Final Program Feb. 27, 2014 Drug Screening

Transcription

Final Program Feb. 27, 2014 Drug Screening
Feb. 27, 2014
Drug Screening : from Phenotypes to Molecular Modeling
Final Program
Drug Screening : from Phenotypes to Molecular Modeling
1
Feb. 27, 2014
Drug Screening : from Phenotypes to Molecular Modeling
Welcome to the first Rhône-Alpes meeting promoting the chemical biology topic : «Drug
Screening : from Phenotypes to Molecular Modeling»
A few time ago Dr. Ronald Frank, Coordinator of the EU-OPENSCREEN European project wrote :
«Why investigate the biological effects of chemical substances?
Chemical substances can kill bacteria (but also man!), can block the spread of viruses, hinder the
growth of cancer cells, protect plant growth, and many more things. Nature has produced a virtually infinite
variety of molecules in environments ranging from the depths of the oceans, to rain forests, to natural oil
sources. Additionally, chemists have been continuing to develop novel, artificial compounds. This has
produced an immense reservoir of potential drugs that might, for instance, prevent or treat diseases – if only
scientists could determine the various effects of these many substances.
Across the world, scientists are seeking substances that can solve particular health challenges in a
targeted way. This is taking place in private and public laboratories that have, so far, worked mainly in an
isolated manner. Only joining forces and overcoming this fragmentation will allow us to learn everything we
can about each substance –to understand the mechanisms behind its activity and to recognize potential
hazardous effects as early as possible»
Chemical Biology is an evolving interdisciplinary research field that studies biological processes
using chemical techniques and tools. Chemical entities are introduced into biological systems as nonnatural building blocks, tags, enzyme substrates or ligands (binders); they then selectively modify cellular
target molecules to become activated, inhibited or labelled for removal (pull-down) or visualisation.
One major focus is on developing chemical substances to be used as probes that interact with
defined sites on the surfaces of target cellular molecules, such as proteins, and thereby selectively modulate
their biological function. When applied to cells or organisms, the consequences of this perturbation, such as
proliferation, differentiation, death etc., are studied in molecular detail. Such chemical probes help
scientists to reveal the exact role of cellular components in the complex network of cellular processes and
responses. Simultaneously, the value of a compound as potential effective reagent and future product is
made apparent.
Today we are pleased to welcome in Villeurbanne, France about 80 participants from very different
fields belonging to chemistry, cell biology, physical chemistry, etc. We hope that this meeting will help them
in discussing their research projects and, hopefully, construct new collaboratives ideas for the best of the
chemical biology development in the Rhône-Alpes Region.
Jean-Marc Lancelin, Université Lyon 1, Marie-Odile Fauvarques, CEA-DSV Grenoble
Co-organizers
Drug Screening : from Phenotypes to Molecular Modeling
2
Acknowledgements We thank ARC 1 Santé and Région Rhône-Alpes for a significant financial support, Ecole
Doctorale de Chimie de Lyon and INSA Lyon for hosting the conference and the logistic support.
How to come to the conference theater?
The meeting is located at :
Amphithéatre Emilie du Châtelet
SCD Doc'INSA 31 avenue Jean Capelle, 69621 Villeurbanne, France
Tél: +33(0)4 72 43 81 40 - Fax : +33(0)4 72 43 85 02
Index «1» on the map below :
http://scd.docinsa.insa-lyon.fr/plan-dacces-aux-bibliotheques
http://www.insa-lyon.fr/en/coming-insa-lyon :
Drug Screening : from Phenotypes to Molecular Modeling
3
Access by train or by plane:
From Lyon-St Exupéry airport: Airport ↔ Lyon in less than 30 minutes : http://www.rhonexpress.fr/
From Part-Dieu train station:
Take the T1 Tramway towards “IUT Feyssine” and get off at “INSA-Einstein”. About 15 min transportation. Tickets
are available from the vending machines at each tram stops.
From Perrache train station:
Take the Line A metro towards “Laurent Bonnevay” and get off at "Charpennes", then take the T1 Tramway towards
“IUT Feyssine” and get off at "INSA-Einstein".
Access from the highway: Via "Rocade Est" ring road: exit 1B then "Croix Luizet", follow "la Doua", then "Domaine Scientifique de la Doua". Via the Boulevard Laurent BONNEVAY: exit 6 "Porte de Croix Luizet", then follow direction "Campus de la
Doua" (road access map at http://www.insa-lyon.fr/files/rte/PlanAccesINSA_route.pdf
Public Transport of Lyon (TCL)
All the information to travel in public transports on: www.tcl.fr
Drug Screening : from Phenotypes to Molecular Modeling
4
Final Program
8h30-9h : Registration
9h-9h15 : Meeting opening (Jean-Marc Lancelin, Marie-Odile Fauvarque)
9h15-10h15 :
Plenary lecture 1
Gianni De Fabritiis, In-silico ligand binding assays: poses, affinities and kinetics
10h15 -10h45 : Coffee break and poster installations
Oral presentations session 1
10h45 - 11h15 :
Olivier Walker, Ligand affinity for proteins explored by NMR and molecular metadynamics
11h15 -11h45 :
Maxime Prost, Tagging live cells which express specific peptidase activity with solid-state fluorescence.
11h45-12h15 :
Maurice Médebielle, Difluoromethylbenzoxazole pyrimidine thioether (DFMB) derivatives as novel nonnucleoside HIV-1 reverse transcriptase inhibitors.
Complimentary buffet, poster session and unformal discussions
14h-15h :
Plenary lecture 2
Philippe Masson, Introduction to Drug Discovery & A case study of phenotypic screening.
Oral présentations session 2
15h-15h30 :
Laurent Guyon, Gscore, a Robust Cell-by-cell Score for Sensitive and Specific Hit Discovery in High Content
Screening.
15h30-16h :
Thierry Lomberget, Chemical synthesis and in cellulo tubulin polymerization inhibition evaluation: a winning
combination for the discovery of new anticancer agents, deriving from combretastatin A-4
Drug Screening : from Phenotypes to Molecular Modeling
5
Coffee break
16h20 -16h50 :
Morgane Champleboux, Treating yeast infections with new innovative chromatin targets.
16h50-17h20 :
Dimitrios Skoufias, STLC-resistant cell lines as tools to classify chemically divergent Eg5 targeting agents
according to their mode of action and target specificity.
17h20 : Concluding remarks and meeting closing.
Drug Screening : from Phenotypes to Molecular Modeling
6
Plenary lectures
Drug Screening : from Phenotypes to Molecular Modeling
7
Plenary Lecture 1
In-silico ligand binding assays: poses, affinities and kinetics
Gianni De Fabritiis
Computational Biophysics Laboratory (GRIB-IMIM),Universitat Pompeu Fabra, Barcelona
Biomedical Research Park (PRBB),C/ Dr. Aiguader 88, 08003, Barcelona, Spain. Tel. +34 93 316
0537 (0506) Fax. +34 93 316 0550 Office: 492.02
Unit page: http://grib.imim.es Lab page: http://multiscalelab.org
Understanding kinetics and thermodynamics properties of protein-ligand interactions by
computer simulations is of critical importance for biomedical research. Recently, we have been
able to quantitatively reconstruct the complete binding process of several molecular systems in
terms of binding poses, kinetics, affinities and pathways of binding. The methodology is based on
performing high-throughput molecular dynamics simulations of free ligand binding with the aim
of recovering binding poses with accuracy of less than 2 Å RMSD compared to crystal structures
and associated kinetics. Furthermore we obtain secondary binding sites which can be of
importance fragment based drug design approaches. We assess in this talk the current
capabilities of the methodology and its accuracy and precision on a set of protein-ligand systems
which demonstrate the potential for drug design.
References
http://scholar.google.com/citations?user=-_kX4kMAAAAJ&hl=en
Drug Screening : from Phenotypes to Molecular Modeling
8
Plenary Lecture 2
Introduction to Drug Discovery & A case study of phenotypic screening
Philippe Masson
INVENTIVA
50, rue de Dijon, 21121 Daix
France
[email protected]
http://www.inventivapharma.com
Drug Screening : from Phenotypes to Molecular Modeling
9
Introduction to Drug Discovery & a Case Study of Phenotypic Screening
Philippe Masson, Head of Biology, screening and Compound Management, Inventiva Pharma
The first part of the presentation will focus on drug discovery concepts and challenges. Despite
huge investments Pharmaceutical industry productivity is decreasing. Better understanding of the
biological mechanisms and innovation is probably one key for future success. After decades of
target-based screening, more biology-integrated assays like phenotypic screening could be a
valuable approach.
In a second part, an example of phenotypic screening to find anti-fibrotic compounds for treating
chronic kidney disease (CKD) will be presented. CKD is a major public health problem mainly
causes by diabetes and hypertension. A common feature of these CKD is the fibrotic status which
settles down progressively over years. Using a high-content screening approach, renal fibroblast
treated by TGFβ1, a major fibrotic factor, clearly induced a fibrotic response based on the
increase of extracellular matrix deposition, fibroblast differentiation, and proliferation. These
effects are blocked, in dose dependent manner, by an Alk5 inhibitor. 51K compounds were
screened at 3 µM using this four colour assay. 47 compounds, not toxic, were founded to block,
in dose dependent manner, the three fibrotic parameters all together induced by TGFβ1. 21
compounds distributed into 6 families (≥ 2 hits) and 26 singletons displaying IC50 values from
>30µM to 0.1µM. Activity of these 47 hits was confirmed by qPCR on Fibronectin1 and Acta2
genes, indicating that the compounds do not act through post translational modification of
fibronectin and αSMA. In order to check the activity of compounds in a human fibroblastic
context, MRC5 cells treated with TGFβ1 were incubated with the 47 hits. qPCR analyses
performed on fibronectin1, acta2, collagen Iα1 and collagen IVα1, showed that 11 hits were still
able to block in human cells the induction by TGFβ1 on these four genes. Interestingly some of
these hits did not block the TGFβ1 binding or TGFβ receptor kinase activities. These data
support the fact that a phenotypic screening can deliver innovating hits that act on conserved
TGFβ1 pathways without directly involving the TGFβ receptors.
In a third part, slides about Inventiva a new Partnering Research Company (PRO) will be
presented.
Communications
Drug Screening : from Phenotypes to Molecular Modeling
10
Using NMR and molecular dynamics as a microscope for life science. Application to the determination of protein-­‐ligand affinity Olivier WALKER Institut des Sciences Analytiques, 5 rue de la Doua, 69100 Villeurbane, France Molecular interactions are of prime importance to ensure communication within cells. Capturing theses processes requires the use of a method sensitive to both structure and dynamics at an atomic level. As a suitable method, NMR can probe dynamics processes in the liquid state through observables averaged over different time scales. To fill the gap, CPU accelerated molecular dynamics (MD) can provide a valuable piece of information to interpret and decipher NMR data. Through different examples, we will see how NMR, MD and funnel metadynamics(1,2) can help to understand protein-­‐ligand interactions and affinity. A) B) Figure 1: (Left) definition of the funnel used to explore binding modes of a small compound to the SH3 domain of STAM2, (Right) Free energy as a function of the projection on the z axis and distance from z axis of the center of mass of the ligand, where z is the axis of the funnel.
Reference: 1. Limongelli, V., Bonomi, M., and Parrinello, M. (2013) Funnel metadynamics as accurate binding free-­‐energy method. Proc Natl Acad Sci U S A 110, 6358-­‐6363 2. Harvey,M., Giupponi, G. and De Fabritiis,G. (2009) ACEMD: Accelerated molecular dynamics simulations in the microseconds timescale, J. Chem. Theory and Comput. 5, 1632 TAGGING LIVE CELLS WHICH EXPRESS SPECIFIC PEPTIDASE
ACTIVITY WITH SOLID-STATE FLUORESCENCE
1,2
Maxime PROST,
2
2
Laurence CANAPLE, Jacques SAMARUT, Jens HASSERODT
1
1
Laboratoire de Chimie, ENS de Lyon, Lyon, France
Institut de Génomique Fonctionnel de Lyon, ENS de Lyon, Lyon, France
[email protected], http://www.ens-lyon.fr/CHIMIE/, http://igfl.ens-lyon.fr/
2
Detecting a specific enzyme activity has long been of great interest because it is applied in fields as
diverse as histology, biotechnology or medical diagnostics. However current probes usually suffer
from a lack of robustness (false positive signal), from swift degradation by photobleaching, and from
poor sensitivity which makes them unsuitable for precise enzymatic activity localization.[1]
To overcome the above mentioned problems, we have developed three-component fluorogenic probes
which allow very precise and sensitive localization of a specific active enzyme by releasing a unique
solid-state fluorophore (Figure 1).
Figure 2 : Imaging Leucine Amino
Figure 1 : Principle of the three-component probes
Peptidase in HeLa cells (25 µM for 2 h)
[2]
This phenolic fluorophore, known as ELF-97 alcohol, is only fluorescent in solid state, possesses an
unusually large Stokes shift, is totally photostable, and can be turned off if its phenolic proton is
replaced by another group. However, this compound has not been widely used because of the
instability of the chemical link between the fluorophore and the enzyme-susceptible portion of the
probe. We have overcome this issue by incorporating a smart spacer ensuring complete probe stability.
After catalytic cleavage, a metastable intermediate is generated that cyclizes thereby releasing the
fluorophore (Figure 1).
The simple four-step synthesis[3] allowed us to create a variety of fluorogenic probes incorporating
several spacers, fluorophores, and enzyme-susceptible trigger units. The resulting constructs were first
characterized in vitro against their specific purified enzymes, and the most promising ones tested in
cellulo. Thus, we were able to tag HeLa cells expressing LecuineAminoPeptidase (Figure 2) with
micrograins of fluorescent solid.[4]
References:
[1] E. Boonacker, C. J.F. Van Noorden J. Histochem. Cytochem. 2001, 49, 1473
[2] V.B. Paragas, J.A. Kramer, C. Fox, R.P. Haugland, V.L. Singer J. Microscopy 2002, 206, 106
[3] O. Thorn-Seshold, M. Vargas-Sanchez, S. McKeon, J. Hasserodt Chem. Commun. 2012, 48, 625
[4] M. Prost, L. Canaple, J Samarut, J. Hasserodt, Submitted
Difluoromethylbenzoxazole pyrimidine thioether (DFMB) derivatives as
novel non-nucleoside HIV-1 reverse transcriptase inhibitors
a)
J. Boyer,a) A.-M. Menot,a) R. Terreux,b) E. Arnoult,c) J. Unge,d) D. Jochmans,e) J.
Guillemont,c) M. Médebiellea)*
Université Claude Bernard Lyon 1 (UCBL), Institut de Chimie et Biochimie Moléculaire et Supramoléculaire
(ICBMS), UMR CNRS – UCBL – INSA Lyon 5246, Equipe « Synthèse de Molécules d’Intérêt Thérapeutique
(SMITH) », 43 bd du 11 Novembre 1918, Villeurbanne, France
b)
Université Claude Bernard Lyon 1 (UCBL), Laboratoire B.I.S.I, UMR CNRS - UCBL 5086, Equipe « Bases
Moléculaires et Structurales des Systèmes Infectieux (BMSSI) », Lyon, France
c)
Chemistry Lead Antimicrobial Research, Janssen-Cilag/Tibotec, Campus de Maigremont BP615, Val de Reuil
Cedex, France
d)
MAX-Lab, Lund University, P.O. Box 118, Lund, Sweden
e)
Tibotec-Virco BVBA, A division of Janssen Pharmaceutical Companies of Johnson & Johnson,
Turnhoutseweg 30, Beerse, Belgium
*[email protected]
This presentation reports our past and current efforts towards the synthesis and
antiviral properties of new difluoromethylbenzoxazole (DFMB) pyrimidine thioether
derivatives as non-nucleoside HIV-1 reverse transcriptase inhibitors. By use of
combination of structural biology study, docking and traditional medicinal chemistry,
several members of this novel class were synthesized using single electron transfer chain
process (radical nucleophilic substitution, SRN1) and were found to be potent against wildtype HIV-1 reverse transcriptase, with low cytotoxicity but with moderate activity against
resistant drug-resistant strains. One promising compound DFMB2 showed a significant
EC50 value close to 6.4 nM against wild-type IIIB, a moderate EC50 value close to 54 µM
against resistant double mutant (K103N + Y181C) but an excellent selectivity index >
15477 (CC50 > 100 µM) (Figure 1) [1].
Figure 1. Difluoromethylbenzoxazole (DFMB) pyrimidine thioethers as novel NNRTIs
Optimisation of these molecules toward activity on NNRTI-resistant HIV are now
being pursued, with other modifications on the benzoxazole and pyrimidine rings based on
our structural biology, current and new antiviral data and new molecular docking studies.
References
[1] J. Boyer, E. Arnoult, M. Médebielle, J. Guillemont, J. Unge, D. Jochmans, J. Med.
Chem., 54. (2011) 7974-7985.
Gscore, a Robust Cell-by-cell Score for Sensitive and Specific
Hit Discovery in High Content Screening
Laurent GUYON1,2,3, Christian LAJAUNIE4,5,6, Frédéric FER1,2,3, Ricky BHAJUN1,2,3, Mélissa MARY1,2,3, Eric
SULPICE1,2,3, Guillaume PINNA7, Anna CAMPALANS8, J. Pablo RADICELLA8, Stéphanie COMBE1,2,3, Patricia
OBEID1,2,3, Jean-Philippe VERT4,5,6, Xavier GIDROL1,2,3
1
2
CEA, Laboratoire BGE, iRTSV, 17 rue des Martyrs, F-38054 Grenoble cedex 9, France
3
4
Université Grenoble-Alpes, F-38000 Grenoble, France
INSERM, U1038, F-38054 Grenoble cedex 9, France
Centre for Computational Biology - CBIO, Mines ParisTech, 35 rue Saint-Honoré, Fontainebleau, F-77300 France
5
Institut Curie, 26 rue d'Ulm, Paris, F-75248 France
6
7
8
INSERM, U900, Paris, F-75248 France
CEA, Plateforme ARN interference PArI, F- 91191 Gif-sur-Yvette, France
CEA, Institute of Cellular and Molecular Radiobiology, F-96265 Fontenay aux Roses, France
[email protected]
Keywords: High Content Screening (HCS), scoring, hit finding, False Positive reduction.
High Content Screening (HCS) has enabled great advances both in oncology and biology. It
consists in visualizing phenotypes modification of cells after perturbation. This perturbation is
generally achieved either by chemical compounds or RNA interference in a highly parallel manner
(in 384 well-plates for example). HCS experiments produce huge amount of data, typically tables of
millions of rows and tens of columns; each row corresponding to a cell whose phenotype is
characterized with different metrics (each column). After analysis, hits, which are the biggest
modifiers of the cell phenotype, are extracted.
A method of choice, Zscore, which averages the fluorescence for each cell modified by a given
compound, has proven great efficiency when fluorescence is used to monitor the phenotype.
However it faces a few drawbacks:
1. Zscore is very sensitive to artifacts (aberrant fluorescence of just one cell will affect the score)
2. Low cell number for a given treatment will strongly increase Zscore variability
3. Zscore is associated to a pValue only when fluorescence distribution is Gaussian
In situations where the consequences of perturbation are strong and overtake such “artifacts”,
Zscore find hits with a low False Discovery Rate. However in conditions closer to the physiological
reality (such as rare cells or cells from patient), one need to overcome these pitfalls. Thus, in an
attempt to improve the potential of discovery of HCS, we developed a so called Gscore, which is
based on the rank of the fluorescence and takes into account the number of cells in a given
treatment with an appropriate model.
I will show the advantages of the score compared to the widely used Zscore in various situations
using virtual screen and real screening data, including a screen of gene knock-down reagents
(druggable collection, targeting more than 7000 genes).
CHEMICAL SYNTHESIS AND IN CELLULO TUBULIN
POLYMERIZATION INHIBITION EVALUATION: A WINNING
COMBINATION FOR THE DISCOVERY OF NEW ANTICANCER
AGENTS, DERIVING FROM COMBRETASTATIN A-4
Thierry Lombergeta, Cong Viet Doa, Caroline Baretteb, Thi Thanh Binh Nguyena, MarieOdile Fauvarqueb, Evelyne Colombc, Marek Haftekc and Roland Barreta
a
Université de Lyon, Université Lyon 1, Faculté de Pharmacie - ISPB, EA 4446
Biomolécules, Cancer et Chimiorésistances, SFR Santé Lyon-Est CNRS UMS3453 INSERM US7, 8 avenue Rockefeller, F-69373 Lyon cedex 08, France
b
iRTSV - LBGE - Gen&Chem - Centre de Criblage des Molécules Bio-Actives - CMBA
U1038 INSERM/CEA/UJF CEA Grenoble 17, rue des Martyrs, 38054 Grenoble Cedex 09
c
Université Lyon 1, Faculté de Médecine et de Pharmacie, EA 4169 « Fundamental, clinical
and therapeutic aspects of the skin barrier », 8 avenue Rockefeller, F-69373 Lyon cedex 08
Combretastatin A-4 (CA-4) is a natural cis stilbenoid, first isolated from the south african
willow tree Combretum caffrum.1 This compound have shown remarkable antiproliferative
activities against cancer cells, by inhibiting tubulin polymerization; some water-soluble
derivatives currently undergo clinical studies.
Heterocyclic analogs 12 and 2 having thiophenes or benzo[b]thiophenes instead of B ring
were prepared and evaluated for their in cellulo tubulin polymerization inhibition3 and their
antiproliferative activities. The presence of the sulfur benzoheterocycle proved to have a
crucial effect since all the thiophene derivatives were not active. The influence of the
attachment position will also be presented: benzo[b]thiophenes having iso-vinylene (i.e.
isocombretastatin analogs) or cis-methylene 3,4,5-trimethoxybenzenes at position 2 were
more active than the 3-regioisomers.
3
2
MeO
MeO
A
B
OMe
CA-4
OH
OMe
3
MeO
OMe
S
MeO
OMe
1
2
S
MeO
OMe
2
1
Pettit, G. R.; Singh. S. B.; Hamel, E. ; Lin, C. M. ; Alberts, D. S.; Garcia-Kendall, D. Experientia
1989, 45, 209-211.
2
Nguyen, T. T. B.; Lomberget, T.; Tran, N. C.; Colomb, E.; Nachtergaele, L.; Thoret, S.; Dubois, J.;
Guillaume, J.; Abdayem, R.; Haftek, M.; Barret, R. Bioorg. Med. Chem. Lett. 2012, 22, 7227-7231.
3
Vassal, E.; Barette, C.; Fonrose, X.; Dupont, R.; Sans-Soleilhac, E.; Lafanechère, L. J. Biomol.
Screening 2006, 11, 377-389.
Treating yeast infections with new innovative chromatin targets
Morgane Champleboux1, Flore Mietton1, Elena Ferri4, Didier Spittler2, Muriel Cornet3,
Charles McKenna4, Carlo Petosa2 and Jérôme Govin1
1. Institute of Research in Life Sciences and Technologies, Department Large Scale Biology,
17 Rue des Martyrs, 38054 Genoble Cedex 9
2. Institut de Biologie Structural, 41, rue Jules Horowitz, 38027 Grenoble Cedex 1
3. Laboratoire TIMC-TheREx Domaine de la Merci, 38706 La Tronche
4. University of Southern California, Department of Chemistry, 3620 McClintock Avenue,
Los Angeles, CA 90089-1062 USA
C. albicans is the most prevalent human fungal pathogen and is responsible for the
most deaths. With only four drug classes available to treat invasive fungal infection, there is
an urgent need to find new therapeutic agent, to overcome the emergence of drug-resistant
strains, problems related to the toxicity and narrow activity spectrum of existing drug.
Bromodomain proteins are chromatin-associated factors that regulate gene
transcription and chromatin remodelling. They recognize short peptides acetylated on lysine
residues, and are involved in many processes like cancer development, infection and
reproduction. Recently, several efforts of academic groups and biopharmaceutical companies
have led to the discovery of several potent and selective human bromodomain inhibitors, with
promising outcomes in cancers and human pathologies.
This project explores the functional role of bromodomain proteins in C. albicans, and
their potential as therapeutic targets. We investigate their role in C. albicans biology, and
develop an ambitious program to identify yeast-specific bromodomain inhibitors.
.
Title : STLC-­‐resistant cell lines as tools to classify chemically divergent Eg5 targeting agents according to their mode of action and target specificity Isabel Garcia-­‐Saez, Salvatore DeBonis, Rose-­‐Laure Indorato, Françoise Lacroix, Dimitrios Skoufias Institut de Biologie Structurale, UMR5075 (CNRS-­‐CEA-­‐UJF). Grenoble 38027, France The microtubule based kinesin Eg5, also known as KIF11, has been recognized as a valid target for the development of new class of anti-­‐mitotic inhibitors targeting components of the mitotic spindle with a potential cancer chemotherapeutic value (1). To date, a number of chemically distinct small molecules targeting different binding pockets of Eg5 protein are under evaluation in clinical trials with better responses achieved when hematological malignancies were targeted. One of the Eg5 inhibitors, ARRY-­‐520 is set to enter late-­‐stage clinical testing in several hundred human patients with relapsed or refractory multiple myeloma. We have been addressing the issue of specificity and resistance to Eg5 inhibitors and in particular to S-­‐Trityl_L-­‐Cysteine (STLC) that we have previously identified based on enzymatic screening. Based on the structure of the STLC-­‐Eg5 complex, we have been successful in developing drug resistant cell lines as tools to classify chemically divergent Eg5 targeting agents according to their mode of action and target specificity (2,3). We have also identified Eg5 mutants encountered in a number of selected drug resistant clones of colon carcinoma tumor cells and we are carrying out structure activity studies in one of such mutant. Interestingly, this mutation, instead of being localized in the known allosteric drug-­‐binding pocket of Eg5, is in the nucleotide-­‐binding site. Our current biochemical data coupled with cell-­‐based assays support the hypothesis that the mutant, in the absence of the inhibitor, is in a rigor state (high friction mode) and free of nucleotide. When the mutant is expressed in cells, heavily crosslinked MTs are formed. Strikingly, the MT bundles are released in the presence of the inhibitor. The drug resistant cell lines can therefore be used as a filter to distinguish Eg5 loop L5 binding drugs without prior structural information. Additionally, the cells can be used to analyze whether inhibitors of Eg5 are specific to this potential drug target or whether they bind to additional protein targets in dividing cells. One additional outcome of our results is the proposal of a double hit strategy for the same target protein, e.g. an exposure of tumor cells to a combination of ATP competitive and an ATP uncompetitive Eg5 targeting drugs as a possible treatment strategy to minimize or slow the development of drug resistance due to mutations in one of the drug binding sites. (1) Good JA, Skoufias DA, Kozielski F. Elucidating the functionality of kinesins with small molecule probes. 2011 Seminars in Cell and Developmental Biology 22(9): 935-­‐945. (2) Tcherniuk S, R van Lis, F Kozielski, DA Skoufias. 2010 Mutations in the human kinesin Eg5 that confer resistance to monastrol and S-­‐Trityl-­‐L-­‐Cysteine in tumor derived cell lines. Biochemical Pharmacology 79(6):864-­‐72. (3) Indorato RL, DeBonis S, Kozielski F, Garcia-­‐Saez I, Skoufias DA. STLC-­‐resistant cell lines as tools to classify chemically divergent Eg5 targeting agents according to their mode of action and target specificity Biochem Pharmacol. 2013 Nov 15;86(10):1441-­‐51. Poster abstracts
Drug Screening : from Phenotypes to Molecular Modeling
Screening of new anti-virulence molecules by targeting iron metabolism in bacteria
Aynur Ahmadova1, Caroline Barette2, Marie-Odile Fauvarque2, Sophie Mathieu1, Julien
Perard1, Cheickna Cissé1, Mohamed Ould Abeih1, Eve de Rosny3, Jacques Covès3, Serge
Crouzy1 and Isabelle Michaud-Soret1
[email protected], [email protected]
1
CEA, iRTSV, LCBM (Chemistry and Biology of Metals Laboratory), 38054, Grenoble, France
CNRS, UMR 5249, 38000, Grenoble, France
Univ. Grenoble Alpes, UMR 5249, 38000, Grenoble, France
2
CEA, iRTSV, BGE, GEN&CHEM, CMBA (Center of screening for BioActive Molecules), 38054, Grenoble,
France
INSERM, U1038, 38000, Grenoble, France
Univ. Grenoble Alpes, UMR_S 1038, 38000, Grenoble, France
3
Institut de Biologie Structurale, UMR 5075 CNRS-CEA-UJF-Grenoble-1, 6, rue Jules Horowitz, 38000
Grenoble,France
New antimicrobials targeting virulence traits in bacteria could offer a number of
advantages over the conventional antibiotics, such as preserving the host endogenous
microbiota and exerting less pressure on bacterial survival, which may result in decreased
resistance. Our objective is to develop new anti-virulence molecules targeting FUR protein
(Ferric Uptake Regulator), a global transcriptional regulator that senses iron status and
controls the expression of genes involved in iron homeostasis, virulence and oxidative stress.
Ubiquitous in Gram negative bacteria (and in some Gram positive) and absent in eukaryotes,
FUR is an interesting anti-virulence target, as iron acquisition is one of the major virulence
determinants of bacteria through synthesis and secretion of siderophores and toxins.
Moreover, the inactivation of fur gene in various pathogens leads to decrease of their
virulence [1].
By applying peptide aptamers technology we identified four molecules (F1-F4)
interacting with E. coli FUR and able to decrease pathogenic E. coli strain virulence in a fly
infection model [2]. These combinatory molecules are constituted from variable 13-aminoacid peptide loop attached at both ends to a scaffold protein (thioredoxin A from E. coli).
Furthemore, the aptamer F4 was shown to interact with FUR proteins from other pathogens in
yeast two-hybrid assay.
We performed the high-throughput screening of 17,680 chemical compounds from
Prestwick and ChemBridge libraries, applying an automated luminescence LexA-based yeast
two-hybrid test, in order to identify molecules able to disrupt FUR-peptide aptamer complex
and interact with FUR from Pseudomonas aeruginosa, Fransicella tularensis and E. coli. We
suppose that peptide aptamers and small molecules binding to the same molecular surface on
FUR protein should trigger the same biological effects, and thus such small molecules will be
potential inhibitors of FUR protein.
From Prestwick library one compound active on the FUR from Pseudomonas
aeruginosa was identified. Upon analysis of hits from ChemBridge library we retained 33
compounds with good specificity profile (i.e. molecules that produced inhibitions of unrelated
protein-protein interaction were discarded). The in-vitro and in-vivo studies of FUR proteins
inhibition by selected compounds are in progress and results will be presented.
[1] Wang et al., Vaccine 27, 2009
[2] Abed, N. et al. (2007) Mol Cell Proteomics 6, 2110-21
Title : STLC-­‐resistant cell lines as tools to classify chemically divergent Eg5 targeting agents according to their mode of action and target specificity Isabel Garcia-­‐Saez, Salvatore DeBonis, Rose-­‐Laure Indorato, Françoise Lacroix, Dimitrios Skoufias Institut de Biologie Structurale, UMR5075 (CNRS-­‐CEA-­‐UJF). Grenoble 38027, France The microtubule based kinesin Eg5, also known as KIF11, has been recognized as a valid target for the development of new class of anti-­‐mitotic inhibitors targeting components of the mitotic spindle with a potential cancer chemotherapeutic value (1). To date, a number of chemically distinct small molecules targeting different binding pockets of Eg5 protein are under evaluation in clinical trials with better responses achieved when hematological malignancies were targeted. One of the Eg5 inhibitors, ARRY-­‐520 is set to enter late-­‐stage clinical testing in several hundred human patients with relapsed or refractory multiple myeloma. We have been addressing the issue of specificity and resistance to Eg5 inhibitors and in particular to S-­‐Trityl_L-­‐Cysteine (STLC) that we have previously identified based on enzymatic screening. Based on the structure of the STLC-­‐Eg5 complex, we have been successful in developing drug resistant cell lines as tools to classify chemically divergent Eg5 targeting agents according to their mode of action and target specificity (2,3). We have also identified Eg5 mutants encountered in a number of selected drug resistant clones of colon carcinoma tumor cells and we are carrying out structure activity studies in one of such mutant. Interestingly, this mutation, instead of being localized in the known allosteric drug-­‐binding pocket of Eg5, is in the nucleotide-­‐binding site. Our current biochemical data coupled with cell-­‐based assays support the hypothesis that the mutant, in the absence of the inhibitor, is in a rigor state (high friction mode) and free of nucleotide. When the mutant is expressed in cells, heavily crosslinked MTs are formed. Strikingly, the MT bundles are released in the presence of the inhibitor. The drug resistant cell lines can therefore be used as a filter to distinguish Eg5 loop L5 binding drugs without prior structural information. Additionally, the cells can be used to analyze whether inhibitors of Eg5 are specific to this potential drug target or whether they bind to additional protein targets in dividing cells. One additional outcome of our results is the proposal of a double hit strategy for the same target protein, e.g. an exposure of tumor cells to a combination of ATP competitive and an ATP uncompetitive Eg5 targeting drugs as a possible treatment strategy to minimize or slow the development of drug resistance due to mutations in one of the drug binding sites. (1) Good JA, Skoufias DA, Kozielski F. Elucidating the functionality of kinesins with small molecule probes. 2011 Seminars in Cell and Developmental Biology 22(9): 935-­‐945. (2) Tcherniuk S, R van Lis, F Kozielski, DA Skoufias. 2010 Mutations in the human kinesin Eg5 that confer resistance to monastrol and S-­‐Trityl-­‐L-­‐Cysteine in tumor derived cell lines. Biochemical Pharmacology 79(6):864-­‐72. (3) Indorato RL, DeBonis S, Kozielski F, Garcia-­‐Saez I, Skoufias DA. STLC-­‐resistant cell lines as tools to classify chemically divergent Eg5 targeting agents according to their mode of action and target specificity Biochem Pharmacol. 2013 Nov 15;86(10):1441-­‐51. Screening for bioactive molecules at the CMBA platform
Emmanuelle Soleilhac1, Caroline Barette1, Magda Mortier1, Catherine Pillet1, Laurence Lafanechère2
et Marie-Odile Fauvarque1
1
Université Grenoble-Alpes, F-38000 Grenoble; CEA-DSV-iRTSV-BGE-Gen&Chem-CMBA, F38054 Grenoble; INSERM, U1038, F-38054 Grenoble, France.
2
Institut Albert Bonniot, CRI, INSERM/Université Grenoble-Alpes U823, équipe Polarité,
développement et cancer, F-38706 La Tronche Cedex, France.
Bioactive molecules are chemicals modifying the activity of a biological target in vitro or in vivo.
Such molecules are critical for both drug discovery and the development of chemical tools to study
protein function in a temporal and putatively reversible manner. On the one hand, molecules selected
from biochemical or enzymatic in vitro assays are used to disrupt full, or part, of protein activity. On
the other hand, phenotypic screening based on in cellulo assays greatly enhances the probability to
select molecule active in a living organism. Moreover, subsequent determination of their target can
provide new information on proteins and signalling pathways regulating physiological or pathological
processes such as cell growth and differentiation, inflammation, cancerogenesis… The CMBA
screening platform is an open facility managing about 30,000 compounds which helps in developing
robust miniaturized assays and performs large scale automated screens (HTS). The CMBA also
develops high content analysis (HCA) of complex cell phenotypes by automated microscopy. This
methodology is typically used for the high content screening (HCS) of small collection of structurally
or functionally related molecules. Beside our activity of service, we aim at searching for molecules
specifically interfering with members of the deubiquitinating enzymes family (DUBs) which
mutations are associated with various diseases including chronic inflammation, cancer and
neurodegeneration. To date, most of the hundred known DUBs have unknown substrates and only a
handle of molecules targeting DUBs have been published. We address this issue by combining
genetics, cell biology and chemogenomics approaches.
Ongoing collaborative projects at the CMBA platform
Caroline Barette1, Emmanuelle Soleilhac1, Magda Mortier1, Catherine Pillet1, Stéphane Ségard2,
Céline Charavay2, Marie-Odile Fauvarque1 and collaborators
1
Université Grenoble-Alpes, F-38000 Grenoble; CEA-DSV-iRTSV-BGE-Gen&Chem-CMBA, F38054 Grenoble; INSERM, U1038, F-38054 Grenoble, France.
2
Université Grenoble-Alpes, F-38000 Grenoble; CEA-DSV-iRTSV-BGE-GIPSE, F-38054 Grenoble;
INSERM, U1038, F-38054 Grenoble, France.
This poster will briefly present ongoing screening projects with members of academic (or private)
laboratories. It will emphasize the fact that CMBA activity covers a large number of different research
fields in BioEnergy, Plant cell biology, Cancer or Infectiology (anti-bacterial, anti-viral or antifungal). In these projects, members of the platform provide their expertise in the setting of extremely
various kinds of experimental in vitro or in cellulo assays adapted for HTS (High-Throughput
Screening) or HCS (High-Content Screening). In addition, data analysis takes advantage of an inhouse computing program, called TAMIS, that has been developed in close partnership with the
GIPSE.
Magnetogenic small-­‐molecule probes for the detection of (bio-­‐)chemical analytes Fayçal Touti, Jacek Kolanowksi, Corentin Gondrand, Jens Hasserodt Chemistry Laboratory, University of Lyon – ENS, Lyon, France Binary ferrous complexes can be made to respond to an analyte exercising a chemical or catalytic reactivity by [1]
changing their electronic spin from zero to two. This effectively results in the sample developing a significant magnetic susceptibility by an off-­‐on mode. Our new line of magnetogenic probes operate at physiological conditions [2]
(water and pH 7), an important advance also in the field of coordination chemistry. Their binary response (total conversion) relies on the sping-­‐loaded quality of their aminal-­‐based pendant arm, i.e. in an energy-­‐rich molecular moiety whose fragmentation is inhibited by a blocking group on its periphery that can react with the targeted chemical analyte. We have demonstrated this to work with a chemical reactant (H2, Pd/C), and two enzymes: penicillin [2] amidase and nitroreductase. We also present for the first time a new pair of bispidine ferrous complexes that have [3]
a true magnetic off-­‐on relationship and that will serve for the design of an alternative magnetogenic probe system. We are in fact the first to report a low-­‐spin iron(II) complex (four in total) based on the bispidine platform. [1] (a) J. Hasserodt; J. L. Kolanowski; F. Touti “Magnetogenesis in Water Induced by a Chemical Analyte”, invited review article, Angew. Chem. Int. Ed. 2014, 53, 60-­‐73; (b) J. Hasserodt “Contrast Agents for Magnetic Resonance Imaging“ WO2005094903 (2005); (c) V. Stavila, M. Allali, L. Canaple, Y Stortz, C. Franc, P. Maurin, O. Beuf, O. Dufay, J. Samarut, M. Janier, J. Hasserodt New J. Chem. 2008, 32, 428-­‐435; (d) L. Canaple, O. Beuf, M. Armenccan, J. Hasserodt, J. Samarut, M. Janier “Fast Screening of Paramagnetic Molecules in Zebrafish Embryos by MRI” NMR in Biomed. 2008, 21, 129-­‐137; (e) Touti, F., Singh, A. K., Maurin, P., Canaple, L., Beuf, O., Samarut, J., Hasserodt, J., J. Med. Chem. 2011, 54, 4274–4278 ; (f) Touti, F., Maurin, P., Canaple, L., Beuf, O., Hasserodt, J., Inorg. Chem. 2012, 51, 31–33. [2] F. Touti, P. Maurin, J. Hasserodt, “Magnetogenesis under physiological conditions with probes that report on (bio-­‐)chemical stimuli”, Angew. Chem. Int. Ed. 2013, 52, 4654-­‐4658. [3] J. Kolanowski, E. Jeanneau, R. Steinhoff, J. Hasserodt, “Bispidine Platform Grants Full Control over Magnetic State of Ferrous Chelates in Water”, Chem. – Eur. J. 2013, 8839-­‐8849. Toward the discovery of the first small molecule Protein-Protein Interaction
Inhibitor for Casein Kinase 2
Benoît Bestgena,c, Irina Kufarevab, Renaud Prudentc, Ruben Abagyanb, Claude Cochetc, Matthias
Engeld, Thierry Lombergeta and Marc Le Borgne a.
a
Université de Lyon, Université Lyon 1, Faculté de Pharmacie - ISPB, EA 4446 Biomolécules, Cancer et
Chimiorésistances, SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, 8 avenue Rockefeller, F-69373 Lyon
cedex 08, France
b
The Scripps Research Institute, 10550 N Torrey Pines Rd., La Jolla, CA 92037, USA.
c
Institut National de la Santé et de la Recherche Médicale, U873, Transduction du Signal, Commissariat à l’Energie
Atomique, 17 rue des Martyrs, Grenoble, F-38054, France.
d
Pharmaceutical and Medicinal Chemistry, Saarland University, P.O. Box 151150, D-66041 Saarbrücken, Germany
Protein kinases are key regulators of cell signaling and their inhibition is one of the major
interesting axes in the research for new cancer treatments. Casein Kinase 2 (CK2) is involved in
numerous cell pathways that promote cell survival, enhance anti-apoptotic signals, support the
neovascularization and stabilize the oncokinome. Moreover cancer cells are considered as “CK2addict” as a high level of CK2 expression is correlated with a bad prognosis for patients.1 For all
these reasons, CK2 is an important target for the development of new cancer chemotherapies.
CK2 is a unique and ubiquitous protein kinase composed of two catalytic (α or/and α’)
and two regulatory (β) subunits. CK2α is constitutively active but the complex with CK2β
changes the in vitro and in vivo substrate selectivity. Live cell imaging studies have demonstrated
that both subunits can move independently within living cells. Their dynamic association or
dissociation is a key element in the regulation of CK2-dependent cell pathway: an unbalanced
expression of α and β subunits have been observed in a large variety of tumor cells. A large
number of studies underlined the importance of the β regulatory subunit for cell viability,
embryonic development, epithelial to mesenchymal transition.2 An effective inhibitor of the α/β
interaction is also necessary to further investigate the in vivo role of the β subunit.
A cyclic peptide was previously described as the first β antagonist but no in cellulo
efficiency was observed.3 By using a virtual screening approach, followed by medicinal
chemistry work and completed with mechanistic studies, the first small molecule inhibitor of the
α/β protein/protein interaction was identified.
(1) Ruzzene, M. and al. Addiction to Protein Kinase CK2: A Common Denominator of Diverse Cancer Cells? Biochim. Biophys. Acta 2010, 1804, 499–504. (2) Deshiere, A. and al. Regulation of Epithelial to Mesenchymal Transition: CK2β on Stage. Mol. Cell. Biochem. 2011, 356, 11–20. (3) Laudet, B. and al. Structure-­‐Based Design of Small Peptide Inhibitors of Protein Kinase CK2 Subunit Interaction. Biochem. J. 2007, 408, 363. NAD biosynthesis in prokaryotes: A Target for antibacterial agents
Debora Reichmann, Alice Chan, Olivier Hamelin, Caroline Barette, Sandrine Ollagnier de
Choudens
Nicotinamide adenine dinucleotide (NAD) is an essential cofactor playing a crucial role in
several biological redox reactions1. In the NAD biosynthesis a common precursor exists
among all organisms, the quinolinc acid (QA). Interestingly the pathway to generate QA
differs between prokaryotes and eukaryotes. In most eukaryotes and some bacteria the
degradation of tryptophan occurs whereas in prokaryotes and plants L-aspartate and
dihydroxyacetone phosphate (DHAP) are involved. First, L-aspartate is converted by the Laspartate oxidase NadB (flavoprotein) to iminoaspartate (IA) followed by a condensation
reaction with DHAP to form QA, carried out by the quinolinate synthase NadA. In addition to
this de novo pathway most organisms are able to use a salvage pathway where NAD is
recycled. In contrast, for two pathogens Mycobacterium leprae and Heliobacter pylori no
such salvage pathway exists2. Therefore in these pathogens, proteins in the de novo pathway
are interesting targets for antibacterical agents. The quest for an inhibitor, efficiently in vivo
and in vitro, included high throughput screenings (HTS) and chemical production of product
analogs. The results obtained for both approaches will be presented. The inhibitor 4,5dithiohydroxyphthalic acid (DTHPA) was found to inactivate NadA both in vitro and in vivo3.
1
Begley T.P. et al., Vitam. Horm. 2001, 61 : 103-19
Gerdes S.Y. J Bacteriol 2002, 184: 4555-4572
3
Chan A. et al., Ang. Chem. 2012, 51 :7711-4
2
Non-nucleoside inhibitors of NS5B polymerase derived from the naturally
occurring aurones: potential agents against Hepatitis C virus infection
Meguellati, A.1; Peuchmaur, M.1; Haudecoeur, R.1 ; Ahmed-Belkacem, A.2; Brillet,
R.2; Pawlotsky, J.-M2,3 and Boumendjel, A.1
1
Université de Grenoble, CNRS UMR 5063, Département de Pharmacochimie Moléculaire,
Département de Pharmacochimie Moléculaire, BP 53; 38041 Grenoble - FRANCE
2
3
INSERM U955, Hôpital Henri Mondor, 94010 Créteil, FRANCE
Département de Virologie, Hôpital Henri Mondor, Université Paris-Est, 51 avenue du Maréchal de Lattre de
Tassigny, 94010 Créteil, FRANCE
E-mail: [email protected]
Hepatitis C virus (HCV) infection is a global public health problem. The World Health
Organisation (WHO) estimates that 170 million people are infected worldwide. The current
therapeutical treatments consist of a combination of pegylated interferon alpha (peg-IFN) and
Ribavirin (RBV). Unfortunately, the response rate is low, especially among patients infected
by HCV genotype 1, the most frequent genotype.
The HCV RNA-dependent RNA polymerase NS5B constitutes an interesting target
because of its key role in viral replication and being not functional in mammalian cells.We
recently identified through a screening process that the naturally occurring 2benzylidenebenzofuran-3-ones, namely (aurones) as new inhibitors of NS5B. The aurone
active site, identified by site-directed mutagenesis, is located in Thumb Pocket I of HCV
RdRp. Molecular docking studies were used to determine how aurones bind to NS5B and to
predict their range of inhibitory activity. Several aurones were found to have potent inhibitory
effects on HCV RdRp, with excellent selectivity index (inhibition activity versus cellular
cytotoxicity). More very recent promising results obtained on aurones dimers will be
presented. The potent NS5B inhibitory activity combined with their low toxicity make
aurones attractive drug candidates against HCV infection.
OH
OH
HO
O
OH
O
aurone
aurone structure docked in the
Thumb Pocket I of NS5B
[1] Haudecoeur, R. et al.J. Med. Chem. 2011, 54, 5395-402.
Contact imaging plate reader for parallelized time-lapse screening
Vincent Haguet1,2,3, Itebeddine Ghorbel1,2,3, Xavier Gidrol1,2,3
1
CEA, DSV, iRTSV, BGE, 17 rue des Martyrs, 38054 Grenoble Cedex 9, France
INSERM, U1038, Grenoble, France
3
UJF-Grenoble 1, Grenoble, France
2
We developed a small-size plate reader providing time-lapse microscopy of live cell cultures in a
standard microtiter plate to achieve screens of cells inside a CO2 incubator. The imaging
instrument is based on a contact imaging architecture, i.e., the image of cells is numerically
recorded by an image sensor placed under the sample, in the absence of any intermediary
objective or other optical part. An array of 96 image sensors were arranged on a printed circuit
board so that each well of a flat-bottomed 96-well microtiter plate can be imaged by the image
sensor placed underneath (Figure 1). The resulting images of the cultures are 2.8x2.1-mm large
with 1.75-µm pixel-size resolution, which offers a very large field of view of the cells with a ~10x
magnification.
Time-lapse image acquisitions of cell cultures reveal dynamics of cell behavior, such as the
number of cells as well as the position and shape of every cell at every time point. Screening the
evolution of these parameters allows investigating the dynamic effect of drugs or the environment
on cellular mechanisms, optimizing the culture conditions, as well as potentially uncovering
previously unknown behavior of the cell line.
We recently realized with the plate reader a test screen for cell migrations using 96 wound
healing assays (Figure 2). Dynamics of the wound closures were obtained by parallelized timelapse contact imaging microscopy and specifically developed automated image analysis. The
performance of global segmentation was validated on a set of images showing wounds in
confluent epithelial cell cultures. Automated wound localization was compared with manual
segmentation performed by seven cell biology experts by determining the root-mean-square error
between the segmented interfaces and region-oriented analysis. Evaluations of intra and interbiologist variabilities showed that automated segmentations are as accurate and robust as the
cell biologist’s ones.
B
A
Figure 1. Printed circuit board equipped with an array of
96 image sensors (A) to visualize cell populations in a
standard 96-well microtiter plate (B).
Figure 2. Global view of 96 “horizontal” wounds
formed in 96 cultures of prostate cancer cells
PC3 for a cell migration assay.
Lyon 1 University – ICBMS Compound Library:
Preservation and valorization of an academic chemical heritage
Arnaud Comte1
1
Lyon 1 University, CNRS, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS)
UMR5246, bat. Curien, 43 Bd du 11 Novembre 1918, 69622 Villeurbanne Cedex, France,
email : [email protected], website : www.icbms.fr/chimiotheque
The compound library aims to create added scientific value at the interface of chemistry and
biology from the wealth of chemically diverse compounds produced by the department
laboratories in the course of their research. It was created in 2001 and offers a collection of
nearly 3000 organic compounds with high structural diversity and drug like properties1, most of
them not being purchasable through chemicals suppliers. They are available in solid form or as
DMSO solutions, formatted in standard 96-well microplates suitable for manual or automated
screening.
Our work consists of collecting compounds from synthetic chemistry groups (mainly ICBMS
and ISPB, Institut des Sciences Pharmaceutiques et Biologiques de Lyon) and find
collaborations with both academic and industrial partners interested in particular biological
targets to screen the library. Promising hit compounds can lead to new pharmacological tools or
future drug candidates through structure optimization process, molecular modeling and
analogues synthesis.
Référence:
1
1. C.A. Lipinski , F. Lombardo, B.W. Dominy, P.J. Feeney; Adv. Drug. Deliv. Rev. 2001, 46:3-26.
SYNTHESIS AND BIOLOGICAL PROPERTIES OF MACROLACTAM ANALOGS OF THE NATURAL PRODUCT
MACROLIDE (-)-A26771B
Sophie Canova1, Renaud Lépine2, Amber Thys3, Anne Baron1, Didier Roche1
1 Edelris, 115 Avenue Lacassagne, 69003 Lyon, France; 2 Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France; 3 Galapagos NV, Generaal de Wittelaan L11 A3, 2800 Mechelen, Belgium
E-mail: [email protected]
Natural product modifications: the side chain
O
O
NH
O
O
O
O
O
LiOH
CCl3
O
NHBoc
O
O
88%
O
O
suffers from poor pharmacokinetic properties which translates into a
O
O
2
1
R1
4, R 1= Me, 35% (2 steps)
3
unstable
lack of in vivo activity.
NHBoc
48%
OM s
13
O
OH
OH
O
98%
O
O
O
O
O
CuI, C5 H 9MgBr
NHBoc
BocHN
been identified as an attractive target in the development of new
antibiotics [1]. Despite its attractive biological properties, (-)-A26771B
MsCl, Et3 N
OH
O
O
quant.
OH
O
(-)-A26771B (1)
17
quant.
1
>32
16
4
>32
16
>32
2
2
4
>32
2
16
8
2
>32
2
n.d.
>32
8
>32
2
0.5
8
>32
2
2
1
3
>32
2
8
16
1
>32
1
2
2
32
1
2
2
4
>64
2
2
32
4
>64
1
0.5
1
16
1
2
2
5
>64
4
2
32
4
>64
0.5
0.5
1
16
1
2
4
IV381-221
pneumoniae Pen9
ATCC49619
Streptococcus
pneumoniae
Streptococcus
aureus Oxford
+10%SHb
Staphylococcus
Staphylococcus
aureus ATCC25923
Staphylococcus
aureus ATCC25923
Staphylococcus
aureus Sa2 MRSAc
aureus ATCC13709
27853
Staphylococcus
Pseudomonas
aeruginosa ATCC
influenzae LS2
Efflux knock-out
31517
Haemophilus
influenzae ATCC
faecium 1
Haemophilus
faecalis
ATCC29212
ATCC 25922
Enterococcus
O
H
[1] Michel, K. H.; Demarco, P. V.; Nagarajan, R. J. Antibiot. 1977, 30, 571-575.
[2] (a) Hase, T. A.; Nylund, E. L. Tet. Lett. 1979, 28 , 2633-2636. (b) Tatsuta, K. ; Amemiya, Y. ; Kanemura,
Y. ; Kinoshita, M. Bull. Chem. Soc. Jpn. 1982, 55 , 3248-3253. (c) Trost, B.M. ; Brickner S. J. J. Am. Chem.
Soc. 1983, 105 , 568-575. (d) Bestmann, H. J.; Schobert, R. Angew. Chem. 1985, 97, 784-785. (e) Arai, K.;
Rawlings B. J. ; Yoshisawa, Y. ; Vederas, J. C. J. Am. Chem. Soc. 1989, 111, 3391-3399. (f) Quinkert, G.;
Kueber, F.; Knauf, W.; Wacker, M.; Koch, U.; Becker, H.; Nestler, H. P.; Duerner, G.; Zimmermann, G. Helv.
Chim. Act. 1991, 74, 1853-923. (g) Sinha, Subhash C.; Sinha-Bagchi, A.; Keinan, E. J. Org. Chem. 1993, 58 ,
7789-7796. (h) Kobayashi, Y.; Nakano, M.; Biju Kumar, G.; Kishihara, K. J. Org. Chem. 1998, 63, 75057515. (i) Nagarajan, M. Tet. Lett. 1999, 40, 1207-1210. (j) Kobayashi, Y.; Okui, H. J. Org. Chem. 2000, 65 ,
612-615. (k) Lee, W. W.; Shin H. J.; Chang, S. Tet. Asymm . 2001, 12, 29-31. (l) Gebauer, J.; Blechert, S. J.
Org. Chem. 2006, 71, 2021-2025.
© Copyright 2012 Galapagos NV
O
O
O
O
10
O
8
aureus Oxford
+10%SHb
Staphylococcus
Staphylococcus
aureus ATCC25923
Staphylococcus
aureus ATCC25923
aureus Sa2 MRSAc
Staphylococcus
aureus ATCC13709
27853
Staphylococcus
Pseudomonas
aeruginosa ATCC
influenzae LS2
Efflux knock-out
1
>32
16
4
>32
16
>32
2
2
4
>32
2
16
4
>64
2
2
32
4
>64
1
0.5
1
16
1
2
2
6
>64
>64
>64
>64
2
>64
1
2
1
16
1
2
1
7
>64
4
4
32
2
>64
1
2
1
32
1
4
2
8
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
9a
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
9b
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
10
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
>64
11b
>32
>32
>32
>32
>32
>32
>32
>32
>32
>32
>32
>32
>32
12
>32
4
4
>32
n.d.
>32
2
2
4
>32
2
4
2
IV381-221
pneumoniae Pen9
ATCC49619
pneumoniae
16
Streptococcus
Streptococcus
aureus Oxford
+10%SHb
Staphylococcus
Staphylococcus
aureus ATCC25923
Staphylococcus
Staphylococcus
27853
Staphylococcus
Pseudomonas
aeruginosa ATCC
influenzae LS2
Efflux knock-out
31517
Haemophilus
influenzae ATCC
faecium 1
Enterococcus
faecalis
ATCC29212
ATCC 25922
Enterococcus
>32
16
4
>32
16
>32
2
2
4
>32
4
>64
2
2
32
4
>64
1
0.5
1
16
1
2
2
>64
2
0.5
2
0.25
>64
1
1
1
8
1
2
8
>64
2
0.5
2
0.25
>64
0.5
0.5
1
8
0.5
2
8
21b
2
8
1
21a
Table 3. The overall MIC profile has been improved compared to both the macrolactone analog
and the natural product.
9a, 9b, X = O (30%)
11b, R = Ac
influenzae ATCC
Haemophilus
faecalis
ATCC29212
ATCC 25922
Enterococcus
BACTERIAL STRAIN
8, X = CH 2 (14%)
11a, R = H
Ac2 O, Et3 N
46% (2 steps)
31517
12
m-CPBA, DCM
85%
aureus ATCC25923
O
X
O
IV381-221
O
O
O
O
O
O
O
7, X = SO2
TBHP, Triton B
PhMe
4%
pneumoniae Pen9
O
O
O
6, X = S
BACTERIAL STRAIN
1. MeLi, CuI, -40°C
2. PhSeBr, -40°C- rt
3. H2O2, pyridine
(separated by flash chromatography)
O
Me3 SOI, DBU, CH 3CN
or
Streptococcus
O
O
21a and 21b
O
4
O
R
Escherichia. coli
N
O
H
N
PhO
O
O
O
1
O
via synthesis of lactam derivatives to improve the metabolic
References
18
H
N
X
O
NH2
faecium 1
O
H
N
O
56%
OH
62%
Enterococcus
N
O
O
PhO
NH
The goal of the project was to explore further the structure-activity
stability.
19
1. TFA
2.HATU, HOBt, DIPEA, DMAP
25%
PhSH, DCM
O
O
2
and access the first analogs,
O
20
Escherichia. coli
O
O
O
Objective
O
1. LiOH
2. MeI, Ag2 O
35% (2 steps)
88%
O
O
O
ATCC49619
O
O
CCl 3
pneumoniae
O
Streptococcus
NH
O
Haemophilus
O
and potentially toxicological perspective (general Michael acceptor).
via a semisynthetic strategy to replace the reactive functionality
O
O
Natural product modifications: the conjugated system
functionality could be a risk from a chemical and metabolic stability
biological profile and pharmacokinetic properties :
O
66%
quant.
O
O
O
anticipated that the sensitivity of the 4-oxygenated 2-enoic carboxyl
relationship (SAR) in order to identify candidates with improved
BocHN
1. NBS, NaHCO3
2. pyridine
O
HO
possessing a highly oxidized γ-oxo-δ-hydroxy-αβ-unsaturated carboxyl
covered [2], limited SAR was available from literature data. We
BocHN
NaClO2
O
H 2, Pd/C
AcOEt
97%
Table 1. Activity was retained whatever the modification performed on the side chain.
(-)-A26771B is a structurally unique 16 member ring macrolactone
system. Although the total synthesis of (-)-A26771B has been well
16
BocHN
Haemophilus
O
Enterococcus
BACTERIAL STRAIN
O
Escherichia. coli
O
O
O
15
O
O
83%
OMe
NaH, MeI
O
H
5, R 1= Ac, 46% (2 steps)
14
OH
C 5H 9MgBr
Grubbs I (15 m mol%)
ratio 14/16 = 1/5
aureus Sa2 MRSAc
Natural product (-)-A26771B produced by Penicillium turbatum has
Synthesis of Lactam analogs
MeI, Ag2O
or
Ac2O, pyridine
aureus ATCC13709
Introduction
Table 2. The modification of the conjugated system was found to be detrimental for the biological
profile of this natural product family
Conclusion
To conclude, we achieved the synthesis of various analogs of natural product (-)-A26771B. The SAR has
been established and emphasizes the role of the sensitive 4-oxygenated 2-enoic carboxyl functionality in
the antibiotic activity. The first synthesis of macrolactam analogs was achieved via a new cross-metathesis
approach. These compounds revealed a more pronounced antibacterial activity and an improved
metabolic stability as compared to the natural product (-)-A26771B.
Alternative splicing and resistance to cancer targeted therapies
Benoit-Pilven C.1,*, Rey A.1,*, Tranchevent LC.1, Mortada H.1 , Chautard E.1 , Neil-Bernet H.1 ,
Corbo L.1 , Eymin B.2 , and Auboeuf D.1
1
2
Cancer Research Centre of Lyon, Lyon, France
Institut Albert Bonniot, Grenoble, France
Targeted therapies are commonly used to treat cancer but they often fail due to resistance to
the treatment of some tumours. Resistance can happen via different mechanisms, and alternative
splicing appears to be one of them. Indeed, alternative splicing is the process of creating distinct
proteins from a single gene, and recent reports indicate that therapeutic targets often produce
isoforms that do not respond to the targeted therapy.
We propose to better define the role of alternative splicing in cancer drug resistance with a
systems biology approach and experimental validations on breast and lung cancer cell lines. The
main objectives are to develop a computational method that will help users to analyse the role of
alternative splicing of therapeutic targets in resistance and to develop a proof-of-concept in cancer
cell lines by predicting which cell lines exhibit de novo resistance to a given treatment, owing to
alternative splicing.
The first outcome is a web interface freely available for users to analyse the therapeutic
target variants up to the protein level to assess the effect of splicing on protein domains and
therefore on function. Another outcome is an experimentally validated method to predict which cell
lines are resistant to which treatment.
This represents a first step towards the development of more advanced methods that will
take into account all possible alterations observed in human cancers in order to refine the population
stratification for clinical trials and possibly in the future modify the way cancer is treated with
multiple targeted therapies.
Drug Screening : from Phenotypes to Molecular Modeling
Villeurbanne, France, February 27, 2014
List of Participants
Nom participant
ACH
AGUIRRE
AHMAOVA
AUBOEUF
BARETTE
BARON
BELOEIL
BENOIT-­‐PILVEN
BESCOND
BESTGEN
BOUCINHA
BRAÏKI
CALA
CANOVA
CARRIQUE
CECCHINI
CHAMPLEBOUX
CHOW
CLEUZIAT
COMTE
CORNACIU
CORTEJADE
DANIELE
DE CHASSEY
DE CROZALS
DEJEAN
DELCROS
ETHEVE
ETIENNE
ETTOUATI
FAUVARQUE
FEDORYSHCHAK
GARCIA-­‐SAEZ
GHOSEZ
GONDRAND
GRENIER
GUILLIERE
GURAGOSSIAN
GUYON
HAGUET
HASSERODT
HENRARD
HOLOGNE
HOUSSET
IQBAL
JABOT
JOSEPH
LAFANECHÈRE
LANCELIN
LE BORGNE
LÉCINE
LOMBERGET
LUNEAU
LUNVEN
MARQUEZ
MÉDEBIELLE
MEGUELLATI
MIKAELIAN
MOTTO-­‐ROS
NGUYEN
PAILLIER
POLENA
PROST
REICHMANN
RENAUD
REXHEPAJ
REY
RIVALTA
ROBERT
ROCHE
RONOT
SKOUFIAS
SOLEILHAC
STEBE
STRAZEWSKI
SULPICE
TOMÉ
TRANCHEVENT
TRAORE
TROUSSICOT
VERNET
VIALLET
WALKER
YTRE-­‐ARNE
Prénom participant
Delphine
Clémentine
Aynur
Didier
Caroline
Anne
Laurent
Clara
Amandine
Benoît
Lilia
Anissa
Olivier
Sophie
Loic
Tiphaine
Morgane
Melissa
Philippe
Arnaud
Irina
Aurelie
Gaëlle
Benoit
Gabriel
Emmanuel
Jean-­‐guy
Loic
Christelle
Laurent
Marie-­‐odile
Roman
Isabel
Léon
Corentin
Benjamin
Florence
Nathalie
Laurent
Vincent
Jens
Denis
Maggy
Dominique
Muhammad
Claire
Benoît
Laurence
Jean-­‐marc
Marc
Patrick
Thierry
Dominique
Laurent
Jose
Maurice
Amel
Ivan
Vincent
Kim-­‐anh
Celine
Helena
Maxime
Debora
Prudent
Elton
Amandine
Ivan
Xavier
Didier
Xavier
Dimitrios
Emmanuelle
Pierre nicolas
Pierre
Eric
Catarina
Léon-­‐charles
Mohamed
Laura
Audrey
Jean
Olivier
Mari
E-­‐mail
ACH
[email protected]­‐lyon1.fr
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
olivier.cala@univ-­‐lyon1.fr
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
arnaud.comte@univ-­‐lyon1.fr
[email protected]
[email protected]
[email protected]
[email protected]
gabriel.de-­‐[email protected]­‐lyon1.fr
[email protected]
jean-­‐[email protected]
[email protected]
[email protected]
laurent.ettouati@univ-­‐lyon1.fr
[email protected]
roman.fedoryshchak@ens-­‐lyon.fr
[email protected]
[email protected]­‐bordeaux.fr
corentin.gondrand@ens-­‐lyon.fr
[email protected]
florence.guilliere@univ-­‐lyon1.fr
[email protected]
[email protected]
[email protected]
jens.hasserodt@ens-­‐lyon.fr
[email protected]
maggy.hologne@univ-­‐lyon1.fr
[email protected]
[email protected]
[email protected]
benoit.joseph@univ-­‐lyon1.fr
laurence.lafanechere@ujf-­‐grenoble.fr
jean-­‐marc.lancelin@univ-­‐lyon1.fr
marc.le-­‐borgne@univ-­‐lyon1.fr
[email protected]
thierry.lomberget@univ-­‐lyon1.fr
luneau@univ-­‐lyon1.fr
[email protected]
[email protected]
maurice.medebielle@univ-­‐lyon1.fr
marine.peuchmaur@ujf-­‐grenoble.fr
[email protected]
vincent.motto-­‐ros@univ-­‐lyon1.fr
marine.peuchmaur@ujf-­‐grenoble.fr
[email protected]
[email protected]
maxime.prost@ens-­‐lyon.fr
[email protected]
laurence.lafanechere@ujf-­‐grenoble.fr
[email protected]
amandine-­‐[email protected]
ivan.rivalta@ens-­‐lyon.fr
[email protected]
[email protected]
xavier.ronot@ujf-­‐grenoble.fr
[email protected]
[email protected]
pierre.stebe@ens-­‐lyon.fr
strazewski@univ-­‐lyon1.fr
[email protected]
[email protected]
leon-­‐[email protected]
[email protected]
laura.troussicot@univ-­‐lyon1.fr
laurence.lafanechere@ujf-­‐grenoble.fr
Jean.viallet@UJF-­‐Grenoble.fr
olivier.walker@univ-­‐lyon1.fr
[email protected]
Civilité Participant
Delphine
Mlle
Mme
M.
Dr
Dr
Dr
Mlle
Mlle
M.
Mme
Mlle
Dr
Dr
M.
Mlle
Mlle
Mlle
M.
M.
Mlle
Mlle
Mme
Dr
M.
Dr
M.
M.
Dr
Dr
Dr
M.
Dr
Prof
M.
M.
Dr
Mlle
M.
Dr
Prof
Dr
Mme
M.
M.
Mlle
Prof
Dr
Prof
Prof
M.
Dr
Prof
M.
Dr
Dr
Mlle
M.
M.
Mlle
Mme
Mlle
M.
Mlle
Dr
M.
Mlle
Dr
Dr
M.
Prof
M.
Dr
M.
Prof
M.
Mlle
M.
M.
Mlle
Mlle
M.
Dr
Mme
Societe
Fonction
Adresse
CP
Ville
Doctorante
Bat 308G, 43 Bd du 11 Nov 1918
[email protected]­‐lyon1.fr
Mlle
LAGEP
Institut des sciences analytiques Doctorante
5 rue de la Doua
69100 Villeurbanne
LCBM, iRTSV, CEA Grenoble
Post-­‐Doc
Laboratoire de Chimie et Biologie des 38054
Métaux Grenoble
UMR CEA -­‐ UJF -­‐ CNRS n° 5249 iRTSV (institut de Recherches en technolo
CRCL
chef d'équipe
28 rue Laennec
69373 Lyon
CEA/ INSERM/ UJF
Resonsable opérationnelle plate-­‐forme iRTSV-­‐ U1038 de criblage -­‐ LBGE d-­‐ e Gm
en&Chem
olécules 38054 Grenoble Cedex 09
EDELRIS
Responsable Business Development
115 Avenue Lacassagne
69003 LYON
BIOASTER
Chargé de mission scientifique321 Av. Jean Jaurès
69007 Lyon
CRCL
doctorante
28 rue Laennec
69373 Lyon
cea
stagière Master 2
17 rue des martyrs
38000 grenoble
Université Lyon 1 -­‐ EA4446
PhD student
Faculté de Pharmacie -­‐ ISPB, 8 Avenue 69373
Rockefeller
Lyon Cedex 08
Bioaster
Ingénieur en bioinformatique 321 ave Jean Jaurès
69007 Lyon
ENS LYON
ASSISTANTE DE RECHERCHE 46 allée d'Italie
69364 Lyon CEDEX 07
Institut des sciences analytiques IR
5 rue de la doua
69100 Villeurbanne
EDELRIS
Team Leader
115 Avenue Lacassagne
69003 LYON
IBCP-­‐UMR 5086 CNRS-­‐UCBL
Doctorant
7 Passage du Vercors
69007 Lyon
bioMerieux
PhD student
chemin de l'orme
69280 Marcy l'étoile
iRTSV/BGE/EDyP
Doctorante
17 Rue des Martyrs
38000 Grenoble
Institut des Sciences Analytiques Doctorante
5 rue de la Doua
69100 Villeurbanne
BIOASTER
Directeur des Programmes de 321 Recherche
avenue Jean Jaurès
69007 Lyon
ICBMS UMR5246
Responsable Chimiothèque ICBMS
43 bd du 11 Novembre 1918
69622 Villeurbanne
EMBL
Postdoctoral fellow
BP 181, 6 rue Jules Horowitz
38042 Grenoble
ISA
doctorante
5 rue de la doua
69100 Villeurbanne
CNRS
Ingénieur de Recherche
5 rue de la doua
69100 Villeurbanne
Inserm
senior scientist
321 avenue Jean Jaurès
69007 Lyon
Institut des sciences analytiques Doctorant
5 rue de la Doua
69100 Villeurbanne
CALIXAR
CEO
7 PASSAGE DU VERCORS
69007 LYON
Centre Léon Bérard-­‐ 28 rue Laennec69008 LYON
CRCL-­‐INSERM 1052
Chargé de Recherche
IBCP
Doctorant
7 passages du Vercors
69367 Lyon
sans emploi
Chef de Projet SI
NA
NA
LYON
UCB Lyon1 -­‐ Faculté de Pharmacie Maître de conférences
8 avenue Rockefeller
69373 LYON
iRTSV/BGE/Gen&Chem, 17 rue des M38054
artyrsGrenoble
CEA-­‐Grenoble, iRTSV
Chercheur
ENS de Lyon
Master student
15 parvis René Descartes
69007 Lyon
Institut de Biologie Structurale
CR1
6, rue Jules Horowitz
38027 Grenoble cedex 1
IECB
Professeur Emérite
2 rue Robert Escarpit
33607 Pessac Cedex
Laboratoire de Chimie de l'ENS de Lyon
Doctorant
46, allée d'Italie
69007 Lyon
Université de Genève
Stagiaire de Master 2
30 Quai Ernest Ansermet
1211 Genève
Institut des Sciences Analytiques Maître de conférences
5 rue de la Doua
69100 Villeurbanne
Institut des Sciences Analytiques Doctorante
5 rue de la Doua
69100 Villeurbanne
CEA / INSERM / UJF
Chercheur
17 rue des Martyrs
38054 GRENOBLE Cedex 9
CEA Grenoble
Chercheur
BGE, 17 rue des Martyrs
38054 Grenoble Cedex 9
ENS de Lyon
Professeur
Laboratoire de Chimie, 46 allée d'Italie
69364 Lyon
DHC Consulting
Principal
Le Beaulieu
38320 Brie et Angonnes
UCBL Lyon 1
Maître de conférence
5 rue de la doua
69100 Villeurbanne
IBS
Chercheur
6 rue Jules Horowitz
38000 Grenoble
LAGEP
Doctorant
F-­‐69622 Villeurbanne, France
69622 Villeurbanne, Lyon
Institut des Sciences Analytiques Doctorante
5 rue de la Doua
69100 Villeurbanne
ICBMS, Université Claude Bernard -­‐ Enseignant-­‐Chercheur
Lyon 1
43 Boulevard du 11 novembre 1918
F-­‐69622 Villeurbanne
CNRS
Directrice de Recherche
IAB Rond-­‐Point de la Chantourne 38700 La Tronche
Institut des Sciences Analytiques Professeur
5, rue de la Doua
69100 Volleurbanne
EA 4446 B2C
Directeur
8 avenue Rockefeller
69373 Lyon
INSERM U1111/CIRI
Chercheur
321 Avenue Jean Jaures
69002 LYON
EA4446 -­‐ B2C -­‐ Biomolécules, Cancer Maître et Chimiorésistances
de Conférences -­‐ HDR ISPB Faculté de Pharmacie 8, avenue 69373
Rockefeller
LYON cedex 8
UCBL
Professeur
Campus Scientifique de La Doua
69622 Villeurbanne
Département de Pharmacochime MDoctorant
oléculaire
470 rue de la chimie, Bâtiment E, BP53
38400 Saint Martin d'Hères
EMBL, Grenoble
Team leader
6 rue Jules Horowitz
38000 Grenoble
Institut de Chimie et Biochimie Moléculaire Directeur et dSe upramoléculaire recherche CNRS(ICBMS)
Université Claude Bernard Lyon 1, Bâtiment 69622 Villeurbanne
Curien, 43 bd du 11 Novembre 1918
Département de Pharmacochimie MDoctorante
oléculaire
470 rue de la chimie
38400 Saint-­‐Martin-­‐d'Hères
CRCL Lyon
CR1
28 rue Laennec
69008 LYON
ILM
Maître de Conférences
Bat Kaslter, Campus de la doua, Bd 169622
1 Novembre
Villeurbannes
Département de Pharmacochimie MEtudiante oléculaireM2
470 rue de la chimie
38400 Saint-­‐Martin-­‐d'Hères
GENEL
Sales&Marketing Manager
17 rue des Martyrs
38000 GRENOBLE
UMRS1036 -­‐ BCI/iRTSV -­‐ CEA Grenoble
Post-­‐Doctorante
17, rue des Martyrs
38000 Grenoble
ENS de Lyon
PhD Student (3rd year)
46 allée d'Italie
69007 Lyon
CEA Grenoble iRTSV/LCBM
Post-­‐doc
17 rue des martyrs
38054 Grenoble
INSERM
Post-­‐doctorant
IAB Rond-­‐Point de la Chantourne 38700 La Tronche
Curie institute
Bioinformatic project managerRue de l'Ulm 26
75015 Paris
CRCL
Doctorant
28 rue Laennec
69008 Lyon
ENS-­‐Lyon
CR
46 allee d'italie
69007 Lyon
CNRS IBCP
Ingénieur IE1
7 passage du Vercors
69367 LYON
Edelris
VP Strategic Innovation
115 avenue Lacassagne
69003 Lyon
ECOLE PRATIQUE DES HAUTES ETUDES
DIRECTEUR D'ETUDES
LABORATOIRE CACYS, UFR MEDECINE 38700
ET PHARMACIE
LA TRONCHE
Institut de Biologie Structurale
Chercheur
6, rue Jules Horowitz
38000 Grenoble
iRTSV/BGE/ Equipe Gen&Chem
Ingénieur Chercheur CEA
CEA Grenoble 17 rue des Martyrs 38054 GRENOBLE
ENS Lyon
doctorant
46 place d'Italie
69007 Lyon
UCBL
Enseignant-­‐Chercheur
43 bvd du 11 novembre 1918
69622 Villeurbanne
CEA
Chercheur
17 rue des martyrs
38054 Grenoble
IBS
Doctorante
6 rue Jules Horowitz
38000 Grenoble
CRCL
Post-­‐doc
28, rue Laennec
69008 Lyon
UJF
Doctorant
470 rue de la chimie
38400 st martin d'hères
Institut des Sciences Analytiques de Doctorante
Lyon
5 Rue de la Doua
69100 VILLEURBANNE
INSERM
Assistant Ingénieur
IAB Rond-­‐Point de la Chantourne 38700 La Tronche
UJF
Enseignant Chercheur
IAB CRI INSERM UJF U823
38000 Grenoble
Institut de Sciences Analytiques
Maître de Conférence
5 rue de la Doua
69100 Villeurbanne
EMBL
Visiting PhD Student
EMBL Grenoble Outstation, 6 Rue Jules 38042
Horowitz, Grenoble
BP181
Drug Screening : from Phenotypes to Molecular Modeling
Villeurbanne, France, February 27, 2014
List of Participants
CEA
17 rue des martyrs
38054 Grenoble
France
Drug Screening : from Phenotypes to Molecular Modeling

Documents pareils