Network


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

Hotspot


Dive into the research topics where Melissa Yarde is active.

Publication


Featured researches published by Melissa Yarde.


Journal of Medicinal Chemistry | 2014

Discovery of the CCR1 antagonist, BMS-817399, for the treatment of rheumatoid arthritis.

Joseph B. Santella; Daniel S. Gardner; John V. Duncia; Hong Wu; Murali T. G. Dhar; Cullen L. Cavallaro; Andrew J. Tebben; Percy H. Carter; Joel C. Barrish; Melissa Yarde; Stephanie W. Briceno; Mary Ellen Cvijic; R. Robert Grafstrom; Richard Liu; Sima R. Patel; Andrew Watson; Guchen Yang; Anne Rose; Rodney Vickery; Janet Caceres-Cortes; Christian Caporuscio; Daniel M. Camac; Javed Khan; Yongmi An; William R. Foster; Paul Davies; John Hynes

High-affinity, functionally potent, urea-based antagonists of CCR1 have been discovered. Modulation of PXR transactivation has revealed the selective and orally bioavailable CCR1 antagonist BMS-817399 (29), which entered clinical trials for the treatment of rheumatoid arthritis.


Journal of Medicinal Chemistry | 2016

Potent and Selective Agonists of Sphingosine 1-Phosphate 1 (S1P1): Discovery and SAR of a Novel Isoxazole Based Series

Scott H. Watterson; Junqing Guo; Steve Spergel; Charles M. Langevine; Robert V. Moquin; Ding Ren Shen; Melissa Yarde; Mary Ellen Cvijic; Dana Banas; Richard Liu; Suzanne J. Suchard; Kathleen M. Gillooly; Tracy L. Taylor; Sandra Rex-Rabe; David J. Shuster; Kim W. McIntyre; Georgia Cornelius; Celia D’Arienzo; Anthony Marino; Praveen Balimane; Bethanne M. Warrack; Luisa Salter-Cid; Murray McKinnon; Joel C. Barrish; Percy H. Carter; William J. Pitts; Jenny Xie; Alaric J. Dyckman

Sphingosine 1-phosphate (S1P) is the endogenous ligand for the sphingosine 1-phosphate receptors (S1P1-5) and evokes a variety of cellular responses through their stimulation. The interaction of S1P with the S1P receptors plays a fundamental physiological role in a number of processes including vascular development and stabilization, lymphocyte migration, and proliferation. Agonism of S1P1, in particular, has been shown to play a significant role in lymphocyte trafficking from the thymus and secondary lymphoid organs, resulting in immunosuppression. This article will detail the discovery and SAR of a potent and selective series of isoxazole based full agonists of S1P1. Isoxazole 6d demonstrated impressive efficacy when administered orally in a rat model of arthritis and in a mouse experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis.


Journal of Medicinal Chemistry | 2016

Discovery and Structure–Activity Relationship (SAR) of a Series of Ethanolamine-Based Direct-Acting Agonists of Sphingosine-1-phosphate (S1P1)

John L. Gilmore; James E. Sheppeck; Scott H. Watterson; Lauren Haque; Parag Mukhopadhyay; Andrew J. Tebben; Michael A. Galella; Ding Ren Shen; Melissa Yarde; Mary Ellen Cvijic; Virna Borowski; Kathleen M. Gillooly; Tracy L. Taylor; Kim W. McIntyre; Bethanne M. Warrack; Paul Levesque; Julia P. Li; Georgia Cornelius; Celia D’Arienzo; Anthony Marino; Praveen Balimane; Luisa Salter-Cid; Joel C. Barrish; William J. Pitts; Percy H. Carter; Jenny Xie; Alaric J. Dyckman

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite that regulates a multitude of physiological processes such as lymphocyte trafficking, cardiac function, vascular development, and inflammation. Because of the ability of S1P1 receptor agonists to suppress lymphocyte egress, they have great potential as therapeutic agents in a variety of autoimmune diseases. In this article, the discovery of selective, direct acting S1P1 agonists utilizing an ethanolamine scaffold containing a terminal carboxylic acid is described. Potent S1P1 agonists such as compounds 18a and 19a which have greater than 1000-fold selectivity over S1P3 are described. These compounds efficiently reduce blood lymphocyte counts in rats through 24 h after single doses of 1 and 0.3 mpk, respectively. Pharmacodynamic properties of both compounds are discussed. Compound 19a was further studied in two preclinical models of disease, exhibiting good efficacy in both the rat adjuvant arthritis model (AA) and the mouse experimental autoimmune encephalomyelitis model (EAE).


Bioorganic & Medicinal Chemistry Letters | 2013

The discovery of BMS-457, a potent and selective CCR1 antagonist

Daniel S. Gardner; Joseph B. Santella; John V. Duncia; Percy H. Carter; T. G. Murali Dhar; Hong Wu; Weiwei Guo; Cullen L. Cavallaro; Katy Van Kirk; Melissa Yarde; Stephanie W. Briceno; R. Robert Grafstrom; Richard Liu; Sima R. Patel; Andrew J. Tebben; Dan Camac; Javed Khan; Andrew Watson; Guchen Yang; Anne Rose; William R. Foster; Mary Ellen Cvijic; Paul Davies; John Hynes

A series of compounds which exhibited good human CCR1 binding and functional potency was modified resulting in the discovery of a novel series of high affinity, functionally potent antagonists of the CCR1 receptor. Issues of PXR activity, ion-channel potency, and poor metabolic stability were addressed by the addition of a hydroxyl group to an otherwise lipophilic area in the molecule resulting in the discovery of preclinical candidate BMS-457 for the treatment of rheumatoid arthritis.


Journal of Biomolecular Screening | 2016

384-Well Multiplexed Luminex Cytokine Assays for Lead Optimization

Huaping Tang; Reshma Panemangalore; Melissa Yarde; Litao Zhang; Mary Ellen Cvijic

Cytokines serve as a major mechanism of communication between immune cells and are the functional molecules at the end of immune pathways. Abnormalities in cytokines are involved in a wide variety of diseases, including chronic inflammation, autoimmune diseases, and cancer. Cytokines are not only direct targets of therapeutics but also important biomarkers for assessing drug efficacy and safety. Traditionally, enzyme-linked immunosorbent assays (ELISA) were most popular for identifying and quantifying cytokines. However, ELISA is expensive, labor intensive, and low throughput. Here, we report the development of a miniaturized Luminex (Austin, TX) assay platform to establish a panel of high-throughput, multiplexed assays for measuring cytokines in human whole blood. The miniaturized 384-well Luminex assay uses <25% of the assay reagents compared with the 96-well assay. The development and validation of the 384-well Luminex cytokine assays enabled high-throughput screening of compounds in primary cells using cytokines as physiologically relevant readouts. Furthermore, this miniaturized multiplexed technology platform allows for high-throughput biomarker profiling of biofluids from animal studies and patient samples for translational research.


Journal of Biomolecular Screening | 2014

Integrating High-Content Analysis into a Multiplexed Screening Approach to Identify and Characterize GPCR Agonists

Yingjie Zhu; John Watson; Mengjie Chen; Ding Ren Shen; Melissa Yarde; Michele Agler; Neil T. Burford; Andrew Alt; Sukhanya Jayachandra; Mary Ellen Cvijic; Litao Zhang; Alaric J. Dyckman; Jenny Xie; Jonathan O’Connell; Martyn Banks; Andrea Weston

G protein–coupled receptors (GPCRs) are one of the most popular and proven target classes for therapeutic intervention. The increased appreciation for allosteric modulation, receptor oligomerization, and biased agonism has led to the development of new assay platforms that seek to capitalize on these aspects of GPCR biology. High-content screening is particularly well suited for GPCR drug discovery given the ability to image and quantify changes in multiple cellular parameters, to resolve subcellular structures, and to monitor events within a physiologically relevant environment. Focusing on the sphingosine-1-phosphate (S1P1) receptor, we evaluated the utility of high-content approaches in hit identification efforts by developing and applying assays to monitor β-arrestin translocation, GPCR internalization, and GPCR recycling kinetics. Using these approaches in combination with more traditional GPCR screening assays, we identified compounds whose unique pharmacological profiles would have gone unnoticed if using a single platform. In addition, we identified a compound that induces an atypical pattern of β-arrestin translocation and GPCR recycling kinetics. Our results highlight the value of high-content imaging in GPCR drug discovery efforts and emphasize the value of a multiassay approach to study pharmacological properties of compounds of interest.


Bioorganic & Medicinal Chemistry Letters | 2016

Identification and synthesis of potent and selective pyridyl-isoxazole based agonists of sphingosine-1-phosphate 1 (S1P1)

Junqing Guo; Scott H. Watterson; Steven H. Spergel; James Kempson; Charles M. Langevine; Ding Ren Shen; Melissa Yarde; Mary Ellen Cvijic; Dana Banas; Richard Liu; Suzanne J. Suchard; Kathleen M. Gillooly; Tracy L. Taylor; Sandra Rex-Rabe; David J. Shuster; Kim W. McIntyre; Georgia Cornelius; Celia D’Arienzo; Anthony Marino; Praveen Balimane; Luisa Salter-Cid; Murray McKinnon; Joel C. Barrish; Percy H. Carter; William J. Pitts; Jenny Xie; Alaric J. Dyckman

The synthesis and structure-activity relationship (SAR) of a series of pyridyl-isoxazole based agonists of S1P1 are discussed. Compound 5b provided potent in vitro activity with selectivity, had an acceptable pharmacokinetic profile, and demonstrated efficacy in a dose dependent manner when administered orally in a rodent model of arthritis.


Bioorganic & Medicinal Chemistry Letters | 2018

Identification of bicyclic hexafluoroisopropyl alcohol sulfonamides as retinoic acid receptor-related orphan receptor gamma (RORγ/RORc) inverse agonists. Employing structure-based drug design to improve pregnane X receptor (PXR) selectivity

Hua Gong; David S. Weinstein; Zhonghui Lu; James J.-W. Duan; Sylwia Stachura; Lauren Haque; Ananta Karmakar; Hemalatha Hemagiri; Dhanya Kumar Raut; Arun Kumar Gupta; Javed Khan; Dan Camac; John S. Sack; Andrew T. Pudzianowski; Dauh-Rurng Wu; Melissa Yarde; Ding-Ren Shen; Virna Borowski; Jenny Xie; Huadong Sun; Celia D'Arienzo; Marta Dabros; Michael A. Galella; Faye Wang; Carolyn A. Weigelt; Qihong Zhao; William R. Foster; John E. Somerville; Luisa Salter-Cid; Joel C. Barrish

We disclose the optimization of a high throughput screening hit to yield benzothiazine and tetrahydroquinoline sulfonamides as potent RORγt inverse agonists. However, a majority of these compounds showed potent activity against pregnane X receptor (PXR) and modest activity against liver X receptor α (LXRα). Structure-based drug design (SBDD) led to the identification of benzothiazine and tetrahydroquinoline sulfonamide analogs which completely dialed out LXRα activity and were less potent at PXR. Pharmacodynamic (PD) data for compound 35 in an IL-23 induced IL-17 mouse model is discussed along with the implications of a high Ymax in the PXR assay for long term preclinical pharmacokinetic (PK) studies.


Cancer Research | 2014

Abstract 1643: BMS-983970, an oral pan-Notch inhibitor for the treatment of cancer

Ashvinikumar V. Gavai; Yufen Zhao; Daniel O'Malley; Brian E. Fink; Claude A. Quesnelle; Derek J. Norris; Libing Chen; Soong-Hoon Kim; Wen-Ching Han; Patrice Gill; Weifang Shan; Aaron Balog; Andrew J. Tebben; Richard Rampulla; Dauh-Rurng Wu; Yingru Zhang; Arvind Mathur; Haiqing Wang; Zheng Yang; Qian Ruan; Robin Moore; David Rodrigues; Asoka Ranasinghe; Celia D'Arienzo; Ching Kim Tye; Ching Su; Gerry Everlof; Melissa Yarde; Mary Ellen Cvijic; Krista Menard

Deregulation of the Notch pathway has been shown to be oncogenic in numerous tissue types including T-cell acute lymphoblastic leukemia (T-ALL), breast cancer, non-small cell lung cancer, and colorectal carcinoma. Notch signal activation can cause uncontrolled proliferation, restrict differentiation leading to increased self-renewal capacity, evasion of apoptosis, and enhancement of angiogenesis and metastasis. There is increasing evidence that Notch plays a role in the maintenance and survival of cancer stem cells. γ-Secretase mediates the Notch signaling pathway by releasing the Notch intracellular domain (NICD) which translocates to the nucleus and binds to the transcription factor CSL to activate transcription of various target genes. BMS-906024 is a potent pan-Notch inhibitor that demonstrated robust anti-tumor activity at tolerated doses in multiple tumor xenograft models. It is being evaluated in Phase 1 clinical studies. BMS-906024 is being administered IV (once weekly) in the clinic and the projected human efficacious dose is 4 - 6 mg. Based on the preclinical data, the projected human half-life of BMS-906024 is in the 37 h - 124 h range. This presentation will describe further structure-activity relationships in the 1,4-benzodiazepinone series that culminated in the identification of BMS-983970 as an oral-pan-Notch inhibitor. Pharmacokinetic properties and in vivo evaluation of BMS-983970 in T-ALL and solid tumor xenograft models will be presented. Citation Format: Ashvinikumar V. Gavai, Yufen Zhao, Daniel O9Malley, Brian Fink, Claude Quesnelle, Derek Norris, Libing Chen, Soong-Hoon Kim, Wen-Ching Han, Patrice Gill, Weifang Shan, Aaron Balog, Andrew Tebben, Richard Rampulla, Dauh-Rurng Wu, Yingru Zhang, Arvind Mathur, Haiqing Wang, Zheng Yang, Qian Ruan, Robin Moore, David Rodrigues, Asoka Ranasinghe, Celia D9Arienzo, Ching Kim Tye, Ching Su, Gerry Everlof, Melissa Yarde, Mary Ellen Cvijic, Krista Menard, Mei-Li Wen, George Trainor, Bruce Fischer, John Hunt, Gregory Vite, Richard Westhouse, Francis Lee. BMS-983970, an oral pan-Notch inhibitor for the treatment of cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1643. doi:10.1158/1538-7445.AM2014-1643


Cancer Research | 2010

Abstract 5782: Androgen receptor antagonists: Lead optimization and preclinical pharmacology

Ashvin Gavai; Derek J. Norris; Aaron Balog; Joel F. Austin; Weifang Shan; Yufen Zhao; Wen-Ching Han; Lisa Sharma; Andrew Nation; Dauh-Rurng Wu; Arvind Mathur; Litai Zhang; Mary Ellen Cvijic; Melissa Yarde; Donald Apanovitch; Celeste Twamley; Yuwei Tang; Benjamin M. Johnson; Paul A. Elzinga; Georgia Cornelius; Mary T. Obermeier; William R. Foster; Punit Marathe; Janet Dell-John; Gennaro Dito; Gregg Masters; Cheryl A. Rizzo; Liang Schweizer; Maria Jure-Kunkel; Ricardo M. Attar

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC The development and progression of prostate cancer is known to be dependent on androgens and their signaling mediated by the androgen receptor (AR). The primary therapeutic intervention involves using agents that lower serum testosterone (e.g., LHRH agonists), often in concert with an AR antagonist, such as bicalutamide. Despite a favorable initial anti-tumor response, most patients progress to the advanced hormone-refractory disease. The development of resistance to anti-androgen therapy has been shown to be associated with an increase in the levels of both AR mRNA and protein. This observation supports the concept that an AR antagonist with a significant improvement in potency as compared to bicalutamide and a broader spectrum of in vivo anti-tumor activity, including the bicalutamide-refractory human prostate tumor xenografts, may provide a significant clinical advantage in the treatment of advanced prostate cancer. This presentation will describe structure-activity relationships in a novel tetracyclic series of androgen receptor antagonists leading up to the identification of BMS-779333. It is a potent AR full antagonist, which exhibited broad spectrum efficacy in four human prostate tumor xenograft models. BMS-779333 did not exhibit agonist activity for AR mutant isoforms. Tumors that failed bicalutamide treatment were shown to retain their sensitivity to respond to BMS-779333. Transcriptomic changes in LuCaP-35 tumors treated with BMS-779333 were closer to castration than with other drug treatments. Based on its overall profile, BMS-779333 was selected for further preclinical evaluation. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5782.

Collaboration


Dive into the Melissa Yarde'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge