217 results on '"R. Kiplin Guy"'
Search Results
2. Similarly efficacious anti-malarial drugs SJ733 and pyronaridine differ in their ability to remove circulating parasites in mice
- Author
-
Arya SheelaNair, Aleksandra S. Romanczuk, Rosemary A. Aogo, Rohit Nemai Haldar, Lianne I. M. Lansink, Deborah Cromer, Yandira G. Salinas, R. Kiplin Guy, James S. McCarthy, Miles P. Davenport, Ashraful Haque, and David S. Khoury
- Subjects
SJ733 ,Pyronaridine ,Artesunate ,Plasmodium berghei ,Parasite clearance ,Arctic medicine. Tropical medicine ,RC955-962 ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Artemisinin-based combination therapy (ACT) has been a mainstay for malaria prevention and treatment. However, emergence of drug resistance has incentivised development of new drugs. Defining the kinetics with which circulating parasitized red blood cells (pRBC) are lost after drug treatment, referred to as the “parasite clearance curve”, has been critical for assessing drug efficacy; yet underlying mechanisms remain partly unresolved. The clearance curve may be shaped both by the rate at which drugs kill parasites, and the rate at which drug-affected parasites are removed from circulation. Methods In this context, two anti-malarials, SJ733, and an ACT partner drug, pyronaridine were compared against sodium artesunate in mice infected with Plasmodium berghei (strain ANKA). To measure each compound’s capacity for pRBC removal in vivo, flow cytometric monitoring of a single cohort of fluorescently-labelled pRBC was employed, and combined with ex vivo parasite culture to assess parasite maturation and replication. Results These three compounds were found to be similarly efficacious in controlling established infection by reducing overall parasitaemia. While sodium artesunate acted relatively consistently across the life-stages, single-dose SJ733 elicited a biphasic effect, triggering rapid, partly phagocyte-dependent removal of trophozoites and schizonts, followed by arrest of residual ring-stages. In contrast, pyronaridine abrogated maturation of younger parasites, with less pronounced effects on mature parasites, while modestly increasing pRBC removal. Conclusions Anti-malarials SJ733 and pyronaridine, though similarly efficacious in reducing overall parasitaemia in mice, differed markedly in their capacity to arrest replication and remove pRBC from circulation. Thus, similar parasite clearance curves can result for anti-malarials with distinct capacities to inhibit, kill and clear parasites.
- Published
- 2022
- Full Text
- View/download PDF
3. Selecting an anti-malarial clinical candidate from two potent dihydroisoquinolones
- Author
-
Yizhe Chen, Fangyi Zhu, Jared Hammill, Gloria Holbrook, Lei Yang, Burgess Freeman, Karen L. White, David M. Shackleford, Kathleen G. O’Loughlin, Susan A. Charman, Jon C. Mirsalis, and R. Kiplin Guy
- Subjects
Candidate selection ,Physicochemical properties ,In vitro and in vivo metabolism ,Bioavailability ,Dose proportional exposure ,Arctic medicine. Tropical medicine ,RC955-962 ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background The ongoing global malaria eradication campaign requires development of potent, safe, and cost-effective drugs lacking cross-resistance with existing chemotherapies. One critical step in drug development is selecting a suitable clinical candidate from late leads. The process used to select the clinical candidate SJ733 from two potent dihydroisoquinolone (DHIQ) late leads, SJ733 and SJ311, based on their physicochemical, pharmacokinetic (PK), and toxicity profiles is described. Methods The compounds were tested to define their physicochemical properties including kinetic and thermodynamic solubility, partition coefficient, permeability, ionization constant, and binding to plasma proteins. Metabolic stability was assessed in both microsomes and hepatocytes derived from mice, rats, dogs, and humans. Cytochrome P450 inhibition was assessed using recombinant human cytochrome enzymes. The pharmacokinetic profiles of single intravenous or oral doses were investigated in mice, rats, and dogs. Results Although both compounds displayed similar physicochemical properties, SJ733 was more permeable but metabolically less stable than SJ311 in vitro. Single dose PK studies of SJ733 in mice, rats, and dogs demonstrated appreciable oral bioavailability (60–100%), whereas SJ311 had lower oral bioavailability (mice 23%, rats 40%) and higher renal clearance (10–30 fold higher than SJ733 in rats and dogs), suggesting less favorable exposure in humans. SJ311 also displayed a narrower range of dose-proportional exposure, with plasma exposure flattening at doses above 200 mg/kg. Conclusion SJ733 was chosen as the candidate based on a more favorable dose proportionality of exposure and stronger expectation of the ability to justify a strong therapeutic index to regulators.
- Published
- 2021
- Full Text
- View/download PDF
4. Combining SJ733, an oral ATP4 inhibitor of Plasmodium falciparum, with the pharmacokinetic enhancer cobicistat: An innovative approach in antimalarial drug development
- Author
-
Aditya H. Gaur, John C. Panetta, Amber M. Smith, Ronald H. Dallas, Burgess B. Freeman, III, Tracy B. Stewart, Li Tang, Elizabeth John, Kristen C. Branum, Nehali D. Patel, Shelley Ost, Ryan N. Heine, Julie L. Richardson, Jared T. Hammill, Lidiya Bebrevska, Fabian Gusovsky, Noritsugu Maki, Toshiharu Yanagi, Patricia M. Flynn, James S. McCarthy, Stephan Chalon, and R. Kiplin Guy
- Subjects
SJ733 ,Antimalarial ,Pharmacoboost ,Pharmacokinetics ,Pharmacodynamics ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: SJ733, a newly developed inhibitor of P. falciparum ATP4, has a favorable safety profile and rapid antiparasitic effect but insufficient duration to deliver a single-dose cure of malaria. We investigated the safety, tolerability, and pharmacokinetics of a multidose SJ733 regimen and a single-dose pharmacoboost approach using cobicistat to inhibit CYP3A4, thereby increasing exposure. Methods: Two multidose unboosted cohorts (n = 9) (SJ733, 300 mg and 600 mg daily for 3 days) followed by three single-dose boosted cohorts combining SJ733 (n = 18) (75-, 300-, or 600-mg single dose) with cobicistat (150-mg single dose) as a pharmacokinetic booster were evaluated in healthy volunteers (ClinicalTrials.gov: NCT02661373). Findings: All participants tolerated SJ733 well, with no serious adverse events (AEs), dose-limiting toxicity, or clinically significant electrocardiogram or laboratory test findings. All reported AEs were Grade 1, clinically insignificant, and considered unlikely or unrelated to SJ733. Compared to unboosted cohorts, the SJ733/cobicistat-boosted cohorts showed a median increase in area under the curve and maximum concentration of 3·9 × and 2·6 ×, respectively, and a median decrease in the ratio of the major CYP3A-produced metabolite SJ506 to parent drug of 4·6 × . Incorporating these data in a model of parasite dynamics indicated that a 3-day regimen of SJ733/cobicistat (600 mg/150 mg daily) relative to a single 600-mg dose ± cobicistat would increase parasite clearance from 106 to 1012 parasites/µL. Interpretation: The multidose and pharmacoboosted approaches to delivering SJ733 were well-tolerated and significantly increased drug exposure and prediction of cure. This study supports the further development of SJ733 and demonstrates an innovative pharmacoboost approach for an antimalarial. Funding: Global Health Innovative Technology Fund, Medicines for Malaria Venture, National Institutes of Health, and American Lebanese Syrian Associated Charities.
- Published
- 2022
- Full Text
- View/download PDF
5. Author Correction: Proteasome inhibition targets the KMT2A transcriptional complex in acute lymphoblastic leukemia
- Author
-
Jennifer L. Kamens, Stephanie Nance, Cary Koss, Beisi Xu, Anitria Cotton, Jeannie W. Lam, Elizabeth A. R. Garfinkle, Pratima Nallagatla, Amelia M. R. Smith, Sharnise Mitchell, Jing Ma, Duane Currier, William C. Wright, Kanisha Kavdia, Vishwajeeth R. Pagala, Wonil Kim, LaShanale M. Wallace, Ji-Hoon Cho, Yiping Fan, Aman Seth, Nathaniel Twarog, John K. Choi, Esther A. Obeng, Mark E. Hatley, Monika L. Metzger, Hiroto Inaba, Sima Jeha, Jeffrey E. Rubnitz, Junmin Peng, Taosheng Chen, Anang A. Shelat, R. Kiplin Guy, and Tanja A. Gruber
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
6. A high-throughput screen indicates gemcitabine and JAK inhibitors may be useful for treating pediatric AML
- Author
-
Christina D. Drenberg, Anang Shelat, Jinjun Dang, Anitria Cotton, Shelley J. Orwick, Mengyu Li, Jae Yoon Jeon, Qiang Fu, Daelynn R. Buelow, Marissa Pioso, Shuiying Hu, Hiroto Inaba, Raul C. Ribeiro, Jeffrey E. Rubnitz, Tanja A. Gruber, R. Kiplin Guy, and Sharyn D. Baker
- Subjects
Science - Abstract
Pediatric AML is traditionally treated with chemotherapy and stem cell transplant but some subsets of patients have a poor response to therapy. Here, the authors perform a high throughput screen and identify several FDA approved drugs that might be useful in treating this disease.
- Published
- 2019
- Full Text
- View/download PDF
7. Discovery of a Diaminopyrimidine FLT3 Inhibitor Active against Acute Myeloid Leukemia
- Author
-
Jamie A. Jarusiewicz, Jae Yoon Jeon, Michele C. Connelly, Yizhe Chen, Lei Yang, Sharyn D. Baker, and R. Kiplin Guy
- Subjects
Chemistry ,QD1-999 - Published
- 2017
- Full Text
- View/download PDF
8. Open Source Drug Discovery: Highly Potent Antimalarial Compounds Derived from the Tres Cantos Arylpyrroles
- Author
-
Alice E. Williamson, Paul M. Ylioja, Murray N. Robertson, Yevgeniya Antonova-Koch, Vicky Avery, Jonathan B. Baell, Harikrishna Batchu, Sanjay Batra, Jeremy N. Burrows, Soumya Bhattacharyya, Felix Calderon, Susan A. Charman, Julie Clark, Benigno Crespo, Matin Dean, Stefan L. Debbert, Michael Delves, Adelaide S. M. Dennis, Frederik Deroose, Sandra Duffy, Sabine Fletcher, Guri Giaever, Irene Hallyburton, Francisco-Javier Gamo, Marinella Gebbia, R. Kiplin Guy, Zoe Hungerford, Kiaran Kirk, Maria J. Lafuente-Monasterio, Anna Lee, Stephan Meister, Corey Nislow, John P. Overington, George Papadatos, Luc Patiny, James Pham, Stuart A. Ralph, Andrea Ruecker, Eileen Ryan, Christopher Southan, Kumkum Srivastava, Chris Swain, Matthew J. Tarnowski, Patrick Thomson, Peter Turner, Iain M. Wallace, Timothy N. C. Wells, Karen White, Laura White, Paul Willis, Elizabeth A. Winzeler, Sergio Wittlin, and Matthew H. Todd
- Subjects
Chemistry ,QD1-999 - Published
- 2016
- Full Text
- View/download PDF
9. Shared Consensus Machine Learning Models for Predicting Blood Stage Malaria Inhibition.
- Author
-
Andreas Verras, Chris L. Waller, Peter Gedeck, Darren V. S. Green, Thierry Kogej, Anandkumar Raichurkar, Manoranjan Panda, Anang A. Shelat, Julie Clark, R. Kiplin Guy, George Papadatos, and Jeremy N. Burrows
- Published
- 2017
- Full Text
- View/download PDF
10. Data from Targeting Histone Demethylases in MYC-Driven Neuroblastomas with Ciclopirox
- Author
-
Andrew M. Davidoff, Taosheng Chen, R. Kiplin Guy, Stephen White, Douglas R. Green, Junmin Peng, Chunxu Qu, Yong Cheng, Ashutosh Mishra, Jie J. Zheng, Zhenmei Li, Yinan Wu, Anang Shelat, Sourav Das, Brandon Young, Vincent A. Boyd, Su Sien Ong, Yingdi Wang, Ruoning Wang, Amit C. Nathwani, Pollyanna Goh, Ju Bao, Wenwei Lin, Jonathan Low, Jesse Davidson, Junfang Zhou, Rodrigo B. Interiano, Alaa M. AlTahan, Dongli Hu, Sandra Milasta, and Jun Yang
- Abstract
Histone lysine demethylases facilitate the activity of oncogenic transcription factors, including possibly MYC. Here we show that multiple histone demethylases influence the viability and poor prognosis of neuroblastoma cells, where MYC is often overexpressed. We also identified the approved small-molecule antifungal agent ciclopirox as a novel pan-histone demethylase inhibitor. Ciclopirox targeted several histone demethylases, including KDM4B implicated in MYC function. Accordingly, ciclopirox inhibited Myc signaling in parallel with mitochondrial oxidative phosphorylation, resulting in suppression of neuroblastoma cell viability and inhibition of tumor growth associated with an induction of differentiation. Our findings provide new insights into epigenetic regulation of MYC function and suggest a novel pharmacologic basis to target histone demethylases as an indirect MYC-targeting approach for cancer therapy. Cancer Res; 77(17); 4626–38. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Bromodomain-Selective BET Inhibitors Are Potent Antitumor Agents against MYC-Driven Pediatric Cancer
- Author
-
Philip M. Potter, Anang A. Shelat, Stephen W. White, R. Kiplin Guy, Daniel Savic, Taosheng Chen, Martine F. Roussel, Marie Morfouace, Richard E. Lee, Jun Yang, Vincent A. Boyd, Nagakumar Bharatham, Brandon M. Young, Jeanine E. Price, Jonathan A. Low, Kevin W. Freeman, Rachelle R. Olsen, William R. Shadrick, Zhenmei Li, Geoffrey Neale, Sourav Das, M. Brett Waddell, Michele Connelly, Sergio C. Chai, Barbara Jonchere, Nancy E. Martinez, Lyudmila Tsurkan, Mi-Kyung Yun, Liying Chi, and P. Jake Slavish
- Abstract
Inhibition of members of the bromodomain and extraterminal (BET) family of proteins has proven a valid strategy for cancer chemotherapy. All BET identified to date contain two bromodomains (BD; BD1 and BD2) that are necessary for recognition of acetylated lysine residues in the N-terminal regions of histones. Chemical matter that targets BET (BETi) also interact via these domains. Molecular and cellular data indicate that BD1 and BD2 have different biological roles depending upon their cellular context, with BD2 particularly associated with cancer. We have therefore pursued the development of BD2-selective molecules both as chemical probes and as potential leads for drug development. Here we report the structure-based generation of a novel series of tetrahydroquinoline analogs that exhibit >50-fold selectivity for BD2 versus BD1. This selective targeting resulted in engagement with BD-containing proteins in cells, resulting in modulation of MYC proteins and downstream targets. These compounds were potent cytotoxins toward numerous pediatric cancer cell lines and were minimally toxic to nontumorigenic cells. In addition, unlike the pan BETi (+)-JQ1, these BD2-selective inhibitors demonstrated no rebound expression effects. Finally, we report a pharmacokinetic-optimized, metabolically stable derivative that induced growth delay in a neuroblastoma xenograft model with minimal toxicity. We conclude that BD2-selective agents are valid candidates for antitumor drug design for pediatric malignancies driven by the MYC oncogene.Significance:This study presents bromodomain-selective BET inhibitors that act as antitumor agents and demonstrates that these molecules have in vivo activity towards neuroblastoma, with essentially no toxicity.
- Published
- 2023
- Full Text
- View/download PDF
12. Figure S1-S6, Table S1-S2 from Targeting Histone Demethylases in MYC-Driven Neuroblastomas with Ciclopirox
- Author
-
Andrew M. Davidoff, Taosheng Chen, R. Kiplin Guy, Stephen White, Douglas R. Green, Junmin Peng, Chunxu Qu, Yong Cheng, Ashutosh Mishra, Jie J. Zheng, Zhenmei Li, Yinan Wu, Anang Shelat, Sourav Das, Brandon Young, Vincent A. Boyd, Su Sien Ong, Yingdi Wang, Ruoning Wang, Amit C. Nathwani, Pollyanna Goh, Ju Bao, Wenwei Lin, Jonathan Low, Jesse Davidson, Junfang Zhou, Rodrigo B. Interiano, Alaa M. AlTahan, Dongli Hu, Sandra Milasta, and Jun Yang
- Abstract
Figure S1 shows siRNA screening in neuroblastoma cells. Figure S2 shows the dentification of ciclopirox that targets KDM4B. Figure S3 shows the CPX effect on other histone methyl marks. Figure S4 shows that CPX targets the Myc pathway. Figure S5 shows that CPX inhibits oxidative phosphorylation. Figure S6 shows the CPX effect on BE2C xenografts with KDM4B knockdown. Table S1 shows the sequences of the focused KDM siRNA library. Table S2 shows the primers for real time PCR.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figures 1-3 from An Antagonist of Dishevelled Protein-Protein Interaction Suppresses β-Catenin–Dependent Tumor Cell Growth
- Author
-
David M. Jablons, R. Kiplin Guy, Jie Zheng, Geoffrey Neale, Lillian R. Edmondson, Iwao Mikami, Biao He, Jufang Shan, Kazutsugu Uematsu, Zhidong Xu, Liang You, and Naoaki Fujii
- Abstract
Supplementary Figures 1-3 from An Antagonist of Dishevelled Protein-Protein Interaction Suppresses β-Catenin–Dependent Tumor Cell Growth
- Published
- 2023
- Full Text
- View/download PDF
14. Amino-Substituted 3-Aryl- and 3-Heteroarylquinolines as Potential Antileishmanial Agents
- Author
-
Kristin L. Begley, Amy L. Rice, Jean-Claude Dujardin, Jonah Rector, Vivek M. Rangnekar, Corinne M. Fargo, David S. Watt, Yizhe Chen, Diana Ortiz, Liliia M. Kril, Vitaliy M. Sviripa, Ho Shin Kim, Scott M. Landfear, Malgorzata A. Domagalska, Jared T. Hammill, Chunming Liu, and R. Kiplin Guy
- Subjects
Leishmania donovani ,Leishmaniasis, Cutaneous ,Pharmacology ,Article ,Structure-Activity Relationship ,Cutaneous leishmaniasis ,In vivo ,Drug Discovery ,parasitic diseases ,medicine ,Animals ,Amastigote ,Leishmania ,Mice, Inbred BALB C ,biology ,Molecular Structure ,Chemistry ,Leishmaniasis ,medicine.disease ,biology.organism_classification ,Trypanocidal Agents ,Microsomes, Liver ,Quinolines ,Molecular Medicine ,Antimonial ,Protozoa ,Female - Abstract
Leishmaniasis, a disease caused by protozoa of the Leishmania species, afflicts roughly 12 million individuals worldwide. Most existing drugs for leishmaniasis are toxic, expensive, difficult to administer, and subject to drug resistance. We report a new class of antileishmanial leads, the 3-arylquinolines, that potently block proliferation of the intramacrophage amastigote form of Leishmania parasites with good selectivity relative to the host macrophages. Early lead 34 was rapidly acting and possessed good potency against L. mexicana (EC50 = 120 nM), 30-fold selectivity for the parasite relative to the macrophage (EC50 = 3.7 μM), and also blocked proliferation of Leishmania donovani parasites resistant to antimonial drugs. Finally, another early lead, 27, which exhibited reasonable in vivo tolerability, impaired disease progression during the dosing period in a murine model of cutaneous leishmaniasis. These results suggest that the arylquinolines provide a fruitful departure point for the development of new antileishmanial drugs.
- Published
- 2021
15. Discovery of novel, orally bioavailable, antileishmanial compounds using phenotypic screening.
- Author
-
Diana Ortiz, W Armand Guiguemde, Jared T Hammill, Angela K Carrillo, Yizhe Chen, Michele Connelly, Kayla Stalheim, Carolyn Elya, Alex Johnson, Jaeki Min, Anang Shelat, David C Smithson, Lei Yang, Fangyi Zhu, R Kiplin Guy, and Scott M Landfear
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Leishmaniasis is a parasitic infection that afflicts approximately 12 million people worldwide. There are several limitations to the approved drug therapies for leishmaniasis, including moderate to severe toxicity, growing drug resistance, and the need for extended dosing. Moreover, miltefosine is currently the only orally available drug therapy for this infection. We addressed the pressing need for new therapies by pursuing a two-step phenotypic screen to discover novel, potent, and orally bioavailable antileishmanials. First, we conducted a high-throughput screen (HTS) of roughly 600,000 small molecules for growth inhibition against the promastigote form of the parasite life cycle using the nucleic acid binding dye SYBR Green I. This screen identified approximately 2,700 compounds that inhibited growth by over 65% at a single point concentration of 10 μM. We next used this 2700 compound focused library to identify compounds that were highly potent against the disease-causing intra-macrophage amastigote form and exhibited limited toxicity toward the host macrophages. This two-step screening strategy uncovered nine unique chemical scaffolds within our collection, including two previously described antileishmanials. We further profiled two of the novel compounds for in vitro absorption, distribution, metabolism, excretion, and in vivo pharmacokinetics. Both compounds proved orally bioavailable, affording plasma exposures above the half-maximal effective concentration (EC50) concentration for at least 12 hours. Both compounds were efficacious when administered orally in a murine model of cutaneous leishmaniasis. One of the two compounds exerted potent activity against trypanosomes, which are kinetoplastid parasites related to Leishmania species. Therefore, this compound could help control multiple parasitic diseases. The promising pharmacokinetic profile and significant in vivo efficacy observed from our HTS hits highlight the utility of our two-step phenotypic screening strategy and strongly suggest that medicinal chemistry optimization of these newly identified scaffolds will lead to promising candidates for an orally available anti-parasitic drug.
- Published
- 2017
- Full Text
- View/download PDF
16. Discovery of Novel Antimalarial Compounds Enabled by QSAR-Based Virtual Screening.
- Author
-
Liying Zhang, Denis Fourches, Alexander Sedykh, Hao Zhu 0012, Alexander Golbraikh, Sean Ekins, Julie Clark, Michele C. Connelly, Martina Sigal, Dena Hodges, Armand Guiguemde, R. Kiplin Guy, and Alexander Tropsha
- Published
- 2013
- Full Text
- View/download PDF
17. Selecting an anti-malarial clinical candidate from two potent dihydroisoquinolones
- Author
-
Kathleen O’Loughlin, Yizhe Chen, Lei Yang, Fangyi Zhu, Jared T. Hammill, Karen L. White, Gloria Holbrook, David M. Shackleford, Susan A. Charman, Jon C. Mirsalis, R. Kiplin Guy, and Burgess B. Freeman
- Subjects
0301 basic medicine ,In vitro and in vivo metabolism ,lcsh:Arctic medicine. Tropical medicine ,Bioavailability ,Physicochemical properties ,lcsh:RC955-962 ,Biological Availability ,Pharmacology ,Dose proportional exposure ,Heterocyclic Compounds, 4 or More Rings ,lcsh:Infectious and parasitic diseases ,03 medical and health sciences ,Antimalarials ,Mice ,0302 clinical medicine ,Therapeutic index ,Dogs ,Pharmacokinetics ,Animals ,Humans ,lcsh:RC109-216 ,030212 general & internal medicine ,biology ,Chemistry ,Research ,Cytochrome P450 ,Isoquinolines ,Blood proteins ,In vitro ,Rats ,030104 developmental biology ,Infectious Diseases ,Toxicity ,biology.protein ,Microsome ,Hepatocytes ,Microsomes, Liver ,Parasitology ,Candidate selection - Abstract
Background The ongoing global malaria eradication campaign requires development of potent, safe, and cost-effective drugs lacking cross-resistance with existing chemotherapies. One critical step in drug development is selecting a suitable clinical candidate from late leads. The process used to select the clinical candidate SJ733 from two potent dihydroisoquinolone (DHIQ) late leads, SJ733 and SJ311, based on their physicochemical, pharmacokinetic (PK), and toxicity profiles is described. Methods The compounds were tested to define their physicochemical properties including kinetic and thermodynamic solubility, partition coefficient, permeability, ionization constant, and binding to plasma proteins. Metabolic stability was assessed in both microsomes and hepatocytes derived from mice, rats, dogs, and humans. Cytochrome P450 inhibition was assessed using recombinant human cytochrome enzymes. The pharmacokinetic profiles of single intravenous or oral doses were investigated in mice, rats, and dogs. Results Although both compounds displayed similar physicochemical properties, SJ733 was more permeable but metabolically less stable than SJ311 in vitro. Single dose PK studies of SJ733 in mice, rats, and dogs demonstrated appreciable oral bioavailability (60–100%), whereas SJ311 had lower oral bioavailability (mice 23%, rats 40%) and higher renal clearance (10–30 fold higher than SJ733 in rats and dogs), suggesting less favorable exposure in humans. SJ311 also displayed a narrower range of dose-proportional exposure, with plasma exposure flattening at doses above 200 mg/kg. Conclusion SJ733 was chosen as the candidate based on a more favorable dose proportionality of exposure and stronger expectation of the ability to justify a strong therapeutic index to regulators.
- Published
- 2021
18. Screening and Development of New Inhibitors of FtsZ from M. Tuberculosis.
- Author
-
Bini Mathew, Judith Varady Hobrath, Larry Ross, Michele C Connelly, Hava Lofton, Malini Rajagopalan, R Kiplin Guy, and Robert C Reynolds
- Subjects
Medicine ,Science - Abstract
A variety of commercial analogs and a newer series of Sulindac derivatives were screened for inhibition of M. tuberculosis (Mtb) in vitro and specifically as inhibitors of the essential mycobacterial tubulin homolog, FtsZ. Due to the ease of preparing diverse analogs and a favorable in vivo pharmacokinetic and toxicity profile of a representative analog, the Sulindac scaffold may be useful for further development against Mtb with respect to in vitro bacterial growth inhibition and selective activity for Mtb FtsZ versus mammalian tubulin. Further discovery efforts will require separating reported mammalian cell activity from both antibacterial activity and inhibition of Mtb FtsZ. Modeling studies suggest that these analogs bind in a specific region of the Mtb FtsZ polymer that differs from human tubulin and, in combination with a pharmacophore model presented herein, future hybrid analogs of the reported active molecules that more efficiently bind in this pocket may improve antibacterial activity while improving other drug characteristics.
- Published
- 2016
- Full Text
- View/download PDF
19. Synthesis and Structure–Activity Relationship of Dual-Stage Antimalarial Pyrazolo[3,4-b]pyridines
- Author
-
Marc O. Anderson, Martina Sigal, Ashley Cheung, Jared T. Hammill, Anna S Kashtanova, Korina Eribez, Lauren Loop, Horacio Lazaro, Irene García-Barbazán, Cory A. Chaplan, Kenya Yniguez, Benigno Crespo, Alisje Churchyard, Mofolusho O. Falade, Grant Koch, Briana Belanger, Celine DiBernardo, Jake Baum, Julia E Tryhorn, Amy L. Rice, Katie Kim, Joshua J. Kimball, R. Kiplin Guy, Steven Wilkinson, Benoît Laleu, Nimisha Mittal, Rei Takahashi, Elizabeth A. Winzeler, Francisco-Javier Gamo, Scott Eagon, and Kevin J Ahn
- Subjects
biology ,Chemistry ,Plasmodium falciparum ,medicine.disease ,biology.organism_classification ,Virology ,In vitro ,Docking (molecular) ,Drug Discovery ,Toxicity ,Gametocyte ,medicine ,Molecular Medicine ,Structure–activity relationship ,Parasite hosting ,Malaria - Abstract
Malaria remains one of the most deadly infectious diseases, causing hundreds of thousands of deaths each year, primarily in young children and pregnant mothers. Here, we report the discovery and derivatization of a series of pyrazolo[3,4-b]pyridines targeting Plasmodium falciparum, the deadliest species of the malaria parasite. Hit compounds in this series display sub-micromolar in vitro activity against the intraerythrocytic stage of the parasite as well as little to no toxicity against the human fibroblast BJ and liver HepG2 cell lines. In addition, our hit compounds show good activity against the liver stage of the parasite but little activity against the gametocyte stage. Parasitological profiles, including rate of killing, docking, and molecular dynamics studies, suggest that our compounds may target the Qo binding site of cytochrome bc1.
- Published
- 2020
- Full Text
- View/download PDF
20. Bromodomain-Selective BET Inhibitors Are Potent Antitumor Agents against MYC-Driven Pediatric Cancer
- Author
-
Barbara Jonchere, Jonathan Low, Michele Connelly, Liying Chi, Sergio C. Chai, R. Kiplin Guy, Kevin W. Freeman, Brandon Young, Jeanine E. Price, Mi-Kyung Yun, Marie Morfouace, Anang A. Shelat, Geoffrey Neale, Rachelle R. Olsen, Nagakumar Bharatham, Daniel Savic, Stephen W. White, P. Jake Slavish, Vincent A. Boyd, Lyudmila Tsurkan, Jun J. Yang, Nancy E. Martinez, Zhenmei Li, Martine F. Roussel, Philip M. Potter, M. Brett Waddell, William R. Shadrick, Richard E. Lee, Taosheng Chen, and Sourav Das
- Subjects
0301 basic medicine ,Cancer Research ,Antineoplastic Agents ,Context (language use) ,Mice, SCID ,Article ,Proto-Oncogene Proteins c-myc ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Cell Line, Tumor ,Neoplasms ,Neuroblastoma ,medicine ,Animals ,Humans ,Child ,Cytotoxicity ,Oncogene ,Chemistry ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Pediatric cancer ,Bromodomain ,030104 developmental biology ,Oncology ,Drug development ,Drug Design ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Transcription Factors - Abstract
Inhibition of members of the bromodomain and extraterminal (BET) family of proteins has proven a valid strategy for cancer chemotherapy. All BET identified to date contain two bromodomains (BD; BD1 and BD2) that are necessary for recognition of acetylated lysine residues in the N-terminal regions of histones. Chemical matter that targets BET (BETi) also interact via these domains. Molecular and cellular data indicate that BD1 and BD2 have different biological roles depending upon their cellular context, with BD2 particularly associated with cancer. We have therefore pursued the development of BD2-selective molecules both as chemical probes and as potential leads for drug development. Here we report the structure-based generation of a novel series of tetrahydroquinoline analogs that exhibit >50-fold selectivity for BD2 versus BD1. This selective targeting resulted in engagement with BD-containing proteins in cells, resulting in modulation of MYC proteins and downstream targets. These compounds were potent cytotoxins toward numerous pediatric cancer cell lines and were minimally toxic to nontumorigenic cells. In addition, unlike the pan BETi (+)-JQ1, these BD2-selective inhibitors demonstrated no rebound expression effects. Finally, we report a pharmacokinetic-optimized, metabolically stable derivative that induced growth delay in a neuroblastoma xenograft model with minimal toxicity. We conclude that BD2-selective agents are valid candidates for antitumor drug design for pediatric malignancies driven by the MYC oncogene. Significance: This study presents bromodomain-selective BET inhibitors that act as antitumor agents and demonstrates that these molecules have in vivo activity towards neuroblastoma, with essentially no toxicity.
- Published
- 2020
- Full Text
- View/download PDF
21. 8-Triazolylpurines: Towards Fluorescent Inhibitors of the MDM2/p53 Interaction.
- Author
-
Mariell Pettersson, David Bliman, Jimmy Jacobsson, Jesper R Nilsson, Jaeki Min, Luigi Iconaru, R Kiplin Guy, Richard W Kriwacki, Joakim Andréasson, and Morten Grøtli
- Subjects
Medicine ,Science - Abstract
Small molecule nonpeptidic mimics of α-helices are widely recognised as protein-protein interaction (PPIs) inhibitors. Protein-protein interactions mediate virtually all important regulatory pathways in a cell, and the ability to control and modulate PPIs is therefore of great significance to basic biology, where controlled disruption of protein networks is key to understanding network connectivity and function. We have designed and synthesised two series of 2,6,9-substituted 8-triazolylpurines as α-helix mimetics. The first series was designed based on low energy conformations but did not display any biological activity in a biochemical fluorescence polarisation assay targeting MDM2/p53. Although solution NMR conformation studies demonstrated that such molecules could mimic the topography of an α-helix, docking studies indicated that the same compounds were not optimal as inhibitors for the MDM2/p53 interaction. A new series of 8-triazolylpurines was designed based on a combination of docking studies and analysis of recently published inhibitors. The best compound displayed low micromolar inhibitory activity towards MDM2/p53 in a biochemical fluorescence polarisation assay. In order to evaluate the applicability of these compounds as biologically active and intrinsically fluorescent probes, their absorption/emission properties were measured. The compounds display fluorescent properties with quantum yields up to 50%.
- Published
- 2015
- Full Text
- View/download PDF
22. Identification of Selective Inhibitors of the Plasmodium falciparum Hexose Transporter PfHT by Screening Focused Libraries of Anti-Malarial Compounds.
- Author
-
Diana Ortiz, W Armand Guiguemde, Alex Johnson, Carolyn Elya, Johanna Anderson, Julie Clark, Michele Connelly, Lei Yang, Jaeki Min, Yuko Sato, R Kiplin Guy, and Scott M Landfear
- Subjects
Medicine ,Science - Abstract
Development of resistance against current antimalarial drugs necessitates the search for novel drugs that interact with different targets and have distinct mechanisms of action. Malaria parasites depend upon high levels of glucose uptake followed by inefficient metabolic utilization via the glycolytic pathway, and the Plasmodium falciparum hexose transporter PfHT, which mediates uptake of glucose, has thus been recognized as a promising drug target. This transporter is highly divergent from mammalian hexose transporters, and it appears to be a permease that is essential for parasite viability in intra-erythrocytic, mosquito, and liver stages of the parasite life cycle. An assay was developed that is appropriate for high throughput screening against PfHT based upon heterologous expression of PfHT in Leishmania mexicana parasites that are null mutants for their endogenous hexose transporters. Screening of two focused libraries of antimalarial compounds identified two such compounds that are high potency selective inhibitors of PfHT compared to human GLUT1. Additionally, 7 other compounds were identified that are lower potency and lower specificity PfHT inhibitors but might nonetheless serve as starting points for identification of analogs with more selective properties. These results further support the potential of PfHT as a novel drug target.
- Published
- 2015
- Full Text
- View/download PDF
23. Design, Synthesis and Evaluation of 2,5-Diketopiperazines as Inhibitors of the MDM2-p53 Interaction.
- Author
-
Mariell Pettersson, Maria Quant, Jaeki Min, Luigi Iconaru, Richard W Kriwacki, M Brett Waddell, R Kiplin Guy, Kristina Luthman, and Morten Grøtli
- Subjects
Medicine ,Science - Abstract
The transcription factor p53 is the main tumour suppressor in cells and many cancer types have p53 mutations resulting in a loss of its function. In tumours that retain wild-type p53 function, p53 activity is down-regulated by MDM2 (human murine double minute 2) via a direct protein-protein interaction. We have designed and synthesised two series of 2,5-diketopiperazines as inhibitors of the MDM2-p53 interaction. The first set was designed to directly mimic the α-helical region of the p53 peptide, containing key residues in the i, i+4 and i+7 positions of a natural α-helix. Conformational analysis indicated that 1,3,6-trisubstituted 2,5-diketopiperazines were able to place substituents in the same spatial orientation as an α-helix template. The key step of the synthesis involved the cyclisation of substituted dipeptides. The other set of tetrasubstituted 2,5-diketopiperazines were designed based on structure-based docking studies and the Ugi multicomponent reaction was used for the synthesis. This latter set comprised the most potent inhibitors which displayed micromolar IC50-values in a biochemical fluorescence polarisation assay.
- Published
- 2015
- Full Text
- View/download PDF
24. Discovery of Novel Pyrazolo-pyridone DCN1 Inhibitors Controlling Cullin Neddylation
- Author
-
Bhuvanesh Singh, Jonah Rector, Jaeki Min, Brenda A. Schulman, R. Kiplin Guy, Jared T. Hammill, Ho Shin Kim, Daniel C. Scott, and Yizhe Chen
- Subjects
NEDD8 Protein ,Pyridones ,Glycine ,Cyclopentanes ,01 natural sciences ,NEDD8 ,Article ,Structure-Activity Relationship ,03 medical and health sciences ,Protein Domains ,Cell Line, Tumor ,Protein Interaction Mapping ,Drug Discovery ,Humans ,Structure–activity relationship ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Intracellular Signaling Peptides and Proteins ,Fibroblasts ,Cullin Proteins ,Amides ,Small molecule ,0104 chemical sciences ,Squamous carcinoma ,Cell biology ,010404 medicinal & biomolecular chemistry ,Pyrimidines ,Pevonedistat ,Drug Design ,Ubiquitin-Conjugating Enzymes ,Carcinoma, Squamous Cell ,biology.protein ,Pyrazoles ,Molecular Medicine ,CUL1 ,Neddylation ,Reactive Oxygen Species ,Cullin - Abstract
Chemical control of cullin neddylation is attracting increased attention based largely on the successes of the NEDD8-activating enzyme (E1) inhibitor Pevonedistat. Recently reported chemical probes enable selective and time-dependent inhibition of downstream members of the neddylation tri-enzymatic cascade including the co-E3, DCN1. In this work, we report the optimization of a novel class of small molecule inhibitors of the DCN1-UBE2M interaction. Rational X-ray co-structure enabled optimization afforded a 25-fold improvement in potency relative to the initial screening hit. The potency gains are largely attributed to additional hydrophobic interactions mimicking the N-terminal acetyl group that drives binding of UBE2M to DCN1. The compounds inhibit the protein-protein interaction, block NEDD8 transfer in biochemical assays, engage DCN1 in cells, and selectively reduce the steady-state neddylation of Cul1 and Cul3 in two squamous carcinoma cell lines harboring DCN1 amplification.
- Published
- 2019
- Full Text
- View/download PDF
25. A high-throughput screen indicates gemcitabine and JAK inhibitors may be useful for treating pediatric AML
- Author
-
Jeffrey E. Rubnitz, Anang A. Shelat, Daelynn R. Buelow, Shelley Orwick, Tanja A. Gruber, Marissa S. Pioso, Shuiying Hu, Mengyu Li, Hiroto Inaba, Qiang Fu, Christina D. Drenberg, Sharyn D. Baker, Raul C. Ribeiro, Jinjun Dang, R. Kiplin Guy, Anitria Cotton, and Jae Yoon Jeon
- Subjects
0301 basic medicine ,Oncology ,Male ,Myeloid ,medicine.medical_treatment ,General Physics and Astronomy ,02 engineering and technology ,Deoxycytidine ,Acute megakaryoblastic leukemia ,Mice ,Bone Marrow ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Child ,lcsh:Science ,Bone Marrow Transplantation ,Multidisciplinary ,Cytarabine ,021001 nanoscience & nanotechnology ,3. Good health ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Cabazitaxel ,Child, Preschool ,Female ,Taxoids ,0210 nano-technology ,Whole-Body Irradiation ,medicine.drug ,Adult ,medicine.medical_specialty ,Science ,Mice, Transgenic ,General Biochemistry, Genetics and Molecular Biology ,Disease-Free Survival ,Article ,Paediatric cancer ,03 medical and health sciences ,Young Adult ,Targeted therapies ,In vivo ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Janus Kinase Inhibitors ,Cancer models ,Chemotherapy ,Haematological cancer ,Leukemia, Experimental ,business.industry ,Infant ,General Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,Gemcitabine ,High-Throughput Screening Assays ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Q ,business - Abstract
Improvement in survival has been achieved for children and adolescents with AML but is largely attributed to enhanced supportive care as opposed to the development of better treatment regimens. High risk subtypes continue to have poor outcomes with event free survival rates, Pediatric AML is traditionally treated with chemotherapy and stem cell transplant but some subsets of patients have a poor response to therapy. Here, the authors perform a high throughput screen and identify several FDA approved drugs that might be useful in treating this disease.
- Published
- 2019
26. Antimalarial activity of 2,6-dibenzylidenecyclohexanone derivatives
- Author
-
Brandon Nguyen, Julia Elizabeth Glidden, Scott Eagon, Kasey Fitzsimmons, Jared T. Hammill, Kara Thompson, Jarvis Law, Mofolusho O. Falade, Steven Wilkinson, Amy L. Rice, Celine DiBernardo, Natalie Sienko, R. Kiplin Guy, Aashrita Manjunath, and Joshua J. Kimball
- Subjects
Clinical Biochemistry ,Plasmodium falciparum ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,Plasmodium ,Antimalarials ,Structure-Activity Relationship ,Parasitic Sensitivity Tests ,parasitic diseases ,Drug Discovery ,medicine ,Humans ,Molecular Biology ,Cell Proliferation ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Plasmodium parasite ,Treatment options ,Chloroquine ,Fibroblasts ,biology.organism_classification ,medicine.disease ,Virology ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Molecular Medicine ,Human erythrocytes ,Malaria - Abstract
Malaria remains one of the deadliest diseases on the planet, infecting an estimated 229 million individuals in 2019 with more than 400,000 associated deaths, primarily in young children and pregnant women. 1 Malaria is caused by Plasmodium parasite infection of the liver and blood, with P. falciparum accounting for the vast majority of deaths. 2 The complexity of the Plasmodium life cycle3 coupled with mounting resistance to current treatments4 underscores the need for new treatment options. As a part of this discovery effort, we recently identified a number of active compounds in a high-throughput screen designed to identify molecules that arrest the growth of P. falciparum in co-culture with human erythrocytes. 5 One compound displaying notable activity was compound 1, a 2,6-dibenzylidenecyclohexanone shown in.Fig 1
- Published
- 2021
27. Improvement of Oral Bioavailability of Pyrazolo-Pyridone Inhibitors of the Interaction of DCN1/2 and UBE2M
- Author
-
Bhuvanesh Singh, Yizhe Chen, William Pistel, R. Kiplin Guy, Amy L. Rice, Daniel C. Scott, Jared T. Hammill, Ho Shin Kim, and Brenda A. Schulman
- Subjects
Pyridines ,Administration, Oral ,Pharmacology ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,01 natural sciences ,NEDD8 ,03 medical and health sciences ,Mice ,Structure-Activity Relationship ,Ubiquitin ,Drug Stability ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,030304 developmental biology ,Cell Proliferation ,chemistry.chemical_classification ,0303 health sciences ,Binding Sites ,biology ,Intracellular Signaling Peptides and Proteins ,0104 chemical sciences ,Bioavailability ,010404 medicinal & biomolecular chemistry ,Enzyme ,chemistry ,Cell culture ,Drug Design ,Ubiquitin-Conjugating Enzymes ,biology.protein ,Molecular Medicine ,Pyrazoles ,Neddylation ,Cullin ,Half-Life - Abstract
The cullin-RING ubiquitin ligases (CRLs) are ubiquitin E3 enzymes that play a key role in controlling proteasomal degradation and are activated by neddylation. We previously reported inhibitors that target CRL activation by disrupting the interaction of defective in cullin neddylation 1 (DCN1), a CRL neddylation co-E3, and UBE2M, a neddylation E2. Our first-generation inhibitors possessed poor oral bioavailability and fairly rapid clearance that hindered the study of acute inhibition of DCN-controlled CRL activity in vivo. Herein, we report studies to improve the pharmacokinetic performance of the pyrazolo-pyridone inhibitors. The current best inhibitor, 40, inhibits the interaction of DCN1 and UBE2M, blocks NEDD8 transfer in biochemical assays, thermally stabilizes cellular DCN1, and inhibits anchorage-independent growth in a DCN1 amplified squamous cell carcinoma cell line. Additionally, we demonstrate that a single oral 50 mg/kg dose sustains plasma exposures above the biochemical IC90 for 24 h in mice.
- Published
- 2021
28. Additional file 1 of Selecting an anti-malarial clinical candidate from two potent dihydroisoquinolones
- Author
-
Yizhe Chen, Fangyi Zhu, Hammill, Jared, Holbrook, Gloria, Yang, Lei, Burgess Freeman, White, Karen L., Shackleford, David M., O’Loughlin, Kathleen G., Charman, Susan A., Mirsalis, Jon C., and R. Kiplin Guy
- Abstract
Additional file 1: Table S1. Summary of thermodynamic solubility data for ( +)-SJ733 obtained at 37 °C in phosphate buffer, 0.1 N HCl and simulated intestinal fluids. Table S2. Summary of measured CLint, vitro and scaled CLint, vivo of SJ733 and SJ311 (independent replicate experiments to Table 4). Table S3. Metabolic stability parameters for ( +)-SJ733 in human, dog, rat and mouse cryopreserved hepatocytes. Table S4. Rat pharmacokinetic study of S733 after oral administration. Table S5. Murine plasma pharmacokinetic parameters of S733 and SJ311 after intravenous injection. Table S6. Rat pharmacokinetic parameters of S733 and SJ311 after intravenous administration. Table S7. Dog pharmacokinetic parameters of S733 and SJ311 after intravenous administration.
- Published
- 2021
- Full Text
- View/download PDF
29. Anticancer properties of distinct antimalarial drug classes.
- Author
-
Rob Hooft van Huijsduijnen, R Kiplin Guy, Kelly Chibale, Richard K Haynes, Ingmar Peitz, Gerhard Kelter, Margaret A Phillips, Jonathan L Vennerstrom, Yongyuth Yuthavong, and Timothy N C Wells
- Subjects
Medicine ,Science - Abstract
We have tested five distinct classes of established and experimental antimalarial drugs for their anticancer potential, using a panel of 91 human cancer lines. Three classes of drugs: artemisinins, synthetic peroxides and DHFR (dihydrofolate reductase) inhibitors effected potent inhibition of proliferation with IC50s in the nM- low µM range, whereas a DHODH (dihydroorotate dehydrogenase) and a putative kinase inhibitor displayed no activity. Furthermore, significant synergies were identified with erlotinib, imatinib, cisplatin, dasatinib and vincristine. Cluster analysis of the antimalarials based on their differential inhibition of the various cancer lines clearly segregated the synthetic peroxides OZ277 and OZ439 from the artemisinin cluster that included artesunate, dihydroartemisinin and artemisone, and from the DHFR inhibitors pyrimethamine and P218 (a parasite DHFR inhibitor), emphasizing their shared mode of action. In order to further understand the basis of the selectivity of these compounds against different cancers, microarray-based gene expression data for 85 of the used cell lines were generated. For each compound, distinct sets of genes were identified whose expression significantly correlated with compound sensitivity. Several of the antimalarials tested in this study have well-established and excellent safety profiles with a plasma exposure, when conservatively used in malaria, that is well above the IC50s that we identified in this study. Given their unique mode of action and potential for unique synergies with established anticancer drugs, our results provide a strong basis to further explore the potential application of these compounds in cancer in pre-clinical or and clinical settings.
- Published
- 2013
- Full Text
- View/download PDF
30. Identification of Plasmodium falciparum heat shock 90 inhibitors via molecular docking
- Author
-
Jordan Bach, Jared T. Hammill, Mofolusho O. Falade, R. Kiplin Guy, Scott Eagon, Nikalet Everson, and Amy L. Rice
- Subjects
Clinical Biochemistry ,Plasmodium falciparum ,Binding pocket ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,Fibroblast cell line ,Cell Line ,Antimalarials ,Structure-Activity Relationship ,Parasitic Sensitivity Tests ,Drug Discovery ,Virtual screen ,medicine ,Humans ,HSP90 Heat-Shock Proteins ,Molecular Biology ,Cell Proliferation ,Virtual screening ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Fibroblasts ,biology.organism_classification ,Zinc database ,In vitro ,0104 chemical sciences ,Molecular Docking Simulation ,010404 medicinal & biomolecular chemistry ,Shock (circulatory) ,Molecular Medicine ,medicine.symptom - Abstract
A virtual screen was performed to identify anti-malarial compounds targeting Plasmodium falciparum heat shock 90 protein by applying a series of drug-like and commercial availability filters to compounds in the ZINC database, resulting in a virtual library of more than 13 million candidates. The goal of the virtual screen was to identify novel compounds which could serve as a starting point for the development of antimalarials with a mode of action different from anything currently used in the clinic. The screen targeted the ATP binding pocket of the highly conserved Plasmodium heat shock 90 protein, as this protein is critical to the survival of the parasite and has several significant structural differences from the human homolog. The top twelve compounds from the virtual screen were tested in vitro, with all twelve showing no antiproliferative activity against the human fibroblast cell line and three compounds exhibiting single digit or better micromolar antiproliferative activity against the chloroquine-sensitive P. falciparum 3D7 strain.
- Published
- 2020
31. Synthesis and Structure-Activity Relationship of Dual-Stage Antimalarial Pyrazolo[3,4
- Author
-
Scott, Eagon, Jared T, Hammill, Martina, Sigal, Kevin J, Ahn, Julia E, Tryhorn, Grant, Koch, Briana, Belanger, Cory A, Chaplan, Lauren, Loop, Anna S, Kashtanova, Kenya, Yniguez, Horacio, Lazaro, Steven P, Wilkinson, Amy L, Rice, Mofolusho O, Falade, Rei, Takahashi, Katie, Kim, Ashley, Cheung, Celine, DiBernardo, Joshua J, Kimball, Elizabeth A, Winzeler, Korina, Eribez, Nimisha, Mittal, Francisco-Javier, Gamo, Benigno, Crespo, Alisje, Churchyard, Irene, García-Barbazán, Jake, Baum, Marc O, Anderson, Benoît, Laleu, and R Kiplin, Guy
- Subjects
Models, Molecular ,Antimalarials ,Structure-Activity Relationship ,Dose-Response Relationship, Drug ,Molecular Structure ,Parasitic Sensitivity Tests ,Pyridines ,Plasmodium falciparum ,Humans ,Pyrazoles ,Hep G2 Cells ,Cell Line - Abstract
Malaria remains one of the most deadly infectious diseases, causing hundreds of thousands of deaths each year, primarily in young children and pregnant mothers. Here, we report the discovery and derivatization of a series of pyrazolo[3,4
- Published
- 2020
32. University-pharmacy partnerships for COVID-19
- Author
-
Clark Kebodeaux, Vincent J. Venditto, Patricia R. Freeman, R. Kiplin Guy, and Brooke Hudspeth
- Subjects
2019-20 coronavirus outbreak ,Multidisciplinary ,Coronavirus disease 2019 (COVID-19) ,Universities ,business.industry ,Viral Epidemiology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,MEDLINE ,Emergency Responders ,COVID-19 ,Pharmacy ,medicine.disease ,Virology ,Pneumonia ,Pandemic ,medicine ,Humans ,business ,Coronavirus Infections ,Pandemics - Published
- 2020
33. Selecting an anti-malarial clinical candidate from two potent dihydroisoquinolones
- Author
-
Yizhe Chen, Fangyi Zhu, Jared Hammill, Gloria Holbrook, Lei Yang, Burgess Freeman, Karen L White, David M Shackleford, Kathleen G O’Loughlin, Liang Tang, Susan A Charman, Jon C Mirsalis, and R Kiplin Guy
- Abstract
Background: The ongoing global malaria eradication campaign requires development of potent, safe, and cost-effective drugs lacking cross-resistance with existing chemotherapies. One critical step in drug development is selecting a suitable clinical candidate from late leads. Herein we present the process used to select the clinical candidate SJ733 from two potent dihydroisoquinolone (DHIQ) late leads, SJ733 and SJ311, based on their physicochemical, pharmacokinetic (PK), and toxicity profiles. Methods: The compounds were tested to define their physicochemical properties including kinetic and thermodynamic solubility, partition coefficient, permeability, ionization constant, and binding to plasma proteins. Metabolic stability was assessed in both microsomes and hepatocytes derived from mice, rats, dogs, and humans. Cytochrome P450 inhibition was assessed using recombinant human cytochrome enzymes. The pharmacokinetic profiles of single intravenous or oral doses were investigated in mice, rats, and dogs. Results: Although both compounds displayed similar physicochemical properties, SJ733 was more permeable but metabolically less stable than SJ311 in vitro. Single dose PK studies of SJ733 in mice, rats, and dogs demonstrated appreciable oral bioavailability (60-100%), whereas SJ311 had lower oral bioavailability (mice 23%, rats 40%) and higher renal clearance (10-30 fold higher than SJ733 in rats and dogs), suggesting less favorable exposure in humans. SJ311 also displayed a narrower range of dose-proportional exposure, with plasma exposure flattening at doses above 200 mg/kg. Conclusion: SJ733 was chosen as the candidate based on a more favorable dose proportionality of exposure and stronger expectation of the ability to justify a strong therapeutic index to regulators.
- Published
- 2020
- Full Text
- View/download PDF
34. Antimalarial activity of tetrahydro-β-carbolines targeting the ATP binding pocket of the Plasmodium falciparum heat shock 90 protein
- Author
-
Amy L. Rice, Celine DiBernardo, Jordan Bach, Tyler A Sisley, Scott Eagon, R. Kiplin Guy, Mofolusho O. Falade, Sierra D. Durham, Nikalet Everson, Joshua J. Kimball, Dylan R. Pillai, Michael J. Walls, Jared T. Hammill, and Horacio Lazaro
- Subjects
Clinical Biochemistry ,Plasmodium falciparum ,Binding pocket ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,chemistry.chemical_compound ,Antimalarials ,Structure-Activity Relationship ,Adenosine Triphosphate ,Cell Line, Tumor ,Drug Discovery ,medicine ,Structure–activity relationship ,Potency ,Humans ,HSP90 Heat-Shock Proteins ,Molecular Biology ,Clinical treatment ,Cell Proliferation ,Binding Sites ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,biology.organism_classification ,0104 chemical sciences ,Molecular Docking Simulation ,010404 medicinal & biomolecular chemistry ,Shock (circulatory) ,Toxicity ,Molecular Medicine ,medicine.symptom ,Lead compound ,Carbolines - Abstract
A series of tetrahydro-β-carboline derivatives of a lead compound known to target the heat shock 90 protein of Plasmodium falciparum were synthesized and assayed for both potency against the parasite and toxicity against a human cell line. Using a rationalized structure based design strategy, a new lead compound with a potency two orders of magnitude greater than the original lead compound was found. Additional modeling of this new lead compound suggests multiple avenues to further increase potency against this target, potentially paving the path for a therapeutic with a mode of action different than any current clinical treatment.
- Published
- 2020
35. Rapid repurposing of drugs for COVID-19
- Author
-
Rebecca Ellis Dutch, Frank Romanelli, Robert S. DiPaola, and R. Kiplin Guy
- Subjects
0301 basic medicine ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,Antiviral Agents ,03 medical and health sciences ,Betacoronavirus ,0302 clinical medicine ,Drug Delivery Systems ,Pandemic ,medicine ,Humans ,Pandemics ,Repurposing ,Clinical Trials as Topic ,Multidisciplinary ,biology ,business.industry ,SARS-CoV-2 ,Respiratory disease ,Drug Repositioning ,COVID-19 ,medicine.disease ,biology.organism_classification ,Virology ,Drug repositioning ,Pneumonia ,030104 developmental biology ,030220 oncology & carcinogenesis ,business ,Coronavirus Infections - Abstract
The emergence of a new coronaviral respiratory disease calls for repurposing existing drugs
- Published
- 2020
36. On the mechanism of action of SJ-172550 in inhibiting the interaction of MDM4 and p53.
- Author
-
Michal Bista, David Smithson, Aleksandra Pecak, Gabriella Salinas, Katarzyna Pustelny, Jaeki Min, Artur Pirog, Kristin Finch, Michal Zdzalik, Brett Waddell, Benedykt Wladyka, Sylwia Kedracka-Krok, Michael A Dyer, Grzegorz Dubin, and R Kiplin Guy
- Subjects
Medicine ,Science - Abstract
SJ-172550 (1) was previously discovered in a biochemical high throughput screen for inhibitors of the interaction of MDMX and p53 and characterized as a reversible inhibitor (J. Biol. Chem. 2010; 285:10786). Further study of the biochemical mode of action of 1 has shown that it acts through a complicated mechanism in which the compound forms a covalent but reversible complex with MDMX and locks MDMX into a conformation that is unable to bind p53. The relative stability of this complex is influenced by many factors including the reducing potential of the media, the presence of aggregates, and other factors that influence the conformational stability of the protein. This complex mechanism of action hinders the further development of compound 1 as a selective MDMX inhibitor.
- Published
- 2012
- Full Text
- View/download PDF
37. Establishing a Preclinical Multidisciplinary Board for Brain Tumors
- Author
-
Burgess B. Freeman, Jason Dapper, Hope Elizabeth Terhune, Sabrina Terranova, Michael DeCuypere, Anang A. Shelat, Martine F. Roussel, Nidal Boulos, R. Kiplin Guy, Birgit Nimmervoll, Clinton F. Stewart, Thomas E. Merchant, Amar Gajjar, Karen Wright, Christopher Calabrese, Yogesh T. Patel, Richard J. Gilbertson, Arzu Onar-Thomas, Brandon Bianski, Nimmervoll, Birgit [0000-0002-3324-092X], Gilbertson, Richard [0000-0001-7539-9472], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Oncology ,Ependymoma ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Brain tumor ,Mice, Nude ,Deoxycytidine ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Repurposing ,Chemotherapy ,Brain Neoplasms ,business.industry ,Choroid plexus carcinoma ,medicine.disease ,Gemcitabine ,Drug repositioning ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Neurosurgery ,business ,medicine.drug - Abstract
Purpose: Curing all children with brain tumors will require an understanding of how each subtype responds to conventional treatments and how best to combine existing and novel therapies. It is extremely challenging to acquire this knowledge in the clinic alone, especially among patients with rare tumors. Therefore, we developed a preclinical brain tumor platform to test combinations of conventional and novel therapies in a manner that closely recapitulates clinic trials. Experimental Design: A multidisciplinary team was established to design and conduct neurosurgical, fractionated radiotherapy and chemotherapy studies, alone or in combination, in accurate mouse models of supratentorial ependymoma (SEP) subtypes and choroid plexus carcinoma (CPC). Extensive drug repurposing screens, pharmacokinetic, pharmacodynamic, and efficacy studies were used to triage active compounds for combination preclinical trials with “standard-of-care” surgery and radiotherapy. Results: Mouse models displayed distinct patterns of response to surgery, irradiation, and chemotherapy that varied with tumor subtype. Repurposing screens identified 3-hour infusions of gemcitabine as a relatively nontoxic and efficacious treatment of SEP and CPC. Combination neurosurgery, fractionated irradiation, and gemcitabine proved significantly more effective than surgery and irradiation alone, curing one half of all animals with aggressive forms of SEP. Conclusions: We report a comprehensive preclinical trial platform to assess the therapeutic activity of conventional and novel treatments among rare brain tumor subtypes. It also enables the development of complex, combination treatment regimens that should deliver optimal trial designs for clinical testing. Postirradiation gemcitabine infusion should be tested as new treatments of SEP and CPC. Clin Cancer Res; 24(7); 1654–66. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
38. Oxazole and thiazole analogs of sulindac for cancer prevention
- Author
-
Bini Mathew, R. Kiplin Guy, Michele Connelly, Robert C. Reynolds, and Judith V. Hobrath
- Subjects
Male ,0301 basic medicine ,Spectrometry, Mass, Electrospray Ionization ,NSAIDs ,Colorectal cancer ,Proton Magnetic Resonance Spectroscopy ,Antineoplastic Agents ,Familial adenomatous polyposis ,03 medical and health sciences ,chemistry.chemical_compound ,Sulindac ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,medicine ,Humans ,cancer ,Thiazole ,Oxazoles ,Chromatography, High Pressure Liquid ,Oxazole ,Pharmacology ,heterocycles ,Cancer prevention ,Anti-Inflammatory Agents, Non-Steroidal ,oxazole ,Cancer ,medicine.disease ,digestive system diseases ,3. Good health ,Thiazoles ,030104 developmental biology ,chemistry ,Drug Design ,030220 oncology & carcinogenesis ,Cancer research ,Heterografts ,Molecular Medicine ,Female ,Chromatography, Thin Layer ,Drug Screening Assays, Antitumor ,thiazole ,Research Article ,medicine.drug - Abstract
Aim: Experimental and epidemiological studies and clinical trials suggest that nonsteroidal anti-inflammatory drugs possess antitumor potential. Sulindac, a widely used nonsteroidal anti-inflammatory drug, can prevent adenomatous colorectal polyps and colon cancer, especially in patients with familial adenomatous polyposis. Sulindac sulfide amide (SSA) is an amide-linked sulindac sulfide analog that showed in vivo antitumor activity in a human colon tumor xenograft model. Results/methodology: A new analog series with heterocyclic rings such as oxazole or thiazole at the C-2 position of sulindac was prepared and screened against prostate, colon and breast cancer cell lines to probe the effect of these novel substitutions on the activity of sulindac analogs. Conclusion: In general, replacement of the amide function of SSA analogs had a negative impact on the cell lines tested. A small number of hits incorporating rigid oxazole or thiazole groups in the sulindac scaffold in place of the amide linkage show comparable activity to our lead agent SSA., Graphical abstract
- Published
- 2018
- Full Text
- View/download PDF
39. Amine Containing Analogs of Sulindac for Cancer Prevention
- Author
-
Judith V. Hobrath, Bini Mathew, R. Kiplin Guy, Michele Connelly, and Robert C. Reynolds
- Subjects
0301 basic medicine ,NSAIDs ,Colorectal cancer ,Pharmaceutical Science ,Sulfonamide ,Familial adenomatous polyposis ,03 medical and health sciences ,chemistry.chemical_compound ,Sulindac ,0302 clinical medicine ,In vivo ,Amide ,Drug Discovery ,medicine ,Cancer ,COX-independent ,Pharmacology ,chemistry.chemical_classification ,medicine.disease ,digestive system diseases ,3. Good health ,Anticancer ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Amine gas treating ,Medicinal Chemistry ,Reverse amide ,medicine.drug - Abstract
Background:Sulindac belongs to the chemically diverse family of Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) that effectively prevent adenomatous colorectal polyps and colon cancer, especially in patients with familial adenomatous polyposis. Sulindac sulfide amide (SSA), an amide analog of sulindac sulfide, shows insignificant COX-related activity and toxicity while enhancing anticancer activityin vitroand demonstratingin vivoxenograft activity.Objective:Develop structure-activity relationships in the sulindac amine series and identify analogs with promising anticancer activities.Method:A series of sulindac amine analogs were designed and synthesized and then further modified in a “libraries from libraries” approach to produce amide, sulfonamide and N,N-disubstituted sulindac amine sub-libraries. All analogs were screened against three cancer cell lines (prostate, colon and breast).Results:Several active compounds were identifiedviain vitrocancer cell line screening with the most potent compound (26) in the nanomolar range.Conclusion:Compound26and analogs showing the most potent inhibitory activity may be considered for further design and optimization efforts as anticancer hit scaffolds.
- Published
- 2018
- Full Text
- View/download PDF
40. Exploiting a water network to achieve enthalpy-driven, bromodomain-selective BET inhibitors
- Author
-
Philip M. Potter, Sergio C. Chai, Brett Waddell, Marie Morfouace, Anang A. Shelat, Nagakumar Bharatham, R. Kiplin Guy, William R. Shadrick, Brandon Young, Peter J. Slavish, Richard E. Lee, Jonathan Low, Stefan Knapp, Cynthia Tallant, Vincent A. Boyd, Michele Connelly, Taosheng Chen, and Martine F. Roussel
- Subjects
0301 basic medicine ,BRD4 ,Cell Survival ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Article ,BET inhibitor ,Structure-Activity Relationship ,03 medical and health sciences ,Cell Line, Tumor ,Drug Discovery ,Humans ,Structure–activity relationship ,Molecular Biology ,Dose-Response Relationship, Drug ,Molecular Structure ,Hydrogen bond ,Chemistry ,Organic Chemistry ,Proteins ,Water ,Isothermal titration calorimetry ,Pediatric cancer ,Bromodomain ,030104 developmental biology ,Quinolines ,Thermodynamics ,Molecular Medicine ,Selectivity - Abstract
Within the last decade, the Bromodomain and Extra-Terminal domain family (BET) of proteins have emerged as promising drug targets in diverse clinical indications including oncology, auto-immune disease, heart failure, and male contraception. The BET family consists of four isoforms (BRD2, BRD3, BRD4, and BRDT/BRDT6) which are distinguished by the presence of two tandem bromodomains (BD1 and BD2) that independently recognize acetylated-lysine (KAc) residues and appear to have distinct biological roles. BET BD1 and BD2 bromodomains differ at five positions near the substrate binding pocket: the variation in the ZA channel induces different water networks nearby. We designed a set of congeneric 2- and 3-heteroaryl substituted tetrahydroquinolines (THQ) to differentially engage bound waters in the ZA channel with the goal of achieving bromodomain selectivity. SJ830599 (9) showed modest, but consistent, selectivity for BRD2-BD2. Using isothermal titration calorimetry, we showed that the binding of all THQ analogs in our study to either of the two bromodomains was enthalpy driven. Remarkably, the binding of 9 to BRD2-BD2 was marked by negative entropy and was entirely driven by enthalpy, consistent with significant restriction of conformational flexibility and/or engagement with bound waters. Co-crystallography studies confirmed that 9 did indeed stabilize a water-mediated hydrogen bond network. Finally, we report that 9 retained cytotoxicity against several pediatric cancer cell lines with EC50 values comparable to BET inhibitor (BETi) clinical candidates.
- Published
- 2018
- Full Text
- View/download PDF
41. Ventromorphins: A New Class of Small Molecule Activators of the Canonical BMP Signaling Pathway
- Author
-
David B. Finkelstein, Karen Vrijens, Taosheng Chen, Jose Grenet, Jaeki Min, Wilson K. Clements, R. Kiplin Guy, Anang A. Shelat, Wenwei Lin, Martine F. Roussel, Dana Farmer, and Jamie R. Genthe
- Subjects
0301 basic medicine ,animal structures ,Cellular differentiation ,Smad Proteins ,Biology ,Bone morphogenetic protein ,Biochemistry ,Article ,Cell Line ,Myoblasts ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,Chalcones ,Osteogenesis ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,BMP signaling pathway ,Zebrafish ,Osteoblasts ,Activator (genetics) ,Gene Expression Profiling ,Cell Differentiation ,General Medicine ,biology.organism_classification ,Molecular biology ,BMPR2 ,030104 developmental biology ,Gene Expression Regulation ,Bone Morphogenetic Proteins ,embryonic structures ,Molecular Medicine ,Signal transduction ,C2C12 ,Signal Transduction - Abstract
Here we describe three new small-molecule activators of BMP signaling found by high throughput screening of a library of ~600,000 small molecules. Using a cell-based luciferase assay in the BMP4-responsive human cervical carcinoma clonal cell line, C33A-2D2, we identified three compounds with similar chemotypes that each ventralize zebrafish embryos and stimulate increased expression of the BMP target genes, bmp2b and szl. Because these compounds ventralize zebrafish embryos, we have termed them “ventromorphins.” As expected for BMP pathway activators, they induce the differentiation of C2C12 myoblasts to osteoblasts. Affymetrix RNA analysis confirmed the differentiation results and showed that ventromorphins treatment elicits a response similar to BMP4 treatment. Unlike Isoliquiritigenin (SJ000286237), a flavone that maximally activates the pathway after 24 hours of treatment, all three ventromorphins induced SMAD1/5/8 phosphorylation within 30 minutes of treatment and achieved peak activity within 1 hour, indicating that their responses are consistent with directly activated BMP signaling.
- Published
- 2017
- Full Text
- View/download PDF
42. Performance of a docking/molecular dynamics protocol for virtual screening of nutlin-class inhibitors of Mdmx
- Author
-
Kristin Finch, Jaeki Min, R. Kiplin Guy, Nagakumar Bharatham, Donald Bashford, Michael A. Dyer, and Anand Mayasundari
- Subjects
0301 basic medicine ,MDMX ,Antineoplastic Agents ,Cell Cycle Proteins ,Computational biology ,Molecular Dynamics Simulation ,01 natural sciences ,Molecular Docking Simulation ,Article ,Piperazines ,03 medical and health sciences ,chemistry.chemical_compound ,Computational chemistry ,Proto-Oncogene Proteins ,Materials Chemistry ,Humans ,Physical and Theoretical Chemistry ,Spectroscopy ,Virtual screening ,Binding Sites ,Imidazoles ,Nuclear Proteins ,Nutlin ,AutoDock ,Computer Graphics and Computer-Aided Design ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Improved performance ,030104 developmental biology ,chemistry ,Docking (molecular) ,Drug Screening Assays, Antitumor ,Hydrophobic and Hydrophilic Interactions ,Protein Binding - Abstract
A virtual screening protocol involving docking and molecular dynamics has been tested against the results of fluorescence polarization assays testing the potency of a series of compounds of the nutlin class for inhibition of the interaction between p53 and Mdmx, an interaction identified as a driver of certain cancers. The protocol uses a standard docking method (AutoDock) with a cutoff based on the AutoDock score (ADscore), followed by molecular dynamics simulation with a cutoff based on root-mean-square-deviation (RMSD) from the docked pose. An analysis of the experimental and computational results shows modest performance of ADscore alone, but dramatically improved performance when RMSD is also used.
- Published
- 2017
- Full Text
- View/download PDF
43. Safety, tolerability, pharmacokinetics, and antimalarial efficacy of a novel Plasmodium falciparum ATP4 inhibitor SJ733: a first-in-human and induced blood-stage malaria phase 1a/b trial
- Author
-
Amber M. Smith, Stephan Chalon, Ryan N Heine, Tracy B Stewart, James S. McCarthy, Robbin Christensen, Lidiya Bebrevska, Elizabeth John, Ronald H. Dallas, Fabian Gusovsky, John C. Panetta, Yvonne Van Gessel, Kristen Branum, Branko Mitasev, R. Kiplin Guy, Aditya H. Gaur, Shelley Ost, Burgess B. Freeman, John Woodford, Nehali Patel, Li Tang, Jörg J. Möhrle, Patricia M. Flynn, and Julie Richardson
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Erythrocytes ,030231 tropical medicine ,030106 microbiology ,Plasmodium falciparum ,Gastroenterology ,Heterocyclic Compounds, 4 or More Rings ,03 medical and health sciences ,Antimalarials ,H(+)-K(+)-Exchanging ATPase ,Young Adult ,0302 clinical medicine ,Pharmacokinetics ,Oral administration ,Internal medicine ,medicine ,Humans ,Dosing ,Malaria, Falciparum ,Adverse effect ,Volunteer ,Life Cycle Stages ,biology ,business.industry ,Proton Pump Inhibitors ,Middle Aged ,biology.organism_classification ,Isoquinolines ,Infectious Diseases ,Treatment Outcome ,Tolerability ,Case-Control Studies ,Cohort ,Female ,business - Abstract
Summary Background (+)-SJ000557733 (SJ733) is a novel, orally bioavailable inhibitor of Plasmodium falciparum ATP4. In this first-in-human and induced blood-stage malaria phase 1a/b trial, we investigated the safety, tolerability, pharmacokinetics, and antimalarial activity of SJ733 in humans. Methods The phase 1a was a single-centre, dose-escalation, first-in-human study of SJ733 allowing modifications to dose increments and dose-cohort size on the basis of safety and pharmacokinetic results. The phase 1a took place at St Jude Children's Research Hospital and at the University of Tennessee Clinical Research Center (Memphis, TN, USA). Enrolment in more than one non-consecutive dose cohort was allowed with at least 14 days required between doses. Participants were fasted in seven dose cohorts and fed in one 600 mg dose cohort. Single ascending doses of SJ733 (75, 150, 300, 600, 900, or 1200 mg) were administered to participants, who were followed up for 14 days after SJ733 dosing. Phase 1a primary endpoints were safety, tolerability, and pharmacokinetics of SJ733, and identification of an SJ733 dose to test in the induced blood-stage malaria model. The phase 1b was a single-centre, open-label, volunteer infection study using the induced blood-stage malaria model in which fasted participants were intravenously infected with blood-stage P falciparum and subsequently treated with a single dose of SJ733. Phase 1b took place at Q-Pharm (Herston, QLD, Australia) and was initiated only after phase 1a showed that exposure exceeding the threshold minimum exposure could be safely achieved in humans. Participants were inoculated on day 0 with P falciparum-infected human erythrocytes (around 2800 parasites in the 150 mg dose cohort and around 2300 parasites in the 600 mg dose cohort), and parasitaemia was monitored before malaria inoculation, after inoculation, immediately before SJ733 dosing, and then post-dose. Participants were treated with SJ733 within 24 h of reaching 5000 parasites per mL or at a clinical score higher than 6. Phase 1b primary endpoints were calculation of a parasite reduction ratio (PRR48) and parasite clearance half-life, and safety and tolerability of SJ733 (incidence, severity, and drug-relatedness of adverse events). In both phases of the trial, SJ733 hydrochloride salt was formulated as a powder blend in capsules containing 75 mg or 300 mg for oral administration. Healthy men and women (of non-childbearing potential) aged 18–55 years were eligible for both studies. Both studies are registered with ClinicalTrials.gov ( NCT02661373 for the phase 1a and NCT02867059 for the phase 1b). Findings In the phase 1a, 23 healthy participants were enrolled and received one to three non-consecutive doses of SJ733 between March 14 and Dec 7, 2016. SJ733 was safe and well tolerated at all doses and in fasted and fed conditions. 119 adverse events were recorded: 54 (45%) were unrelated, 63 (53%) unlikely to be related, and two (2%) possibly related to SJ733. In the phase 1b, 17 malaria-naive, healthy participants were enrolled. Seven participants in the 150 mg dose cohort were inoculated and dosed with SJ733. Eight participants in the 600 mg dose cohort were inoculated, but two participants could not be dosed with SJ733. Two additional participants were subsequently inoculated and dosed with SJ733. SJ733 exposure increased proportional to the dose through to the 600 mg dose, then was saturable at higher doses. Fasted participants receiving 600 mg exceeded the target area under the concentration curve extrapolated to infinity (AUC0–∞) of 13 000 μg × h/L (median AUC0–∞ 24 283 [IQR 16 135–31 311] μg × h/L, median terminal half-life 17·4 h [IQR 16·1–24·0], and median timepoint at which peak plasma concentration is reached 1·0 h [0·6–1·3]), and this dose was tested in the phase 1b. All 15 participants dosed with SJ733 had at least one adverse event. Of the 172 adverse events recorded, 128 (74%) were mild. The only adverse event attributed to SJ733 was mild bilateral foot paraesthesia that lasted 3·75 h and resolved spontaneously. The most common adverse events were related to malaria. Based on parasite clearance half-life, the derived log10 PRR48 and corresponding parasite clearance half-lives were 2·2 (95% CI 2·0–2·5) and 6·47 h (95% CI 5·88–7·18) for 150 mg, and 4·1 (3·7–4·4) and 3·56 h (3·29–3·88) for 600 mg. Interpretation The favourable pharmacokinetic, tolerability, and safety profile of SJ733, and rapid antiparasitic effect support its development as a fast-acting component of combination antimalarial therapy. Funding Global Health Innovative Technology Fund, Medicines for Malaria Venture, and the American Lebanese Syrian Associated Charities.
- Published
- 2019
44. Evaluation of artemisinins for the treatment of acute myeloid leukemia
- Author
-
Anang A. Shelat, R. Kiplin Guy, Shuiying Hu, Yiping Fan, Christina D. Drenberg, Shelley Orwick, Jassada Buaboonnam, Lie Li, Sharyn D. Baker, and Jeffrey E. Rubnitz
- Subjects
0301 basic medicine ,Cancer Research ,Artemisinins ,Cell Survival ,medicine.medical_treatment ,Artesunate ,Dihydroartemisinin ,Apoptosis ,Mice, Inbred Strains ,Pharmacology ,Toxicology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Animals ,Humans ,Medicine ,Pharmacology (medical) ,Artemisinin ,Chemotherapy ,business.industry ,Cytarabine ,Myeloid leukemia ,Xenograft Model Antitumor Assays ,High-Throughput Screening Assays ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Lysosomes ,Reactive Oxygen Species ,business ,medicine.drug - Abstract
Investigate antileukemic activity of artemisinins, artesunate (ART), and dihydroartemisinin (DHA), in combination with cytarabine, a key component of acute myeloid leukemia (AML) chemotherapy using in vitro and in vivo models. Using ten human AML cell lines, we conducted a high-throughput screen to identify antimalarial agents with antileukemic activity. We evaluated effects of ART and DHA on cell viability, cytotoxicity, apoptosis, lysosomal integrity, and combination effects with cytarabine in cell lines and primary patient blasts. In vivo pharmacokinetic studies and efficacy of single-agent ART or combination with cytarabine were evaluated in three xenograft models. ART and DHA had the most potent activity in a panel of AML cell lines, with selectivity toward samples harboring MLL rearrangements and FLT3-ITD mutations. Combination of ART or DHA was synergistic with cytarabine. Single-dose ART (120 mg/kg) produced human equivalent exposures, but multiple dose daily administration required for in vivo efficacy was not tolerated. Combination treatment produced initial regression, but did not prolong survival in vivo. The pharmacology of artemisinins is problematic and should be considered in designing AML treatment strategies with currently available agents. Artemisinins with improved pharmacokinetic properties may offer therapeutic benefit in combination with conventional therapeutic strategies in AML.
- Published
- 2016
- Full Text
- View/download PDF
45. Optimization of a Novel Series of Ataxia-Telangiectasia Mutated Kinase Inhibitors as Potential Radiosensitizing Agents
- Author
-
Clementine Feau, Brandon Young, Yizhe Chen, Kexiao Guo, Gloria Holbrook, Jaeki Min, Michele Connelly, R. Kiplin Guy, Andrew Lemoff, Fangyi Zhu, Praveen Kumar Suryadevara, and Michael B. Kastan
- Subjects
0301 basic medicine ,Radiation-Sensitizing Agents ,Ataxia Telangiectasia Mutated Proteins ,In Vitro Techniques ,Pharmacology ,Substrate Specificity ,Colony-Forming Units Assay ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Delivery Systems ,Pharmacokinetics ,Drug Discovery ,Quinazoline ,Animals ,Humans ,Structure–activity relationship ,Potency ,Kinase ,Chemistry ,Mice, Inbred C57BL ,030104 developmental biology ,Biochemistry ,Drug Design ,MCF-7 Cells ,Microsomes, Liver ,Quinazolines ,Molecular Medicine ,Female ,Radiosensitizing Agent ,Selectivity - Abstract
We previously reported a novel inhibitor of the ataxia-telangiectasia mutated (ATM) kinase, which is a target for novel radiosensitizing drugs. While our initial lead, compound 4, was relatively potent and nontoxic, it exhibited poor stability to oxidative metabolism and relatively poor selectivity against other kinases. The current study focused on balancing potency and selectivity with metabolic stability through structural modification to the metabolized site on the quinazoline core. We performed extensive structure-activity and structure-property relationship studies on this quinazoline ATM kinase inhibitor in order to identify structural variants with enhanced selectivity and metabolic stability. We show that, while the C-7-methoxy group is essential for potency, replacing the C-6-methoxy group considerably improves metabolic stability without affecting potency. Promising analogues 20, 27g, and 27n were selected based on in vitro pharmacology and evaluated in murine pharmacokinetic and tolerability studies. Compound 27g possessed significantly improve pharmacokinetics relative to that of 4. Compound 27g was also significantly more selective against other kinases than 4. Therefore, 27g is a good candidate for further development as a potential radiosensitizer.
- Published
- 2016
- Full Text
- View/download PDF
46. Seeking the Elusive Long-Acting Ozonide: Discovery of Artefenomel (OZ439)
- Author
-
Ho Shin Kim, R. Kiplin Guy, and Jared T. Hammill
- Subjects
Plasmodium ,Adamantane ,Artefenomel ,Pharmacology ,01 natural sciences ,Antimalarials ,chemistry.chemical_compound ,parasitic diseases ,Drug Discovery ,medicine ,Animals ,Humans ,Ozonide ,Cost of goods ,Dosing ,Artemisinin ,Arterolane ,010405 organic chemistry ,Drug discovery ,Malaria ,Peroxides ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Long acting ,chemistry ,Molecular Medicine ,medicine.drug - Abstract
The majority of frontline therapies for the treatment of malaria are combination drugs containing artemisinin (or its semisynthetic analogs), known as artemisinin combination therapies (ACTs). While generally efficacious, ACTs and the first generation fully synthetic ozonide, arterolane (OZ277, 1), suffer from rapid clearance requiring 3-day dosing regimens. Extensive structure-activity studies led to the discovery of a second-generation ozonide, artefenomel (OZ439, 2), which has overcome this limitation, maintaining the rapid onset of action and potent activity of the artemisinin derivatives while exhibiting greatly improved pharmacokinetics, low projected cost of goods, prophylactic activity, and the potential for a single dose cure.
- Published
- 2017
- Full Text
- View/download PDF
47. Evaluation of 1,1-cyclopropylidene as a thioether isostere in the 4-thio-thienopyrimidine (TTP) series of antimalarials
- Author
-
Michael C. Tran, R. Kiplin Guy, Christopher W. Davis, Jared T. Hammill, Paul R. Rablen, Robert D. Barrows, Mofolusho O. Falade, Thomas J. Emge, Spencer Knapp, and Amy L. Rice
- Subjects
Cyclopropanes ,Models, Molecular ,Steric effects ,Ketone ,Isostere ,Cyclopropanation ,Stereochemistry ,Plasmodium falciparum ,Clinical Biochemistry ,Pharmaceutical Science ,Thio ,Protonation ,Crystallography, X-Ray ,01 natural sciences ,Biochemistry ,Antimalarials ,Structure-Activity Relationship ,chemistry.chemical_compound ,Parasitic Sensitivity Tests ,Thioether ,Drug Discovery ,Molecular Biology ,Cells, Cultured ,Density Functional Theory ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Organic Chemistry ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Pyrimidines ,chemistry ,Intramolecular force ,Molecular Medicine - Abstract
The 4-(heteroarylthio)thieno[2,3-d]pyrimidine (TTP) series of antimalarials, represented by 1 and 17, potently inhibit proliferation of the 3D7 strain of P. falciparum (EC50 70–100 nM), but suffer from oxidative metabolism. The 1,1-cyclopropylidene isosteres 6 and 16 were designed to obviate this drawback. They were prepared by a short route that features a combined Peterson methylenation / cyclopropanation transformation of, e. g., ketone 7. Isosteres 6 and 16 possess significantly attenuated antimalarial potency relative to parents 1 and 17. This outcome can be rationalized based on the increased out-of-plane steric demands of the latter two. In support of this hypothesis, the relatively flat ketone 7 retains some of the potency of 1, even though it appears to be a comparatively inferior mimic with respect to electronics and bond lengths and angles. We also demonstrate crystallographically and computationally an apparent increase in the strength of the intramolecular sulfur hole interaction of 1 upon protonation.
- Published
- 2020
- Full Text
- View/download PDF
48. Phenotypic Screens Reveal Posaconazole as a Rapidly Acting Amebicidal Combination Partner for Treatment of Primary Amoebic Meningoencephalitis
- Author
-
Christopher A. Rice, Dennis E. Kyle, Beatrice L. Colon, and R. Kiplin Guy
- Subjects
0301 basic medicine ,Drug ,Posaconazole ,Time Factors ,Phenotypic screening ,media_common.quotation_subject ,Antiprotozoal Agents ,Central Nervous System Protozoal Infections ,Pharmacology ,Azithromycin ,03 medical and health sciences ,Major Articles and Brief Reports ,Inhibitory Concentration 50 ,Mice ,0302 clinical medicine ,Amphotericin B ,parasitic diseases ,Drug Discovery ,medicine ,Immunology and Allergy ,Animals ,Humans ,030212 general & internal medicine ,Fluconazole ,Naegleria fowleri ,media_common ,Miltefosine ,biology ,business.industry ,United States Food and Drug Administration ,Meningoencephalitis ,Drug Synergism ,Triazoles ,biology.organism_classification ,medicine.disease ,United States ,Disease Models, Animal ,Drug Combinations ,030104 developmental biology ,Infectious Diseases ,Phenotype ,Female ,business ,medicine.drug - Abstract
Naegleria fowleri is the causative agent of primary amoebic meningoencephalitis (PAM), which is fatal in >97% of cases. In this study, we aimed to identify new, rapidly acting drugs to increase survival rates. We conducted phenotypic screens of libraries of Food and Drug Administration-approved compounds and the Medicines for Malaria Venture Pathogen Box and validated 14 hits (defined as a 50% inhibitory concentration of
- Published
- 2018
49. Identification of Toll-like receptor signaling inhibitors based on selective activation of hierarchically acting signaling proteins
- Author
-
Hans Häcker, Julie A. Pollock, Wenwei Lin, Taosheng Chen, Vanessa Redecke, Yizhe Chen, R. Kiplin Guy, Sirish K. Ippagunta, Naina Sharma, Kazuki Tawaratsumida, Jaeki Min, Anthony A. High, and John A. Katzenellenbogen
- Subjects
0301 basic medicine ,TIRAP ,Phenotypic screening ,Quantitative proteomics ,Drug Evaluation, Preclinical ,Biochemistry ,Article ,Small Molecule Libraries ,Inhibitory Concentration 50 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Piperidines ,Animals ,Humans ,Protein Interaction Maps ,Receptor ,Molecular Biology ,Cell Proliferation ,Toll-like receptor ,Dose-Response Relationship, Drug ,biology ,Toll-Like Receptors ,HEK 293 cells ,Signal transducing adaptor protein ,Cell Biology ,Ubiquitin ligase ,Cell biology ,HEK293 Cells ,RAW 264.7 Cells ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Pyrazoles ,Protein Binding ,Signal Transduction - Abstract
Toll-like receptors (TLRs) recognize various pathogen- and host tissue-derived molecules and initiate inflammatory immune responses. Exaggerated or prolonged TLR activation, however, can promote etiologically diverse diseases, such as bacterial sepsis, metabolic and autoimmune diseases, or stroke. Despite the apparent medical need, no small molecule drugs against TLR-pathways are clinically available. This may be because of the complex signaling mechanisms of TLRs, which are governed by a series of protein interactions initiated by Toll-interleukine-1 receptor (TIR)-domains found in TLRs and the cytoplasmic adapter proteins TIRAP and MyD88. Activation/oligomerization of TLRs and consecutive oligomerization of MyD88 and/or TIRAP leads to the recruitment of IRAK family members and the ubiquitin ligase TRAF6. Here, we developed a phenotypic drug screening system based on inducible dimerization of TIRAP, MyD88 and TRAF6, that ranked hits according to the position of action in the hierarchy of the TLR signaling cascade. From a bioactive compound library, we identified methyl-piperidino-pyrazole (MPP) as a TLR-specific inhibitor. Structure-activity relationship analysis, quantitative proteomics, protein interaction assays, and cellular thermal shift assays strongly suggest that MPP targets the TIR domain of MyD88. Chemical evolution of the original MPP-scaffold generated compounds with selectivity for distinct TLRs that interfered with specific TIR interactions. Administration of a MPP-analog to mice protected against LPS-mediated TNFα release. These results validate this phenotypic screening approach and implicate the MPP scaffold as starting point for anti-inflammatory drug development.
- Published
- 2018
- Full Text
- View/download PDF
50. Cysteine modifiers suggest an allosteric inhibitory site on the CAL PDZ domain
- Author
-
Alexandre A. Pletnev, Maria Pellegrini, David C. Smithson, Sahar Al-Ayyoubi, R. Kiplin Guy, Andrew V. Grassetti, Patrick R. Cushing, Yu Zhao, Scott A. Gerber, and Dean R. Madden
- Subjects
0301 basic medicine ,Scaffold protein ,Allosteric regulation ,PDZ domain ,protein-protein interactions ,Biophysics ,Biochemistry ,Protein–protein interaction ,03 medical and health sciences ,NMR spectroscopy ,high-throughput screen ,Molecular Biology ,Research Articles ,X-ray crystallography ,biology ,Chemistry ,covalent allostery ,Cell Biology ,Transmembrane protein ,Cell biology ,A-site ,030104 developmental biology ,DLG1 ,biology.protein ,Research Article ,Cysteine - Abstract
Protein–protein interactions have become attractive targets for both experimental and therapeutic interventions. The PSD-95/Dlg1/ZO-1 (PDZ) domain is found in a large family of eukaryotic scaffold proteins that plays important roles in intracellular trafficking and localization of many target proteins. Here, we seek inhibitors of the PDZ protein that facilitates post-endocytic degradation of the cystic fibrosis (CF) transmembrane conductance regulator (CFTR): the CFTR-associated ligand (CAL). We develop and validate biochemical screens and identify methyl-3,4-dephostatin (MD) and its analog ethyl-3,4-dephostatin (ED) as CAL PDZ inhibitors. Depending on conditions, MD can bind either covalently or non-covalently. Crystallographic and NMR data confirm that MD attacks a pocket at a site distinct from the canonical peptide-binding groove, and suggests an allosteric connection between target residue Cys319 and the conserved Leu291 in the GLGI motif. MD and ED thus appear to represent the first examples of small-molecule allosteric regulation of PDZ:peptide affinity. Their mechanism of action may exploit the known conformational plasticity of the PDZ domains and suggests that allosteric modulation may represent a strategy for targeting of this family of protein–protein binding modules.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.