michieli; H. de Haard

Several lines of experimental evidence suggest that Hepatocyte Growth Factor (HGF) and its receptor MET play an important role in breast cancer progression and drug resistance. To date, targeted MET inhibitors in clinical development have primarily shown cytostatic rather than cytotoxic effects. Development of a cytotoxic MET inhibitor would serve to complement standard breast cancer therapy, especially when administered in the adjuvant/neo-adjuvant setting. We have developed ARGX-111, a human antibody antagonist of MET function. ARGX-111 blocks both HGF-dependent and -independent signaling, down-regulates tumor cell surface expression of MET and kills MET-overexpressing cells by enhanced antibody-dependent cellular cytotoxicity (ADCC). ARGX-111 was shown to be more efficacious than an ADCC-inactive control antibody in both HGF-dependent and -independent tumor xenograft models. ADCC reporter assays confirmed the cytotoxic effects of ARGX-111 in patient-derived primary tumor specimens, including MET-expressing breast cancer stem-like cells. In an orthotopic mouse model of metastatic mammary carcinoma (MDA-MB-231), adjuvant or neo-adjuvant treatment with ARGX-111 was significantly more effective in depleting circulating tumor cells (CTCs) and suppressing the development of bone and lung metastases than the ADCC-inactive control. Taken together, these results provide a rationale for clinical investigation of ARGX-111 in the early breast cancer setting. An ongoing Phase 1 study (NCT02055066) is examining the effects of ARGX-111 on CTCs, alongside the assessment of its safety and efficacy. Citation Format: Morello V, Hultberg A, De Jonge N, Huyghe L, Hanssens V, Brouckaert P, Saunders M, Dreier T, Thibault A, Rolfo C, Aftimos P, Awada A, Michieli P, de Haard H. ARGX-111 depletes MET-expressing circulating tumor cells via enhanced ADCC, resulting in inhibition of metastasis. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P2-05-17.

Collaboration


Dive into the Alain Thibault's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

William D. Figg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Charles E. Myers

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael R. Cooper

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ahmad Awada

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Philippe Aftimos

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge