Network


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

Hotspot


Dive into the research topics where Kasey Rivas is active.

Publication


Featured researches published by Kasey Rivas.


The Journal of Infectious Diseases | 2014

A Specific Inhibitor of PfCDPK4 Blocks Malaria Transmission: Chemical-genetic Validation

Kayode K. Ojo; Richard T. Eastman; Ramasubbarao Vidadala; Zhongsheng Zhang; Kasey Rivas; Ryan Choi; Justin D. Lutz; Molly C. Reid; Anna M. W. Fox; Matthew A. Hulverson; Mark Kennedy; Nina Isoherranen; Laura M. Kim; Kenneth M. Comess; Dale J. Kempf; Christophe L. M. J. Verlinde; Xin-Zhuan Su; Stefan H. I. Kappe; Dustin J. Maly; Erkang Fan; Wesley C. Van Voorhis

Malaria parasites are transmitted by mosquitoes, and blocking parasite transmission is critical in reducing or eliminating malaria in endemic regions. Here, we report the pharmacological characterization of a new class of malaria transmission-blocking compounds that acts via the inhibition of Plasmodia CDPK4 enzyme. We demonstrate that these compounds achieved selectivity over mammalian kinases by capitalizing on a small serine gatekeeper residue in the active site of the Plasmodium CDPK4 enzyme. To directly confirm the mechanism of action of these compounds, we generated P. falciparum parasites that express a drug-resistant methionine gatekeeper (S147 M) CDPK4 mutant. Mutant parasites showed a shift in exflagellation EC50 relative to the wild-type strains in the presence of compound 1294, providing chemical-genetic evidence that CDPK4 is the target of the compound. Pharmacokinetic analyses suggest that coformulation of this transmission-blocking agent with asexual stage antimalarials such as artemisinin combination therapy (ACT) is a promising option for drug delivery that may reduce transmission of malaria including drug-resistant strains. Ongoing studies include refining the compounds to improve efficacy and toxicological properties for efficient blocking of malaria transmission.


The Journal of Infectious Diseases | 2013

A novel Calcium Dependent Protein Kinase Inhibitor as a lead compound for treating Cryptosporidiosis

Alejandro Castellanos-Gonzalez; A. Clinton White; Kayode K. Ojo; Rama Subba Rao Vidadala; Zhongsheng Zhang; Molly C. Reid; Anna M. W. Fox; Katelyn R. Keyloun; Kasey Rivas; Ayesha Irani; Sara M. Dann; Erkang Fan; Dustin J. Maly; Wesley C. Van Voorhis

Cryptosporidium parasites infect intestinal cells, causing cryptosporidiosis. Despite its high morbidity and association with stunting in the developing world, current therapies for cryptosporidiosis have limited efficacy. Calcium-dependent protein kinases (CDPKs) are essential enzymes in the biology of protozoan parasites. CDPK1 was cloned from the genome of Cryptosporidium parvum, and potent and specific inhibitors have been developed based on structural studies. In this study, we evaluated the anti-Cryptosporidium activity of a novel CDPK1 inhibitor, 1294, and demonstrated that 1294 significantly reduces parasite infection in vitro, with a half maximal effective concentration of 100 nM. Pharmacokinetic studies revealed that 1294 is well absorbed, with a half-life supporting daily administration. Oral therapy with 1294 eliminated Cryptosporidium parasites from 6 of 7 infected severe combined immunodeficiency-beige mice, and the parasites did not recur in these immunosuppressed mice. Mice treated with 1294 had less epithelial damage, corresponding to less apoptosis. Thus, 1294 is an important lead for the development of drugs for treatment of cryptosporidiosis.


Journal of Medicinal Chemistry | 2016

Development of an Orally Available and Central Nervous System (CNS) Penetrant Toxoplasma gondii Calcium-Dependent Protein Kinase 1 (TgCDPK1) Inhibitor with Minimal Human Ether-a-go-go-Related Gene (hERG) Activity for the Treatment of Toxoplasmosis

Rama Subba Rao Vidadala; Kasey Rivas; Kayode K. Ojo; Matthew A. Hulverson; Jennifer A. Zambriski; Igor Bruzual; Tracey L. Schultz; Wenlin Huang; Zhongsheng Zhang; Suzanne Scheele; Amy E. DeRocher; Ryan Choi; Lynn K. Barrett; Latha Kallur Siddaramaiah; Wim G. J. Hol; Erkang Fan; Ethan A. Merritt; Marilyn Parsons; Gail M. Freiberg; Kennan Marsh; Dale J. Kempf; Vern B. Carruthers; Nina Isoherranen; J. Stone Doggett; Wesley C. Van Voorhis; Dustin J. Maly

New therapies are needed for the treatment of toxoplasmosis, which is a disease caused by the protozoan parasite Toxoplasma gondii. To this end, we previously developed a potent and selective inhibitor (compound 1) of Toxoplasma gondii calcium-dependent protein kinase 1 (TgCDPK1) that possesses antitoxoplasmosis activity in vitro and in vivo. Unfortunately, 1 has potent human ether-a-go-go-related gene (hERG) inhibitory activity, associated with long Q-T syndrome, and consequently presents a cardiotoxicity risk. Here, we describe the identification of an optimized TgCDPK1 inhibitor 32, which does not have a hERG liability and possesses a favorable pharmacokinetic profile in small and large animals. 32 is CNS-penetrant and highly effective in acute and latent mouse models of T. gondii infection, significantly reducing the amount of parasite in the brain, spleen, and peritoneal fluid and reducing brain cysts by >85%. These properties make 32 a promising lead for the development of a new antitoxoplasmosis therapy.


Chemistry & Biology | 2009

Structural basis for binding and selectivity of antimalarial and anticancer ethylenediamine inhibitors to protein farnesyltransferase.

Michael A. Hast; Steven Fletcher; Christopher G. Cummings; Erin E. Pusateri; Michelle A. Blaskovich; Kasey Rivas; Michael H. Gelb; Wesley C. Van Voorhis; Said M. Sebti; Andrew D. Hamilton; Lorena S. Beese

Protein farnesyltransferase (FTase) catalyzes an essential posttranslational lipid modification of more than 60 proteins involved in intracellular signal transduction networks. FTase inhibitors have emerged as a significant target for development of anticancer therapeutics and, more recently, for the treatment of parasitic diseases caused by protozoan pathogens, including malaria (Plasmodium falciparum). We present the X-ray crystallographic structures of complexes of mammalian FTase with five inhibitors based on an ethylenediamine scaffold, two of which exhibit over 1000-fold selective inhibition of P. falciparum FTase. These structures reveal the dominant determinants in both the inhibitor and enzyme that control binding and selectivity. Comparison to a homology model constructed for the P. falciparum FTase suggests opportunities for further improving selectivity of a new generation of antimalarial inhibitors.


Journal of Medicinal Chemistry | 2008

Potent, Plasmodium-selective farnesyltransferase inhibitors that arrest the growth of malaria parasites: structure-activity relationships of ethylenediamine-analogue scaffolds and homology model validation.

Steven Fletcher; Christopher G. Cummings; Kasey Rivas; William P. Katt; Carrie Hornéy; Frederick S. Buckner; Debopam Chakrabarti; Saı̈d M. Sebti; Michael H. Gelb; Wesley C. Van Voorhis; Andrew D. Hamilton

New chemotherapeutics are urgently needed to combat malaria. We previously reported on a novel series of antimalarial, ethylenediamine-based inhibitors of protein farnesyltransferase (PFT). In the current study, we designed and synthesized a series of second generation inhibitors, wherein the core ethylenediamine scaffold was varied in order to examine both the homology model of Plasmodium falciparum PFT (PfPFT) and our predicted inhibitor binding mode. We identified several PfPFT inhibitors (PfPFTIs) that are selective for PfPFT versus the mammalian isoform of the enzyme (up to 136-fold selectivity), that inhibit the malarial enzyme with IC50 values down to 1 nM, and that block the growth of P. falciparum in infected whole cells (erythrocytes) with ED50 values down to 55 nM. The structure-activity data for these second generation, ethylenediamine-inspired PFT inhibitors were rationalized by consideration of the X-ray crystal structure of mammalian PFT and the homology model of the malarial enzyme.


Antimicrobial Agents and Chemotherapy | 2007

Efficacy, Pharmacokinetics, and Metabolism of Tetrahydroquinoline Inhibitors of Plasmodium falciparum Protein Farnesyltransferase

Wesley C. Van Voorhis; Kasey Rivas; Pravin Bendale; Laxman Nallan; Carolyn P. Hornéy; Lynn K. Barrett; Kevin D. Bauer; Brian P. Smart; Sudha Ankala; Oliver Hucke; Christophe L. M. J. Verlinde; Debopam Chakrabarti; Corey Strickland; Kohei Yokoyama; Frederick S. Buckner; Andrew D. Hamilton; David K. Williams; Louis J. Lombardo; David M. Floyd; Michael H. Gelb

ABSTRACT New antimalarials are urgently needed. We have shown that tetrahydroquinoline (THQ) protein farnesyltransferase (PFT) inhibitors (PFTIs) are effective against the Plasmodium falciparum PFT and are effective at killing P. falciparum in vitro. Previously described THQ PFTIs had limitations of poor oral bioavailability and rapid clearance from the circulation of rodents. In this paper, we validate both the Caco-2 cell permeability model for predicting THQ intestinal absorption and the in vitro liver microsome model for predicting THQ clearance in vivo. Incremental improvements in efficacy, oral absorption, and clearance rate were monitored by in vitro tests; and these tests were followed up with in vivo absorption, distribution, metabolism, and excretion studies. One compound, PB-93, achieved cure when it was given orally to P. berghei-infected rats every 8 h for a total of 72 h. However, PB-93 was rapidly cleared, and dosing every 12 h failed to cure the rats. Thus, the in vivo results corroborate the in vitro pharmacodynamics and demonstrate that 72 h of continuous high-level exposure to PFTIs is necessary to kill plasmodia. The metabolism of PB-93 was demonstrated by a novel technique that relied on double labeling with a radiolabel and heavy isotopes combined with radiometric liquid chromatography and mass spectrometry. The major liver microsome metabolite of PB-93 has the PFT Zn-binding N-methyl-imidazole removed; this metabolite is inactive in blocking PFT function. By solving the X-ray crystal structure of PB-93 bound to rat PFT, a model of PB-93 bound to malarial PFT was constructed. This model suggests areas of the THQ PFTIs that can be modified to retain efficacy and protect the Zn-binding N-methyl-imidazole from dealkylation.


Journal of Medicinal Chemistry | 2008

2-Oxotetrahydroquinoline-Based Antimalarials with High Potency and Metabolic Stability

Vivek J. Bulbule; Kasey Rivas; Christophe L. M. J. Verlinde; Wesley C. Van Voorhis; Michael H. Gelb

We report a series of novel inhibitors of protein farnesyltransferase based on the 2-oxotetrahydroquinoline scaffold. We developed an efficient synthesis of these compounds. These compounds show selective inhibtion of the malaria versus human farnesyltransferase and inhibit the growth of the malaria parasite in the low nanomolar range. Some of the compounds are at least an order of magnitude more stable to metabolic degradation than the corresponding tetrahydroquinolines.


The Journal of Infectious Diseases | 2017

Bumped-Kinase Inhibitors for Cryptosporidiosis Therapy

Matthew A. Hulverson; Sumiti Vinayak; Ryan Choi; Deborah A. Schaefer; Alejandro Castellanos-Gonzalez; Rama Subba Rao Vidadala; Carrie F. Brooks; Gillian T. Herbert; Dana P. Betzer; Grant R. Whitman; Hayley Sparks; Samuel L.M. Arnold; Kasey Rivas; Lynn K. Barrett; A. Clinton White; Dustin J. Maly; Michael W. Riggs; Boris Striepen; Wesley C. Van Voorhis; Kayode K. Ojo

Bumped kinase inhibitors (BKIs) of Cryptosporidium parvum calcium-dependent protein kinase 1 (CpCDPK1) are leading candidates for treatment of cryptosporidiosis-associated diarrhea. Potential cardiotoxicity related to anti-human ether-à-go-go potassium channel (hERG) activity of the first-generation anti-Cryptosporidium BKIs triggered further testing for efficacy. A luminescence assay adapted for high-throughput screening was used to measure inhibitory activities of BKIs against C. parvum in vitro. Furthermore, neonatal and interferon γ knockout mouse models of C. parvum infection identified BKIs with in vivo activity. Additional iterative experiments for optimum dosing and selecting BKIs with minimum levels of hERG activity and frequencies of other safety liabilities included those that investigated mammalian cell cytotoxicity, C. parvum proliferation inhibition in vitro, anti-human Src inhibition, hERG activity, in vivo pharmacokinetic data, and efficacy in other mouse models. Findings of this study suggest that fecal concentrations greater than parasite inhibitory concentrations correlate best with effective therapy in the mouse model of cryptosporidiosis, but a more refined model for efficacy is needed.


PLOS ONE | 2016

Biochemical Screening of Five Protein Kinases from Plasmodium falciparum against 14,000 Cell-Active Compounds

Gregory J. Crowther; Heidi Hillesland; Katelyn R. Keyloun; Molly C. Reid; Maria Jose Lafuente-Monasterio; Sonja Ghidelli-Disse; Stephen E. Leonard; Panqing He; Jackson C. Jones; Mallory M. Krahn; Jack S. Mo; Kartheek S. Dasari; Anna M. W. Fox; Markus Boesche; Majida El Bakkouri; Kasey Rivas; Didier Leroy; Raymond Hui; Gerard Drewes; Dustin J. Maly; Wesley C. Van Voorhis; Kayode K. Ojo

In 2010 the identities of thousands of anti-Plasmodium compounds were released publicly to facilitate malaria drug development. Understanding these compounds’ mechanisms of action—i.e., the specific molecular targets by which they kill the parasite—would further facilitate the drug development process. Given that kinases are promising anti-malaria targets, we screened ~14,000 cell-active compounds for activity against five different protein kinases. Collections of cell-active compounds from GlaxoSmithKline (the ~13,000-compound Tres Cantos Antimalarial Set, or TCAMS), St. Jude Children’s Research Hospital (260 compounds), and the Medicines for Malaria Venture (the 400-compound Malaria Box) were screened in biochemical assays of Plasmodium falciparum calcium-dependent protein kinases 1 and 4 (CDPK1 and CDPK4), mitogen-associated protein kinase 2 (MAPK2/MAP2), protein kinase 6 (PK6), and protein kinase 7 (PK7). Novel potent inhibitors (IC50 < 1 μM) were discovered for three of the kinases: CDPK1, CDPK4, and PK6. The PK6 inhibitors are the most potent yet discovered for this enzyme and deserve further scrutiny. Additionally, kinome-wide competition assays revealed a compound that inhibits CDPK4 with few effects on ~150 human kinases, and several related compounds that inhibit CDPK1 and CDPK4 yet have limited cytotoxicity to human (HepG2) cells. Our data suggest that inhibiting multiple Plasmodium kinase targets without harming human cells is challenging but feasible.


ACS Infectious Diseases | 2017

Biochemical and Structural Characterization of Selective Allosteric Inhibitors of the Plasmodium falciparum Drug Target, Prolyl-tRNA-synthetase.

Stephen N. Hewitt; David M. Dranow; Benjamin G. Horst; Jan Abendroth; Barbara Forte; Irene Hallyburton; Chimed Jansen; Beatriz Baragaña; Ryan Choi; Kasey Rivas; Matthew A. Hulverson; Mitchell Dumais; Thomas E. Edwards; Donald D. Lorimer; Alan H. Fairlamb; David W. Gray; Kevin D. Read; Adele M. Lehane; Kiaran Kirk; Peter J. Myler; Amy K. Wernimont; Chris Walpole; Robin Stacy; Lynn K. Barrett; Ian H. Gilbert; Wesley C. Van Voorhis

Plasmodium falciparum (Pf) prolyl-tRNA synthetase (ProRS) is one of the few chemical-genetically validated drug targets for malaria, yet highly selective inhibitors have not been described. In this paper, approximately 40,000 compounds were screened to identify compounds that selectively inhibit PfProRS enzyme activity versus Homo sapiens (Hs) ProRS. X-ray crystallography structures were solved for apo, as well as substrate- and inhibitor-bound forms of PfProRS. We identified two new inhibitors of PfProRS that bind outside the active site. These two allosteric inhibitors showed >100 times specificity for PfProRS compared to HsProRS, demonstrating this class of compounds could overcome the toxicity related to HsProRS inhibition by halofuginone and its analogues. Initial medicinal chemistry was performed on one of the two compounds, guided by the cocrystallography of the compound with PfProRS, and the results can instruct future medicinal chemistry work to optimize these promising new leads for drug development against malaria.

Collaboration


Dive into the Kasey Rivas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kayode K. Ojo

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dustin J. Maly

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ryan Choi

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Erkang Fan

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Debopam Chakrabarti

University of Central Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge