Network


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

Hotspot


Dive into the research topics where Nancy Su is active.

Publication


Featured researches published by Nancy Su.


Journal of Biological Chemistry | 2008

Effects of the JAK2 Inhibitor, AZ960, on Pim/BAD/BCL-xL Survival Signaling in the Human JAK2 V617F Cell Line SET-2

Joseph M. Gozgit; Geraldine Bebernitz; Pankaj Patil; Minwei Ye; Julie Parmentier; Jiaquan Wu; Nancy Su; Tao Wang; Stephanos Ioannidis; Audrey Davies; Dennis Huszar; Michael Zinda

The Janus-associated kinase 2 (JAK2) V617F mutation is believed to play a critical role in the pathogenesis of polycythemia vera, essential thrombocythemia, and idiopathic myelofibrosis. We have characterized a novel small molecule JAK2 inhibitor, AZ960, and used it as a tool to investigate the consequences of JAK2 V617F inhibition in the SET-2 cell line. AZ960 inhibits JAK2 kinase with a Ki of 0.00045 μm in vitro and treatment of TEL-JAK2 driven Ba/F3 cells with AZ960 blocked STAT5 phosphorylation and potently inhibited cell proliferation (GI50 = 0.025 μm). AZ960 demonstrated selectivity for TEL-JAK2-driven STAT5 phosphorylation and cell proliferation when compared with cell lines driven by similar fusions of the other JAK kinase family members. In the SET-2 human megakaryoblastic cell line, heterozygous for the JAK2 V617F allele, inhibition of JAK2 resulted in decreased STAT3/5 phosphorylation and inhibition of cell proliferation (GI50 = 0.033 μm) predominately through the induction of mitochondrial-mediated apoptosis. We provide evidence that JAK2 inhibition induces apoptosis by direct and indirect regulation of the anti-apoptotic protein BCL-xL. Inhibition of JAK2 blocked BCL-XL mRNA expression resulting in a reduction of BCL-xL protein levels. Additionally, inhibition of JAK2 resulted in decreased PIM1 and PIM2 mRNA expression. Decreased PIM1 mRNA corresponded with a decrease in Pim1 protein levels and inhibition of BAD phosphorylation at Ser112. Finally, small interfering RNA-mediated suppression of BCL-xL resulted in apoptotic cell death similar to the phenotype observed following JAK2 inhibition. These results suggest a model in which JAK2 promotes cell survival by signaling through the Pim/BAD/BCL-xL pathway.


ACS Chemical Biology | 2013

Discovery and mechanistic study of a small molecule inhibitor for motor protein KIFC1.

Jiaquan Wu; Keith Mikule; Wenxian Wang; Nancy Su; Philip Petteruti; Farzin Gharahdaghi; Erin Code; Xiahui Zhu; Kelly Jacques; Zhongwu Lai; Bin Yang; Michelle Lamb; Claudio Chuaqui; Nicholas Keen; Huawei Chen

Centrosome amplification is observed in many human cancers and has been proposed to be a driver of both genetic instability and tumorigenesis. Cancer cells have evolved mechanisms to bundle multiple centrosomes into two spindle poles to avoid multipolar mitosis that can lead to chromosomal segregation defects and eventually cell death. KIFC1, a kinesin-14 family protein, plays an essential role in centrosomal bundling in cancer cells, but its function is not required for normal diploid cell division, suggesting that KIFC1 is an attractive therapeutic target for human cancers. To this end, we have identified the first reported small molecule inhibitor AZ82 for KIFC1. AZ82 bound specifically to the KIFC1/microtubule (MT) binary complex and inhibited the MT-stimulated KIFC1 enzymatic activity in an ATP-competitive and MT-noncompetitive manner with a Ki of 0.043 μM. AZ82 effectively engaged with the minus end-directed KIFC1 motor inside cells to reverse the monopolar spindle phenotype induced by the inhibition of the plus end-directed kinesin Eg5. Treatment with AZ82 caused centrosome declustering in BT-549 breast cancer cells with amplified centrosomes. Consistent with genetic studies, our data confirmed that KIFC1 inhibition by a small molecule holds promise for targeting cancer cells with amplified centrosomes and provided evidence that functional suppression of KIFC1 by inhibiting its enzymatic activity could be an effective means for developing cancer therapeutics.


Nature Chemical Biology | 2016

Inhibition of Mcl-1 through covalent modification of a noncatalytic lysine side chain

Gizem Akçay; Matthew A. Belmonte; Brian Aquila; Claudio Chuaqui; Alexander Hird; Michelle L. Lamb; Philip Rawlins; Nancy Su; Sharon Tentarelli; Neil Grimster; Qibin Su

Targeted covalent inhibition of disease-associated proteins has become a powerful methodology in the field of drug discovery, leading to the approval of new therapeutics. Nevertheless, current approaches are often limited owing to their reliance on a cysteine residue to generate the covalent linkage. Here we used aryl boronic acid carbonyl warheads to covalently target a noncatalytic lysine side chain, and generated to our knowledge the first reversible covalent inhibitors for Mcl-1, a protein-protein interaction (PPI) target that has proven difficult to inhibit via traditional medicinal chemistry strategies. These covalent binders exhibited improved potency in comparison to noncovalent congeners, as demonstrated in biochemical and cell-based assays. We identified Lys234 as the residue involved in covalent modification, via point mutation. The covalent binders discovered in this study will serve as useful starting points for the development of Mcl-1 therapeutics and probes to interrogate Mcl-1-dependent biological phenomena.


ACS Medicinal Chemistry Letters | 2015

Pyrimidinone Nicotinamide Mimetics as Selective Tankyrase and Wnt Pathway Inhibitors Suitable for in Vivo Pharmacology

Jeffrey W. Johannes; Lynsie Almeida; Bernard Barlaam; P. Ann Boriack-Sjodin; Robert Casella; Rosemary A. Croft; Allan Dishington; Lakshmaiah Gingipalli; Chungang Gu; Janet Hawkins; Jane L. Holmes; Tina Howard; Jian Huang; Stephanos Ioannidis; Steven Kazmirski; Michelle L. Lamb; Thomas M. McGuire; Jane E. Moore; Derek Ogg; Anil Patel; Kurt Gordon Pike; Timothy Pontz; Graeme R. Robb; Nancy Su; Haiyun Wang; Xiaoyun Wu; Hai-Jun Zhang; Yue Zhang; Xiaolan Zheng; Tao Wang

The canonical Wnt pathway plays an important role in embryonic development, adult tissue homeostasis, and cancer. Germline mutations of several Wnt pathway components, such as Axin, APC, and ß-catenin, can lead to oncogenesis. Inhibition of the poly(ADP-ribose) polymerase (PARP) catalytic domain of the tankyrases (TNKS1 and TNKS2) is known to inhibit the Wnt pathway via increased stabilization of Axin. In order to explore the consequences of tankyrase and Wnt pathway inhibition in preclinical models of cancer and its impact on normal tissue, we sought a small molecule inhibitor of TNKS1/2 with suitable physicochemical properties and pharmacokinetics for hypothesis testing in vivo. Starting from a 2-phenyl quinazolinone hit (compound 1), we discovered the pyrrolopyrimidinone compound 25 (AZ6102), which is a potent TNKS1/2 inhibitor that has 100-fold selectivity against other PARP family enzymes and shows 5 nM Wnt pathway inhibition in DLD-1 cells. Moreover, compound 25 can be formulated well in a clinically relevant intravenous solution at 20 mg/mL, has demonstrated good pharmacokinetics in preclinical species, and shows low Caco2 efflux to avoid possible tumor resistance mechanisms.


ACS Medicinal Chemistry Letters | 2012

Potent and Selective Inhibitors of CK2 Kinase Identified through Structure-Guided Hybridization

James E. Dowling; Claudio Chuaqui; Timothy Pontz; Paul Lyne; Nicholas A. Larsen; Michael Howard Block; Huawei Chen; Nancy Su; Allan Wu; Daniel John Russell; Hannah Pollard; John W. Lee; Bo Peng; Kumar Thakur; Qing Ye; Tao Zhang; Patrick Brassil; Vicki Racicot; Larry Bao; Christopher R. Denz; Emma L. Cooke

In this paper we describe a series of 3-cyano-5-aryl-7-aminopyrazolo[1,5-a]pyrimidine hits identified by kinase-focused subset screening as starting points for the structure-based design of conformationally constrained 6-acetamido-indole inhibitors of CK2. The synthesis, SAR, and effects of this novel series on Akt signaling and cell proliferation in vitro are described.


Journal of Medicinal Chemistry | 2014

Discovery of Potent KIFC1 Inhibitors Using a Method of Integrated High-Throughput Synthesis and Screening

Bin Yang; Michelle Lamb; Tao Zhang; Edward J. Hennessy; Gurmit Grewal; Li Sha; Mark Zambrowski; Michael Howard Block; James E. Dowling; Nancy Su; Jiaquan Wu; Tracy L. Deegan; Keith Mikule; Wenxian Wang; Rüdiger Kaspera; Claudio Chuaqui; Huawei Chen

KIFC1 (HSET), a member of the kinesin-14 family of motor proteins, plays an essential role in centrosomal bundling in cancer cells, but its function is not required for normal diploid cell division. To explore the potential of KIFC1 as a therapeutic target for human cancers, a series of potent KIFC1 inhibitors featuring a phenylalanine scaffold was developed from hits identified through high-throughput screening (HTS). Optimization of the initial hits combined both design-synthesis-test cycles and an integrated high-throughput synthesis and biochemical screening method. An important aspect of this integrated method was the utilization of DMSO stock solutions of compounds registered in the corporate compound collection as synthetic reactants. Using this method, over 1500 compounds selected for structural diversity were quickly assembled in assay-ready 384-well plates and were directly tested after the necessary dilutions. Our efforts led to the discovery of a potent KIFC1 inhibitor, AZ82, which demonstrated the desired centrosome declustering mode of action in cell studies.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of AZ0108, an orally bioavailable phthalazinone PARP inhibitor that blocks centrosome clustering

Jeffrey W. Johannes; Lynsie Almeida; Kevin Daly; Andrew D. Ferguson; Shaun Grosskurth; Huiping Guan; Tina Howard; Stephanos Ioannidis; Steven Kazmirski; Michelle Lamb; Nicholas A. Larsen; Paul Lyne; Keith Mikule; Claude Ogoe; Bo Peng; Philip Petteruti; Jon Read; Nancy Su; Mark Sylvester; Scott Throner; Wenxian Wang; Xin Wang; Jiaquan Wu; Qing Ye; Yan Yu; Xiaolan Zheng; David Scott

The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZenecas collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.


ACS Medicinal Chemistry Letters | 2017

Structure Based Design of Non-Natural Peptidic Macrocyclic Mcl-1 Inhibitors

Jeffrey W. Johannes; Stephanie Bates; Carl Beigie; Matthew A. Belmonte; John Breen; Shenggen Cao; Paolo A. Centrella; Matthew A. Clark; John W. Cuozzo; Christoph E. Dumelin; Andrew D. Ferguson; Sevan Habeshian; David Hargreaves; Camil Joubran; Steven Kazmirski; Anthony D. Keefe; Michelle L. Lamb; Haiye Lan; Yunxia Li; Hao Ma; Scott Mlynarski; Martin J. Packer; Philip Rawlins; Daniel W. Robbins; Haidong Shen; Eric A. Sigel; Holly H. Soutter; Nancy Su; Dawn M. Troast; Haiyun Wang

Mcl-1 is a pro-apoptotic BH3 protein family member similar to Bcl-2 and Bcl-xL. Overexpression of Mcl-1 is often seen in various tumors and allows cancer cells to evade apoptosis. Here we report the discovery and optimization of a series of non-natural peptide Mcl-1 inhibitors. Screening of DNA-encoded libraries resulted in hit compound 1, a 1.5 μM Mcl-1 inhibitor. A subsequent crystal structure demonstrated that compound 1 bound to Mcl-1 in a β-turn conformation, such that the two ends of the peptide were close together. This proximity allowed for the linking of the two ends of the peptide to form a macrocycle. Macrocyclization resulted in an approximately 10-fold improvement in binding potency. Further exploration of a key hydrophobic interaction with Mcl-1 protein and also with the moiety that engages Arg256 led to additional potency improvements. The use of protein-ligand crystal structures and binding kinetics contributed to the design and understanding of the potency gains. Optimized compound 26 is a <3 nM Mcl-1 inhibitor, while inhibiting Bcl-2 at only 5 μM and Bcl-xL at >99 μM, and induces cleaved caspase-3 in MV4-11 cells with an IC50 of 3 μM after 6 h.


ChemMedChem | 2018

Structure-Based Design of Selective Noncovalent CDK12 Inhibitors

Jeffrey W. Johannes; Christopher R. Denz; Nancy Su; Allan Wu; Anna C. Impastato; Scott Mlynarski; Jeffrey G. Varnes; D. Bryan Prince; Justin Cidado; Ning Gao; Malcolm Haddrick; Natalie H. Jones; Shaobin Li; Xiuwei Li; Yang Liu; Toan B. Nguyen; Nichole O'Connell; Emma Rivers; Daniel W. Robbins; Ronald Tomlinson; Tieguang Yao; Xiahui Zhu; Andrew D. Ferguson; Michelle Lamb; John Irvin Manchester; Sylvie Guichard

Cyclin‐dependent kinase (CDK) 12 knockdown via siRNA decreases the transcription of DNA‐damage‐response genes and sensitizes BRCA wild‐type cells to poly(ADP‐ribose) polymerase (PARP) inhibition. To recapitulate this effect with a small molecule, we sought a potent, selective CDK12 inhibitor. Crystal structures and modeling informed hybridization between dinaciclib and SR‐3029, resulting in lead compound 5 [(S)‐2‐(1‐(6‐(((6,7‐difluoro‐1H‐benzo[d]imidazol‐2‐yl)methyl)amino)‐9‐ethyl‐9H‐purin‐2‐yl)piperidin‐2‐yl)ethan‐1‐ol]. Further structure‐guided optimization delivered a series of selective CDK12 inhibitors, including compound 7 [(S)‐2‐(1‐(6‐(((6,7‐difluoro‐1H‐benzo[d]imidazol‐2‐yl)methyl)amino)‐9‐isopropyl‐9H‐purin‐2‐yl)piperidin‐2‐yl)ethan‐1‐ol]. Profiling of this compound across CDK9, 7, 2, and 1 at high ATP concentration, single‐point kinase panel screening against 352 targets at 0.1 μm, and proteomics via kinase affinity matrix technology demonstrated the selectivity. This series of compounds inhibits phosphorylation of Ser2 on the C‐terminal repeat domain of RNA polymerase II, consistent with CDK12 inhibition. These selective compounds were also acutely toxic to OV90 as well as THP1 cells.


ACS Medicinal Chemistry Letters | 2017

Correction to “Structure Based Design of Non-Natural Peptidic Macrocyclic Mcl-1 Inhibitors”

Jeffrey W. Johannes; Stephanie Bates; Carl Beigie; Matthew A. Belmonte; John Breen; Shenggen Cao; Paolo A. Centrella; Matthew A. Clark; John W. Cuozzo; Christoph E. Dumelin; Andrew D. Ferguson; Sevan Habeshian; David Hargreaves; Camil Joubran; Steven Kazmirski; Anthony D. Keefe; Michelle L. Lamb; Haiye Lan; Yunxia Li; Hao Ma; Scott Mlynarski; Martin J. Packer; Philip Rawlins; Daniel W. Robbins; Haidong Shen; Eric A. Sigel; Holly H. Soutter; Nancy Su; Dawn M. Troast; Haiyun Wang

[This corrects the article DOI: 10.1021/acsmedchemlett.6b00464.].

Collaboration


Dive into the Nancy Su's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge