Payman Amiri
Novartis
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Payman Amiri.
Journal of Medicinal Chemistry | 2008
Savithri Ramurthy; Sharadha Subramanian; Mina Aikawa; Payman Amiri; Abran Costales; Jeff Dove; Susan Fong; Johanna M. Jansen; Barry Levine; Sylvia Ma; Christopher Mcbride; Jonah Michaelian; Teresa Pick; Daniel J. Poon; Sandhya Girish; Cynthia Shafer; Darrin Stuart; Leonard Sung; Paul A. Renhowe
A series of arylaminobenzimidazoles was designed and synthesized as Raf kinase inhibitors. Exploration of the structure-activity relationship resulted in compounds that are potent in vitro and show desirable in vivo properties.
ACS Medicinal Chemistry Letters | 2015
Alexei Karpov; Payman Amiri; Cornelia Bellamacina; Marie-Helene Bellance; Werner Breitenstein; Dylan Daniel; Regis Denay; Doriano Fabbro; César Fernández; Inga Galuba; Stephanie Guerro-Lagasse; Sascha Gutmann; Linda Hinh; Wolfgang Jahnke; Julia Klopp; Albert Lai; Mika Lindvall; Sylvia Ma; Henrik Möbitz; Sabina Pecchi; Gabriele Rummel; Kevin Shoemaker; Joerg Trappe; Charles Voliva; Sandra W. Cowan-Jacob; Andreas Marzinzik
The discovery of inhibitors targeting novel allosteric kinase sites is very challenging. Such compounds, however, once identified could offer exquisite levels of selectivity across the kinome. Herein we report our structure-based optimization strategy of a dibenzodiazepine hit 1, discovered in a fragment-based screen, yielding highly potent and selective inhibitors of PAK1 such as 2 and 3. Compound 2 was cocrystallized with PAK1 to confirm binding to an allosteric site and to reveal novel key interactions. Compound 3 modulated PAK1 at the cellular level and due to its selectivity enabled valuable research to interrogate biological functions of the PAK1 kinase.
ACS Medicinal Chemistry Letters | 2015
Teresa E. Williams; Sharadha Subramanian; Joelle Verhagen; Christopher Mcbride; Abran Costales; Leonard Sung; William R. Antonios-Mccrea; Maureen Mckenna; Alicia Louie; Savithri Ramurthy; Barry Levine; Cynthia Shafer; Timothy D. Machajewski; Paul A. Renhowe; Brent A. Appleton; Payman Amiri; James Chou; Darrin Stuart; Kimberly Aardalen; Daniel Poon
Abrogation of errant signaling along the MAPK pathway through the inhibition of B-RAF kinase is a validated approach for the treatment of pathway-dependent cancers. We report the development of imidazo-benzimidazoles as potent B-RAF inhibitors. Robust in vivo efficacy coupled with correlating pharmacokinetic/pharmacodynamic (PKPD) and PD-efficacy relationships led to the identification of RAF265, 1, which has advanced into clinical trials.
Journal of Medicinal Chemistry | 2017
Gisele Nishiguchi; Alice Rico; Huw Tanner; Robert Aversa; Benjamin Taft; Sharadha Subramanian; Lina Setti; Matthew Burger; Lifeng Wan; Victoriano Tamez; Aaron Smith; Yan Lou; Paul A. Barsanti; Brent A. Appleton; Mulugeta Mamo; Laura Tandeske; Ina Dix; John E. Tellew; Shenlin Huang; Lesley A. Mathews Griner; Vesselina G. Cooke; Anne Van Abbema; Hanne Merritt; Sylvia Ma; Kalyani Gampa; Fei Feng; Jing Yuan; Yingyun Wang; Jacob R Haling; Sepideh Vaziri
RAS oncogenes have been implicated in >30% of human cancers, all representing high unmet medical need. The exquisite dependency on CRAF kinase in KRAS mutant tumors has been established in genetically engineered mouse models and human tumor cells. To date, many small molecule approaches are under investigation to target CRAF, yet kinase-selective and cellular potent inhibitors remain challenging to identify. Herein, we describe 14 (RAF709) [ Aversa , Biaryl amide compounds as kinase inhibitors and their preparation . WO 2014151616, 2014 ], a selective B/C RAF inhibitor, which was developed through a hypothesis-driven approach focusing on drug-like properties. A key challenge encountered in the medicinal chemistry campaign was maintaining a balance between good solubility and potent cellular activity (suppression of pMEK and proliferation) in KRAS mutant tumor cell lines. We investigated the small molecule crystal structure of lead molecule 7 and hypothesized that disruption of the crystal packing would improve solubility, which led to a change from N-methylpyridone to a tetrahydropyranyl oxy-pyridine derivative. 14 proved to be soluble, kinase selective, and efficacious in a KRAS mutant xenograft model.
ACS Medicinal Chemistry Letters | 2014
Sharadha Subramanian; Abran Costales; Teresa E. Williams; Barry Levine; Christopher Mcbride; Daniel Poon; Payman Amiri; Paul A. Renhowe; Cynthia Shafer; Darrin Stuart; Joelle Verhagen; Savithri Ramurthy
Benzimidazole reverse amides were designed and synthesized as Pan RAF kinase inhibitors. Investigation of the structure-activity relationship of the compounds revealed that they were potent in vitro and exhibited desirable in vivo properties.
Journal of Medicinal Chemistry | 2016
Wooseok Han; Yu Ding; Yongjin Xu; Keith B. Pfister; Shejin Zhu; Bob Warne; Mike Doyle; Mina Aikawa; Payman Amiri; Brent A. Appleton; Darrin Stuart; Abdallah Fanidi; Cynthia Shafer
The discovery of a highly potent and selective small molecule inhibitor 9 for in vitro target validation of MNK1/2 kinases is described. The aminopyrazine benzimidazole series was derived from an HTS hit and optimized by utilization of a docking model, conformation analysis, and binding pocket comparison against antitargets.
Cancer Research | 2018
Wenlin Shao; Yuji Mishina; Yun Feng; Giordano Caponigro; Vesselina G. Cooke; Stacy Rivera; Yingyun Wang; Fang Shen; Joshua Korn; Lesley A. Mathews Griner; Gisele Nishiguchi; Alice Rico; John Tellew; Jacob R. Haling; Robert Aversa; Valery Polyakov; Richard Zang; Mohammad Hekmat-Nejad; Payman Amiri; Mallika Singh; Nicholas Keen; Michael P. Dillon; Emma Lees; Savithri Ramurthy; William R. Sellers; Darrin Stuart
Resistance to the RAF inhibitor vemurafenib arises commonly in melanomas driven by the activated BRAF oncogene. Here, we report antitumor properties of RAF709, a novel ATP-competitive kinase inhibitor with high potency and selectivity against RAF kinases. RAF709 exhibited a mode of RAF inhibition distinct from RAF monomer inhibitors such as vemurafenib, showing equal activity against both RAF monomers and dimers. As a result, RAF709 inhibited MAPK signaling activity in tumor models harboring either BRAFV600 alterations or mutant N- and KRAS-driven signaling, with minimal paradoxical activation of wild-type RAF. In cell lines and murine xenograft models, RAF709 demonstrated selective antitumor activity in tumor cells harboring BRAF or RAS mutations compared with cells with wild-type BRAF and RAS genes. RAF709 demonstrated a direct pharmacokinetic/pharmacodynamic relationship in in vivo tumor models harboring KRAS mutation. Furthermore, RAF709 elicited regression of primary human tumor-derived xenograft models with BRAF, NRAS, or KRAS mutations with excellent tolerability. Our results support further development of inhibitors like RAF709, which represents a next-generation RAF inhibitor with unique biochemical and cellular properties that enables antitumor activities in RAS-mutant tumors.Significance: In an effort to develop RAF inhibitors with the appropriate pharmacological properties to treat RAS mutant tumors, RAF709, a compound with potency, selectivity, and in vivo properties, was developed that will allow preclinical therapeutic hypothesis testing, but also provide an excellent probe to further unravel the complexities of RAF kinase signaling. Cancer Res; 78(6); 1537-48. ©2018 AACR.
Cancer Research | 2016
Wenlin Shao; Yuji Mishina; Yun Feng; Giordano Caponigro; Savithri Ramurthy; Vesselina G. Cooke; Lesley A. Mathews Griner; Gisele Nishiguchi; Alice Rico; Ben Taft; Matthew Burger; Huw Tanner; Valery Polyakov; Brent A. Appleton; John Tellew; Richard Zang; Mohammad Hekmat-Nejad; Payman Amiri; Mallika Singh; Darrin Stuart
The mitogen-activated protein kinase (MAPK) signaling pathway is frequently activated in human cancers due to genetic alterations that can occur at multiple nodes in the pathway, the most prevalent of which are mutations in RAS or BRAF. While BRAFV600 mutant tumors are effectively treated with existing RAF inhibitors, RAS mutant cancers and tumors expressing atypical BRAF mutants remain an unmet medical need. Emerging biology has demonstrated that the CRAF kinase functions as a critical mediator of mutant KRAS-driven cell proliferation and tumor development. CRAF was also shown to be the mediator of feedback-mediated pathway reactivation following MEK inhibitor treatment in KRAS mutant cancers. Hence selective inhibitors that potently inhibit the activity of CRAF could be both effective in blocking mutant RAS-driven tumorigenesis and in alleviating feedback activation. We have developed a type II ATP-competitive inhibitor that inhibits both B- and CRAF kinase activities at picomolar IC50 values in biochemical assays with high selectivity profile against a panel of 456 human kinases. The inhibitor not only inhibits MAPK signaling activity in tumor models harboring BRAFV600 mutation, but also inhibits mutant N- and KRAS-driven signaling with minimum paradoxical activation, likely due to its activity in inhibiting both RAF monomers and dimers with similar potencies. Correspondingly, profiling data of the inhibitor in a panel of 480 human cancer cell lines shows that it has higher antitumor activities in cell lines harboring BRAF or RAS mutations as compared to those that are wild-type. The inhibitor is orally bioavailable, it demonstrates a direct PK/PD relationship and causes tumor regression in multiple cell line and primary human tumor derived xenograft models that have BRAF, NRAS or KRAS mutations with good tolerability. Thus, we have developed a next generation RAF inhibitor with unique biochemical and cellular properties that enables its antitumor activities in RAS mutant tumors. Citation Format: Wenlin Shao, Yuji Mishina, Yun Feng, Giordano Caponigro, Savithri Ramurthy, Vesselina Cooke, Lesley Griner, Gisele Nishiguchi, Alice Rico, Ben Taft, Matthew Burger, Huw Tanner, Valery Polyakov, Brent Appleton, John Tellew, Richard Zang, Mohammad Hekmat-Nejad, Payman Amiri, Mallika Singh, Darrin Stuart. Development of a highly selective B/CRAF kinase inhibitor that exhibits antitumor activities in RAS and BRAF mutant tumors with minimal paradoxical activation. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 330.
ACS Medicinal Chemistry Letters | 2014
Sharadha Subramanian; Abran Costales; Teresa E. Williams; Barry Levine; Christopher Mcbride; Daniel Poon; Payman Amiri; Paul A. Renhowe; Cynthia Shafer; Darrin Stuart; Joelle Verhagen; Savithri Ramurthy
[This corrects the article DOI: 10.1021/ml5002272.].
Archive | 2003
Paul A. Renhowe; Savithri Ramurthy; Payman Amiri; Barry Haskell Levine; Daniel J. Poon; Sharadha Subramanian; Leonard Sung; Wendy J. Fantl