Network


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

Hotspot


Dive into the research topics where Lynsie Almeida is active.

Publication


Featured researches published by Lynsie Almeida.


Journal of Medicinal Chemistry | 2011

Discovery of 5-Chloro-N2-[(1S)-1-(5-Fluoropyrimidin-2-Yl) Ethyl]-N4-(5-Methyl-1H-Pyrazol-3-Yl)Pyrimidine-2,4-Diamine (Azd1480) as a Novel Inhibitor of the Jak/Stat Pathway

Stephanos Ioannidis; Michelle L. Lamb; Tao Wang; Lynsie Almeida; Michael Howard Block; Audrey Davies; Bo Peng; Mei Su; Hai-Jun Zhang; Ethan Hoffmann; Caroline Rivard; Isabelle Green; Tina Howard; Hannah Pollard; Jon Read; Marat Alimzhanov; Geraldine A. Bebernitz; Kirsten Bell; Minwei Ye; Dennis Huszar; Michael Zinda

The myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia, and idiopathic myelofibrosis are a heterogeneous but related group of hematological malignancies characterized by clonal expansion of one or more myeloid lineages. The discovery of the Jak2 V617F gain of function mutation highlighted Jak2 as a potential therapeutic target in the MPNs. Herein, we disclose the discovery of a series of pyrazol-3-yl pyrimidin-4-amines and the identification of 9e (AZD1480) as a potent Jak2 inhibitor. 9e inhibits signaling and proliferation of Jak2 V617F cell lines in vitro, demonstrates in vivo efficacy in a TEL-Jak2 model, has excellent physical properties and preclinical pharmacokinetics, and is currently being evaluated in Phase I clinical trials.


Journal of Medicinal Chemistry | 2012

Discovery of Checkpoint Kinase Inhibitor (S)-5-(3-Fluorophenyl)-N-(piperidin-3-yl)-3-ureidothiophene-2-carboxamide (AZD7762) by Structure-Based Design and Optimization of Thiophenecarboxamide Ureas.

Oza; Susan Ashwell; Lynsie Almeida; Patrick Brassil; Jason Breed; Chun Deng; Thomas Gero; Michael Grondine; C Horn; Stephanos Ioannidis; D Liu; Paul Lyne; Nicholas John Newcombe; Martin Pass; Jon Read; S Ready; S Rowsell; Mei Su; Dorin Toader; Melissa Vasbinder; Dingwei Yu; Yan Yu; Y Xue; S Zabludoff; James W. Janetka

Checkpoint kinases CHK1 and CHK2 are activated in response to DNA damage that results in cell cycle arrest, allowing sufficient time for DNA repair. Agents that lead to abrogation of such checkpoints have potential to increase the efficacy of such compounds as chemo- and radiotherapies. Thiophenecarboxamide ureas (TCUs) were identified as inhibitors of CHK1 by high throughput screening. A structure-based approach is described using crystal structures of JNK1 and CHK1 in complex with 1 and 2 and of the CHK1-3b complex. The ribose binding pocket of CHK1 was targeted to generate inhibitors with excellent cellular potency and selectivity over CDK1and IKKβ, key features lacking from the initial compounds. Optimization of 3b resulted in the identification of a regioisomeric 3-TCU lead 12a. Optimization of 12a led to the discovery of the clinical candidate 4 (AZD7762), which strongly potentiates the efficacy of a variety of DNA-damaging agents in preclinical models.


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 | 2008

Discovery of a novel class of 2-ureido thiophene carboxamide checkpoint kinase inhibitors.

James W. Janetka; Lynsie Almeida; Susan Ashwell; Patrick Brassil; Kevin Daly; Chun Deng; Thomas Gero; Roberta Glynn; Candice Horn; Stephanos Ioannidis; Paul Lyne; Nicholas John Newcombe; Vibha Oza; Martin Pass; Stephanie Springer; Mei Su; Dorin Toader; Melissa Vasbinder; Dingwei Yu; Yan Yu; Sonya Zabludoff

Checkpoint kinase-1 (Chk1, CHEK1) is a Ser/Thr protein kinase that mediates the cellular response to DNA-damage. A novel class of 2-ureido thiophene carboxamide urea (TCU) Chk1 inhibitors is described. Inhibitors in this chemotype were optimized for cellular potency and selectivity over Cdk1.


Bioorganic & Medicinal Chemistry Letters | 2010

Replacement of pyrazol-3-yl amine hinge binder with thiazol-2-yl amine: Discovery of potent and selective JAK2 inhibitors.

Stephanos Ioannidis; Michelle Lamb; Lynsie Almeida; Huiping Guan; Bo Peng; Geraldine Bebernitz; Kirsten Bell; Marat Alimzhanov; Michael Zinda

Thiazol-2-yl amine was identified as an isosteric replacement for pyrazol-3-yl amine during our efforts to identify potent and selective JAK2 inhibitors. The rationale, synthesis and biological evaluation of several analogs is reported, along with the in vivo evaluation of the lead compounds.


Bioorganic & Medicinal Chemistry Letters | 2009

Discovery of pyrazol-3-ylamino pyrazines as novel JAK2 inhibitors.

Stephanos Ioannidis; Michelle Lamb; Audrey Davies; Lynsie Almeida; Mei Su; Geraldine Bebernitz; Minwei Ye; Kirsten Bell; Marat Alimzhanov; Michael Zinda

The design, synthesis and biological evaluation of a series of pyrazol-3-ylamino pyrazines as potent and selective JAK2 kinase inhibitors is reported, along with the pharmacokinetic and pharmacodynamic properties of lead compounds.


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.


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.


Bioorganic & Medicinal Chemistry Letters | 2011

In vitro and in vivo evaluation of 6-aminopyrazolyl-pyridine-3-carbonitriles as JAK2 kinase inhibitors.

Tao Wang; Stephanos Ioannidis; Lynsie Almeida; Michael Howard Block; Audrey Davies; Michelle Lamb; David Scott; Mei Su; Hai-Jun Zhang; Marat Alimzhanov; Geraldine Bebernitz; Kirsten Bell; Michael Zinda

Synthesis and biological evaluation of a series of 6-aminopyrazolyl-pyridine-3-carbonitriles as JAK2 kinase inhibitors was reported. Biochemical screening, followed by profile optimization, resulted in JAK2 inhibitors exhibiting good kinase selectivity, pharmacokinetic properties, physical properties and pharmacodynamic effects.


Cancer Research | 2011

Abstract 5196: The discovery of small molecule inhibitors of Tankyrases 1 and 2, which modulate Axin homeostasis and inhibit Wnt signaling in vivo

Emma-Louise Cooke; Marat Alimzhanov; Lynsie Almeida; Larry Bao; Vahe Bedian; Jeffrey L. Brown; Chun Deng; Christopher R. Denz; Lakshmaiah Gingipalli; Jeffrey W. Johannes; Steven Kazmirski; Michelle Lamb; Zhong-Ying Liu; Vicki Racicot; Nancy Su; Haiyun Wang; Tao Wang; Xiaoyun Wu; Lihua Yu; Hai-Jun Zhang; Weijia Zheng; Paul Lyne

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Deregulated Wnt signaling has been implicated in a wide range of cancer types, both through truncating mutations in the tumor suppressor protein Adenomatous Polyposis Coli (APC), most prevalent in colorectal cancer, as well as overexpression of Wnt ligands and receptors. Canonical Wnt signaling involves the regulated degradation of the beta catenin protein. In the absence of Wnt signaling, cytosolic beta catenin levels are maintained at low levels, as a result of its phosphorylation-dependent ubiquitination and subsequent proteasomal degradation. This degradation is mediated by the ‘destruction complex’, whose constituents include Glycogen Synthase Kinase 3 (GSK3β), Casein Kinase 1 (CK1) and the scaffolding proteins Axin and APC. The efficient assembly of this complex depends upon the steady state levels of its components, such as Axin, which when overexpressed has been shown to decrease beta catenin levels and inhibit Wnt signaling. Recent reports suggest that the poly-ADP-ribosylating Tankyrase enzymes can bind to and regulate Axin levels, hence providing promising targets for treating Wnt-dependent tumors. We have identified potent, low nM small molecule inhibitors of the Wnt pathway, which inhibit Tankyrases 1 and 2, stabilize Axin, deplete beta catenin protein levels and modulate a set of Wnt-regulated genes in colorectal cell lines in a similar manner as seen when using siRNA to beta catenin. Furthermore, using siRNAs to Tankyrase 1 and 2 we have shown that co-depletion of both enzymes is required for Axin stabilization and Wnt pathway inhibition. We have also successfully demonstrated that our lead molecules cause prolonged Axin stabilization and Wnt pathway inhibition in vivo following oral BID dosing in colon xenografts carrying APC mutations. However, in contrast to previous reports, using molecular tools and our lead compounds (e.g. inducible Axin cell lines, compound rescue experiments using siRNAs to Axin 1/2), we have shown that Axin stabilization has no effect on the viability/proliferation of APC mutant colorectal cell lines, under conditions where siRNA to beta catenin was able to cause significant growth inhibition. Some potential mechanistic rationales for this lack of phenotypic effect will be presented. Understanding the consequences of Tankyrase inhibition in vivo in relevant disease models will be important to establish the clinical utility of these molecules in colorectal cancer and beyond. 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 5196. doi:10.1158/1538-7445.AM2011-5196

Collaboration


Dive into the Lynsie Almeida'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