Network


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

Hotspot


Dive into the research topics where Deqiang Niu is active.

Publication


Featured researches published by Deqiang Niu.


Cancer Discovery | 2013

Discovery of a Mutant-Selective Covalent Inhibitor of EGFR that Overcomes T790M-Mediated Resistance in NSCLC

Annette O Walter; Robert Tjin Tham Sjin; Henry J Haringsma; Kadoaki Ohashi; Jing Sun; Kwangho Lee; Aleksander Dubrovskiy; Matthew T. Labenski; Zhendong Zhu; Zhigang Wang; Michael Sheets; Thia St Martin; Russell Karp; Dan van Kalken; Prasoon Chaturvedi; Deqiang Niu; M. Nacht; Russell C. Petter; William F. Westlin; Kevin Lin; Sarah S. Jaw-Tsai; Mitch Raponi; Terry Van Dyke; Jeff Etter; Zoe Weaver; William Pao; Juswinder Singh; Andrew Simmons; Thomas Harding; Andrew E. Allen

UNLABELLED Patients with non-small cell lung cancer (NSCLC) with activating EGF receptor (EGFR) mutations initially respond to first-generation reversible EGFR tyrosine kinase inhibitors. However, clinical efficacy is limited by acquired resistance, frequently driven by the EGFR(T790M) mutation. CO-1686 is a novel, irreversible, and orally delivered kinase inhibitor that specifically targets the mutant forms of EGFR, including T790M, while exhibiting minimal activity toward the wild-type (WT) receptor. Oral administration of CO-1686 as single agent induces tumor regression in EGFR-mutated NSCLC tumor xenograft and transgenic models. Minimal activity of CO-1686 against the WT EGFR receptor was observed. In NSCLC cells with acquired resistance to CO-1686 in vitro, there was no evidence of additional mutations or amplification of the EGFR gene, but resistant cells exhibited signs of epithelial-mesenchymal transition and demonstrated increased sensitivity to AKT inhibitors. These results suggest that CO-1686 may offer a novel therapeutic option for patients with mutant EGFR NSCLC. SIGNIFICANCE We report the preclinical development of a novel covalent inhibitor, CO-1686, that irreversibly and selectively inhibits mutant EGFR, in particular the T790M drug-resistance mutation, in NSCLC models. CO-1686 is the fi rst drug of its class in clinical development for the treatment of T790M-positive NSCLC, potentially offering potent inhibition of mutant EGFR while avoiding the on-target toxicity observed with inhibition of the WT EGFR.


Nature Chemical Biology | 2011

Selective irreversible inhibition of a protease by targeting a noncatalytic cysteine

Margit Hagel; Deqiang Niu; Thia St. Martin; Michael Sheets; Lixin Qiao; Hugues Bernard; Russell Karp; Zhendong Zhu; Matthew T. Labenski; Prasoon Chaturvedi; Mariana Nacht; William F. Westlin; Russell C. Petter; Juswinder Singh

Designing selective inhibitors of proteases has proven problematic, in part because pharmacophores that confer potency exploit the conserved catalytic apparatus. We developed a fundamentally different approach by designing irreversible inhibitors that target noncatalytic cysteines that are structurally unique to a target in a protein family. We have successfully applied this approach to the important therapeutic target HCV protease, which has broad implications for the design of other selective protease inhibitors.


Molecular Cancer Therapeutics | 2014

In vitro and In vivo Characterization of Irreversible Mutant-Selective EGFR Inhibitors that are Wild-type Sparing

Robert Tjin Tham Sjin; Kwangho Lee; Annette O. Walter; Aleksandr Dubrovskiy; Michael Sheets; Thia St Martin; Matthew T. Labenski; Zhendong Zhu; Richland Wayne Tester; Russell Karp; Aravind Prasad Medikonda; Prasoon Chaturvedi; Yixuan Ren; Henry J. Haringsma; Jeff Etter; Mitch Raponi; Andrew Simmons; Thomas C. Harding; Deqiang Niu; M. Nacht; William F. Westlin; Russell C. Petter; Andrew M. Allen; Juswinder Singh

Patients with non–small cell lung carcinoma (NSCLC) with activating mutations in epidermal growth factor receptor (EGFR) initially respond well to the EGFR inhibitors erlotinib and gefitinib. However, all patients relapse because of the emergence of drug-resistant mutations, with T790M mutations accounting for approximately 60% of all resistance. Second-generation irreversible EGFR inhibitors are effective against T790M mutations in vitro, but retain affinity for wild-type EGFR (EGFRWT). These inhibitors have not provided compelling clinical benefit in T790M-positive patients, apparently because of dose-limiting toxicities associated with inhibition of EGFRWT. Thus, there is an urgent clinical need for therapeutics that overcome T790M drug resistance while sparing EGFRWT. Here, we describe a lead optimization program that led to the discovery of four potent irreversible 2,4-diaminopyrimidine compounds that are EGFR mutant (EGFRmut) selective and have been designed to have low affinity for EGFRWT. Pharmacokinetic and pharmacodynamic studies in H1975 tumor–bearing mice showed that exposure was dose proportional resulting in dose-dependent EGFR modulation. Importantly, evaluation of normal lung tissue from the same animals showed no inhibition of EGFRWT. Of all the compounds tested, compound 3 displayed the best efficacy in EGFRL858R/T790M-driven tumors. Compound 3, now renamed CO-1686, is currently in a phase I/II clinical trial in patients with EGFRmut-advanced NSCLC that have received prior EGFR-directed therapy. Mol Cancer Ther; 13(6); 1468–79. ©2014 AACR.


Journal of Medicinal Chemistry | 2013

Discovery of a potent and isoform-selective targeted covalent inhibitor of the lipid kinase PI3Kα.

Mariana Nacht; Lixin Qiao; Michael Sheets; Thia St. Martin; Matthew T. Labenski; Hormoz Mazdiyasni; Russell Karp; Zhendong Zhu; Prasoon Chaturvedi; Deepa Bhavsar; Deqiang Niu; William F. Westlin; Russell C. Petter; Aravind Prasad Medikonda; Juswinder Singh

PI3Kα has been identified as an oncogene in human tumors. By use of rational drug design, a targeted covalent inhibitor 3 (CNX-1351) was created that potently and specifically inhibits PI3Kα. We demonstrate, using mass spectrometry and X-ray crystallography, that the selective inhibitor covalently modifies PI3Kα on cysteine 862 (C862), an amino acid unique to the α isoform, and that PI3Kβ, -γ, and -δ are not covalently modified. 3 is able to potently (EC(50) < 100 nM) and specifically inhibit signaling in PI3Kα-dependent cancer cell lines, and this leads to a potent antiproliferative effect (GI(50) < 100 nM). A covalent probe, 8 (CNX-1220), which selectively bonds to PI3Kα, was used to investigate the duration of occupancy of 3 with PI3Kα in vivo. This is the first report of a PI3Kα-selective inhibitor, and these data demonstrate the biological impact of selectively targeting PI3Kα.


Journal of Immunology | 2015

The Tec Kinase–Regulated Phosphoproteome Reveals a Mechanism for the Regulation of Inhibitory Signals in Murine Macrophages

Giacomo Tampella; Hannah M. Kerns; Deqiang Niu; Swati Singh; Socheath Khim; Katherine A. Bosch; Meghan E. Garrett; Albanus O. Moguche; Erica Evans; Beth Browning; Tahmina A. Jahan; M. Nacht; Alejandro Wolf-Yadlin; Alessandro Plebani; Jessica A. Hamerman; David J. Rawlings; Richard G. James

Previous work has shown conflicting roles for Tec family kinases in regulation of TLR-dependent signaling in myeloid cells. In the present study, we performed a detailed investigation of the role of the Tec kinases Btk and Tec kinases in regulating TLR signaling in several types of primary murine macrophages. We demonstrate that primary resident peritoneal macrophages deficient for Btk and Tec secrete less proinflammatory cytokines in response to TLR stimulation than do wild-type cells. In contrast, we found that bone marrow–derived and thioglycollate-elicited peritoneal macrophages deficient for Btk and Tec secrete more proinflammatory cytokines than do wild-type cells. We then compared the phosphoproteome regulated by Tec kinases and LPS in primary peritoneal and bone marrow–derived macrophages. From this analysis we determined that Tec kinases regulate different signaling programs in these cell types. In additional studies using bone marrow–derived macrophages, we found that Tec and Btk promote phosphorylation events necessary for immunoreceptor-mediated inhibition of TLR signaling. Taken together, our results are consistent with a model where Tec kinases (Btk, Tec, Bmx) are required for TLR-dependent signaling in many types of myeloid cells. However, our data also support a cell type–specific TLR inhibitory role for Btk and Tec that is mediated by immunoreceptor activation and signaling via PI3K.


Molecular Cancer Therapeutics | 2011

Abstract C189: CO-1686, an orally available, mutant-selective inhibitor of the epidermal growth factor receptor (EGFR), causes tumor shrinkage in non-small cell lung cancer (NSCLC) with T790M mutations.

Annette Walter; Robert Tjin; Henry J Haringsma; Kevin Lin; Alex Dubrovskiy; Kwangho Lee; Thia St. Martin; Russell Karp; Zhendong Zhu; Deqiang Niu; Mariana Nacht; Russell C. Petter; William F. Westlin; Juswinder Singh; Mitch Raponi; Andrew E. Allen

Introduction: Non-small cell lung cancer (NSCLC) patients with activating EGFR mutations initially respond well to EGFR tyrosine kinase inhibitors. However, clinical efficacy is limited by the development of resistance. The most common mechanism of resistance is a second site mutation within exon 20 of EGFR (T790M), observed in ∼50% of cases. Our goal was to develop a mutant-selective EGFR inhibitor that potently inhibits activating EGFR mutations as well as the T790M resistance mutation while sparing wild-type EGFR for the treatment of NSCLC patients. Such a drug has the potential to effectively treat first- and second-line NSCLC patients with EGFR mutations without causing the dose limiting toxicities associated with EGFR kinase inhibitors currently in clinical development. Experimental procedures: Using structure-based drug design, we identified CO-1686, a covalent, irreversible small molecule, which selectively inhibits mutant EGFR. We assessed antitumor activity of CO-1686 both in vitro and in vivo in two NSCLC cell lines harboring EGFR mutations: H1975 (EGFR L858R/T790M) and HCC827 (EGFR delE746-A750). We evaluated inhibition of EGFR phosphorylation and downstream signaling by immunoblot analysis in cells and tissue samples. IHC staining on skin samples was performed to address effects on wild-type EGFR. Results: CO-1686 is a potent inhibitor of cell proliferation and EGFR signaling in NSCLC cells harboring the single activating mutation EGFR delE746-A750 as well as the double mutation EGFR L858R/T790M. When administered orally, CO-1686 (3 − 100 mg/kg) significantly suppresses tumor growth of H1975 cells (L858R/T790M) in a dose-dependent manner causing tumor regressions at the highest dose (100 mg/kg) without affecting body weight. Erlotinib at the same dose exhibits no effect against H1975 xenografts. In HCC827 (delE746-A750) xenografts, both agents cause tumor shrinkage. In both NSCLC mouse models, inhibition of EGFR phosphorylation in tumors correlate with the observed anti-tumor activity, while no effect on EGFR signaling is observed in normal lung or skin tissues with CO-1686 treatment, confirming that CO-1686 does not inhibt wild-type EGFR. Conclusions: Our results establish CO-1686 as a mutant-selective, wild-type sparing EGFR inhibitor with in vivo efficacy against tumors with activating EGFR mutations as well as the resistance mutation T790M. These data suggest that treatment with CO-1686 as a single agent can overcome T790M-mediated drug resistance in NSCLC. This hypothesis will be tested clinically. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr C189.


Cancer Research | 2011

Abstract 4482: Discovery of an irreversible PI3Kα-specific Inhibitor

Lixin Qiao; Mariana Nacht; Michael Sheets; Thia St. Martin; Matthew T. Labenski; Hormoz Mazdiyasni; Zhendong Zhu; Prasoon Chaturvedi; Deepa Bhavsar; Deqiang Niu; William F. Westlin; Russell C. Petter; Juswinder Singh

The PI3K pathway, which regulates cell growth, proliferation and survival, is activated in many types of human tumors by mutational activation of PI3Kα, PTEN inactivation, or activation of upstream receptor tyrosine kinases. Several PI3K inhibitors are currently in clinical development, but most are pan-PI3K inhibitors. Tumor biology data suggests that targeting PI3Kα specifically should be efficacious, and there may be advantages to not disrupting other members of the complex PI3K signaling cascade. Using structure-based drug design (SBDD), we have discovered a series of small molecules that selectively inhibit PI3Kα via the formation of an irreversible covalent bond. In addition to the enhanced pharmacodynamics achieved through permanent silencing of targets, covalent inhibitors also exhibit key advantages in terms of isoform selectivity and translational biomarker opportunities. SBDD was used to design small molecules which bond to an amino acid present uniquely in the α-isoform of PI3K. Mass spectrometry verified covalent bond formation to PI3Kα but not to the other isoforms. PI3K enzyme activity was measured using an HTRF assay. PI3Kα inhibition was evaluated in SKOV-3 cells by measuring P-AktSer473 levels. Washout experiments were performed to assess prolonged duration of action in cells. Effects on cell proliferation were assessed in cell lines bearing different mutations, such as SKOV-3 (PIK3CA H1047R), PC3 (PTEN-null) and others. SKOV-3 xenograft studies were performed in mice to measure P-AktSer473 inhibition and tumor growth inhibition in vivo. A biotinylated covalent probe molecule specific for PI3Kα was used to verify and quantitate target occupancy by the covalent inhibitor, both in vitro and ex vivo. Using trypsin digestion and MS-MS analysis, we confirmed that the designed inhibitors covalently bonded to the protein at the desired amino acid that is unique to PI3Kα. The covalent bonding resulted in potent inhibition of the PI3Kα enzyme activity and inhibition of P-AktSer473 in cells (EC50 Selective irreversible inhibitors of PI3Kα demonstrate prolonged duration of action and activity in vivo. This targeted approach should yield a first-in-class selective covalent PI3Kα inhibitor with clinical advantages. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4482. doi:10.1158/1538-7445.AM2011-4482


Archive | 2009

Heteroaryl compounds and uses thereof

Juswinder Singh; Russell C. Petter; Richland Wayne Tester; Arthur F. Kluge; Hormoz Mazdiyasni; William F. Westlin; Deqiang Niu; Lixin Qiao


Archive | 2009

2,4-disubstituted pyrimidines useful as kinase inhibitors

Juswinder Singh; Russell C. Petter; Richland Wayne Tester; Arthur F. Kluge; Hormoz Mazdiyasni; William F. Westlin; Deqiang Niu; Lixin Qiao


Archive | 2012

Pi3 kinase inhibitors and uses thereof

Deqiang Niu; Russell C. Petter; Juswinder Singh; Arthur F. Kluge; Hormoz Mazdiyasni; Zhendong Zhu; Lixin Qiao; Kevin Kuntz

Collaboration


Dive into the Deqiang Niu's collaboration.

Researchain Logo
Decentralizing Knowledge