Network


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

Hotspot


Dive into the research topics where Stephane Leonce is active.

Publication


Featured researches published by Stephane Leonce.


Bioorganic & Medicinal Chemistry Letters | 2001

Novel antitumor artemisinin derivatives targeting G1 phase of the cell cycle

Ying Li; Feng Shan; Jin-Ming Wu; Guang-Shao Wu; Jian Ding; Dong Xiao; Wei-Yi Yang; Ghanem Atassi; Stephane Leonce; Daniel-Henri Caignard; Pierre Renard

Modification of artemisinin structure led us to the discovery of a novel class of antitumor compounds. These artemisinin derivatives containing cyano and aryl groups showed potent antiproliferative effect in vitro against P388 and A549 cells. This activity was reflected in P388 murine leukemia by an accumulation of cells in G1 phase, and induction of apoptosis.


Bioorganic & Medicinal Chemistry Letters | 2003

Synthesis and pharmacological evaluation of novel non-lactone analogues of camptothecin.

Patrick Hautefaye; Bernard Cimetiere; Alain Pierré; Stephane Leonce; John Hickman; William Laine; Christian Bailly; Gilbert Lavielle

Ten novel camptothecin (CPT) derivatives devoid of the lactone function in the E-ring were synthesized and evaluated as anticancer agents. Several of these CPT analogues bearing a five-membered E-ring are potent inhibitors of the DNA relaxation and cleavage reactions catalyzed by topoisomerase I and exhibit promising cytotoxic activities with IC(50) values in the nM range. This is the first successful design of lactone-free CPT, providing thus a new avenue to the development of topoisomerase I targeted antitumor agents.


Molecular Cancer Therapeutics | 2013

S49076 is a Novel Kinase Inhibitor of MET, AXL and FGFR with Strong Preclinical Activity Alone and in Association with Bevacizumab

Mike F. Burbridge; Celine Bossard; Carine Saunier; Imre Fejes; Alain Bruno; Stephane Leonce; Gilles Ferry; Georges Da Violante; Fraņcois Bouzom; Valérie Cattan; Anne Jacquet-Bescond; Paolo M. Comoglio; Brian Lockhart; Jean A. Boutin; Alex Cordi; Jean Claude Ortuno; Alain Pierré; John Hickman; Francisco Cruzalegui; Stéphane Depil

Aberrant activity of the receptor tyrosine kinases MET, AXL, and FGFR1/2/3 has been associated with tumor progression in a wide variety of human malignancies, notably in instances of primary or acquired resistance to existing or emerging anticancer therapies. This study describes the preclinical characterization of S49076, a novel, potent inhibitor of MET, AXL/MER, and FGFR1/2/3. S49076 potently blocked cellular phosphorylation of MET, AXL, and FGFRs and inhibited downstream signaling in vitro and in vivo. In cell models, S49076 inhibited the proliferation of MET- and FGFR2-dependent gastric cancer cells, blocked MET-driven migration of lung carcinoma cells, and inhibited colony formation of hepatocarcinoma cells expressing FGFR1/2 and AXL. In tumor xenograft models, a good pharmacokinetic/pharmacodynamic relationship for MET and FGFR2 inhibition following oral administration of S49076 was established and correlated well with impact on tumor growth. MET, AXL, and the FGFRs have all been implicated in resistance to VEGF/VEGFR inhibitors such as bevacizumab. Accordingly, combination of S49076 with bevacizumab in colon carcinoma xenograft models led to near total inhibition of tumor growth. Moreover, S49076 alone caused tumor growth arrest in bevacizumab-resistant tumors. On the basis of these preclinical studies showing a favorable and novel pharmacologic profile of S49076, a phase I study is currently underway in patients with advanced solid tumors. Mol Cancer Ther; 12(9); 1749–62. ©2013 AACR.


European Journal of Medicinal Chemistry | 2001

Synthesis and cytotoxicity of artemisinin derivatives containing cyanoarylmethyl group.

Jin-Ming Wu; Feng Shan; Guang-Shao Wu; Ying Li; Jian Ding; Dong Xiao; Jia-Xian Han; Ghanem Atassi; Stephane Leonce; Daniel-Henri Caignard; Pierre Renard

A series of 12alpha-deoxoartemisinyl cyanoarylmethyl dicarboxylates (4a-4o), dicarboxylic acids 12alpha-deoxoartemisinyl ester cyanoarylmethyl amide (5a-5k), and dicarboxylic acids 12alpha-deoxoartemisinyl ester N-methylcyanoarylmethyl amide (6a-6l), showing moderate cytotoxicity against P388 and L1210 cells were prepared. They induced the significant accumulation of L1210 and P388 cells in the G1 phase of the cell cycle. This mechanism of action was quite different from that of the majority of cytotoxic compounds used in the chemotherapy of cancer. Compound 4b possessed better cytotoxicity than the other compounds.


Bioorganic & Medicinal Chemistry | 2003

Syntheses and antiproliferative activities of rebeccamycin analogues bearing two 7-azaindole moieties

Christelle Marminon; Alain Pierré; Bruno Pfeiffer; Valérie Pérez; Stephane Leonce; Pierre Renard; Michelle Prudhomme

As a part of structure-activity relationship studies on rebeccamycin analogues, compounds containing two aza-indole moieties were synthesized bearing either a methyl group or a hydrogen atom on the imide nitrogen. The azaindole substructures were expected to enhance the cytotoxicity toward tumor cell lines through stronger hydrogen bonding with the target enzyme(s). The cytotoxicities of compounds 8, 10 and 19 against a panel of tumor cell lines were examined and compared with those of rebeccamycin, dechlorinated rebeccamycin 2 and N-methylated analogue A. Their effect on the L1210 cell cycle was also evaluated. Compound 19, having an imide NH function had the strongest cytotoxicity towards L1210 cells and induced the largest accumulation of cells in the G2+M phases of the cell cycle. In contrast to their non-aza analogues, which were cytotoxic for all the cell lines tested, diaza compounds 10 and 19 showed selectivity for some cell lines.


Cancer Research | 2006

Generation of Replication-Dependent Double-Strand Breaks by the Novel N2-G-Alkylator S23906-1

Stephane Leonce; Laurence Kraus-Berthier; Roy M. Golsteyn; Marie-Hélène David-Cordonnier; Christelle Tardy; Amélie Lansiaux; Virginie Poindessous; Annette K. Larsen; Alain Pierré

S23906-1, a new DNA alkylating agent that reacts with the exocyclic 2-NH2 group of guanine residues yielding monofunctional adducts, is currently under clinical evaluation in phase I trials. To investigate the mechanism of action of S23906-1, we compared parental KB-3-1 cells and KB/S23-500 cells that are 15-fold resistant to S23906-1. Cell death induced by 1 micromol/L S23906-1 in KB-3-1 cells was associated with their irreversible arrest in the G2-M phases of the cell cycle followed by apoptosis, whereas a proportion of the resistant KB/S23-500 cells were able to exit from the G2 arrest and divide, leading to a significantly lower rate of apoptosis. The attenuated apoptotic response was associated with decreased Chk2 protein phosphorylation, indicating that the DNA damage signaling pathways are more potently activated in the sensitive cells. However, similar rates of adduct formation and repair were measured in both cell lines. Exposure to S23906-1 induced a higher formation of DNA breaks, measured by the comet assay, in sensitive cells. In agreement, a histone H2AX phosphorylation assay revealed that S23906-1 induced double-strand breaks (DSB) in a dose- and time-dependent manner and that these were more persistent in the parental cells. These DSBs were found mainly in S-phase cells and inhibited by aphidicolin, suggesting that they are DNA replication-mediated DSBs. These results suggest that secondary DNA lesions play an important role in the cytotoxicity of this compound and make histone H2AX phosphorylation an attractive marker for monitoring the efficacy of S23906-1.


Biochemical Pharmacology | 2002

Induction of apoptosis in HL-60 leukemia and B16 melanoma cells by the acronycine derivative S23906-1

Jérôme Kluza; Amélie Lansiaux; Nicole Wattez; Marie-Paule Hildebrand; Stephane Leonce; Alain Pierré; John Hickman; Christian Bailly

The benzoacronycine derivative S23906-1 is a highly potent antitumor agent with a broad spectrum of activity against different human solid tumor xenografts. The marked cytotoxic potential of this drug may be the result of its interaction with DNA but the precise mechanism of action remains unclear at present. We have investigated the induction of apoptosis in human promyelocytic leukemia HL-60 and murine melanoma B16 cells treated with S23906-1. With both cell lines, the drug induces cell cycle perturbations (G2/M arrest) and triggers apoptosis as revealed by the externalization of Annexin V-targeted PS residues at the periphery of the cells. But the biochemical pathways leading to apoptosis are different for the two cancer cell lines. In HL-60 cells, the drug induces significant variations of the Delta Psi(mt), measured by flow cytometry using the fluorochromes JC-1 and cm-X-ros. Activation of caspase-3 and chromatin condensation in HL-60 cells exposed to submicromolar concentrations of S23906-1 for 24hr were also clearly seen by flow cytometry and confocal microscopy experiments. In contrast, the extent of apoptosis induced by S23906-1 was found to be much more limited in B16 cells. No significant variations of Delta Psi(mt) and no cleavage of the fluorescent caspase-3 substrate GDEVDGI (PhiPhiLux-G(1)D(2) probe) could be detected by cytometry in B16 cells exposed to S23906-1. In addition, we characterized the mitochondrial production of reactive oxygen species (ROS) using the probe dihydroethidine (HE) and the variations of the mitochondrial mass using the cardiolipin-interacting probe nonyl acridine orange (NAO). S23906-1 stimulates the production of ROS in both cell lines but the number of mitochondria seems to increase only in drug-treated B16 cells. Collectively these findings identify S23906-1 as a potent inducer of cell apoptosis in the leukemia cells and to a lower extent in the melanoma cells. The results help to understand the downstream cytotoxic actions of this new anticancer agent which is currently undergoing preclinical development.


Bioorganic & Medicinal Chemistry Letters | 2001

Design, synthesis and antiproliferative activity of tripentones: A new series of antitubulin agents

Vincent Lisowski; Cecile Enguehard; Jean-Charles Lancelot; Daniel-Henri Caignard; Stéphanie Lambel; Stephane Leonce; Alain Pierre; Ghanem Atassi; Pierre Renard; Sylvain Rault

Structure-activity relationship studies of a new series of tripentones (thieno[2,3-b]pyrrolizin-8-ones), led us to prepare several derivatives with antiproliferative activities. The most promising 3-(3-hydroxy-4-methoxyphenyl)thieno[2,3-b]pyrrolizin-8-one 20 (leukemia L1210, IC(50)=15 nM) was shown to be a potent inhibitor of tubulin polymerization.


Bioorganic & Medicinal Chemistry Letters | 2002

Synthesis and Biological Evaluation of Aristolactams

Axel Couture; Eric Deniau; Pierre Grandclaudon; Hélène Rybalko-Rosen; Stephane Leonce; Bruno Pfeiffer; Pierre Renard

A variety of aristolactam derivatives were synthesized and evaluated for cytotoxicity. Modulations were carried out on the phenanthrene nucleus and the lactam moiety as well. N-(N-dialkylaminoalkyl) derivatives exhibited interesting cytotoxic activity against the L1210 leukemia cell line.


Anti-Cancer Drugs | 2002

Acronycine derivatives as promising antitumor agents.

Nicolas Guilbaud; Stephane Leonce; François Tillequin; Michel Koch; John Hickman; Alain Pierré

Originally isolated from an Australian plant, acronycine is an antitumor alkaloid with poor water solubility and low potency. The modest antitumor activity of this compound was markedly improved by the total synthesis of original analogs resulting in the selection of S23906-1, a diester derivative of 1,2-dihydrobenzo[b]acronycine. S23906-1 is characterized in vitro by a high potency in clonogenic assays and uncommon cell cycle pertubations. In vivo, this compound demonstrated a selectivity for human solid tumors as compared to murine transplantable tumors. The unique pharmacological profile of S23906-1 was particularly defined by a broad antitumor efficacy when administered i.v. or orally on aggressive orthotopic models of human lung, ovarian and colon models with comparable or better activity than clinically used anticancer drugs. The molecular mechanism of action of S23906-1 could involve DNA alkylation, modulation of cyclin E protein levels and inhibition of DNA synthesis leading to apoptosis. Ongoing preclinical toxicological studies will help to define the potential of this novel agent which is already considered as a valuable candidate for clinical studies.

Collaboration


Dive into the Stephane Leonce's collaboration.

Researchain Logo
Decentralizing Knowledge