Network


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

Hotspot


Dive into the research topics where Minying Pu is active.

Publication


Featured researches published by Minying Pu.


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.


Cancer Research | 2014

IDH1 mutations alter citric acid cycle metabolism and increase dependence on oxidative mitochondrial metabolism.

Alexandra R. Grassian; Seth J. Parker; Shawn M. Davidson; Ajit S. Divakaruni; Courtney R. Green; Xiamei Zhang; Kelly Slocum; Minying Pu; Fallon Lin; Chad Vickers; Carol Joud-Caldwell; Franklin Chung; Hong Yin; Erika Handly; Christopher Sean Straub; Joseph D. Growney; Matthew G. Vander Heiden; Anne N. Murphy; Raymond Pagliarini; Christian M. Metallo

Oncogenic mutations in isocitrate dehydrogenase 1 and 2 (IDH1/2) occur in several types of cancer, but the metabolic consequences of these genetic changes are not fully understood. In this study, we performed (13)C metabolic flux analysis on a panel of isogenic cell lines containing heterozygous IDH1/2 mutations. We observed that under hypoxic conditions, IDH1-mutant cells exhibited increased oxidative tricarboxylic acid metabolism along with decreased reductive glutamine metabolism, but not IDH2-mutant cells. However, selective inhibition of mutant IDH1 enzyme function could not reverse the defect in reductive carboxylation activity. Furthermore, this metabolic reprogramming increased the sensitivity of IDH1-mutant cells to hypoxia or electron transport chain inhibition in vitro. Lastly, IDH1-mutant cells also grew poorly as subcutaneous xenografts within a hypoxic in vivo microenvironment. Together, our results suggest therapeutic opportunities to exploit the metabolic vulnerabilities specific to IDH1 mutation.


International Journal of Cancer | 2010

Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T-cell lymphoma models: Defining molecular mechanisms of resistance.

Wenlin Shao; Joseph D. Growney; Yun Feng; Gregory O'Connor; Minying Pu; Wenjing Zhu; Yung-Mae Yao; Paul Kwon; Stephen Fawell; Peter Atadja

Panobinostat (LBH589) is a highly potent deacetylase inhibitor that has demonstrated clinical efficacy in patients with advanced cutaneous T‐cell lymphoma (CTCL). To gain a better understanding of the compound activity in this tumor type, we investigated the cellular and molecular effects of panobinostat using both in vitro and in vivo models of CTCL. All 4 tested CTCL cell lines exhibited very high sensitivity to panobinostat‐induced growth inhibition. However, only 2 of 4 lines exhibited significant response to the cytotoxic activity of panobinostat. In a CTCL xenograft mouse tumor model, panobinostat treatment resulted in complete tumor regression. The difference in cell sensitivity to panobinostat‐induced death enabled us to further investigate potential mechanisms responsible for tumor sensitivity or resistance. In CTCL cell lines that were insensitive to panobinostat‐induced apoptosis, constitutively activated NF‐κB and high levels of Bcl‐2 were observed. Inhibition of Bcl‐2 sensitized cells to the cytotoxic activity of panobinostat. Conversely, knockdown of Bax diminished the CTCL cell sensitivity. Interestingly, panobinostat could induce cytotoxicity in vorinostat‐resistant CTCL cells by downregulating phosphorylated STAT3 and STAT5 proteins. These studies suggest distinct mechanisms responsible for resistance to different deacetylase inhibitors. We show that the intrinsic apoptotic signaling plays an essential role in mediating panobinostat anticancer activity. Moreover, cancer cell sensitivity to panobinostat treatment may be further improved by combination with inhibition of anti‐apoptotic factors. These data provide preclinical support that panobinostat, as a single agent or in combination with other anticancer agents, is a promising therapy for CTCL.


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 | 2011

Optimization of the in Vitro Cardiac Safety of Hydroxamate-Based Histone Deacetylase Inhibitors

Michael Shultz; Xueying Cao; Christine Hiu-Tung Chen; Young Shin Cho; Nicole R. Davis; Joe Eckman; Jianmei Fan; Alex Fekete; Brant Firestone; Julie Flynn; Jack Green; Joseph D. Growney; Mats Holmqvist; Meier Hsu; Daniel Jansson; Lei Jiang; Paul Kwon; Gang Liu; Franco Lombardo; Qiang Lu; Dyuti Majumdar; Christopher Meta; Lawrence Blas Perez; Minying Pu; Tim Ramsey; Stacy W. Remiszewski; Suzanne Skolnik; Martin Traebert; Laszlo Urban; Vinita Uttamsingh

Histone deacetylase (HDAC) inhibitors have shown promise in treating various forms of cancer. However, many HDAC inhibitors from diverse structural classes have been associated with QT prolongation in humans. Inhibition of the human ether a-go-go related gene (hERG) channel has been associated with QT prolongation and fatal arrhythmias. To determine if the observed cardiac effects of HDAC inhibitors in humans is due to hERG blockade, a highly potent HDAC inhibitor devoid of hERG activity was required. Starting with dacinostat (LAQ824), a highly potent HDAC inhibitor, we explored the SAR to determine the pharmacophores required for HDAC and hERG inhibition. We disclose here the results of these efforts where a high degree of pharmacophore homology between these two targets was discovered. This similarity prevented traditional strategies for mitigating hERG binding/modulation from being successful and novel approaches for reducing hERG inhibition were required. Using a hERG homology model, two compounds, 11r and 25i, were discovered to be highly efficacious with weak affinity for the hERG and other ion channels.


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.


ACS Medicinal Chemistry Letters | 2017

Discovery and Evaluation of Clinical Candidate IDH305, a Brain Penetrant Mutant IDH1 Inhibitor

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

Inhibition of mutant IDH1 is being evaluated clinically as a promising treatment option for various cancers with hotspot mutation at Arg132. Having identified an allosteric, induced pocket of IDH1R132H, we have explored 3-pyrimidin-4-yl-oxazolidin-2-ones as mutant IDH1 inhibitors for in vivo modulation of 2-HG production and potential brain penetration. We report here optimization efforts toward the identification of clinical candidate IDH305 (13), a potent and selective mutant IDH1 inhibitor that has demonstrated brain exposure in rodents. Preclinical characterization of this compound exhibited in vivo correlation of 2-HG reduction and efficacy in a patient-derived IDH1 mutant xenograft tumor model. IDH305 (13) has progressed into human clinical trials for the treatment of cancers with IDH1 mutation.


Cancer Research | 2014

Abstract LB-139: IDH1 mutations alter citric acid cycle metabolism and increase dependence on oxidative mitochondrial metabolism

Alexandra R. Grassian; Seth J. Parker; Shawn M. Davidson; Ajit S. Divakaruni; Courtney R. Green; Xiamei Zhang; Kelly Slocum; Minying Pu; Fallon Lin; Chad Vickers; Carol Joud-Caldwell; Franklin Chung; Hong Yin; Erika Handly; Christopher Sean Straub; Joseph D. Growney; Matt Vander Heiden; Anne N. Murphy; Raymond Pagliarini; Christian M. Metallo

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Mutations in the genes encoding isocitrate dehydrogenase 1 and 2 (IDH1/2) occur in a variety of tumor types, resulting in production of the proposed oncometabolite, 2-hydroxyglutarate (2-HG). How mutant IDH alters central carbon metabolism, though, remains unclear. To address this question, we performed 13C metabolic flux analysis (MFA) on an isogenic cell panel containing heterozygous IDH1/2 mutations. We observe a dramatic and consistent decrease in the ability of IDH1, but not IDH2, mutant cell lines to utilize reductive glutamine metabolism via the carboxylation of α-ketoglutarate to isocitrate. Additionally we find that cells with IDH1 mutations exhibit increased oxidative tricarboxylic acid (TCA) metabolism. Similar metabolic trends were observed in vivo as well, and also in endogenous, non-engineered IDH1/2 mutant cell lines. Interestingly, IDH1-mutant specific inhibitors were unable to reverse the decrease in reductive metabolism, suggesting that this metabolic phenotype is independent of 2-HG. Furthermore, this metabolic reprogramming increases the sensitivity of IDH1 mutant cells to hypoxia or electron transport chain (ETC) inhibition in vitro . IDH1 mutant cells also grow poorly as subcutaneous xenografts within hypoxic in vivo microenvironments. These results suggest that exploiting metabolic defects specific to IDH1 mutant cells could be an interesting avenue to explore therapeutically. Citation Format: Alexandra R. Grassian, Seth Parker, Shawn Davidson, Ajit Divakaruni, Courtney Green, Xiamei Zhang, Kelly Slocum, Minying Pu, Fallon Lin, Chad Vickers, Carol Joud-Caldwell, Franklin Chung, Hong Yin, Erika Handly, Christopher Straub, Joseph D. Growney, Matt Vander Heiden, Anne Murphy, Raymond Pagliarini, Christian Metallo. IDH1 mutations alter citric acid cycle metabolism and increase dependence on oxidative mitochondrial metabolism. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr LB-139. doi:10.1158/1538-7445.AM2014-LB-139


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

Collaboration


Dive into the Minying Pu's collaboration.

Researchain Logo
Decentralizing Knowledge