Network


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

Hotspot


Dive into the research topics where Mark G. Palermo is active.

Publication


Featured researches published by Mark G. Palermo.


Nature | 2016

Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases

Yan Chen; Matthew J. LaMarche; Ho Man Chan; Peter Fekkes; Garcia-Fortanet J; Acker Mg; Brandon Antonakos; Christine Hiu-Tung Chen; Zhouliang Chen; Vesselina G. Cooke; Zhan Deng; Fei F; Brant Firestone; Michelle Fodor; Cary Fridrich; Hui Gao; Denise Grunenfelder; Hao Hx; Jacob J; Samuel Ho; Kathy Hsiao; Zhao B. Kang; Rajesh Karki; Mitsunori Kato; Jay Larrow; La Bonte Lr; Francois Lenoir; Gang Liu; Shumei Liu; Dyuti Majumdar

The non-receptor protein tyrosine phosphatase SHP2, encoded by PTPN11, has an important role in signal transduction downstream of growth factor receptor signalling and was the first reported oncogenic tyrosine phosphatase. Activating mutations of SHP2 have been associated with developmental pathologies such as Noonan syndrome and are found in multiple cancer types, including leukaemia, lung and breast cancer and neuroblastoma. SHP2 is ubiquitously expressed and regulates cell survival and proliferation primarily through activation of the RAS–ERK signalling pathway. It is also a key mediator of the programmed cell death 1 (PD-1) and B- and T-lymphocyte attenuator (BTLA) immune checkpoint pathways. Reduction of SHP2 activity suppresses tumour cell growth and is a potential target of cancer therapy. Here we report the discovery of a highly potent (IC50 = 0.071 μM), selective and orally bioavailable small-molecule SHP2 inhibitor, SHP099, that stabilizes SHP2 in an auto-inhibited conformation. SHP099 concurrently binds to the interface of the N-terminal SH2, C-terminal SH2, and protein tyrosine phosphatase domains, thus inhibiting SHP2 activity through an allosteric mechanism. SHP099 suppresses RAS–ERK signalling to inhibit the proliferation of receptor-tyrosine-kinase-driven human cancer cells in vitro and is efficacious in mouse tumour xenograft models. Together, these data demonstrate that pharmacological inhibition of SHP2 is a valid therapeutic approach for the treatment of cancers.


Journal of Medicinal Chemistry | 2013

Identification of NVP-TNKS656: The Use of Structure-Efficiency Relationships To Generate a Highly Potent, Selective, and Orally Active Tankyrase Inhibitor.

Michael Shultz; Atwood Cheung; Christina A. Kirby; Brant Firestone; Jianmei Fan; Christine Hiu-Tung Chen; Zhouliang Chen; Donovan Noel Chin; Lucian DiPietro; Aleem Fazal; Yun Feng; Pascal D. Fortin; Ty Gould; Bharat Lagu; Huangshu Lei; Francois Lenoir; Dyuti Majumdar; Etienne Ochala; Mark G. Palermo; Ly Luu Pham; Minying Pu; Troy Smith; Travis Stams; Ronald C. Tomlinson; B. Barry Touré; Michael Scott Visser; Run Ming Wang; Nigel J. Waters; Wenlin Shao

Tankyrase 1 and 2 have been shown to be redundant, druggable nodes in the Wnt pathway. As such, there has been intense interest in developing agents suitable for modulating the Wnt pathway in vivo by targeting this enzyme pair. By utilizing a combination of structure-based design and LipE-based structure efficiency relationships, the core of XAV939 was optimized into a more stable, more efficient, but less potent dihydropyran motif 7. This core was combined with elements of screening hits 2, 19, and 33 and resulted in highly potent, selective tankyrase inhibitors that are novel three pocket binders. NVP-TNKS656 (43) was identified as an orally active antagonist of Wnt pathway activity in the MMTV-Wnt1 mouse xenograft model. With an enthalpy-driven thermodynamic signature of binding, highly favorable physicochemical properties, and high lipophilic efficiency, NVP-TNKS656 is a novel tankyrase inhibitor that is well suited for further in vivo validation studies.


Journal of Medicinal Chemistry | 2016

Allosteric Inhibition of SHP2: Identification of a Potent, Selective, and Orally Efficacious Phosphatase Inhibitor

Jorge Garcia Fortanet; Christine Hiu-Tung Chen; Ying-Nan P. Chen; Zhouliang Chen; Zhan Deng; Brant Firestone; Peter Fekkes; Michelle Fodor; Pascal D. Fortin; Cary Fridrich; Denise Grunenfelder; Samuel Ho; Zhao B. Kang; Rajesh Karki; Mitsunori Kato; Nick Keen; Laura R. Labonte; Jay Larrow; Francois Lenoir; Gang Liu; Shumei Liu; Franco Lombardo; Dyuti Majumdar; Matthew John Meyer; Mark G. Palermo; Lawrence Blas Perez; Minying Pu; Timothy Michael Ramsey; William R. Sellers; Michael Shultz

SHP2 is a nonreceptor protein tyrosine phosphatase (PTP) encoded by the PTPN11 gene involved in cell growth and differentiation via the MAPK signaling pathway. SHP2 also purportedly plays an important role in the programmed cell death pathway (PD-1/PD-L1). Because it is an oncoprotein associated with multiple cancer-related diseases, as well as a potential immunomodulator, controlling SHP2 activity is of significant therapeutic interest. Recently in our laboratories, a small molecule inhibitor of SHP2 was identified as an allosteric modulator that stabilizes the autoinhibited conformation of SHP2. A high throughput screen was performed to identify progressable chemical matter, and X-ray crystallography revealed the location of binding in a previously undisclosed allosteric binding pocket. Structure-based drug design was employed to optimize for SHP2 inhibition, and several new protein-ligand interactions were characterized. These studies culminated in the discovery of 6-(4-amino-4-methylpiperidin-1-yl)-3-(2,3-dichlorophenyl)pyrazin-2-amine (SHP099, 1), a potent, selective, orally bioavailable, and efficacious SHP2 inhibitor.


ChemBioChem | 2002

Synthesis and structure-activity relationship of the isoindolinyl benzisoxazolpiperidines as potent, selective, and orally active human dopamine D4 receptor antagonists.

James A. Hendrix; Stephen J. Shimshock; Gregory Michael Shutske; John Dick Tomer; Kevin J. Kapples; Mark G. Palermo; Thomas J. (Roy) Corbett; H M Vargas; Sharon Kafka; Karen M. Brooks; Lynn Laws-Ricker; David K.H. Lee; Inez de Lannoy; Michel Bordeleau; Geihan Rizkalla; Joshua Owolabi; Rajender Kamboj

A new class of potent dopamine D4 antagonists was discovered with selectivity over dopamine D2 and the α‐1 adrenoceptor. The lead compound was discovered by screening our compound collection. The structure–activity relationships of substituted isoindoline rings and the chirality about the hydroxymethyl side chain were explored. The isoindoline analogues showed modest differences in potency and selectivity. The S enantiomer proved to be the more potent enantiomer at the D4 receptor. Several analogues with greater than 100‐fold selectivity for D4 over D2 and the α‐1 adrenoreceptor were discovered. Several selective analogues were active in vivo upon oral or intraperitoneal administration. A chiral synthesis starting from either D‐ or L‐O‐benzylserine is also described.


ACS Medicinal Chemistry Letters | 2018

Optimization of 3-Pyrimidin-4-yl-oxazolidin-2-ones as Orally Bioavailable and Brain Penetrant Mutant IDH1 Inhibitors

Qian Zhao; James R. Manning; James Sutton; Abran Costales; Martin Sendzik; Cynthia Shafer; Julian Levell; Gang Liu; Thomas Caferro; Young Shin Cho; Mark G. Palermo; Gregg Chenail; Julia Dooley; Brian Villalba; Ali Farsidjani; Jinyun Chen; Stephanie Dodd; Ty Gould; Guiqing Liang; Kelly Slocum; Minying Pu; Brant Firestone; Joseph D. Growney; Tycho Heimbach; Raymond Pagliarini

Mutant isocitrate dehydrogenase 1 (IDH1) is an attractive therapeutic target for the treatment of various cancers such as AML, glioma, and glioblastoma. We have evaluated 3-pyrimidin-4-yl-oxazolidin-2-ones as mutant IDH1 inhibitors that bind to an allosteric, induced pocket of IDH1R132H. This Letter describes SAR exploration focused on improving both the in vitro and in vivo metabolic stability of the compounds, leading to the identification of 19 as a potent and selective mutant IDH1 inhibitor that has demonstrated brain penetration and excellent oral bioavailability in rodents. In a preclinical patient-derived IDH1 mutant xenograft tumor model study, 19 efficiently inhibited the production of the biomarker 2-HG.


Cancer Research | 2017

Abstract 2084: Conformational activation and allosteric inhibition of SHP2 in RTK-driven cancers

Michael G. Acker; Ying-Nan P. Chen; Matthew J. LaMarche; Ho Man Chan; Peter Fekkes; Jorge Garcia-Fortanet; Jonathan R. LaRochelle; Brandon Antonakos; C. H. Chen; Zhuoliang Chen; Vesselina G. Cooke; Jason R. Dobson; Zhan Deng; Fei Feng; Brant Firestone; Michelle Fodor; Cary Fridrich; Hui Gao; Huai-Xiang Hao; Jaison Jacob; Samuel Ho; Kathy Hsiao; Zhao B. Kang; Rajesh Karki; Mitsunori Kato; Jay Larrow; Laura R. La Bonte; Gang Liu; Shumei Liu; Dyuti Majumdar

The non-receptor protein tyrosine phosphatase (PTP) SHP2 is an important component of RTK signaling in response to growth factor stimulus and sits just upstream of the RAS-MAPK signaling cascade. The first oncogenic phosphatase to be identified, SHP2 is dysregulated in multiple human diseases including the developmental disorders Noonan and Leopard syndromes, as well as leukemia, lung cancer and neuroblastoma where aberrant activity of SHP2 leads to uncontrolled MAPK signaling. Cancer-associated activating mutations in SHP2 impart an “auto-on” state of the enzyme, boosting basal activity by shifting the equilibrium away from the auto-inhibited state. Reduction of SHP2 activity through genetic knockdown suppresses tumor growth, validating SHP2 as a target for cancer therapy. SHP099, a recently reported potent and selective allosteric inhibitor of SHP2, stabilizes the auto-inhibited form of SHP2 through interactions with the N-terminal SH2 and C-terminal PTP domains of the protein. SHP099 suppresses MAPK signaling in RTK amplified cancers resulting in suppressed proliferation in vitro and inhibition of tumor growth in mouse tumor xenograft models. Together, these data demonstrate the therapeutic potential of SHP2 inhibition in the treatment of cancer and other RAS/MAPK-linked diseases. Citation Format: Michael G. Acker, Ying-Nan P. Chen, Matthew J. LaMarche, Ho Man Chan, Peter Fekkes, Jorge Garcia-Fortanet, Jonathan R. LaRochelle, Brandon Antonakos, Christine Hiu-Tung Chen, Zhuoliang Chen, Vesselina G. Cooke, Jason R. Dobson, Zhan Deng, Fei Feng, Brant Firestone, Michelle Fodor, Cary Fridrich, Hui Gao, Huai-Xiang Hao, Jaison Jacob, Samuel Ho, Kathy Hsiao, Zhao B. Kang, Rajesh Karki, Mitsunori Kato, Jay Larrow, Laura R. La Bonte, Gang Liu, Shumei Liu, Dyuti Majumdar, Matthew J. Meyer, Mark Palermo, Minying Pu, Edmund Price, Subarna Shakya, Michael D. Shultz, Kavitha Venkatesan, Ping Wang, Markus Warmuth, Sarah Williams, Guizhi Yang, Jing Yuan, Ji-Hu Zhang, Ping Zhu, Stephen C. Blacklow, Timothy Ramsey, Nicholas J. Keen, William R. Sellers, Travis Stams, Pascal D. Fortin. Conformational activation and allosteric inhibition of SHP2 in RTK-driven cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2084. doi:10.1158/1538-7445.AM2017-2084


Bioorganic & Medicinal Chemistry Letters | 2017

Structure based design of Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors from a phenotypic screen

Daniel Steven Palacios; Erik Meredith; Toshio Kawanami; Christopher Michael Adams; Xin Chen; Veronique Darsigny; Erin Geno; Mark G. Palermo; Daniel Baird; Geoffrey Boynton; Scott A. Busby; Elizabeth George; Chantale T. Guy; Jeffrey Hewett; Laryssa Tierney; Sachin Thigale; Wilhelm Weihofen; Louis Wang; Nicole White; Ming Yin; Upendra A. Argikar

Nicotinamide phosphoribosyltransferase is a key metabolic enzyme that is a potential target for oncology. Utilizing publicly available crystal structures of NAMPT and in silico docking of our internal compound library, a NAMPT inhibitor, 1, obtained from a phenotypic screening effort was replaced with a more synthetically tractable scaffold. This compound then provided an excellent foundation for further optimization using crystallography driven structure based drug design. From this approach, two key motifs were identified, the (S,S) cyclopropyl carboxamide and the (S)-1-N-phenylethylamide that endowed compounds with excellent cell based potency. As exemplified by compound 27e such compounds could be useful tools to explore NAMPT biology in vivo.


Molecular Cancer Therapeutics | 2013

Abstract C54: Aryl-N-(1H-imidazol-2-yl)-acetamides: Nonpeptidic binders of Bir3 cIAP protein.

Mark G. Palermo; Rohan Eric John Beckwith; Christopher Sean Straub; Kara Herlihy; Yiping Shen; Xiaolu Zhang; Matthew Clapham; Brian Hurley

Inhibitor of Apoptosis Proteins (IAP) negatively regulate cell death through a caspase-3 and caspase-7 activation. IAP inhibitors (IAPi) were derived from the peptide sequence Alanine-Valine-Proline (AVP) based on Smac-protein binding to the BIR-3 domain of IAP protein. These peptidic compounds are low molecular weight and mimic Smac when binding to the BIR3 domain of XIAP, CIAP1, and CIAP2. However, peptides generally suffer from poor permeability that can potentially influence the amount of drugability largely due to the amide backbone and an alternate chemotype was sought. In an effort to find a replacement for AVP scaffolds, an FBS by NMR identified indole-carboxylic acid 1 as a hit in competitive binding studies. The indole scaffold was further refined through FEPOP, virtual screening, and SAR leading to the benzimidazole scaffold which upon further docking studies resulted in 2-aminobenzimidazole 2. In silico tools were utilized to propose a synthetic pharmacophore which resulted in a phenyl-hetero-imidazole tricyclic series peptidomimetic with only one amide moiety. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C54. Citation Format: Mark G. Palermo, Rohan Beckwith, Christopher S. Straub, Kara Herlihy, Yiping Shen, Xiaolu Zhang, Matthew Clapham, Brian Hurley. Aryl-N-(1H-imidazol-2-yl)-acetamides: Nonpeptidic binders of Bir3 cIAP protein. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr C54.


Archive | 2003

Peptide inhibitors of smac protein binding to inhibitor of apoptosis proteins (IAP)

Sushil K. Sharma; Leigh Zawel; Mark G. Palermo; Nagarajan Chandramouli; Kenneth W. Bair


Archive | 2014

3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant idh

Thomas Caferro; Young Shin Cho; Abran Costales; Huangshu Lei; Francois Lenoir; Julian Levell; Gang Liu; Mark G. Palermo; Keith B. Pfister; Martin Sendzik; Cynthia Shafer; Michael Shultz; Troy Smith; James Sutton; Bakary-Barry Toure; Fan Yang; Qian Zhao

Collaboration


Dive into the Mark G. Palermo's collaboration.

Researchain Logo
Decentralizing Knowledge