76 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. 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
10. 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
11. 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
12. 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
13. 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
14. 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
15. 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
16. 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
17. 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
18. 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
19. 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
20. Discovery of novel, orally bioavailable, antileishmanial compounds using phenotypic screening
- Author
-
Yizhe Chen, W. Armand Guiguemde, Diana Ortiz, Fangyi Zhu, Kayla Stalheim, Anang A. Shelat, Michele Connelly, Lei Yang, David C. Smithson, Jaeki Min, Alex G. Johnson, Angela K. Carrillo, Carolyn Elya, R. Kiplin Guy, Scott M. Landfear, and Jared T. Hammill
- Subjects
0301 basic medicine ,Life Cycles ,Chemistry, Pharmaceutical ,Leishmania mexicana ,Drug Evaluation, Preclinical ,Administration, Oral ,Drug resistance ,Protozoology ,Pharmacology ,Mice ,White Blood Cells ,Animal Cells ,Zoonoses ,Drug Discovery ,Medicine and Health Sciences ,Leishmaniasis ,Mice, Inbred BALB C ,Drug discovery ,lcsh:Public aspects of medicine ,Animal Models ,3. Good health ,Phenotype ,Infectious Diseases ,Experimental Organism Systems ,Female ,Protozoan Life Cycles ,Cellular Types ,Research Article ,Amastigotes ,Neglected Tropical Diseases ,medicine.drug ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,Immune Cells ,Phenotypic screening ,Immunology ,030106 microbiology ,Antiprotozoal Agents ,Leishmaniasis, Cutaneous ,Mouse Models ,Library Screening ,Biology ,Research and Analysis Methods ,Microbiology ,Cell Line ,03 medical and health sciences ,Model Organisms ,Cutaneous leishmaniasis ,In vivo ,Parasitic Diseases ,medicine ,Animals ,Humans ,Pharmacokinetics ,Molecular Biology Techniques ,Amastigote ,Molecular Biology ,Molecular Biology Assays and Analysis Techniques ,Miltefosine ,Blood Cells ,Protozoan Infections ,Macrophages ,Promastigotes ,Public Health, Environmental and Occupational Health ,Biology and Life Sciences ,lcsh:RA1-1270 ,Cell Biology ,Tropical Diseases ,medicine.disease ,biology.organism_classification ,030104 developmental biology ,Developmental Biology - 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., Author summary Leishmaniasis, caused by the protozoa of the Leishmania species, represents a spectrum of diseases that afflicts roughly 12 million individuals worldwide. Current drug therapies for this parasitic disease are suboptimal because they are toxic, expensive, difficult to administer, and subject to drug resistance. In order to identify new and improved drug candidates, we screened a large library of small molecules for compounds that inhibit parasitic growth inside mammalian host macrophages, and have low toxicity toward the macrophages. We discovered two compounds that significantly impaired disease progression when administered orally in an animal model of cutaneous leishmaniasis. The promising pharmacokinetic and in vivo efficacy profile of the compounds make them attractive starting points for pharmaceutical development.
- Published
- 2017
21. Open Source Drug Discovery with the Malaria Box Compound Collection for Neglected Diseases and Beyond
- Author
-
Christopher D. Huston, Amornrat Naranuntarat Jensen, Louis Maes, Jordi Mestres, Nao Aki Watanabe, Michael Adsetts Edberg Hansen, Roberto Adelfio, Simon Townson, Didier Leroy, Kate Weatherby, Thomas Spangenberg, Manuela Carrasquilla, Kailash P. Patra, Robert E. Sinden, José Brea, Abhai K. Tripathi, W. Armand Guiguemde, Alan Y. Du, Melanie Wree, Katherine T. Andrews, Kirsten K. Hanson, Tyler B. Hughes, Sundari Suresh, Adele M. Lehane, Sangeeta N. Bhatia, Edward J. Wojcik, Andrew Hemphill, Francielly Morais Rodrigues da Costa, Worathad Chindaudomsate, Joseph M. Vinetz, Ben Gold, Sunyoung Kim, Edgar Vigil, Nuha R. Mansour, Mohamed Abdo Rizk, Patrick Valere Tsouh Fokou, Audrey Burton, Laran T. Jensen, David A. Fidock, Aishah Alsibaee, Filipe Silva Villela, Yesmalie Alemán Resto, Rajarshi Guha, Conor R. Caffrey, José A. Fernández Robledo, Thomas J. Ketas, Luke Mercer, Rob Hooft van Huijsduijnen, Maria Jose Lafuente, Wesley C. Van Voorhis, Lauve R. Y. Tchokouaha, Dee A. Carter, Anjo Theron, Benoît Laleu, Kiaran Kirk, Maurice A. Itoe, Robert P. St.Onge, Celia Quevedo, Andrea Ruecker, Paul Henri Amvam Zollo, Francisco-Javier Gamo, Nathan Lee, Alvine Ngoutane Mfopa, Paul Horrocks, Ikuo Igarashi, Nil Gural, Todd R. Golub, Gordana Panic, Jeremy N. Burrows, Phat Voong Vinh, Annette Kaiser, Fabrice Fekam Boyom, Pietro Alano, Anupam Pradhan, Sandra Duffy, Raj N. Misra, Vidya Prasanna Kumar, Aintzane Alday, Timothy N. C. Wells, María Isabel Loza, Sébastien Kicka, William J. Sullivan, Gregory M. Goldgof, Yo Suzuki, Yolanda Corbett, Sally-Ann Poulsen, Vida Ahyong, George Papadatos, Sujeevi Nawaratna, Rafaela Salgado Ferreira, Takaaki Horii, Imran Ullah, Nathalie Narraidoo, Natalie G. Robinett, Simon V. Avery, Grazia Camarda, Iset Medina Vera, Michael T. Ferdig, Fengwu Li, David Plouffe, Joseph L. DeRisi, Jasmeet Samra, Andreas Spitzmüller, Liqiong Liu, Christopher A. Rice, Thierry Soldati, Serge Maximilian Stamm, Suzanne Gokool, Beatrice L. Colon, Shimaa Abd El-Salam El-Sayed, Mark Baker, Kenneth O. Udenze, Na Le Dang, Katrin Ingram-Sieber, Dennis A. Smith, Rays H. Y. Jiang, Marian P. Brennan, Ani Galstian, Paul Willis, Dennis E. Kyle, Ainhoa Alzualde, Sarah Prats, Sheena McGowan, Vicky M. Avery, Jennifer Keiser, John P. Moore, Dalu Mancama, Gregory J. Crowther, Noemi Cowan, Maria B. Cassera, Valentin Trofimov, David Thomas, David J. Sullivan, Diana Ortiz, Nada Abla, S. Joshua Swamidass, Benjamin Blasco, Hoan Vu, Francesco Silvestrini, Anthony J. Chubb, Pamela M. White, Scott M. Landfear, Isabelle Florent, John H. Adams, Ronald J. Quinn, Andrew F. Wilks, Sandra March, Leonardo Lucantoni, Stephen Baker, Tana Bowling, Joachim Müller, Arantza Muriana, Lauren E. Boucher, Ajit Jadhav, Sukjun Lee, Elizabeth A. Winzeler, Choukri Ben Mamoun, Ulrich Schlecht, Daisy D. Colón-López, Marjorie Schmitt, Myles H. Akabas, Isabelle S Lucet, Stephen N. Hewitt, Naoaki Yokoyama, Carl Nathan, Bakela Nare, Cindy Vallières, Lotfi Bounaadja, Kayode K. Ojo, Wesley Wu, Ken Chih-Chien Cheng, Kathryn F. Tonissen, Michael J. Delves, Brian M. Suzuki, Aristea Lubar, Quentin D. Bickle, Stephan Meister, Silvia Parapini, Manuel Llinás, Ngoc Minh Pham, Seunghyun Moon, R. Kiplin Guy, Donatella Taramelli, Lawrence Ayong, Sarah D'Alessandro, Jürgen Bosch, David Little, Istituto Superiore di Sanita [Rome], Zentrum für Infektiologie [Heidelberg, Germany], Universität Heidelberg [Heidelberg]-Heidelberg University Hospital [Heidelberg], University of Nottingham, UK (UON), Eskitis Institute for Drug Discovery, Griffith University [Brisbane], Institut Pasteur Korea - Institut Pasteur de Corée, Réseau International des Instituts Pasteur (RIIP), Oxford University Clinical Research Unit [Ho Chi Minh City] (OUCRU), Laboratoire de génétique moléculaire et cellulaire, Institut National de la Recherche Agronomique (INRA), Agence Française de Sécurité Sanitaire des Aliments (AFSSA), Faculté des Sciences - Yaoundé I, Université de Yaoundé I, Skaggs School of Pharmacy and Pharmaceutical Sciences [San Diego], University of California [San Diego] (UC San Diego), University of California-University of California, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano [Milano] (UNIMI), Imperial College London, Eck Institute for Global Health, University of Notre Dame [Indiana] (UND), Columbia University Medical Center (CUMC), Columbia University [New York], Interdisciplinary Nanoscience Centre (iNANO), Institute of Parasitology, University of Bern, MS Project - Initiation (MSPRI), Apollo SSC Genève, Keele University, Swiss Tropical and Public Health Institute [Basel], University of Regina, School of Engineering and Science, University of the West of Scotland (UWS), Research School of Biology [Canberra, Australie], Australian National University (ANU), University of Pennsylvania [Philadelphia], Laboratory of Microbiology, Parasitology and Hygiene [Antwerpen] (LMPH), University of Antwerp (UA), Department of Biochemistry and Molecular Biology, Mayo Clinic, Institute for Wine Biotechnology [University of Stellenbosch - Afrique du Sud], Stellenbosch University, Oregon Health and Science University [Portland] (OHSU), Biozentrum, Swiss Institute of Bioinformatics [Lausanne] (SIB), Université de Lausanne (UNIL)-Université de Lausanne (UNIL), Laboratoire d'innovation moléculaire et applications (LIMA), Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Advanced Materials Research Laboratories, Department of Chemistry and Center for Optical ((COMSET), Clemson University, Université de Genève (UNIGE), Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, University of California [Santa Cruz] (UCSC), Department of sanità pubblica-microbiologia-virologia, University of California, Département Réseaux, Information, Multimédia (RIM-ENSMSE), École des Mines de Saint-Étienne (Mines Saint-Étienne MSE), Institut Mines-Télécom [Paris] (IMT)-Institut Mines-Télécom [Paris] (IMT)-Centre G2I, UC San Diego School of Medicine, Medicines for Malaria Venture [Geneva] (MMV), Princeton University, Institute for Medical Engineering and Science, Harvard University--MIT Division of Health Sciences and Technology, Bhatia, Sangeeta N, Bill & Melinda Gates Foundation, Biochemistry, Center for Drug Discovery, Kicka, Sébastien, Soldati, Thierry, Trofimov, Valentin, and Abla, Nada
- Subjects
0301 basic medicine ,Plasmodium ,[SDV]Life Sciences [q-bio] ,Medizin ,Drug Evaluation, Preclinical ,Datasets as Topic ,infected erythrocytes ,stage plasmodium-falciparum ,Q1 ,Gametocytes ,Toxicology ,Pathology and Laboratory Medicine ,Drug Metabolism ,1108 Medical Microbiology ,Animal Cells ,inhibitors ,Drug Discovery ,Medicine and Health Sciences ,Biology (General) ,Genetics ,Protozoans ,biology ,630 Agriculture ,Drug discovery ,transmission ,Malarial Parasites ,Neglected Diseases ,3. Good health ,Chemistry ,1107 Immunology ,ddc:540 ,Physical Sciences ,590 Animals (Zoology) ,Identification (biology) ,Cellular Types ,Medicaments ,0605 Microbiology ,Research Article ,Drug Research and Development ,QH301-705.5 ,Immunology ,Computational biology ,in-vitro ,Microbiology ,target ,Small Molecule Libraries ,03 medical and health sciences ,Antimalarials ,blood ,Virology ,Parasite Groups ,Gametocyte ,medicine ,Parasitic Diseases ,[CHIM]Chemical Sciences ,Humans ,Pharmacokinetics ,Molecular Biology ,Biology ,theileria parasites ,Pharmacology ,Toxicity ,QH ,Malària -- Medicaments ,Organisms ,Biology and Life Sciences ,Plasmodium falciparum ,Cell Biology ,RC581-607 ,biology.organism_classification ,medicine.disease ,Tropical Diseases ,Parasitic Protozoans ,babesia ,Malaria ,030104 developmental biology ,Germ Cells ,Vector (epidemiology) ,identification ,570 Life sciences ,Parasitology ,Human medicine ,Medicinal Chemistry ,Immunologic diseases. Allergy ,Apicomplexa - Abstract
A major cause of the paucity of new starting points for drug discovery is the lack of interaction between academia and industry. Much of the global resource in biology is present in universities, whereas the focus of medicinal chemistry is still largely within industry. Open source drug discovery, with sharing of information, is clearly a first step towards overcoming this gap. But the interface could especially be bridged through a scale-up of open sharing of physical compounds, which would accelerate the finding of new starting points for drug discovery. The Medicines for Malaria Venture Malaria Box is a collection of over 400 compounds representing families of structures identified in phenotypic screens of pharmaceutical and academic libraries against the Plasmodium falciparum malaria parasite. The set has now been distributed to almost 200 research groups globally in the last two years, with the only stipulation that information from the screens is deposited in the public domain. This paper reports for the first time on 236 screens that have been carried out against the Malaria Box and compares these results with 55 assays that were previously published, in a format that allows a meta-analysis of the combined dataset. The combined biochemical and cellular assays presented here suggest mechanisms of action for 135 (34%) of the compounds active in killing multiple life-cycle stages of the malaria parasite, including asexual blood, liver, gametocyte, gametes and insect ookinete stages. In addition, many compounds demonstrated activity against other pathogens, showing hits in assays with 16 protozoa, 7 helminths, 9 bacterial and mycobacterial species, the dengue fever mosquito vector, and the NCI60 human cancer cell line panel of 60 human tumor cell lines. Toxicological, pharmacokinetic and metabolic properties were collected on all the compounds, assisting in the selection of the most promising candidates for murine proof-of-concept experiments and medicinal chemistry programs. The data for all of these assays are presented and analyzed to show how outstanding leads for many indications can be selected. These results reveal the immense potential for translating the dispersed expertise in biological assays involving human pathogens into drug discovery starting points, by providing open access to new families of molecules, and emphasize how a small additional investment made to help acquire and distribute compounds, and sharing the data, can catalyze drug discovery for dozens of different indications. Another lesson is that when multiple screens from different groups are run on the same library, results can be integrated quickly to select the most valuable starting points for subsequent medicinal chemistry efforts., Author Summary Malaria leads to the loss of over 440,000 lives annually; accelerating research to discover new candidate drugs is a priority. Medicines for Malaria Venture (MMV) has distilled over 25,000 compounds that kill malaria parasites in vitro into a group of 400 representative compounds, called the "Malaria Box". These Malaria Box sets were distributed free-of-charge to research laboratories in 30 different countries that work on a wide variety of pathogens. Fifty-five groups compiled >290 assay results for this paper describing the many activities of the Malaria Box compounds. The collective results suggest a potential mechanism of action for over 130 compounds against malaria and illuminate the most promising compounds for further malaria drug development research. Excitingly some of these compounds also showed outstanding activity against other disease agents including fungi, bacteria, other single-cellular parasites, worms, and even human cancer cells. The results have ignited over 30 drug development programs for a variety of diseases. This open access effort was so successful that MMV has begun to distribute another set of compounds with initial activity against a wider range of infectious agents that are of public health concern, called the Pathogen Box, available now to scientific labs all over the world (www.PathogenBox.org).
- Published
- 2016
- Full Text
- View/download PDF
22. Monitoring ligand-induced protein ordering in drug discovery
- Author
-
Jaeki Min, Kristin Finch, Richard W. Kriwacki, Michael A. Dyer, David Ban, Lie Min, R. Kiplin Guy, Lyra Griffiths, Christy R. Grace, Nagakumar Bharatham, Anand Mayasundari, and Donald Bashford
- Subjects
0301 basic medicine ,Models, Molecular ,MDMX ,Magnetic Resonance Spectroscopy ,Stereochemistry ,Protein Conformation ,Plasma protein binding ,010402 general chemistry ,01 natural sciences ,Article ,Piperazines ,03 medical and health sciences ,Structure-Activity Relationship ,Protein structure ,Structural Biology ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Humans ,Molecular Biology ,Drug discovery ,Chemistry ,Imidazoles ,Proto-Oncogene Proteins c-mdm2 ,Ligand (biochemistry) ,Small molecule ,0104 chemical sciences ,030104 developmental biology ,Protein Multimerization ,Tumor Suppressor Protein p53 ,Heteronuclear single quantum coherence spectroscopy ,Protein Binding - Abstract
While the gene for p53 is mutated in many human cancers causing loss of function, many others maintain a wild-type gene but exhibit reduced p53 tumor suppressor activity through overexpression of the negative regulators, Mdm2 and/or MdmX. For the latter mechanism of loss of function, the activity of endogenous p53 can be restored through inhibition of Mdm2 or MdmX with small molecules. We previously reported a series of compounds based upon the Nutlin-3 chemical scaffold that bind to both MdmX and Mdm2 [Vara, B. A. et al. (2014) Organocatalytic, diastereo- and enantioselective synthesis of nonsymmetric cis-stilbene diamines: A platform for the preparation of single-enantiomer cis-imidazolines for protein-protein inhibition. J. Org. Chem. 79, 6913-6938]. Here we present the first solution structures based on data from NMR spectroscopy for MdmX in complex with four of these compounds and compare them with the MdmX:p53 complex. A p53-derived peptide binds with high affinity (Kd value of 150nM) and causes the formation of an extensive network of hydrogen bonds within MdmX; this constitutes the induction of order within MdmX through ligand binding. In contrast, the compounds bind more weakly (Kd values from 600nM to 12μM) and induce an incomplete hydrogen bond network within MdmX. Despite relatively weak binding, the four compounds activated p53 and induced p21(Cip1) expression in retinoblastoma cell lines that overexpress MdmX, suggesting that they specifically target MdmX and/or Mdm2. Our results document structure-activity relationships for lead-like small molecules targeting MdmX and suggest a strategy for their further optimization in the future by using NMR spectroscopy to monitor small-molecule-induced protein order as manifested through hydrogen bond formation.
- Published
- 2016
23. Screening and Development of New Inhibitors of FtsZ from M-Tuberculosis
- Author
-
Robert C. Reynolds, Judith V. Hobrath, Hava Lofton, Malini Rajagopalan, Bini Mathew, Michele Connelly, Larry J. Ross, and R. Kiplin Guy
- Subjects
0301 basic medicine ,Antitubercular Agents ,lcsh:Medicine ,Biochemistry ,Polymerization ,Mice ,Pharmaceutical Sciences ,Sulindac ,Medicine and Health Sciences ,lcsh:Science ,Crystallography ,Multidisciplinary ,biology ,Organic Compounds ,Antimicrobials ,Physics ,Chemical Reactions ,Drugs ,respiratory system ,Condensed Matter Physics ,3. Good health ,Actinobacteria ,Chemistry ,Physical Sciences ,Crystal Structure ,Pharmacophore ,Antibacterial activity ,Research Article ,medicine.drug ,030106 microbiology ,Microbial Sensitivity Tests ,macromolecular substances ,Sulfides ,Microbiology ,Cell Line ,Mycobacterium tuberculosis ,03 medical and health sciences ,Bacterial Proteins ,Tubulins ,In vivo ,Microbial Control ,medicine ,Animals ,Solid State Physics ,FtsZ ,Pharmacology ,Bacteria ,lcsh:R ,Organic Chemistry ,Organisms ,Chemical Compounds ,Biology and Life Sciences ,Proteins ,Polymer Chemistry ,biology.organism_classification ,Farmaceutiska vetenskaper ,Amides ,In vitro ,Cytoskeletal Proteins ,030104 developmental biology ,Tubulin ,biology.protein ,Antibacterials ,lcsh:Q ,Colchicine - 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
24. An Automated Approach to Efficiently Reformat a Large Collection of Compounds
- Author
-
Anang A. Shelat, R. Kiplin Guy, Taosheng Chen, Sergio C. Chai, and Jimmy Cui
- Subjects
dynamic ,compound ,business.industry ,Computer science ,Process (engineering) ,Compound management ,library ,Timeline ,Commercial Sources ,Biochemistry ,Data science ,Automation ,Article ,Genetics ,Molecular Medicine ,reformat ,business ,Software engineering ,Molecular Biology ,Throughput (business) ,high throughput screening - Abstract
Large-scale screening of small organic compounds has become a standard and essential practice in the early discovery of chemical entities with potential therapeutic use. To effectively support high-throughput screening campaigns, compound collections have to be in suitable formats, which requires a process known as compound reformatting. Here we report our approach to reformat the newly-established chemical repository of a large-scale screening facility at St. Jude Children's Research Hospital, which comprises more than half a million compounds, mostly from commercial sources. We highlight the timeline for a reformatting process, the importance of standardizing the operational procedures, and the advantages and disadvantages of using automation. The end result of our reformatting process is the concurrent generation of copies for long-term storage, screening, and "cherry-picking"; all of which facilitate compound management and high- throughput screening.
- Published
- 2011
25. Chemical genetics of Plasmodium falciparum
- Author
-
Wesley C. Van Voorhis, John S. Lazo, Paula L. McGinley, Michele Connelly, Abhai K. Tripathi, Margaret A. Phillips, Isaac P. Forquer, Gregory J. Crowther, R. Kiplin Guy, Sandra Duffy, María Belén Jiménez-Díaz, David Bouck, Paul H. Davis, María Santos Martínez, Vicky M. Avery, Julie Clark, Elizabeth R. Sharlow, Farah El Mazouni, Joseph W. Fowble, Michael K. Riscoe, Steve Castro, Anang A. Shelat, Iñigo Angulo-Barturen, David S. Roos, David J. Sullivan, Ian Bathurst, Emily Wilson, W. Armand Guiguemde, Fangyi Zhu, Jiri Gut, Santiago Ferrer, Pradipsinh K. Rathod, David C. Smithson, Philip J. Rosenthal, and Joseph L. DeRisi
- Subjects
Erythrocytes ,Drug Resistance ,Drug resistance ,Mice ,0302 clinical medicine ,Drug Discovery ,2.2 Factors relating to the physical environment ,Artemisinin ,Aetiology ,Phylogeny ,0303 health sciences ,Multidisciplinary ,Drug discovery ,Preclinical ,3. Good health ,Phenotype ,Infectious Diseases ,Drug development ,5.1 Pharmaceuticals ,Combination ,Development of treatments and therapeutic interventions ,Infection ,Chemical genetics ,medicine.drug ,Biotechnology ,Falciparum ,General Science & Technology ,030231 tropical medicine ,Plasmodium falciparum ,Computational biology ,Biology ,Article ,Cell Line ,Small Molecule Libraries ,03 medical and health sciences ,Antimalarials ,Rare Diseases ,Drug Therapy ,parasitic diseases ,medicine ,Genetics ,Animals ,Humans ,030304 developmental biology ,Reproducibility of Results ,medicine.disease ,biology.organism_classification ,Malaria ,Vector-Borne Diseases ,Orphan Drug ,Good Health and Well Being ,Parasitology ,Immunology ,Drug Evaluation - Abstract
Malaria caused by Plasmodium falciparum is a disease that is responsible for 880,000 deaths per year worldwide. Vaccine development has proved difficult and resistance has emerged for most antimalarial drugs. To discover new antimalarial chemotypes, we have used a phenotypic forward chemical genetic approach to assay 309,474 chemicals. Here we disclose structures and biological activity of the entire library-many of which showed potent in vitro activity against drug-resistant P. falciparum strains-and detailed profiling of 172 representative candidates. A reverse chemical genetic study identified 19 new inhibitors of 4 validated drug targets and 15 novel binders among 61 malarial proteins. Phylochemogenetic profiling in several organisms revealed similarities between Toxoplasma gondii and mammalian cell lines and dissimilarities between P. falciparum and related protozoans. One exemplar compound displayed efficacy in a murine model. Our findings provide the scientific community with new starting points for malaria drug discovery.
- Published
- 2010
26. Pentacyclic nitrofurans that rapidly kill nifurtimox-resistant trypanosomes
- Author
-
Adaris Rodriguez-Cortes, Susan Wyllie, Richard E. Lee, Alan H. Fairlamb, David F. Bruhn, Angela K. Carrillo, R. Kiplin Guy, and Rakesh
- Subjects
0301 basic medicine ,Microbiology (medical) ,ATP Binding Cassette Transporter, Subfamily B ,medicine.drug_class ,Nitrofurans ,Trypanosoma brucei brucei ,Drug Resistance ,Drug resistance ,Microbial Sensitivity Tests ,Trypanosoma brucei ,Microbiology ,Cell Line ,03 medical and health sciences ,In vivo ,parasitic diseases ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Nifurtimox ,Nitrofuran ,Trypanocidal agent ,Original Research ,Pharmacology ,biology ,biology.organism_classification ,Trypanocidal Agents ,In vitro ,3. Good health ,Kinetics ,030104 developmental biology ,Infectious Diseases ,Trypanosoma ,medicine.drug - Abstract
OBJECTIVES In response to reports of Trypanosoma brucei resistance to the nitroaromatic drug nifurtimox, we evaluated the potential of antituberculosis nitrofuran isoxazolines as inhibitors of trypanosome growth. METHODS The susceptibility of T. brucei brucei was assessed in vitro. The lowest effective concentration to inhibit growth (EC90) against drug-susceptible and -resistant parasites, time-kill kinetics, reversibility of inhibition and propensity for P-glycoprotein-mediated exclusion from the blood-brain barrier were determined. RESULTS Nitrofuran isoxazolines were potent inhibitors of T. brucei brucei proliferation at nanomolar concentrations, with pentacyclic nitrofurans being 100-fold more potent than nifurtimox. Activity was sustained against nifurtimox-resistant parasites, suggesting the possibility of a unique mechanism of activation and potential for use in the treatment of drug-resistant infections. Exposure of parasites to the maximum concentrations of Compound 15 achieved in vivo with oral dosing yielded >2 logs of irreversible killing in
- Published
- 2015
27. Design, Synthesis and Evaluation of 2,5-Diketopiperazines as Inhibitors of the MDM2-p53 Interaction
- Author
-
Jaeki Min, M. Brett Waddell, Maria Quant, R. Kiplin Guy, Richard W. Kriwacki, Mariell Pettersson, Kristina Luthman, Luigi I. Iconaru, and Morten Grøtli
- Subjects
Models, Molecular ,Stereochemistry ,lcsh:Medicine ,Peptide ,Chemistry Techniques, Synthetic ,Diketopiperazines ,Protein Structure, Secondary ,Protein–protein interaction ,chemistry.chemical_compound ,Protein structure ,Peptide synthesis ,Humans ,Binding site ,lcsh:Science ,chemistry.chemical_classification ,Multidisciplinary ,Binding Sites ,lcsh:R ,Rational design ,Proto-Oncogene Proteins c-mdm2 ,Biochemistry ,chemistry ,Docking (molecular) ,Drug Design ,lcsh:Q ,Tumor Suppressor Protein p53 ,Research Article ,Protein Binding - 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 alpha-helical region of the p53 peptide, containing key residues in the i, i+4 and i+7 positions of a natural alpha-helix. Conformational analysis indicated that 1,3,6-trisubstituted 2,5-diketopiperazines were able to place substituents in the same spatial orientation as an alpha-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
28. 8-Triazolylpurines: Towards Fluorescent Inhibitors of the MDM2/p53 Interaction
- Author
-
Jesper R. Nilsson, Joakim Andréasson, Jaeki Min, Jimmy Jacobsson, Richard W. Kriwacki, Morten Grøtli, Mariell Pettersson, David Bliman, Luigi I. Iconaru, and R. Kiplin Guy
- Subjects
Models, Molecular ,Multidisciplinary ,Molecular Structure ,lcsh:R ,Molecular Conformation ,lcsh:Medicine ,Biological activity ,Proto-Oncogene Proteins c-mdm2 ,Nuclear magnetic resonance spectroscopy ,Plasma protein binding ,Fluorescence ,Small molecule ,Biochemistry ,Docking (molecular) ,Purines ,Biophysics ,Molecule ,lcsh:Q ,Tumor Suppressor Protein p53 ,lcsh:Science ,Fluorescence anisotropy ,Research Article ,Protein Binding - 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
29. Potent Plasmodium falciparum Gametocytocidal Activity of Diaminonaphthoquinones, Lead Antimalarial Chemotypes Identified in an Antimalarial Compound Screen
- Author
-
Praveen Kumar Suryadevara, Kim C. Williamson, W. Armand Guiguemde, Jaeki Min, Maxim I. Maron, Takeshi Tanaka, R. Kiplin Guy, Michele Connelly, and David S. Barnett
- Subjects
Drug ,media_common.quotation_subject ,Population ,Plasmodium falciparum ,Drug Evaluation, Preclinical ,Drug resistance ,Pharmacology ,Plasmodium ,Antimalarials ,Structure-Activity Relationship ,Gametocyte ,medicine ,Humans ,Pharmacology (medical) ,Experimental Therapeutics ,education ,media_common ,education.field_of_study ,biology ,Transmission (medicine) ,Imidazoles ,Hep G2 Cells ,biology.organism_classification ,medicine.disease ,Infectious Diseases ,Malaria ,Naphthoquinones - Abstract
Forty percent of the world's population is threatened by malaria, which is caused by Plasmodium parasites and results in an estimated 200 million clinical cases and 650,000 deaths each year. Drug resistance has been reported for all commonly used antimalarials and has prompted screens to identify new drug candidates. However, many of these new candidates have not been evaluated against the parasite stage responsible for transmission, gametocytes. If Plasmodium falciparum gametocytes are not eliminated, patients continue to spread malaria for weeks after asexual parasite clearance. Asymptomatic individuals can also harbor gametocyte burdens sufficient for transmission, and a safe, effective gametocytocidal agent could also be used in community-wide malaria control programs. Here, we identify 15 small molecules with nanomolar activity against late-stage gametocytes. Fourteen are diaminonaphthoquinones (DANQs), and one is a 2-imino-benzo[d]imidazole (IBI). One of the DANQs identified, SJ000030570, is a lead antimalarial candidate. In contrast, 94% of the 650 compounds tested are inactive against late-stage gametocytes. Consistent with the ineffectiveness of most approved antimalarials against gametocytes, of the 19 novel compounds with activity against known anti-asexual-stage targets, only 3 had any strong effect on gametocyte viability. These data demonstrate the distinct biology of the transmission stages and emphasize the importance of screening for gametocytocidal activity. The potent gametocytocidal activity of DANQ and IBI coupled with their efficacy against asexual parasites provides leads for the development of antimalarials with the potential to prevent both the symptoms and the spread of malaria.
- Published
- 2015
30. NALP3 inflammasome upregulation and CASP1 cleavage of the glucocorticoid receptor cause glucocorticoid resistance in leukemia cells
- Author
-
John C. Panetta, Anand Mayasundari, Mary V. Relling, Ching-Hon Pui, Alessandra Zanut, Barthelemy Diouf, J. Kevin Hicks, Thomas L. Dunwell, Seth E. Karol, Gabriele Stocco, Mark R. Wilkinson, Daniel Savic, Farida Latif, Audrey Giordanengo, Steven W. Paugh, Prajwal Gurung, Christian A. Fernandez, Daniel J. Crona, Charles G. Mullighan, William L. Carroll, R. Kiplin Guy, Jaeki Min, Richard M. Myers, William E. Thierfelder, Rob Pieters, Erik J. Bonten, Thirumala-Devi Kanneganti, Kristine R. Crews, Yiping Fan, David R. Coss, Cheng Cheng, William E. Evans, Wenjian Yang, Joy J. Bianchi, Sima Jeha, Marcelo Actis, Antonio M. Ferreira, R. K. Subbarao Malireddi, Monique L. den Boer, Colton Smith, Deepa Bhojwani, Lucas T. Laudermilk, J. Robert McCorkle, Linda Holmfeldt, Laura B. Ramsey, Paugh, Steven W., Bonten, Erik J., Savic, Daniel, Ramsey, Laura B., Thierfelder, William E., Gurung, Prajwal, Malireddi, R. K. Subbarao, Actis, Marcelo, Mayasundari, Anand, Min, Jaeki, Coss, David R., Laudermilk, Lucas T., Panetta, John C., Mccorkle, J. Robert, Fan, Yiping, Crews, Kristine R., Stocco, Gabriele, Wilkinson, Mark R., Ferreira, Antonio M., Cheng, Cheng, Yang, Wenjian, Karol, Seth E., Fernandez, Christian A., Diouf, Barthelemy, Smith, Colton, Hicks, J. Kevin, Zanut, Alessandra, Giordanengo, Audrey, Crona, Daniel, Bianchi, Joy J., Holmfeldt, Linda, Mullighan, Charles G., Den Boer, Monique L., Pieters, Rob, Jeha, Sima, Dunwell, Thomas L., Latif, Farida, Bhojwani, Deepa, Carroll, William L., Pui, Ching Hon, Myers, Richard M., Guy, R. Kiplin, Kanneganti, Thirumala Devi, Relling, Mary V., Evans, William E., and Pediatrics
- Subjects
Transcription, Genetic ,Inflammasomes ,Drug Resistance ,NALP3 ,Drug Screening Assays ,glucocorticoids, pharmacogenomics, epigenomics ,0302 clinical medicine ,Glucocorticoid receptor ,Glucocorticoid ,Receptors ,Tumor Cells, Cultured ,Child ,Leukemic ,0303 health sciences ,Cultured ,glucocorticoids ,biology ,Gene Expression Regulation, Leukemic ,Caspase 1 ,Inflammasome ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Adolescent ,Antineoplastic Agents, Hormonal ,Base Sequence ,Carrier Proteins ,Child, Preschool ,DNA Methylation ,Drug Resistance, Neoplasm ,Drug Screening Assays, Antitumor ,HEK293 Cells ,Humans ,Infant ,Infant, Newborn ,NLR Family, Pyrin Domain-Containing 3 Protein ,Neoplasm Recurrence, Local ,Prednisolone ,Proteolysis ,Receptors, Glucocorticoid ,Up-Regulation ,3. Good health ,Tumor Cells ,Leukemia ,Local ,030220 oncology & carcinogenesis ,Transcription ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug ,medicine.medical_specialty ,Antineoplastic Agents ,NLR Family ,Article ,03 medical and health sciences ,Downregulation and upregulation ,Genetic ,Internal medicine ,Genetics ,medicine ,Preschool ,030304 developmental biology ,pharmacogenomics ,Hormonal ,Antitumor ,medicine.disease ,Newborn ,Pyrin Domain-Containing 3 Protein ,Endocrinology ,Neoplasm Recurrence ,Gene Expression Regulation ,epigenomics ,biology.protein ,Cancer research ,Neoplasm - Abstract
Glucocorticoids are universally used in the treatment of acute lymphoblastic leukemia (ALL), and resistance to glucocorticoids in leukemia cells confers poor prognosis. To elucidate mechanisms of glucocorticoid resistance, we determined the prednisolone sensitivity of primary leukemia cells from 444 patients newly diagnosed with ALL and found significantly higher expression of CASP1 (encoding caspase 1) and its activator NLRP3 in glucocorticoid-resistant leukemia cells, resulting from significantly lower somatic methylation of the CASP1 and NLRP3 promoters. Overexpression of CASP1 resulted in cleavage of the glucocorticoid receptor, diminished the glucocorticoid-induced transcriptional response and increased glucocorticoid resistance. Knockdown or inhibition of CASP1 significantly increased glucocorticoid receptor levels and mitigated glucocorticoid resistance in CASP1-overexpressing ALL. Our findings establish a new mechanism by which the NLRP3-CASP1 inflammasome modulates cellular levels of the glucocorticoid receptor and diminishes cell sensitivity to glucocorticoids. The broad impact on the glucocorticoid transcriptional response suggests that this mechanism could also modify glucocorticoid effects in other diseases.
- Published
- 2015
31. (+)-SJ733, a clinical candidate for malaria that acts through ATP4 to induce rapid host-mediated clearance of Plasmodium
- Author
-
Adele M. Lehane, Anupam Pradhan, David A. Fidock, Dennis E. Kyle, Gloria Holbrook, Amy Matheny, Martina Sigal, Anang A. Shelat, David Waterson, Iñigo Angulo-Barturen, Kathleen O’Loughlin, Jian Liu, Laura M. Sanz, Philip D. Stein, Daniel H. Ebert, Kiaran Kirk, Hongshen Ma, Marcus C. S. Lee, Jon C. Mirsalis, W. Armand Guiguemde, Xiaoyan Deng, Greg Fedewa, Ane Rodríguez-Alejandre, María Belén Jiménez-Díaz, María G. Gómez, Angela K. Carrillo, Mariana Justino de Almeida, Yandira G. Salinas, Vicky M. Avery, David M. Shackleford, Sandra Duffy, Marie-Eve Myrand-Lapierre, Gregory M. Miller, María Santos Martínez, Adelaide S. M. Dennis, Charles C. Kim, R. Kiplin Guy, Julie Clark, Jeremy A. Horst, Aaron N. Endsley, Joseph L. DeRisi, Zheng Wang, Spencer Knapp, Fangyi Zhu, David M. Floyd, Santiago Ferrer, Jonathon Diep, Francisco-Javier Gamo, Susan A. Charman, and Natalie J. Spillman
- Subjects
Models, Molecular ,Plasmodium ,Erythrocytes ,Drug Resistance ,Drug resistance ,Pharmacology ,Corrections ,chemistry.chemical_compound ,Models ,Heterocyclic Compounds ,Drug Discovery ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Aetiology ,Cellular Senescence ,PfATP4 ,Multidisciplinary ,Molecular Structure ,Drug discovery ,Flow Cytometry ,Cell biology ,Infectious Diseases ,PNAS Plus ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Infection ,Cell aging ,Intracellular ,Phenotypic screening ,malaria ,Calcium-Transporting ATPases ,Biology ,Heterocyclic Compounds, 4 or More Rings ,drug discovery ,Antimalarials ,Rare Diseases ,In vivo ,Prevention ,Molecular ,Plasmodium falciparum ,4 or More Rings ,biology.organism_classification ,Isoquinolines ,Cipargamin ,High-Throughput Screening Assays ,Malaria ,Vector-Borne Diseases ,Orphan Drug ,Good Health and Well Being ,chemistry - Abstract
Drug discovery for malaria has been transformed in the last 5 years by the discovery of many new lead compounds identified by phenotypic screening. The process of developing these compounds as drug leads and studying the cellular responses they induce is revealing new targets that regulate key processes in the Plasmodium parasites that cause malaria. We disclose herein that the clinical candidate (+)-SJ733 acts upon one of these targets, ATP4. ATP4 is thought to be a cation-transporting ATPase responsible for maintaining low intracellular Na(+) levels in the parasite. Treatment of parasitized erythrocytes with (+)-SJ733 in vitro caused a rapid perturbation of Na(+) homeostasis in the parasite. This perturbation was followed by profound physical changes in the infected cells, including increased membrane rigidity and externalization of phosphatidylserine, consistent with eryptosis (erythrocyte suicide) or senescence. These changes are proposed to underpin the rapid (+)-SJ733-induced clearance of parasites seen in vivo. Plasmodium falciparum ATPase 4 (pfatp4) mutations that confer resistance to (+)-SJ733 carry a high fitness cost. The speed with which (+)-SJ733 kills parasites and the high fitness cost associated with resistance-conferring mutations appear to slow and suppress the selection of highly drug-resistant mutants in vivo. Together, our data suggest that inhibitors of PfATP4 have highly attractive features for fast-acting antimalarials to be used in the global eradication campaign.
- Published
- 2014
32. Repositioning: the fast track to new anti-malarial medicines?
- Author
-
R. Kiplin Guy, Santiago Ferrer-Bazaga, Tanya Parkinson, Iñigo Angulo-Barturen, Francisco Javier Gamo-Benito, Michele Connelly, Sandra Duffy, Julie Lotharius, Julie Clark, Balachandra Bandodkar, Nikhil Rautela, Jörg J. Möhrle, Pavithra Viswanath, Timothy N. C. Wells, Sowmya Bharath, and Vicky M. Avery
- Subjects
Male ,Drug ,Candidate drug re-profiling ,medicine.drug_class ,Plasmodium berghei ,Medicina ,media_common.quotation_subject ,Plasmodium falciparum ,Anti-malarial drugs ,Biology ,Pharmacology ,Antimalarials ,Mice ,Parasitic Sensitivity Tests ,In vitro ,In vivo ,medicine ,Animals ,Malaria, Falciparum ,Biología y Biomedicina ,media_common ,Mice, Inbred BALB C ,Research ,Drug repositioning ,in vitro ,Protein kinase inhibitor ,biology.organism_classification ,Malaria ,in vivo ,Infectious Diseases ,Humanized mouse ,Parasitology - Abstract
Background Repositioning of existing drugs has been suggested as a fast track for developing new anti-malarial agents. The compound libraries of GlaxoSmithKline (GSK), Pfizer and AstraZeneca (AZ) comprising drugs that have undergone clinical studies in other therapeutic areas, but not achieved approval, and a set of US Food and Drug Administration (FDA)-approved drugs and other bio-actives were tested against Plasmodium falciparum blood stages. Methods Molecules were tested initially against erythrocytic co-cultures of P. falciparum to measure proliferation inhibition using one of the following methods: SYBR®I dye DNA staining assay (3D7, K1 or NF54 strains); [3H] hypoxanthine radioisotope incorporation assay (3D7 and 3D7A strain); or 4’,6-diamidino-2-phenylindole (DAPI) DNA imaging assay (3D7 and Dd2 strains). After review of the available clinical pharmacokinetic and safety data, selected compounds with low μM activity and a suitable clinical profile were tested in vivo either in a Plasmodium berghei four-day test or in the P. falciparum Pf3D70087/N9 huSCID ‘humanized’ mouse model. Results Of the compounds included in the GSK and Pfizer sets, 3.8% (9/238) had relevant in vitro anti-malarial activity while 6/100 compounds from the AZ candidate drug library were active. In comparison, around 0.6% (24/3,800) of the FDA-approved drugs and other bio-actives were active. After evaluation of available clinical data, four investigational drugs, active in vitro were tested in the P. falciparum humanized mouse model: UK-112,214 (PAF-H1 inhibitor), CEP-701 (protein kinase inhibitor), CEP-1347 (protein kinase inhibitor), and PSC-833 (p-glycoprotein inhibitor). Only UK-112,214 showed significant efficacy against P. falciparum in vivo, although at high doses (ED90 131.3 mg/kg [95% CI 112.3, 156.7]), and parasitaemia was still present 96 hours after treatment commencement. Of the six actives from the AZ library, two compounds (AZ-1 and AZ-3) were marginally efficacious in vivo in a P. berghei model. Conclusions Repositioning of existing therapeutics in malaria is an attractive proposal. Compounds active in vitro at μM concentrations were identified. However, therapeutic concentrations may not be effectively achieved in mice or humans because of poor bio-availability and/or safety concerns. Stringent safety requirements for anti-malarial drugs, given their widespread use in children, make this a challenging area in which to reposition therapy.
- Published
- 2014
33. A screening based approach to circumvent tumor microenvironment-driven intrinsic resistance to BCR-ABL+ inhibitors in Ph+ acute lymphoblastic leukemia
- Author
-
Richard T. Williams, Nidal Boulos, Harpreet Singh, Anang A. Shelat, Amandeep Singh, and R. Kiplin Guy
- Subjects
Fusion Proteins, bcr-abl ,Antineoplastic Agents ,Pharmacology ,Biology ,Biochemistry ,Article ,Analytical Chemistry ,Mice ,In vivo ,hemic and lymphatic diseases ,Drug Discovery ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Protein Kinase Inhibitors ,Tumor microenvironment ,Imatinib ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,High-Throughput Screening Assays ,Dasatinib ,Leukemia ,Phenotype ,Nilotinib ,Drug Resistance, Neoplasm ,Neoplastic Stem Cells ,Molecular Medicine ,Cytokines ,Classical pharmacology ,Drug Screening Assays, Antitumor ,Biotechnology ,medicine.drug ,Chronic myelogenous leukemia - Abstract
Signaling by the BCR-ABL fusion kinase drives Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) and chronic myelogenous leukemia (CML). Despite their clinical activity in many patients with CML, the BCR-ABL kinase inhibitors (BCR-ABL-KIs) imatinib, dasatinib, and nilotinib provide only transient leukemia reduction in patients with Ph+ ALL. While host-derived growth factors in the leukemia microenvironment have been invoked to explain this drug resistance, their relative contribution remains uncertain. Using genetically defined murine Ph+ ALL cells, we identified interleukin 7 (IL-7) as the dominant host factor that attenuates response to BCR-ABL-KIs. To identify potential combination drugs that could overcome this IL-7-dependent BCR-ABL-KI-resistant phenotype, we screened a small-molecule library including Food and Drug Administration-approved drugs. Among the validated hits, the well-tolerated antimalarial drug dihydroartemisinin (DHA) displayed potent activity in vitro and modest in vivo monotherapy activity against engineered murine BCR-ABL-KI-resistant Ph+ ALL. Strikingly, cotreatment with DHA and dasatinib in vivo strongly reduced primary leukemia burden and improved long-term survival in a murine model that faithfully captures the BCR-ABL-KI-resistant phenotype of human Ph+ ALL. This cotreatment protocol durably cured 90% of treated animals, suggesting that this cell-based screening approach efficiently identified drugs that could be rapidly moved to human clinical testing.
- Published
- 2013
34. Synthesis and evaluation of methylsulfonylnitrobenzamides (MSNBAs) as inhibitors of the thyroid hormone receptor-coactivator interaction
- Author
-
R. Kiplin Guy, Jong Yeon Hwang, Angela K. Carrillo, Ramy R. Attia, and Michele Connelly
- Subjects
medicine.medical_specialty ,Cell Survival ,Clinical Biochemistry ,Pharmaceutical Science ,Transfection ,Biochemistry ,Article ,MED1 ,Thyroid hormone receptor beta ,Structure-Activity Relationship ,Nuclear Receptor Coactivator 1 ,Internal medicine ,Drug Discovery ,Coactivator ,medicine ,Humans ,Protein Interaction Maps ,Molecular Biology ,Thyroid hormone receptor ,Receptors, Thyroid Hormone ,Chemistry ,Organic Chemistry ,Hep G2 Cells ,Thiazoles ,Endocrinology ,Thyroid hormone receptor alpha ,Hormone receptor ,Nuclear receptor coactivator 3 ,Benzamides ,Nuclear receptor coactivator 2 ,Molecular Medicine - Abstract
We previously identified the methylsulfonylnitrobenzoates (MSNBs) that block the interaction of the thyroid hormone receptor with its obligate transcriptional coactivators and prevent thyroid hormone signaling. As part of our lead optimization work we demonstrated that sulfonylnitrophenylthiazoles (SNPTs), which replace the ester linkage of MSNBs with a thiazole, also inhibited coactivator binding to TR. Here we report that replacement of the ester with an amide (methylsulfonylnitrobenzamides, MSNBA) also provides active TR antagonists.
- Published
- 2013
35. Antimalarial activity of 10-alkyl/aryl esters and – aminoethylethers of artemisinin
- Author
-
R. Kiplin Guy, Martina Sigal, Henk J. Krebs, Theunis T. Cloete, Michele Connelly, David D. N'Da, Amy Orcutt, Julie Clark, 13061372 - Cloete, Theunis Theodorus, and 20883072 - N'Da, David Dago
- Subjects
Cell Survival ,Stereochemistry ,medicine.medical_treatment ,Cytotoxicity ,Plasmodium falciparum ,Dihydroartemisinin ,Artesunate ,Ozonide ,Ether ,Biochemistry ,Cell Line ,Antimalarials ,chemistry.chemical_compound ,Drug Discovery ,parasitic diseases ,medicine ,Humans ,Potency ,Artemether ,Malaria, Falciparum ,Artemisinin ,Molecular Biology ,biology ,Aryl ,Organic Chemistry ,biology.organism_classification ,Artemisinins ,chemistry ,medicine.drug - Abstract
A series of n-alkyl/aryl esters were synthesized and their in vitro antiplasmodial activity was measured alongside that of previously synthesized aminoethylethers of artemisinin ozonides against various strains of Plasmodium falciparum. The cytotoxicity against human cell lines was also assessed. The esters were synthesized in a one-step reaction by derivatization on carbon C-10 of dihydroartemisinin. Both classes were active against both the 3D7 and K1 strains of P. falciparum, with all compounds being significantly more potent than artemether against both strains. The majority of compounds possessed potency either comparable or more than artesunate with a high degree of selectivity towards the parasitic cells. The 10a-n-propyl 11 and 10a-benzyl 18 esters were the most potent of all synthesized ozonides, possessing a moderate ( 3-fold) and significant (22- and 12-fold, respectively) potency increases against the 3D7 and K1 strains, respectively, in comparison with artesunate. http://www.journals.elsevier.com/bioorganic-chemistry/
- Published
- 2013
36. Lead Optimization of Anti-Malarial Propafenone Analogs
- Author
-
Michele Connelly, Lalitha V. Iyer, Anupam Pradhan, Andrew Lemoff, Joseph L. DeRisi, Anna M. Furimsky, Fangyi Zhu, Martina Sigal, Dennis E. Kyle, R. Kiplin Guy, Jon C. Mirsalis, Jason Gow, Julie Clark, Emily Wilson, W. Armand Guiguemde, David J. Lowes, Toufan Parman, and Liang Tang
- Subjects
Plasmodium berghei ,Drug Evaluation, Preclinical ,Administration, Oral ,Parasitemia ,Propafenone ,Pharmacology ,Cardiovascular ,Mice ,Drug Discovery ,Drug Interactions ,Mice, Inbred ICR ,Chemistry ,Chloroquine ,Pharmacology and Pharmaceutical Sciences ,Hep G2 Cells ,Inbred ICR ,Preclinical ,Heart Disease ,Infectious Diseases ,Liver ,Cytochrome P-450 CYP2D6 ,5.1 Pharmaceuticals ,Administration ,Microsomes, Liver ,Molecular Medicine ,Female ,Development of treatments and therapeutic interventions ,medicine.drug ,Oral ,Medicinal & Biomolecular Chemistry ,Article ,Medicinal and Biomolecular Chemistry ,Antimalarials ,Structure-Activity Relationship ,Rare Diseases ,Pharmacokinetics ,In vivo ,Microsomes ,Cytochrome P-450 CYP2D6 Inhibitors ,medicine ,Animals ,Humans ,Animal ,Organic Chemistry ,medicine.disease ,Bioavailability ,Malaria ,Vector-Borne Diseases ,Disease Models, Animal ,Orphan Drug ,Good Health and Well Being ,HEK293 Cells ,Ion channel activity ,Disease Models ,Drug Evaluation - Abstract
Previously reported studies identified analogues of propafenone that had potent antimalarial activity, reduced cardiac ion channel activity, and properties that suggested the potential for clinical development for malaria. Careful examination of the bioavailability, pharmacokinetics, toxicology, and efficacy of this series of compounds using rodent models revealed orally bioavailable compounds that are nontoxic and suppress parasitemia in vivo. Although these compounds possess potential for further preclinical development, they also carry some significant challenges.
- Published
- 2012
37. Targeting the Binding Function 3 (BF3) Site of the Human Androgen Receptor Through Virtual Screening
- Author
-
K.H. Chan, Frank Q. Han, Peter Axerio-Cilies, R. Kiplin Guy, Emma Tomlinson Guns, Nathan A. Lack, Paul S. Rennie, Clementine Feau, Artem Cherkasov, Eric Leblanc, and Peyman Tavassoli
- Subjects
Models, Molecular ,Databases, Factual ,Transcription, Genetic ,Protein Conformation ,In silico ,Quantitative Structure-Activity Relationship ,Pharmacology ,Crystallography, X-Ray ,Ligands ,Article ,Small Molecule Libraries ,Prostate cancer ,Cell Line, Tumor ,Drug Discovery ,medicine ,Humans ,Binding site ,Receptor ,Cytotoxicity ,Virtual screening ,Binding Sites ,Chemistry ,Androgen Antagonists ,medicine.disease ,Androgen receptor ,Mechanism of action ,Receptors, Androgen ,Mutation ,Molecular Medicine ,medicine.symptom - Abstract
The androgen receptor (AR) is the best studied drug target for the treatment of prostate cancer. While there are a number of drugs that target the AR, they all work through the same mechanism of action and are prone to the development of drug resistance. There is a large unmet need for novel AR inhibitors which work through alternative mechanism(s). Recent studies have identified a novel site on the AR called Binding Function 3 (BF3) that is involved into AR transcriptional activity. In order to identify inhibitors that target the BF3 site, we have conducted a large-scale in-silico screen followed by experimental evaluation. A number of compounds were identified that effectively inhibited the AR transcriptional activity with no obvious cytotoxicity. The mechanism of action of these compounds was validated by biochemical assays and x-ray crystallography. These findings lay a foundation for the development of alternative or supplementary therapies capable of combating prostate cancer even in its anti-androgen resistant forms.
- Published
- 2011
38. Synthesis and evaluation of 7-substituted 4-aminoquinoline analogs for antimalarial activity
- Author
-
Jong Yeon Hwang, David J. Lowes, R. Kiplin Guy, W. Armand Guiguemde, Michele Connelly, Martina Sigal, Emily Wilson, Joseph L. DeRisi, Takashi Kawasuji, Fangyi Zhu, and Julie Clark
- Subjects
Chemistry ,Hydrogen bond ,Stereochemistry ,Quinoline ,Plasmodium falciparum ,Substituent ,Drug Resistance ,Ether ,Membranes, Artificial ,Article ,Permeability ,Cell Line ,chemistry.chemical_compound ,Antimalarials ,Structure-Activity Relationship ,Solubility ,4-Aminoquinoline ,Drug Discovery ,Side chain ,Aminoquinolines ,Molecular Medicine ,Structure–activity relationship ,Humans ,Amine gas treating - Abstract
We previously reported that substituted 4-aminoquinolines with a phenyl ether substituent at the 7-position of the quinoline ring and the capability of intramolecular hydrogen bonding between the protonated amine on the side chain and a hydrogen bond acceptor on the amine's alkyl substituents exhibited potent antimalarial activity against the multidrug resistant strain P. falciparum W2. We employed a parallel synthetic method to generate diaryl ether, biaryl, and alkylaryl 4-aminoquinoline analogues in the background of a limited number of side chain variations that had previously afforded potent 4-aminoquinolines. All subsets were evaluated for their antimalarial activity against the chloroquine-sensitive strain 3D7 and the chloroquine-resistant K1 strain as well as for cytotoxicity against mammalian cell lines. While all three arrays showed good antimalarial activity, only the biaryl-containing subset showed consistently good potency against the drug-resistant K1 strain and good selectivity with regard to mammalian cytotoxicity. Overall, our data indicate that the biaryl-containing series contains promising candidates for further study.
- Published
- 2011
39. Development of a New Generation of 4-Aminoquinoline Antimalarial Compounds Using Predictive Pharmacokinetic and Toxicology Models
- Author
-
Michael J. Furniss, Susanna E. LeValley, Peter B. Madrid, Carol E. Green, Sunetra Ray, Joseph L. DeRisi, Linda L. Rausch, R. Kiplin Guy, Jon C. Mirsalis, Paul Catz, and Lalitha V. Iyer
- Subjects
Plasmodium falciparum ,Drug Evaluation, Preclinical ,Pharmacology ,Toxicology ,Article ,Small Molecule Libraries ,chemistry.chemical_compound ,Antimalarials ,Mice ,Pharmacokinetics ,In vivo ,Drug Discovery ,Potency ,Animals ,Antimalarial Agent ,biology ,Chemistry ,biology.organism_classification ,Bioavailability ,Drug development ,4-Aminoquinoline ,Aminoquinolines ,Molecular Medicine ,Half-Life - Abstract
Among the known antimalarial drugs, chloroquine (CQ) and other 4-aminoquinolines have shown high potency and good bioavailability. Yet complications associated with drug resistance necessitate the discovery of effective new antimalarial agents. ADMET prediction studies were employed to evaluate a library of new molecules based on the 4-aminoquinolone-related structure of CQ. Extensive in vitro screening and in vivo pharmacokinetic studies in mice helped to identify two lead molecules, 18 and 4, with promising in vitro therapeutic efficacy, improved ADMET properties, low risk for drug-drug interactions, and desirable pharmacokinetic profiles. Both 18 and 4 are highly potent antimalarial compounds, with IC(50) values of 5.6 and 17.3 nM, respectively, against the W2 (CQ-resistant) strain of Plasmodium falciparum (for CQ, IC(50) = 382 nM). When tested in mice, these compounds were found to have biological half-lives and plasma exposure values similar to or higher than those of CQ; they are therefore desirable candidates to pursue in future clinical trials.
- Published
- 2010
40. Identification and Characterization of the First Small Molecule Inhibitor of MDMX
- Author
-
Antonio M. Ferreira, R. Kiplin Guy, Anang A. Shelat, Michael A. Dyer, Aart G. Jochemsen, Ying Shen, Donald Bashford, Catherine Regni, Brenda A. Schulman, Damon R. Reed, Samantha A. Cicero, Fangyi Zhu, Leggy A. Arnold, David C. Smithson, and Nicholas Mills
- Subjects
MDMX ,High-throughput screening ,Antineoplastic Agents ,Apoptosis ,Acetates ,Biochemistry ,Small Molecule Libraries ,Mice ,Proto-Oncogene Proteins c-mdm2 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Computer Simulation ,Molecular Biology ,biology ,Retinoblastoma ,Cell Biology ,Cell cycle ,medicine.disease ,Small molecule ,protein stability p53 activation ring domain antagonists apoptosis binding cancer cells overexpression retinoblastoma ,High-Throughput Screening Assays ,Cancer research ,biology.protein ,Mdm2 ,Pyrazoles ,Tumor Suppressor Protein p53 - Abstract
The p53 pathway is disrupted in virtually every human tumor. In approximately 50% of human cancers, the p53 gene is mutated, and in the remaining cancers, the pathway is dysregulated by genetic lesions in other genes that modulate the p53 pathway. One common mechanism for inactivation of the p53 pathway in tumors that express wild-type p53 is increased expression of MDM2 or MDMX. MDM2 and MDMX bind p53 and inhibit its function by distinct nonredundant mechanisms. Small molecule inhibitors and small peptides have been developed that bind MDM2 in the p53-binding pocket and displace the p53 protein, leading to p53-mediated cell cycle exit and apoptosis. To date, peptide inhibitors of MDMX have been developed, but no small molecule inhibitors have been reported. We have developed biochemical and cell-based assays for high throughput screening of chemical libraries to identify MDMX inhibitors and identified the first MDMX inhibitor SJ-172550. This compound binds reversibly to MDMX and effectively kills retinoblastoma cells in which the expression of MDMX is amplified. The effect of SJ-172550 is additive when combined with an MDM2 inhibitor. Results from a series of biochemical and structural modeling studies suggest that SJ-172550 binds the p53-binding pocket of MDMX, thereby displacing p53. This lead compound is a useful chemical scaffold for further optimization of MDMX inhibitors that may eventually be used to treat pediatric cancers and various adult tumors that overexpress MDMX or have similar genetic lesions. When combined with selective MDM2 inhibitors, SJ-172550 may also be useful for treating tumors that express wild-type p53.
- Published
- 2010
- Full Text
- View/download PDF
41. Non-competitive androgen receptor inhibition in vitro and in vivo
- Author
-
Marc I. Diamond, Keith R. Yamamoto, Byron Hann, Yong Huang, Jeremy O. Jones, Clementine Feau, Eric C. Bolton, and R. Kiplin Guy
- Subjects
Male ,Cell ,Pharmacology ,Biology ,Cell Line ,Pyrvinium ,chemistry.chemical_compound ,Prostate cancer ,Mice ,Pyrvinium Compounds ,In vivo ,medicine ,Androgen Receptor Antagonists ,Animals ,Humans ,Cell Proliferation ,Harmol ,Multidisciplinary ,Molecular Structure ,Cell growth ,Biological Sciences ,medicine.disease ,Androgen receptor ,Harmine ,medicine.anatomical_structure ,chemistry ,Receptors, Androgen - Abstract
Androgen receptor (AR) inhibitors are used to treat multiple human diseases, including hirsutism, benign prostatic hypertrophy, and prostate cancer, but all available anti-androgens target only ligand binding, either by reduction of available hormone or by competitive antagonism. New strategies are needed, and could have an important impact on therapy. One approach could be to target other cellular mechanisms required for receptor activation. In prior work, we used a cell-based assay of AR conformation change to identify non-ligand inhibitors of AR activity. Here, we characterize 2 compounds identified in this screen: pyrvinium pamoate, a Food and Drug Administration-approved drug, and harmol hydrochloride, a natural product. Each compound functions by a unique, non-competitive mechanism and synergizes with competitive antagonists to disrupt AR activity. Harmol blocks DNA occupancy by AR, whereas pyrvinium does not. Pyrvinium inhibits AR-dependent gene expression in the prostate gland in vivo, and induces prostate atrophy. These results highlight new therapeutic strategies to inhibit AR activity.
- Published
- 2009
42. A High-Throughput Ligand Competition Binding Assay for the Androgen Receptor and other Nuclear Receptors
- Author
-
Clementine Feau, Leggy A. Arnold, Aaron Kosinski, and R. Kiplin Guy
- Subjects
Ligand ,Ligand binding assay ,High-throughput screening ,Receptors, Cytoplasmic and Nuclear ,Biology ,Ligands ,Molecular biology ,Binding, Competitive ,Radioligand Assay ,Article ,Androgen receptor ,Scintillation proximity assay ,Biochemistry ,Nuclear receptor ,Receptors, Androgen ,Androgens ,Receptor - Abstract
Standardized, automated ligand-binding assays facilitate evaluation of endocrine activities of environmental chemicals and identification of antagonists of nuclear receptor ligands. Many current assays rely on fluorescently labeled ligands that are significantly different from the native ligands. The authors describe a radiolabeled ligand competition scintillation proximity assay (SPA) for the androgen receptor (AR) using Ni-coated 384-well FlashPlates® and liganded AR-LBD protein. This highly reproducible, low-cost assay is well suited for automated high-throughput screening. In addition, the authors show that this assay can be adapted to measure ligand affinities for other nuclear receptors (peroxisome proliferation-activated receptor γ, thyroid receptors α and β). ( Journal of Biomolecular Screening 2009:43-48)
- Published
- 2009
43. Parallel Synthesis of 9-Aminoacridines and their Evaluation Against Chloroquine-Resistant Plasmodium falciparum
- Author
-
R. Kiplin Guy, Marc O. Anderson, Joseph L. DeRisi, John Sherrill, Ally P. Liou, Peter B. Madrid, and Jennifer L. Weisman
- Subjects
Erythrocytes ,Stereochemistry ,Clinical Biochemistry ,Plasmodium falciparum ,Drug Resistance ,Pharmaceutical Science ,Biochemistry ,Chemical synthesis ,Article ,chemistry.chemical_compound ,Antimalarials ,Chloroquine ,Diamine ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Biology ,Cells, Cultured ,chemistry.chemical_classification ,biology ,Aminoacridines ,Organic Chemistry ,biology.organism_classification ,Aminacrine ,chemistry ,Acridine ,Molecular Medicine ,Salicylic acid ,Tricyclic ,medicine.drug - Abstract
A parallel synthetic strategy to the 9-aminoacridine scaffold of the classical anti-malarial drug quinacrine (2) is presented. The method features a new route to 9-chloroacridines that utilizes triflates of salicylic acid derivatives, which are commercially available in a variety of substitution patterns. The route allows ready variation of the two diversity elements present in this class of molecules: the tricyclic aromatic heterocyclic core, and the disubstituted diamine sidechain. In this study, a library of 175 compounds was designed, although only 93 of the final products had purities acceptable for screening. Impurity was generally due to incomplete removal of 9-acridones (18), a degradation product of the 9-chloroacridine synthetic intermediates. The library was screened against two strains of Plasmodium falciparum, including a model of the drug-resistant parasite, and six novel compounds were found to have IC(50) values in the low nanomolar range.
- Published
- 2005
44. Parallel Synthesis and Antimalarial Screening of a 4-Aminoquinoline Library
- Author
-
Peter B. Madrid, Joseph L. DeRisi, R. Kiplin Guy, and Nathan Wilson
- Subjects
biology ,Chemistry ,Drug discovery ,Plasmodium falciparum ,Drug Resistance ,General Chemistry ,Drug resistance ,Computational biology ,Pharmacology ,biology.organism_classification ,medicine.disease ,Article ,Malaria ,chemistry.chemical_compound ,Antimalarials ,Causative organism ,4-Aminoquinoline ,parasitic diseases ,Drug Discovery ,medicine ,Humans ,Malaria, Falciparum - Abstract
Due to growing problems with drug resistance, there is an outstanding need for new, cost-effective drugs for the treatment of malaria. The 4-aminoquinolines have provided a number of useful antimalarials, and Plasmodium falciparum, the causative organism for the most deadly form of human malaria, is generally slow to develop resistance to these drugs. Therefore, diverse screening libraries of quinolines continue to be useful for antimalarial drug discovery. We report herein the development of an efficient method for producing libraries of 4-aminoquinolines variant in the side chain portion of the molecule. The effects of these substitutions were evaluated by screening this library for activity against P. falciparum, revealing four potent compounds active against drug-resistant strains.
- Published
- 2004
45. Benzoflavone activators of the cystic fibrosis transmembrane conductance regulator: towards a pharmacophore model for the nucleotide-binding domain
- Author
-
Alan S. Verkman, Wonken Choung, Luis J. V. Galietta, Kolbot By, Gideon O. Berger, R. Kiplin Guy, Hong Yang, Tonghui Ma, Michael H. Nantz, Mark J. Kurth, Mark F. Springsteel, Chao Quan, Anang A. Shelat, Christopher W. Dicus, Springsteel, Mark F, Galietta, Luis J V, Ma, Tonghui, By, Kolbot, Berger, Gideon O, Yang, Hong, Dicus, Christopher W, Choung, Wonken, Quan, Chao, Shelat, Anang A, Guy, R Kiplin, Verkman, A S, Kurth, Mark J, and Nantz, Michael H
- Subjects
Models, Molecular ,congenital, hereditary, and neonatal diseases and abnormalities ,Green Fluorescent Proteins ,Clinical Biochemistry ,Molecular Conformation ,Pharmaceutical Science ,Benzoflavone ,Cystic Fibrosis Transmembrane Conductance Regulator ,Biochemistry ,Flavones ,Green Fluorescent Protein ,chemistry.chemical_compound ,Structure-Activity Relationship ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Apigenin ,Molecular Biology ,Cells, Cultured ,Benzoflavones ,Flavonoids ,chemistry.chemical_classification ,Binding Sites ,biology ,Nucleotides ,Activator (genetics) ,Animal ,Organic Chemistry ,Binding Site ,Luminescent Protein ,Cystic fibrosis transmembrane conductance regulator ,Rats ,Luminescent Proteins ,chemistry ,Cyclic nucleotide-binding domain ,Chloride channel ,biology.protein ,Flavonoid ,Molecular Medicine ,Rat ,Pharmacophore ,Nucleotide ,Human - Abstract
Our previous screen of flavones and related heterocycles for the ability to activate the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel indicated that UCCF-029, a 7,8-benzoflavone, was a potent activator. In the present study, we describe the synthesis and evaluation, using cell-based assays, of a series of benzoflavone analogues to examine structure-activity relationships and to identify compounds having greater potency for activation of both wild type CFTR and a mutant CFTR (G551D-CFTR) that causes cystic fibrosis in some human subjects. Using UCCF-029 as a structural guide, a panel of 77 flavonoid analogues was prepared. Analysis of the panel in FRT cells indicated that benzannulation of the flavone A-ring at the 7,8-position greatly improved compound activity and potency for several flavonoids. Incorporation of a B-ring pyridyl nitrogen either at the 3- or 4-position also elevated CFTR activity, but the influence of this structural modification was not as uniform as the influence of benzannulation. The most potent new analogue, UCCF-339, activated wild-type CFTR with a K(d) of 1.7 microM, which is more active than the previous most potent flavonoid activator of CFTR, apigenin. Several compounds in the benzoflavone panel also activated G551D-CFTR, but none were as active as apigenin. Pharmacophore modeling suggests a common binding mode for the flavones and other known CFTR activators at one of the nucleotide-binding sites, allowing for the rational development of more potent flavone analogues.
- Published
- 2003
46. Targeting the BindingFunction 3 (BF3) Site of theHuman Androgen Receptor through Virtual Screening.
- Author
-
NathanA. Lack, Peter Axerio-Cilies, Peyman Tavassoli, FrankQ. Han, Ka Hong Chan, Clementine Feau, Eric LeBlanc, Emma Tomlinson Guns, R. Kiplin Guy, Paul S. Rennie, and Artem Cherkasov
- Published
- 2011
- Full Text
- View/download PDF
47. Optimization of Propafenone Analogues as Antimalarial Leads.
- Author
-
David J. Lowes, W. Armand Guiguemde, Michele C. Connelly, Fangyi Zhu, Martina S. Sigal, Julie A. Clark, Andrew S. Lemoff, Joseph L. Derisi, Emily B. Wilson, and R. Kiplin Guy
- Published
- 2011
- Full Text
- View/download PDF
48. Synthesis and Evaluation of 7-Substituted 4-Aminoquinoline Analogues for Antimalarial Activity.
- Author
-
Jong Yeon Hwang, Takashi Kawasuji, David J. Lowes, Julie A. Clark, Michele C. Connelly, Fangyi Zhu, W. Armand Guiguemde, Martina S. Sigal, Emily B. Wilson, Joseph L. DeRisi, and R. Kiplin Guy
- Published
- 2011
- Full Text
- View/download PDF
49. Optimization of a Non-Radioactive High-Throughput Assay for Decarboxylase Enzymes.
- Author
-
David C. Smithson, Anang A. Shelat, Jeffrey Baldwin, Margaret A. Phillips, and R. Kiplin Guy
- Subjects
MATHEMATICAL optimization ,HIGH throughput screening (Drug development) ,DECARBOXYLASES ,ENZYME-linked immunosorbent assay ,TARGETED drug delivery ,CHEMICAL reagents ,TRYPANOSOMA brucei ,ENZYME inhibitors - Abstract
Herein, we describe the optimization of a linked enzyme assay suitable for high-throughput screening of decarboxylases, a target family whose activity has historically been difficult to quantify. Our approach uses a commercially available bicarbonate detection reagent to measure decarboxylase activity. The assay is performed in a fully enclosed automated screening system under inert nitrogen atmosphere to minimize perturbation by exogenous CO2. Receiver operating characteristic (ROC) analysis following a pilot screen of a small library of ∼3,600 unique molecules for inhibitors ofTrypanosoma brucei ornithine decarboxylase quantitatively demonstrates that the assay has excellent discriminatory power (area under the curve 0.90 with 95% confidence interval between 0.82 and 0.97). [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
50. Structure-Guided Development of Selective TbcatB Inhibitors.
- Author
-
Jeremy P. Mallari, Anang A. Shelat, Aaron Kosinski, Conor R. Caffrey, Michele Connelly, Fangyi Zhu, James H. McKerrow, and R. Kiplin Guy
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.