Network


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

Hotspot


Dive into the research topics where Qibin Su is active.

Publication


Featured researches published by Qibin Su.


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.


Journal of Medicinal Chemistry | 2014

Discovery of 1-methyl-1H-imidazole derivatives as potent Jak2 inhibitors.

Qibin Su; Stephanos Ioannidis; Claudio Chuaqui; Lynsie Almeida; Marat Alimzhanov; Geraldine A. Bebernitz; Kirsten Bell; Michael Howard Block; Tina Howard; Shan Huang; Dennis Huszar; Jon Read; Caroline Rivard Costa; Jie Shi; Mei Su; Minwei Ye; Michael Zinda

Structure based design, synthesis, and biological evaluation of a novel series of 1-methyl-1H-imidazole, as potent Jak2 inhibitors to modulate the Jak/STAT pathway, are described. Using the C-ring fragment from our first clinical candidate AZD1480 (24), optimization of the series led to the discovery of compound 19a, a potent, orally bioavailable Jak2 inhibitor. Compound 19a displayed a high level of cellular activity in hematopoietic cell lines harboring the V617F mutation and in murine BaF3 TEL-Jak2 cells. Compound 19a demonstrated significant tumor growth inhibition in a UKE-1 xenograft model within a well-tolerated dose range.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of azabenzimidazole derivatives as potent, selective inhibitors of TBK1/IKKε kinases.

Tao Wang; Michael A. Block; Scott Cowen; Audrey Davies; Erik Devereaux; Lakshmaiah Gingipalli; Jeffrey W. Johannes; Nicholas A. Larsen; Qibin Su; Julie A. Tucker; David Whitston; Jiaquan Wu; Hai-Jun Zhang; Michael Zinda; Claudio Chuaqui

The design, synthesis and biological evaluation of a series of azabenzimidazole derivatives as TBK1/IKKε kinase inhibitors are described. Starting from a lead compound 1a, iterative design and SAR exploitation of the scaffold led to analogues with nM enzyme potencies against TBK1/IKKε. These compounds also exhibited excellent cellular activity against TBK1. Further structure-based design to improve selectivity over CDK2 and Aurora B resulted in compounds such as 5b-e. These probe compounds will facilitate study of the complex cancer biology of TBK1 and IKKε.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of novel hedgehog antagonists from cell-based screening: Isosteric modification of p38 bisamides as potent inhibitors of SMO

Bin Yang; Alexander Hird; Daniel John Russell; Benjamin Fauber; Les A. Dakin; Xiaolan Zheng; Qibin Su; Robert Godin; Patrick Brassil; Erik Devereaux; James W. Janetka

Cell-based subset screening of compounds using a Gli transcription factor reporter cell assay and shh stimulated cell differentiation assay identified a series of bisamide compounds as hedgehog pathway inhibitors with good potency. Using a ligand-based optimization strategy, heteroaryl groups were utilized as conformationally restricted amide isosteres replacing one of the amides which significantly increased their potency against SMO and the hedgehog pathway while decreasing activity against p38α kinase. We report herein the identification of advanced lead compounds such as imidazole 11c and 11f encompassing good p38α selectivity, low nanomolar potency in both cell assays, excellent physiochemical properties and in vivo pharmacokinetics.


Expert Opinion on Therapeutic Patents | 2017

Inhibitors of JAK-family kinases: an update on the patent literature 2013-2015, part 1.

Jason Grant Kettle; Annika Åstrand; Matthew Catley; Neil Grimster; Magnus K Nilsson; Qibin Su; Richard Woessner

ABSTRACT Introduction: Janus kinases (JAKs) are a family of four enzymes; JAK1, JAK2, JAK3 and tyrosine kinase 2 (TYK2) that are critical in cytokine signalling and are strongly linked to both cancer and inflammatory diseases. There are currently two launched JAK inhibitors for the treatment of human conditions: tofacitinib for Rheumatoid arthritis (RA) and ruxolitinib for myeloproliferative neoplasms including intermediate or high risk myelofibrosis and polycythemia vera. Areas covered: This review covers patents claiming activity against one or more JAK family members in the period 2013–2015 inclusive, and covers 95 patents from 42 applicants, split over two parts. The authors have ordered recent patents according to the primary applicant’s name, with part 1 covering A through to I. Expert opinion: Inhibition of JAK-family kinases is an area of growing interest, catalysed by the maturity of data on marketed inhibitors ruxolitinib and tofacitinib in late stage clinical trials. Many applicants are pursuing traditional fast-follower strategies around these inhibitors, with a range of chemical strategies adopted. The challenge will be to show sufficient differentiation to the originator compounds, since dose limiting toxicities with such agents appear to be on target and mechanism-related and also considering that such agents may be available as generic compounds by the time follower agents reach market.


Journal of Medicinal Chemistry | 2015

Identification and Optimization of Benzimidazole Sulfonamides as Orally Bioavailable Sphingosine 1-Phosphate Receptor 1 Antagonists with in Vivo Activity.

Edward J. Hennessy; Vibha Oza; Ammar Adam; Kate Byth; Lillian Castriotta; Gurmit Grewal; Geraldine A. Hamilton; Victor Kamhi; Paula Lewis; Danyang Li; Paul Lyne; Linda Öster; Michael T. Rooney; Jamal C. Saeh; Li Sha; Qibin Su; Shengua Wen; Yafeng Xue; Bin Yang

We report here a novel series of benzimidazole sulfonamides that act as antagonists of the S1P1 receptor, identified by exploiting an understanding of the pharmacophore of a high throughput screening (HTS)-derived series of compounds described previously. Lead compound 2 potently inhibits S1P-induced receptor internalization in a cell-based assay (EC50 = 0.05 μM), but has poor physical properties and metabolic stability. Evolution of this compound through structure-activity relationship development and property optimization led to in vivo probes such as 4. However, this compound was unexpectedly found to be a potent CYP3A inducer in human hepatocytes, and thus further chemistry efforts were directed at addressing this liability. By employing a pregnane X receptor (PXR) reporter gene assay to prioritize compounds for further testing in human hepatocytes, we identified lipophilicity as a key molecular property influencing the likelihood of P450 induction. Ultimately, we have identified compounds such as 46 and 47, which demonstrate the desired S1P1 antagonist activity while having greatly reduced risk of CYP3A induction in humans. These compounds have excellent oral bioavailability in preclinical species and exhibit pharmacodynamic effects of S1P1 antagonism in several in vivo models following oral dosing. Relatively modest antitumor activity was observed in multiple xenograft models, however, suggesting that selective S1P1 antagonists would have limited utility as anticancer therapeutics as single agents.


Bioorganic & Medicinal Chemistry Letters | 2016

Identification of azabenzimidazoles as potent JAK1 selective inhibitors.

Melissa Vasbinder; Marat Alimzhanov; Martin Augustin; Geraldine Bebernitz; Kirsten Bell; Claudio Chuaqui; Tracy L. Deegan; Andrew D. Ferguson; Kelly Goodwin; Dennis Huszar; Aarti Kawatkar; Sameer Kawatkar; Jon Read; Jie Shi; Stefan Steinbacher; Holger Steuber; Qibin Su; Dorin Toader; Haixia Wang; Richard Woessner; Allan Wu; Minwei Ye; Michael Zinda

We have identified a class of azabenzimidazoles as potent and selective JAK1 inhibitors. Investigations into the SAR are presented along with the structural features required to achieve selectivity for JAK1 versus other JAK family members. An example from the series demonstrated highly selective inhibition of JAK1 versus JAK2 and JAK3, along with inhibition of pSTAT3 in vivo, enabling it to serve as a JAK1 selective tool compound to further probe the biology of JAK1 selective inhibitors.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of heterocyclic sulfonamides as sphingosine 1-phosphate receptor 1 (S1P1) antagonists

Edward J. Hennessy; Gurmit Grewal; Kate Byth; Victor Kamhi; Danyang Li; Paul Lyne; Vibha Oza; Lucienne Ronco; Michael T. Rooney; Jamal C. Saeh; Qibin Su

We have discovered a novel class of heterocyclic sulfonamides that act as antagonists of the S1P1 receptor. While members of this series identified from a high-throughput screen showed promising levels of potency in a cell-based assay measuring the inhibition of receptor internalization, most compounds were excessively lipophilic and contained an oxidation-prone thioether moiety. As a result, such compounds suffered from poor physical properties and metabolic stability, limiting their utility as in vivo probes. By removing the thioether group and systematically developing an understanding of structure-activity relationships and the effects of lipophilicity on potency within this series, we have been able to identify potent compounds with vastly improved physical properties. A representative enantiopure triazole sulfonamide (33) has measurable bioavailability following a low (3mg/kg) oral dose in rat, highlighting an achievement of the early hit-to-lead efforts for this series.


Journal of Medicinal Chemistry | 2018

Adventures in Scaffold Morphing: Discovery of Fused Ring Heterocyclic Checkpoint Kinase 1 (CHK1) Inhibitors

Bin Yang; Melissa Vasbinder; Alexander Hird; Qibin Su; Haixia Wang; Yan Yu; Dorin Toader; Paul Lyne; Jon Read; Jason Breed; Stephanos Ioannidis; Chun Deng; Michael Grondine; Nancy DeGrace; David Whitston; Patrick Brassil; James W. Janetka

Checkpoint kinase 1 (CHK1) inhibitors are potential cancer therapeutics that can be utilized for enhancing the efficacy of DNA damaging agents. Multiple small molecule CHK1 inhibitors from different chemical scaffolds have been developed and evaluated in clinical trials in combination with chemotherapeutics and radiation treatment. Scaffold morphing of thiophene carboxamide ureas (TCUs), such as AZD7762 (1) and a related series of triazoloquinolines (TZQs), led to the identification of fused-ring bicyclic CHK1 inhibitors, 7-carboxamide thienopyridines (7-CTPs), and 7-carboxamide indoles. X-ray crystal structures reveal a key intramolecular noncovalent sulfur-oxygen interaction in aligning the hinge-binding carboxamide group to the thienopyridine core in a coplanar fashion. An intramolecular hydrogen bond to an indole NH was also effective in locking the carboxamide in the preferred bound conformation to CHK1. Optimization on the 7-CTP series resulted in the identification of lead compound 44, which displayed respectable drug-like properties and good in vitro and in vivo potency.


Journal of Medicinal Chemistry | 2018

Discovery and Optimization of a Novel Series of Highly Selective JAK1 Kinase Inhibitors

Neil Grimster; Erica Anderson; Marat Alimzhanov; Geraldine A. Bebernitz; Kirsten Bell; Claudio Chuaqui; Tracy L. Deegan; Andrew D. Ferguson; Thomas Gero; Andreas Harsch; Dennis Huszar; Aarti Kawatkar; Jason Grant Kettle; Paul Lyne; Jon Read; Caroline Rivard Costa; Linette Ruston; Patricia Schroeder; Jie Shi; Qibin Su; Scott Throner; Dorin Toader; Melissa Vasbinder; Richard Woessner; Haixia Wang; Allan Wu; Minwei Ye; Weijia Zheng; Michael Zinda

Janus kinases (JAKs) have been demonstrated to be critical in cytokine signaling and have thus been implicated in both cancer and inflammatory diseases. The JAK family consists of four highly homologous members: JAK1-3 and TYK2. The development of small-molecule inhibitors that are selective for a specific family member would represent highly desirable tools for deconvoluting the intricacies of JAK family biology. Herein, we report the discovery of a potent JAK1 inhibitor, 24, which displays ∼1000-fold selectivity over the other highly homologous JAK family members (determined by biochemical assays), while also possessing good selectivity over other kinases (determined by panel screening). Moreover, this compound was demonstrated to be orally bioavailable and possesses acceptable pharmacokinetic parameters. In an in vivo study, the compound was observed to dose dependently modulate the phosphorylation of STAT3 (a downstream marker of JAK1 inhibition).

Collaboration


Dive into the Qibin 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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge