Network


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

Hotspot


Dive into the research topics where Janet Gunzner-Toste is active.

Publication


Featured researches published by Janet Gunzner-Toste.


Science Translational Medicine | 2012

Ser1292 Autophosphorylation Is an Indicator of LRRK2 Kinase Activity and Contributes to the Cellular Effects of PD Mutations

Zejuan Sheng; Shuo Zhang; Daisy Bustos; Tracy Kleinheinz; Claire E. Le Pichon; Sara L. Dominguez; Hilda Solanoy; Jason Drummond; Xiaolin Zhang; Xiao Ding; Fang Cai; Qinghua Song; Xianting Li; Zhenyu Yue; Marcel van der Brug; Daniel J. Burdick; Janet Gunzner-Toste; Huifen Chen; Xingrong Liu; Anthony A. Estrada; Zachary Kevin Sweeney; Kimberly Scearce-Levie; John Moffat; Donald S. Kirkpatrick; Haitao Zhu

LRRK2 autophosphorylation on Ser1292 may be a useful indicator of kinase activity, providing a readout for screening candidate LRRK2 inhibitors. LRRK2 Inhibitor Heralds a Happier Song Genetic polymorphisms in the leucine-rich repeat kinase 2 (LRRK2) are the most common causes of familial Parkinson’s disease (PD) and are also linked to idiopathic PD. The most prevalent LRRK2 PD mutation G2019S imbues the kinase with a gain of function, suggesting that blocking LRRK2 activity may be a therapeutic strategy for reversing the pathogenic effects of LRRK2 mutations in PD. However, the mechanistic link between LRRK2 kinase activity and the cellular effects of PD mutations remains elusive, and there has been no reliable way to monitor LRRK2 kinase activity in vivo. Using quantitative mass spectrometry and subsequent phospho-specific antibody approaches, Sheng et al. now report that LRRK2 phosphorylates itself on Ser1292 in vitro and in vivo (Ser1292 autophosphorylation). Five of the six confirmed familial LRRK2 PD mutations increased Ser1292 autophosphorylation when transiently expressed in heterologous cells, suggesting increased Ser1292 autophosphorylation as a common feature of LRRK2 PD mutations. Elimination of the Ser1292 autophosphorylation site abrogated the defects on neurite outgrowth caused by LRRK2 PD mutations in cultured rat embryonic neurons. Using Ser1292 autophosphorylation as the readout of kinase activity, Sheng et al. developed assays to monitor LRRK2 kinase activity in cultured cells and rodents. These assays were used to profile the potencies of hundreds of LRRK2 kinase inhibitors derived from high-throughput compound screening. A potent and selective compound that effectively inhibited LRRK2 kinase activity in mouse brains and reversed cellular effects of LRRK2 PD mutations in cultured primary neurons was identified. Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common cause of familial Parkinson’s disease (PD). Although biochemical studies have shown that certain PD mutations confer elevated kinase activity in vitro on LRRK2, there are no methods available to directly monitor LRRK2 kinase activity in vivo. We demonstrate that LRRK2 autophosphorylation on Ser1292 occurs in vivo and is enhanced by several familial PD mutations including N1437H, R1441G/C, G2019S, and I2020T. Combining two PD mutations together further increases Ser1292 autophosphorylation. Mutation of Ser1292 to alanine (S1292A) ameliorates the effects of LRRK2 PD mutations on neurite outgrowth in cultured rat embryonic primary neurons. Using cell-based and pharmacodynamic assays with phosphorylated Ser1292 as the readout, we developed a brain-penetrating LRRK2 kinase inhibitor that blocks Ser1292 autophosphorylation in vivo and attenuates the cellular consequences of LRRK2 PD mutations in vitro. These data suggest that Ser1292 autophosphorylation may be a useful indicator of LRRK2 kinase activity in vivo and may contribute to the cellular effects of certain PD mutations.


Journal of Medicinal Chemistry | 2012

Discovery of highly potent, selective, and brain-penetrable leucine-rich repeat kinase 2 (LRRK2) small molecule inhibitors.

Anthony A. Estrada; Xingrong Liu; Charles Baker-Glenn; Alan Beresford; Daniel J. Burdick; Mark Stuart Chambers; Bryan K. Chan; Huifen Chen; Xiao Ding; Antonio G. DiPasquale; Sara L. Dominguez; Jennafer Dotson; Jason Drummond; Michael Flagella; Sean P. Flynn; Reina N. Fuji; Andrew Gill; Janet Gunzner-Toste; Seth F. Harris; Timothy P. Heffron; Tracy Kleinheinz; Donna W. Lee; Claire E. Le Pichon; Joseph P. Lyssikatos; Andrew D. Medhurst; John Moffat; Susmith Mukund; Kevin Nash; Kimberly Scearce-Levie; Zejuan Sheng

There is a high demand for potent, selective, and brain-penetrant small molecule inhibitors of leucine-rich repeat kinase 2 (LRRK2) to test whether inhibition of LRRK2 kinase activity is a potentially viable treatment option for Parkinsons disease patients. Herein we disclose the use of property and structure-based drug design for the optimization of highly ligand efficient aminopyrimidine lead compounds. High throughput in vivo rodent cassette pharmacokinetic studies enabled rapid validation of in vitro-in vivo correlations. Guided by this data, optimal design parameters were established. Effective incorporation of these guidelines into our molecular design process resulted in the discovery of small molecule inhibitors such as GNE-7915 (18) and 19, which possess an ideal balance of LRRK2 cellular potency, broad kinase selectivity, metabolic stability, and brain penetration across multiple species. Advancement of GNE-7915 into rodent and higher species toxicity studies enabled risk assessment for early development.


Journal of Medicinal Chemistry | 2012

Discovery of Selective LRRK2 Inhibitors Guided by Computational Analysis and Molecular Modeling

Huifen Chen; Bryan K. Chan; Jason Drummond; Anthony A. Estrada; Janet Gunzner-Toste; Xingrong Liu; Yichin Liu; John Moffat; Daniel Shore; Zachary Kevin Sweeney; Thuy Tran; Shumei Wang; Guiling Zhao; Haitao Zhu; Daniel J. Burdick

Mutations in the genetic sequence of leucine-rich repeat kinase 2 (LRRK2) have been linked to increased LRRK2 activity and risk for the development of Parkinsons disease (PD). Potent and selective small molecules capable of inhibiting the kinase activity of LRRK2 will be important tools for establishing a link between the kinase activity of LRRK2 and PD. In the absence of LRRK2 kinase domain crystal structures, a LRRK2 homology model was developed that provided robust guidance in the hit-to-lead optimization of small molecule LRRK2 inhibitors. Through a combination of molecular modeling, sequence analysis, and matched molecular pair (MMP) activity cliff analysis, a potent and selective lead inhibitor was discovered. The selectivity of this compound could be understood using the LRRK2 homology model, and application of this learning to a series of 2,4-diaminopyrimidine inhibitors in a scaffold hopping exercise led to the identification of highly potent and selective LRRK2 inhibitors that were also brain penetrable.


Journal of Medicinal Chemistry | 2013

Structure-Based Discovery of Novel Amide-Containing Nicotinamide Phosphoribosyltransferase (Nampt) Inhibitors

Xiaozhang Zheng; Paul H. Bauer; Timm Baumeister; Alexandre J. Buckmelter; Maureen Caligiuri; Karl H. Clodfelter; Bingsong Han; Yen-Ching Ho; Nikolai Kley; Jian Lin; Dominic J. Reynolds; Geeta Sharma; Chase Smith; Zhongguo Wang; Peter S. Dragovich; Janet Gunzner-Toste; Bianca M. Liederer; Justin Ly; Thomas O’Brien; Angela Oh; Leslie Wang; Weiru Wang; Yang Xiao; Mark Zak; Guiling Zhao; Po-wai Yuen; Kenneth W. Bair

Crystal structures of several urea- and thiourea-derived compounds in complex with the nicotinamide phosphoribosyltransferase (Nampt) protein were utilized to design a potent amide-containing inhibitor bearing an aza-indole moiety (7, Nampt BC IC50 = 9.0 nM, A2780 cell proliferation IC50 = 10 nM). The Nampt-7 cocrystal structure was subsequently obtained and enabled the design of additional amide-containing inhibitors which incorporated various other fused 6,5-heterocyclic moieties and biaryl sulfone or sulfonamide motifs. Additional modifications of these molecules afforded many potent biaryl sulfone-containing Nampt inhibitors which also exhibited favorable in vitro ADME properties (microsomal and hepatocyte stability, MDCK permeability, plasma protein binding). An optimized compound (58) was a potent inhibitor of multiple cancer cell lines (IC50 <10 nM vs U251, HT1080, PC3, MiaPaCa2, and HCT116 lines), displayed acceptable mouse PK properties (F = 41%, CL = 52.4 mL/min/kg), and exhibited robust efficacy in a U251 mouse xenograft model.


Journal of Medicinal Chemistry | 2013

Structure-Based Identification of Ureas as Novel Nicotinamide Phosphoribosyltransferase (Nampt) Inhibitors

Xiaozhang Zheng; Paul H. Bauer; Timm Baumeister; Alexandre J. Buckmelter; Maureen Caligiuri; Karl H. Clodfelter; Bingsong Han; Yen-Ching Ho; Nikolai Kley; Jian Lin; Dominic J. Reynolds; Geeta Sharma; Chase Smith; Zhongguo Wang; Peter S. Dragovich; Angela Oh; Weiru Wang; Mark Zak; Janet Gunzner-Toste; Guiling Zhao; Po-wai Yuen; Kenneth W. Bair

Nicotinamide phosphoribosyltransferase (Nampt) is a promising anticancer target. Virtual screening identified a thiourea analogue, compound 5, as a novel highly potent Nampt inhibitor. Guided by the cocrystal structure of 5, SAR exploration revealed that the corresponding urea compound 7 exhibited similar potency with an improved solubility profile. These studies also indicated that a 3-pyridyl group was the preferred substituent at one inhibitor terminus and also identified a urea moiety as the optimal linker to the remainder of the inhibitor structure. Further SAR optimization of the other inhibitor terminus ultimately yielded compound 50 as a urea-containing Nampt inhibitor which exhibited excellent biochemical and cellular potency (enzyme IC50 = 0.007 μM; A2780 IC50 = 0.032 μM). Compound 50 also showed excellent in vivo antitumor efficacy when dosed orally in an A2780 ovarian tumor xenograft model (TGI of 97% was observed on day 17).


Journal of Medicinal Chemistry | 2013

Fragment-Based Identification of Amides Derived from trans-2-(Pyridin-3-yl)cyclopropanecarboxylic Acid as Potent Inhibitors of Human Nicotinamide Phosphoribosyltransferase (NAMPT)

Anthony M. Giannetti; Xiaozhang Zheng; Nicholas J. Skelton; Weiru Wang; Brandon J. Bravo; Kenneth W. Bair; Timm Baumeister; Eric Cheng; Lisa Crocker; Yezhen Feng; Janet Gunzner-Toste; Yen-Ching Ho; Rongbao Hua; Bianca M. Liederer; Yongbo Liu; Xiaolei Ma; Thomas O’Brien; Jason Oeh; Deepak Sampath; Youming Shen; Chengcheng Wang; Leslie Wang; Hongxing Wu; Yang Xiao; Po-wai Yuen; Mark Zak; Guiling Zhao; Qiang Zhao; Peter S. Dragovich

Potent, trans-2-(pyridin-3-yl)cyclopropanecarboxamide-containing inhibitors of the human nicotinamide phosphoribosyltransferase (NAMPT) enzyme were identified using fragment-based screening and structure-based design techniques. Multiple crystal structures were obtained of initial fragment leads, and this structural information was utilized to improve the biochemical and cell-based potency of the associated molecules. Many of the optimized compounds exhibited nanomolar antiproliferative activities against human tumor lines in in vitro cell culture experiments. In a key example, a fragment lead (13, KD = 51 μM) was elaborated into a potent NAMPT inhibitor (39, NAMPT IC50 = 0.0051 μM, A2780 cell culture IC50 = 0.000 49 μM) which demonstrated encouraging in vivo efficacy in an HT-1080 mouse xenograft tumor model.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of potent and efficacious urea-containing nicotinamide phosphoribosyltransferase (NAMPT) inhibitors with reduced CYP2C9 inhibition properties.

Janet Gunzner-Toste; Guiling Zhao; Paul H. Bauer; Timm Baumeister; Alexandre J. Buckmelter; Maureen Caligiuri; Karl H. Clodfelter; B Fu; Bingsong Han; Yen-Ching Ho; Nikolai Kley; Xiaorong Liang; Bianca M. Liederer; Jian Lin; S Mukadam; Thomas O'Brien; Angela Oh; Dominic J. Reynolds; Geeta Sharma; Nicholas J. Skelton; Chase Smith; J Sodhi; Weiru Wang; Zhongguo Wang; Yang Xiao; Po-wai Yuen; Mark Zak; Lei Zhang; Xiaozhang Zheng; Kenneth W. Bair

Potent, reversible inhibition of the cytochrome P450 CYP2C9 isoform was observed in a series of urea-containing nicotinamide phosphoribosyltransferase (NAMPT) inhibitors. This unwanted property was successfully removed from the described inhibitors through a combination of structure-based design and medicinal chemistry activities. An optimized compound which did not inhibit CYP2C9 exhibited potent anti-NAMPT activity (17; BC NAMPT IC50=3 nM; A2780 antiproliferative IC50=70 nM), good mouse PK properties, and was efficacious in an A2780 mouse xenograft model. The crystal structure of this compound in complex with the NAMPT protein is also described.


Molecular Cancer Therapeutics | 2017

Modulating Therapeutic Activity and Toxicity of Pyrrolobenzodiazepine Antibody-Drug Conjugates with Self-Immolative Disulfide Linkers

Thomas H. Pillow; Melissa Schutten; Shang-Fan Yu; Rachana Ohri; Jack Sadowsky; Kirsten Achilles Poon; Willy Solis; Fiona Zhong; Geoffrey Del Rosario; Mary Ann T. Go; Jeffrey Lau; Sharon Yee; Jintang He; Luna Liu; Carl Ng; Keyang Xu; Douglas D. Leipold; Amrita V. Kamath; Donglu Zhang; Luke Masterson; Stephen J. Gregson; Philip W. Howard; Fan Fang; Jinhua Chen; Janet Gunzner-Toste; Katherine K. Kozak; Susan D. Spencer; Paul Polakis; Andrew G. Polson; John A. Flygare

A novel disulfide linker was designed to enable a direct connection between cytotoxic pyrrolobenzodiazepine (PBD) drugs and the cysteine on a targeting antibody for use in antibody–drug conjugates (ADCs). ADCs composed of a cysteine-engineered antibody were armed with a PBD using a self-immolative disulfide linker. Both the chemical linker and the antibody site were optimized for this new bioconjugation strategy to provide a highly stable and efficacious ADC. This novel disulfide ADC was compared with a conjugate containing the same PBD drug, but attached to the antibody via a peptide linker. Both ADCs had similar efficacy in mice bearing human tumor xenografts. Safety studies in rats revealed that the disulfide-linked ADC had a higher MTD than the peptide-linked ADC. Overall, these data suggest that the novel self-immolative disulfide linker represents a valuable way to construct ADCs with equivalent efficacy and improved safety. Mol Cancer Ther; 16(5); 871–8. ©2017 AACR.


Journal of Medicinal Chemistry | 2017

Pyrrolobenzodiazepine Dimer Antibody-Drug Conjugates: Synthesis and Evaluation of Non-Cleavable Drug-Linkers.

Stephen J. Gregson; Luke Masterson; Binqing Wei; Thomas H. Pillow; Susan D. Spencer; Gyoung-Dong Kang; Shang-Fan Yu; Helga Raab; Jeffrey Lau; Guangmin Li; Gail Lewis Phillips; Janet Gunzner-Toste; Brian Safina; Rachana Ohri; Martine Darwish; Katherine R. Kozak; Josefa dela Cruz-Chuh; Andrew Polson; John A. Flygare; Philip W. Howard

Three rationally designed pyrrolobenzodiazepine (PBD) drug-linkers have been synthesized via intermediate 19 for use in antibody-drug conjugates (ADCs). They lack a cleavable trigger in the linker and consist of a maleimide for cysteine antibody conjugation, a hydrophilic spacer, and either an alkyne (6), triazole (7), or piperazine (8) link to the PBD. In vitro IC50 values were 11-48 ng/mL in HER2 3+ SK-BR-3 and KPL-4 (7 inactive) for the anti-HER2 ADCs (HER2 0 MCF7, all inactive) and 0.10-1.73 μg/mL (7 inactive) in CD22 3+ BJAB and WSU-DLCL2 for anti-CD22 ADCs (CD22 0 Jurkat, all inactive at low doses). In vivo antitumor efficacy for the anti-HER2 ADCs in Founder 5 was observed with tumor stasis at 0.5-1 mg/kg, 1 mg/kg, and 3-6 mg/kg for 6, 8, and 7, respectively. Tumor stasis at 2 mg/kg was observed for anti-CD22 6 in WSU-DLCL2. In summary, noncleavable PBD-ADCs exhibit potent activity, particularly in HER2 models.


Journal of Medicinal Chemistry | 2017

Discovery of Peptidomimetic Antibody–Drug Conjugate Linkers with Enhanced Protease Specificity

Binqing Wei; Janet Gunzner-Toste; Hui Yao; Tao Wang; Jing Wang; Zijin Xu; Jinhua Chen; John S. Wai; Jim Nonomiya; Siao Ping Tsai; Josefa Chuh; Katherine R. Kozak; Yichin Liu; Shang-Fan Yu; Jeff Lau; Guangmin Li; Gail D. Phillips; Doug Leipold; Amrita Kamath; Dian Su; Keyang Xu; Charles Eigenbrot; Stefan Steinbacher; Rachana Ohri; Helga Raab; Leanna Staben; Guiling Zhao; John A. Flygare; Thomas H. Pillow; Vishal A. Verma

Antibody-drug conjugates (ADCs) have become an important therapeutic modality for oncology, with three approved by the FDA and over 60 others in clinical trials. Despite the progress, improvements in ADC therapeutic index are desired. Peptide-based ADC linkers that are cleaved by lysosomal proteases have shown sufficient stability in serum and effective payload-release in targeted cells. If the linker can be preferentially hydrolyzed by tumor-specific proteases, safety margin may improve. However, the use of peptide-based linkers limits our ability to modulate protease specificity. Here we report the structure-guided discovery of novel, nonpeptidic ADC linkers. We show that a cyclobutane-1,1-dicarboxamide-containing linker is hydrolyzed predominantly by cathepsin B while the valine-citrulline dipeptide linker is not. ADCs bearing the nonpeptidic linker are as efficacious and stable in vivo as those with the dipeptide linker. Our results strongly support the application of the peptidomimetic linker and present new opportunities for improving the selectivity of ADCs.

Collaboration


Dive into the Janet Gunzner-Toste'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
Researchain Logo
Decentralizing Knowledge