Network


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

Hotspot


Dive into the research topics where Pierre-Yves Michellys is active.

Publication


Featured researches published by Pierre-Yves Michellys.


Journal of Medicinal Chemistry | 2013

Synthesis, structure-activity relationships, and in vivo efficacy of the novel potent and selective anaplastic lymphoma kinase (ALK) inhibitor 5-chloro-N2-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (LDK378) currently in phase 1 and phase 2 clinical trials.

Thomas H. Marsilje; Wei Pei; Bei Chen; Wenshuo Lu; Tetsuo Uno; Yunho Jin; Tao Jiang; Sung Joon Kim; Nanxin Li; Markus Warmuth; Yelena Sarkisova; Frank Sun; Auzon Steffy; AnneMarie C. Pferdekamper; Allen Li; Sean B. Joseph; Young Chul Kim; Bo Liu; Tove Tuntland; Xiaoming Cui; Nathanael S. Gray; Ruo Steensma; Yongqin Wan; Jiqing Jiang; Greg Chopiuk; Jie Li; W. Perry Gordon; Wendy Richmond; Kevin Johnson; Jonathan Chang

The synthesis, preclinical profile, and in vivo efficacy in rat xenograft models of the novel and selective anaplastic lymphoma kinase inhibitor 15b (LDK378) are described. In this initial report, preliminary structure-activity relationships (SARs) are described as well as the rational design strategy employed to overcome the development deficiencies of the first generation ALK inhibitor 4 (TAE684). Compound 15b is currently in phase 1 and phase 2 clinical trials with substantial antitumor activity being observed in ALK-positive cancer patients.


Nature | 2016

Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors

Yong Jia; Cai-Hong Yun; Eunyoung Park; Dalia Ercan; Mari Manuia; Jose Juarez; Chunxiao Xu; Kevin Rhee; Ting Chen; Haikuo Zhang; Sangeetha Palakurthi; Jaebong Jang; Gerald Lelais; Michael DiDonato; Badry Bursulaya; Pierre-Yves Michellys; Robert Epple; Thomas H. Marsilje; Matthew McNeill; Wenshuo Lu; Jennifer L. Harris; Steven Bender; Kwok-Kin Wong; Pasi A. Jänne; Michael J. Eck

EGFR tyrosine kinase inhibitors (TKIs) gefitinib, erlotinib and afatinib are approved treatments for non-small cell lung cancers harboring activating mutations in the EGFR kinase1,2, but resistance arises rapidly, most frequently due to the secondary T790M mutation within the ATP-site of the receptor.3,4 Recently developed mutant-selective irreversible inhibitors are highly active against the T790M mutant5,6, but their efficacy can be compromised by acquired mutation of C797, the cysteine residue with which they form a key covalent bond7. All current EGFR TKIs target the ATP-site of the kinase, highlighting the need for therapeutic agents with alternate mechanisms of action. Here we describe rational discovery of EAI045, an allosteric inhibitor that targets selected drug-resistant EGFR mutants but spares the wild type receptor. A crystal structure shows that the compound binds an allosteric site created by the displacement of the regulatory C-helix in an inactive conformation of the kinase. The compound inhibits L858R/T790M-mutant EGFR with low-nanomolar potency in biochemical assays, but as a single agent is not effective in blocking EGFR-driven proliferation in cells due to differential potency on the two subunits of the dimeric receptor, which interact in an asymmetric manner in the active state8. We observe dramatic synergy of EAI045 with cetuximab, an antibody therapeutic that blocks EGFR dimerization9,10, rendering the kinase uniformly susceptible to the allosteric agent. EAI045 in combination with cetuximab is effective in mouse models of lung cancer driven by L858R/T790M EGFR and by L858R/T790M/C797S EGFR, a mutant that is resistant to all currently available EGFR TKIs. More generally, our findings illustrate the utility of purposefully targeting allosteric sites to obtain mutant-selective inhibitors.


Biochemical Journal | 2010

Crystal structure of the ALK (anaplastic lymphoma kinase) catalytic domain.

Christian C. Lee; Yong Jia; Nanxin Li; Xiuying Sun; Kenneth Ng; Eileen Ambing; Mu-Yun Gao; Su Hua; Connie Chen; Sungjoon Kim; Pierre-Yves Michellys; Scott A. Lesley; Jennifer L. Harris; Glen Spraggon

ALK (anaplastic lymphoma kinase) is an RTK (receptor tyrosine kinase) of the IRK (insulin receptor kinase) superfamily, which share an YXXXYY autophosphorylation motif within their A-loops (activation loops). A common activation and regulatory mechanism is believed to exist for members of this superfamily typified by IRK and IGF1RK (insulin-like growth factor receptor kinase-1). Chromosomal translocations involving ALK were first identified in anaplastic large-cell lymphoma, a subtype of non-Hodgkins lymphoma, where aberrant fusion of the ALK kinase domain with the NPM (nucleophosmin) dimerization domain results in autophosphosphorylation and ligand-independent activation. Activating mutations within the full-length ALK kinase domain, most commonly R1275Q and F1174L, which play a major role in neuroblastoma, were recently identified. To provide a structural framework for understanding these mutations and to guide structure-assisted drug discovery efforts, the X-ray crystal structure of the unphosphorylated ALK catalytic domain was determined in the apo, ADP- and staurosporine-bound forms. The structures reveal a partially inactive protein kinase conformation distinct from, and lacking, many of the negative regulatory features observed in inactive IGF1RK/IRK structures in their unphosphorylated forms. The A-loop adopts an inhibitory pose where a short proximal A-loop helix (alphaAL) packs against the alphaC helix and a novel N-terminal beta-turn motif, whereas the distal portion obstructs part of the predicted peptide-binding region. The structure helps explain the reported unique peptide substrate specificity and the importance of phosphorylation of the first A-loop Tyr1278 for kinase activity and NPM-ALK transforming potential. A single amino acid difference in the ALK substrate peptide binding P-1 site (where the P-site is the phosphoacceptor site) was identified that, in conjunction with A-loop sequence variation including the RAS (Arg-Ala-Ser)-motif, rationalizes the difference in the A-loop tyrosine autophosphorylation preference between ALK and IGF1RK/IRK. Enzymatic analysis of recombinant R1275Q and F1174L ALK mutant catalytic domains confirms the enhanced activity and transforming potential of these mutants. The transforming ability of the full-length ALK mutants in soft agar colony growth assays corroborates these findings. The availability of a three-dimensional structure for ALK will facilitate future structure-function and rational drug design efforts targeting this receptor tyrosine kinase.


Cancer Research | 2016

EGF816 Exerts Anticancer Effects in Non–Small Cell Lung Cancer by Irreversibly and Selectively Targeting Primary and Acquired Activating Mutations in the EGF Receptor

Yong Jia; José Juárez; Jie Li; Mari Manuia; Matthew J. Niederst; Celin Tompkins; Noelito Timple; Mei-Ting Vaillancourt; AnneMarie Culazzo Pferdekamper; Elizabeth L. Lockerman; Chun Li; Jennifer Anderson; Carlotta Costa; Debbie Liao; Eric Murphy; Michael DiDonato; Badry Bursulaya; Gerald Lelais; Jordi Barretina; Matthew McNeill; Robert Epple; Thomas H. Marsilje; Nuzhat Pathan; Jeffrey A. Engelman; Pierre-Yves Michellys; Peter McNamara; Jennifer L. Harris; Steven L. Bender; Shailaja Kasibhatla

Non-small cell lung cancer patients carrying oncogenic EGFR mutations initially respond to EGFR-targeted therapy, but later elicit minimal response due to dose-limiting toxicities and acquired resistance. EGF816 is a novel, irreversible mutant-selective EGFR inhibitor that specifically targets EGFR-activating mutations arising de novo and upon resistance acquisition, while sparing wild-type (WT) EGFR. EGF816 potently inhibited the most common EGFR mutations L858R, Ex19del, and T790M in vitro, which translated into strong tumor regressions in vivo in several patient-derived xenograft models. Notably, EGF816 also demonstrated antitumor activity in an exon 20 insertion mutant model. At levels above efficacious doses, EGF816 treatment led to minimal inhibition of WT EGFR and was well tolerated. In single-dose studies, EGF816 provided sustained inhibition of EGFR phosphorylation, consistent with its ability for irreversible binding. Furthermore, combined treatment with EGF816 and INC280, a cMET inhibitor, resulted in durable antitumor efficacy in a xenograft model that initially developed resistance to first-generation EGFR inhibitors via cMET activation. Thus, we report the first preclinical characterization of EGF816 and provide the groundwork for its current evaluation in phase I/II clinical trials in patients harboring EGFR mutations, including T790M.


Journal of Biological Chemistry | 2007

NMR structural studies of interactions of a small, nonpeptidyl tpo mimic with the thrombopoietin receptor extracellular Juxtamembrane and transmembrane domains

Min-Ju Kim; Sang Ho Park; Stanley J. Opella; Thomas H. Marsilje; Pierre-Yves Michellys; H. Martin Seidel; Shin-Shay Tian

Thrombopoietin (Tpo) is a glycoprotein growth factor that supports hematopoietic stem cell survival and expansion and is the principal regulator of megakaryocyte growth and differentiation. Several small, nonpeptidyl molecules have been identified as selective human Tpo receptor (hTpoR) agonists. To understand how the small molecule Tpo mimic SB394725 interacts and activates hTpoR, we performed receptor domain swap and mutagenesis studies. The results suggest that SB394725 interacts specifically with the extracellular juxtamembrane region (JMR) and the transmembrane (TM) domain of hTpoR. Solution and solid-state NMR structural studies using a peptide containing the JMR-TM sequences showed that this region of hTpoR, unexpectedly, consists of two α-helices separated by a few nonhelical residues. SB394725 interacts specifically with His-499 in the TM domain and a few distinct residues in the JMR-TM region and affects several specific C-terminal TM domain residues. The unique structural information provided by these studies both sheds light on the distinctive mechanism of action of SB394725 and provides valuable insight into the mechanism of ligand-induced cytokine receptor activation.


Journal of Medicinal Chemistry | 2016

Discovery of (R,E)-N-(7-Chloro-1-(1-[4-(dimethylamino)but-2-enoyl]azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide (EGF816), a Novel, Potent, and WT Sparing Covalent Inhibitor of Oncogenic (L858R, ex19del) and Resistant (T790M) EGFR Mutants for the Treatment of EGFR Mutant Non-Small-Cell Lung Cancers.

Gerald Lelais; Robert Epple; Thomas H. Marsilje; Yun O. Long; Matthew McNeill; Bei Chen; Wenshuo Lu; Jaganmohan Anumolu; Sangamesh Badiger; Badry Bursulaya; Michael DiDonato; Rina Fong; Jose Juarez; Jie Li; Mari Manuia; Daniel E. Mason; Perry Gordon; Todd Groessl; Kevin Johnson; Yong Jia; Shailaja Kasibhatla; Chun Li; John Isbell; Glen Spraggon; Steven Bender; Pierre-Yves Michellys

Over the past decade, first and second generation EGFR inhibitors have significantly improved outcomes for lung cancer patients with activating mutations in EGFR. However, both resistance through a secondary T790M mutation at the gatekeeper residue and dose-limiting toxicities from wild-type (WT) EGFR inhibition ultimately limit the full potential of these therapies to control mutant EGFR-driven tumors and new therapies are urgently needed. Herein, we describe our approach toward the discovery of 47 (EGF816, nazartinib), a novel, covalent mutant-selective EGFR inhibitor with equipotent activity on both oncogenic and T790M-resistant EGFR mutations. Through molecular docking studies we converted a mutant-selective high-throughput screening hit (7) into a number of targeted covalent EGFR inhibitors with equipotent activity across mutants EGFR and good WT-EGFR selectivity. We used an abbreviated in vivo efficacy study for prioritizing compounds with good tolerability and efficacy that ultimately led to the selection of 47 as the clinical candidate.


Bioorganic & Medicinal Chemistry Letters | 2008

Optimization of small molecule agonists of the thrombopoietin (Tpo) receptor derived from a benzo[a]carbazole hit scaffold

Thomas H. Marsilje; Phil B. Alper; Wenshuo Lu; Daniel Mutnick; Pierre-Yves Michellys; Yun He; Donald S. Karanewsky; Donald Chow; Andrea Gerken; Min-Ju Kim; H. Martin Seidel; Shin-Shay Tian

The lead optimization of a novel series of benzo[a]carbazole-based small molecule agonists of the thrombopoietin (Tpo) receptor is reported. The chemical instability of the dihydro-benzo[a]carbazole lead 2 was successfully addressed in the design and evaluation of compounds which also demonstrated improved potency compared to 2. Members of the scaffold have been identified which are full agonists that demonstrate cellular functional potency <50 nM. Analog 21 demonstrates equivalent efficacy in the human megakaryocyte differentiation (CFU-mega) assay compared to Eltrombopag.


Bioorganic & Medicinal Chemistry Letters | 2014

Syk inhibitors with high potency in presence of blood.

Gebhard Thoma; Joachim Blanz; Peter Dr. Bühlmayer; Peter Drückes; Matthias Kittelmann; Alexander Baxter Smith; Maurice J. van Eis; Eric Vangrevelinghe; Hans-Günter Zerwes; Jianwei Che; Xiaohui He; Yunho Jin; Christian C. Lee; Pierre-Yves Michellys; Tetsuo Uno; Hong Liu

We describe two series of Syk inhibitors which potently abrogate Syk kinase function in enzymatic assays, cellular assays and in primary cells in the presence of blood. Introduction of a 7-aminoindole substituent led to derivatives with good kinase selectivity and little or no hERG channel inhibition (3b, 10c).


Steroids | 2007

Conjugate hydrocyanation of 17-acetyl-11-carbomethoxygona-1,3,5(10),13(17)-tetraenes.

Hélène Pellissier; Pierre-Yves Michellys; Maurice Santelli

The conjugate hydrocyanation of 17-acetylgona-11-carbomethoxy-1,3,5(10),13(17)-tetraenes using diethylaluminum cyanide (Nagata reaction) is reported. This methodology has allowed the introduction of an angular cyano group at the C-13 position of the steroid skeleton. Subsequent reduction of the nitrile group yielded various functionalized steroids. One of them, 22 bears the natural trans/anti/trans stereochemistry and possesses an hydroxyl and aminomethyl functionalities in the positions 11beta and 13beta, respectively. The characteristic (1)H and (13)C NMR spectroscopic features of the synthesized steroids are reported.


Bioorganic & Medicinal Chemistry Letters | 2014

Discovery of structurally novel, potent and orally efficacious GPR119 agonists.

Phil B. Alper; Mihai Azimioara; Christopher Cow; Daniel Mutnick; Victor Nikulin; Pierre-Yves Michellys; Zhiliang Wang; Esther Reding; Michael Paliotti; Jing Li; Dingjiu Bao; Jocelyn Zoll; Young Deuk Kim; Matthew Zimmerman; Todd Groessel; Tove Tuntland; Sean B. Joseph; Peter McNamara; H. Martin Seidel; Robert Epple

Screening hit 5 was identified in a biochemical screen for GPR119 agonists. Compound 5 was structurally novel, displayed modest biochemical activity and no oral exposure, but was structurally distinct from typical GPR119 agonist scaffolds. Systematic optimization led to compound 36 with significantly improved in vitro activity and oral exposure, to elevate GLP1 acutely in an in vivo mouse model at a dose of 10mg/kg.

Collaboration


Dive into the Pierre-Yves Michellys's collaboration.

Top Co-Authors

Avatar

Thomas H. Marsilje

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Robert Epple

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Bei Chen

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenshuo Lu

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Songchun Jiang

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Daniel Mutnick

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christopher Cow

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Researchain Logo
Decentralizing Knowledge