1
|
Turner LD, Nielsen AL, Lin L, Campedelli AJ, Silvaggi NR, Chen JS, Wakefield AE, Allen KN, Janda KD. Use of Crystallography and Molecular Modeling for the Inhibition of the Botulinum Neurotoxin A Protease. ACS Med Chem Lett 2021; 12:1318-1324. [PMID: 34413962 DOI: 10.1021/acsmedchemlett.1c00325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/20/2021] [Indexed: 01/14/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are extremely toxic and have been deemed a Tier 1 potential bioterrorism agent. The most potent and persistent of the BoNTs is the "A" serotype, with strategies to counter its etiology focused on designing small-molecule inhibitors of its light chain (LC), a zinc-dependent metalloprotease. The successful structure-based drug design of inhibitors has been confounded as the LC is highly flexible with significant morphological changes occurring upon inhibitor binding. To achieve greater success, previous and new cocrystal structures were evaluated from the standpoint of inhibitor enantioselectivity and their effect on active-site morphology. Based upon these structural insights, we designed inhibitors that were predicted to take advantage of π-π stacking interactions present in a cryptic hydrophobic subpocket. Structure-activity relationships were defined, and X-ray crystal structures and docking models were examined to rationalize the observed potency differences between inhibitors.
Collapse
Affiliation(s)
- Lewis D. Turner
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Alexander L. Nielsen
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Lucy Lin
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Antonio J. Campedelli
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Nicholas R. Silvaggi
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Jason S. Chen
- Automated Synthesis Facility, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Amanda E. Wakefield
- Department of Biomedical Engineering and Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Karen N. Allen
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Kim D. Janda
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
2
|
Lin L, Olson ME, Sugane T, Turner LD, Tararina MA, Nielsen AL, Kurbanov EK, Pellett S, Johnson EA, Cohen SM, Allen KN, Janda KD. Catch and Anchor Approach To Combat Both Toxicity and Longevity of Botulinum Toxin A. J Med Chem 2020; 63:11100-11120. [PMID: 32886509 PMCID: PMC7581224 DOI: 10.1021/acs.jmedchem.0c01006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Botulinum neurotoxins have remarkable persistence (∼weeks to months in cells), outlasting the small-molecule inhibitors designed to target them. To address this disconnect, inhibitors bearing two pharmacophores-a zinc binding group and a Cys-reactive warhead-were designed to leverage both affinity and reactivity. A series of first-generation bifunctional inhibitors was achieved through structure-based inhibitor design. Through X-ray crystallography, engagement of both the catalytic Zn2+ and Cys165 was confirmed. A second-generation series improved on affinity by incorporating known reversible inhibitor pharmacophores; the mechanism was confirmed by exhaustive dialysis, mass spectrometry, and in vitro evaluation against the C165S mutant. Finally, a third-generation inhibitor was shown to have good cellular activity and low toxicity. In addition to our findings, an alternative method of modeling time-dependent inhibition that simplifies assay setup and allows comparison of inhibition models is discussed.
Collapse
Affiliation(s)
- Lucy Lin
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), Scripps Research, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Margaret E. Olson
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), Scripps Research, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Takashi Sugane
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), Scripps Research, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Lewis D. Turner
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), Scripps Research, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Margarita A. Tararina
- Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, Massachusetts 02118, United States
| | - Alexander L. Nielsen
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), Scripps Research, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Elbek K. Kurbanov
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, Wisconsin 53706, United States
| | - Eric A. Johnson
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, Wisconsin 53706, United States
| | - Seth M. Cohen
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Karen N. Allen
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Kim D. Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), Scripps Research, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
3
|
Thompson JC, Dao WT, Ku A, Rodriguez-Beltran SL, Amezcua M, Palomino AY, Lien T, Salzameda NT. Synthesis and activity of isoleucine sulfonamide derivatives as novel botulinum neurotoxin serotype A light chain inhibitors. Bioorg Med Chem 2020; 28:115659. [PMID: 32828426 DOI: 10.1016/j.bmc.2020.115659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 11/20/2022]
Abstract
The botulinum neurotoxin (BoNT) is the most lethal protein known to man causing the deadly disease botulinum. The neurotoxin, composed of a heavy (HC) and light (LC) chain, work in concert to cause muscle paralysis. A therapeutic strategy to treat individuals infected with the neurotoxin is inhibiting the catalytic activity of the BoNT LC. We report the synthesis, inhibition study and computational docking analysis of novel small molecule BoNT/A LC inhibitors. A structure activity relationship study resulted in the discovery of d-isoleucine functionalized with a hydroxamic acid on the C-terminal and a biphenyl with chlorine at C- 2 connected by a sulfonamide linker at the N-terminus. This compound has a measured IC50 of 0.587 µM for the BoNT/A LC. Computational docking analysis indicates the sulfonamide linker adopts a geometry that is advantageous for binding to the BoNT LC active site. In addition, Arg363 is predicted to be involved in key binding interactions with the scaffold in this study.
Collapse
Affiliation(s)
- Jordan C Thompson
- Department of Chemistry & Biochemistry, California State University, 800 N. State College, Fullerton, CA, 92834, USA
| | - Wendy T Dao
- Department of Chemistry & Biochemistry, California State University, 800 N. State College, Fullerton, CA, 92834, USA
| | - Alex Ku
- Department of Chemistry & Biochemistry, California State University, 800 N. State College, Fullerton, CA, 92834, USA
| | - Sandra L Rodriguez-Beltran
- Department of Chemistry & Biochemistry, California State University, 800 N. State College, Fullerton, CA, 92834, USA
| | - Martin Amezcua
- Department of Chemistry & Biochemistry, California State University, 800 N. State College, Fullerton, CA, 92834, USA
| | - Alejandra Y Palomino
- Department of Chemistry & Biochemistry, California State University, 800 N. State College, Fullerton, CA, 92834, USA
| | - Thanh Lien
- Department of Chemistry & Biochemistry, California State University, 800 N. State College, Fullerton, CA, 92834, USA
| | - Nicholas T Salzameda
- Department of Chemistry & Biochemistry, California State University, 800 N. State College, Fullerton, CA, 92834, USA.
| |
Collapse
|
4
|
Lin L, Olson ME, Eubanks LM, Janda KD. Strategies to Counteract Botulinum Neurotoxin A: Nature's Deadliest Biomolecule. Acc Chem Res 2019; 52:2322-2331. [PMID: 31322847 DOI: 10.1021/acs.accounts.9b00261] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Botulinum neurotoxin serotype A (BoNT/A), marketed commercially as Botox, is the most toxic substance known to man with an estimated intravenous lethal dose (LD50) of 1-2 ng/kg in humans. Despite its widespread use in cosmetic and medicinal applications, no postexposure therapeutics are available for the reversal of intoxication in the event of medical malpractice or bioterrorism. Accordingly, the Centers for Disease Control and Prevention categorizes BoNT/A as a Category A pathogen, posing the highest risk to national security and public health as a result of the ease with which BoNT/A can be weaponized and disseminated. BoNT/A-mediated lethality results from neurons impeded from releasing acetylcholine, which ultimately causes muscle paralysis and possible death by asphyxiation with the loss of diaphragm function. Currently, the only available respite for BoNT/A poisoning is antibody-based therapy; however, this intervention is only effective within 12-24 h postexposure. Small molecule therapeutics remain the only opportunity to reverse BoNT/A intoxication after neuronal poisoning and are urgently needed. Nevertheless, no small molecule BoNT/A inhibitors have reached the clinic or even advanced to clinical trials. This Account highlights the accomplishments and existing challenges facing BoNT/A drug discovery today. Using the comprehensive body of work from our laboratory, we illustrate our nearly two-decade endeavor to discover a clinically relevant BoNT/A inhibitor. Specifically, a discussion on the identification and characterization of new chemical leads, the development of in vitro and in vivo assays, and pertinent discoveries in BoNT/A structural biology related to small molecule inhibition is presented. Lead discovery efforts in our laboratory have leveraged both in vitro high-throughput screening and rational design, and an array of mechanistic strategies for inhibiting BoNT/A has been discovered, including noncovalent inhibition, metal-binding active site inhibition, covalent inhibition, and α- and β-exosite inhibition. We contrast the strengths and weaknesses of each of these mechanistic strategies and propose the most favorable approach for success. Finally, we discuss multiple serendipitous discoveries of antibotulism small molecules with alternative mechanisms of action. Remaining challenges facing clinically relevant BoNT/A inhibition are presented and analyzed, including the current inability to reconcile toxin half-life (months to greater than one year) in neurons with in vivo pharmaceutical lifetimes and reoccurring inconsistencies between in vitro, cellular, and in vivo translation. Our Account of BoNT/A chemical research emphasizes the present accomplishments and critically analyzes the remaining obstacles for drug discovery. Importantly, we call for an increased focus on the discovery of safe and effective covalent inhibitors of BoNT/A that compete with the inherent half-life of the toxin.
Collapse
Affiliation(s)
- Lucy Lin
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The WIRM Institute for Research & Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Margaret E. Olson
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The WIRM Institute for Research & Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Lisa M. Eubanks
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The WIRM Institute for Research & Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Kim D. Janda
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The WIRM Institute for Research & Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
5
|
Chen AY, Adamek RN, Dick BL, Credille CV, Morrison CN, Cohen SM. Targeting Metalloenzymes for Therapeutic Intervention. Chem Rev 2019; 119:1323-1455. [PMID: 30192523 PMCID: PMC6405328 DOI: 10.1021/acs.chemrev.8b00201] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metalloenzymes are central to a wide range of essential biological activities, including nucleic acid modification, protein degradation, and many others. The role of metalloenzymes in these processes also makes them central for the progression of many diseases and, as such, makes metalloenzymes attractive targets for therapeutic intervention. Increasing awareness of the role metalloenzymes play in disease and their importance as a class of targets has amplified interest in the development of new strategies to develop inhibitors and ultimately useful drugs. In this Review, we provide a broad overview of several drug discovery efforts focused on metalloenzymes and attempt to map out the current landscape of high-value metalloenzyme targets.
Collapse
Affiliation(s)
- Allie Y Chen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Rebecca N Adamek
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Benjamin L Dick
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Cy V Credille
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Christine N Morrison
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Seth M Cohen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| |
Collapse
|
6
|
Alam MA. Methods for Hydroxamic Acid Synthesis. CURR ORG CHEM 2019; 23:978-993. [PMID: 32565717 PMCID: PMC7304568 DOI: 10.2174/1385272823666190424142821] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/20/2019] [Accepted: 03/28/2019] [Indexed: 12/26/2022]
Abstract
Substituted hydroxamic acid is one of the most extensively studied pharmacophores because of their ability to chelate biologically important metal ions to modulate various enzymes, such as HDACs, urease, metallopeptidase, and carbonic anhydrase. Syntheses and biological studies of various classes of hydroxamic acid derivatives have been reported in numerous research articles in recent years but this is the first review article dedicated to their synthetic methods and their application for the synthesis of these novel molecules. In this review article, commercially available reagents and preparation of hydroxylamine donating reagents have also been described.
Collapse
Affiliation(s)
- Mohammad A. Alam
- Department of Chemistry and Physics, College of Science and Mathematics, Arkansas State University, Jonesboro, AR 72467, USA
| |
Collapse
|
7
|
Seki H, Xue S, Hixon MS, Pellett S, Remes M, Johnson EA, Janda KD. Toward the discovery of dual inhibitors for botulinum neurotoxin A: concomitant targeting of endocytosis and light chain protease activity. Chem Commun (Camb) 2015; 51:6226-9. [PMID: 25759983 DOI: 10.1039/c5cc00677e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dyngo-4a™ has been found to be an endocytic inhibitor of BoNT/A neurotoxicity through dynamin inhibition. Herein, we demonstrate this molecule to have a previously unrecognized dual activity against BoNT/A, dynamin-protease inhibition. To establish the importance of this dual activity, detailed kinetic analysis of Dyngo-4a's inhibition of BoNT/A metalloprotease as well as cellular and animal toxicity studies have been described. The research presented is the first polypharmacological approach to counteract BoNT/A intoxication.
Collapse
Affiliation(s)
- Hajime Seki
- Departments of Chemistry and Immunology and Microbial Sciences, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | |
Collapse
|
8
|
Burtea A, Salzameda NT. Discovery and SAR study of a sulfonamide hydroxamic acid inhibitor for the botulinum neurotoxin serotype A light chain. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00053f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Through the use of chemical synthesis and high throughput screening, we discovered a sulfonamide hydroxamic acid inhibitor for the botulinum neurotoxin serotype A light chain. A structure activity relationship study of the parent inhibitor resulted in the synthesis of a new inhibitor with an IC50of 0.95 ± 0.60 μM for the BoNT/A LC.
Collapse
Affiliation(s)
- Alexander Burtea
- Department of Chemistry & Biochemistry
- California State University
- Fullerton, USA
| | | |
Collapse
|
9
|
Seki H, Pellett S, Silhár P, Stowe GN, Blanco B, Lardy MA, Johnson EA, Janda KD. Synthesis/biological evaluation of hydroxamic acids and their prodrugs as inhibitors for Botulinum neurotoxin A light chain. Bioorg Med Chem 2013; 22:1208-17. [PMID: 24360826 DOI: 10.1016/j.bmc.2013.11.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/28/2013] [Indexed: 01/03/2023]
Abstract
Botulinum neurotoxin A (BoNT/A) is the most potent toxin known. Unfortunately, it is also a potential bioweapon in terrorism, which is without an approved therapeutic treatment once cellular intoxication takes place. Previously, we reported how hydroxamic acid prodrug carbamates increased cellular uptake, which translated to successful inhibition of this neurotoxin. Building upon this research, we detail BoNT/A protease molecular modeling studies accompanied by the construction of small library of hydroxamic acids based on 2,4-dichlorocinnamic hydroxamic acid scaffold and their carbamate prodrug derivatization along with the evaluation of these molecules in both enzymatic and cellular models.
Collapse
Affiliation(s)
- Hajime Seki
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, WI 53706, United States
| | - Peter Silhár
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - G Neil Stowe
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Beatriz Blanco
- Centro Singular de Investigación en Química Biológica y Materiales Moleculares (CIQUS), Universidad de Santiago de Compostela, calle Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| | - Matthew A Lardy
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Eric A Johnson
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, WI 53706, United States
| | - Kim D Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States; Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States.
| |
Collapse
|
10
|
Silhár P, Eubanks LM, Seki H, Pellett S, Javor S, Tepp WH, Johnson EA, Janda KD. Targeting botulinum A cellular toxicity: a prodrug approach. J Med Chem 2013; 56:7870-9. [PMID: 24127873 DOI: 10.1021/jm400873n] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The botulinum neurotoxin light chain (LC) protease has become an important therapeutic target for postexposure treatment of botulism. Hydroxamic acid based small molecules have proven to be potent inhibitors of LC/A with nanomolar Ki values, yet they lack cellular activity conceivably due to low membrane permeability. To overcome this potential liability, we investigated two prodrug strategies, 1,4,2-dioxazole and carbamate, based on our 1-adamantylacetohydroxamic acid scaffold. The 1,4,2-dioxazole prodrug did not demonstrate cellular activity, however, carbamates exhibited cellular potency with the most active compound displaying an EC50 value of 20 μM. Cellular trafficking studies were conducted using a "fluorescently silent" prodrug that remained in this state until cellular uptake was complete, which allowed for visualization of the drug's release inside neuronal cells. In sum, this research sets the stage for future studies leveraging the specific targeting and delivery of these prodrugs, as well as other antibotulinum agents, into neuronal cells.
Collapse
Affiliation(s)
- Peter Silhár
- Departments of Chemistry and Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, and The Worm Institute for Research and Medicine, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Yang HJ, Xiong FJ, Chen XF, Chen FE. Highly Enantioselective Thiolysis of Prochiral Cyclic Anhydrides Catalyzed by Amino Alcohol Bifunctional Organocatalysts and Its Application to the Synthesis of Pregabalin. European J Org Chem 2013. [DOI: 10.1002/ejoc.201300464] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
12
|
O'Malley S, Sareth S, Jiao GS, Kim S, Thai A, Cregar-Hernandez L, McKasson L, Margosiak SA, Johnson AT. Virtual medicinal chemistry: in silico pre-docking functional group transformation for discovery of novel inhibitors of botulinum toxin serotype A light chain. Bioorg Med Chem Lett 2013; 23:2505-11. [PMID: 23545109 DOI: 10.1016/j.bmcl.2013.03.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/01/2013] [Accepted: 03/07/2013] [Indexed: 10/27/2022]
Abstract
A novel method for applying high-throughput docking to challenging metalloenzyme targets is described. The method utilizes information-based virtual transformation of library carboxylates to hydroxamic acids prior to docking, followed by compound acquisition, one-pot (two steps) chemical synthesis and in vitro screening. In two experiments targeting the botulinum neurotoxin serotype A metalloprotease light chain, hit rates of 32% and 18% were observed.
Collapse
|
13
|
Šilhár P, Silvaggi NR, Pellett S, Čapková K, Johnson EA, Allen KN, Janda KD. Evaluation of adamantane hydroxamates as botulinum neurotoxin inhibitors: synthesis, crystallography, modeling, kinetic and cellular based studies. Bioorg Med Chem 2012; 21:1344-8. [PMID: 23340139 DOI: 10.1016/j.bmc.2012.12.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 12/01/2012] [Accepted: 12/03/2012] [Indexed: 10/27/2022]
Abstract
Botulinum neurotoxins (BoNTs) are the most lethal biotoxins known to mankind and are responsible for the neuroparalytic disease botulism. Current treatments for botulinum poisoning are all protein based and thus have a limited window of treatment opportunity. Inhibition of the BoNT light chain protease (LC) has emerged as a therapeutic strategy for the treatment of botulism as it may provide an effective post exposure remedy. Using a combination of crystallographic and modeling studies a series of hydroxamates derived from 1-adamantylacetohydroxamic acid (3a) were prepared. From this group of compounds, an improved potency of about 17-fold was observed for two derivatives. Detailed mechanistic studies on these structures revealed a competitive inhibition model, with a K(i)=27 nM, which makes these compounds some of the most potent small molecule, non-peptidic BoNT/A LC inhibitors reported to date.
Collapse
Affiliation(s)
- Peter Šilhár
- Department of Chemistry, and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Garner AL, Struss AK, Fullagar JL, Agrawal A, Moreno AY, Cohen SM, Janda KD. 3-Hydroxy-1-alkyl-2-methylpyridine-4(1H)-thiones: Inhibition of the Pseudomonas aeruginosa Virulence Factor LasB. ACS Med Chem Lett 2012. [PMID: 23181168 DOI: 10.1021/ml300128f] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Bacterial resistance coupled to our current arsenal of antibiotics presents us with a growing threat to public health, thus warranting the exploration of alternative antibacterial strategies. In particular, the targeting of virulence factors has been regarded as a "second generation" antibiotic approach. In Pseudomonas aeruginosa, a Zn(2+) metalloprotease virulence factor, LasB or P. aeruginosa elastase, has been implicated in the development of P. aeruginosa-related keratitis, pneumonia and burn infection. Moreover, the enzyme also plays a critical role in swarming and biofilm formation, both of which are processes that have been linked to antibiotic resistance. To further validate the importance of LasB in P. aeruginosa infection, we describe our efforts toward the discovery of non-peptidic small molecule inhibitors of LasB. Using identified compounds, we have confirmed the role that LasB plays in P. aeruginosa swarming and demonstrate the potential for LasB-targeted small molecules in studying antimicrobial resistant P. aeruginosa phenotypes.
Collapse
Affiliation(s)
- Amanda L. Garner
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Anjali K. Struss
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Jessica L. Fullagar
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Arpita Agrawal
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Amira Y. Moreno
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Seth M. Cohen
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Kim D. Janda
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| |
Collapse
|
15
|
Čapek P, Zhang Y, Barlow DJ, Houseknecht KL, Smith GR, Dickerson TJ. Enhancing the Pharmacokinetic Properties of Botulinum Neurotoxin Serotype A Protease Inhibitors Through Rational Design. ACS Chem Neurosci 2011; 2:288-293. [PMID: 21743830 DOI: 10.1021/cn200021q] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Botulinum neurotoxin (BoNT), the etiological agent that causes the neuroparalytic disease botulism, has become a highly studied drug target in light of the potential abuse of this toxin as a weapon of bioterrorism. In particular, small molecule inhibitors of the light chain metalloprotease of BoNT serotype A have received significant attention and a number of small molecule and biologic inhibitors have been reported. However, all small molecules reported have been identified from either primary screens or medicinal chemistry follow-up studies, and the pharmacokinetic profiles of these compounds have not been addressed. In this study, we have removed the pharmacologic liabilities of one of the best compounds reported to date, 2,4-dichlorocinnamate hydroxamic acid, and in the process, uncovered a related class of benzothiophene hydroxamic acids that are significantly more potent inhibitors of the BoNT/A light chain, while also possessing greatly improved ADME properties, with the best compound showing the most potent inhibition of BoNT/A light chain reported (K(i) = 77 nM). Using a strategy of incorporating traditional drug development filters early into the discovery process, potential liabilities in BoNT/A lead compounds have been illuminated and removed, clearing the path for advancement into further pharmacologic optimization and in vivo efficacy testing.
Collapse
Affiliation(s)
- Petr Čapek
- Department of Chemistry and Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Yan Zhang
- Fox Chase Chemical Diversity Center, Doylestown, Pennsylvania, United States
| | - Deborah J. Barlow
- Department of Pharmaceutical Sciences, University of New England College of Pharmacy, Portland, Maine, United States
| | - Karen L. Houseknecht
- Department of Pharmaceutical Sciences, University of New England College of Pharmacy, Portland, Maine, United States
| | - Garry R. Smith
- Fox Chase Chemical Diversity Center, Doylestown, Pennsylvania, United States
| | - Tobin J. Dickerson
- Department of Chemistry and Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
16
|
Thompson AA, Jiao GS, Kim S, Thai A, Cregar-Hernandez L, Margosiak SA, Johnson AT, Han GW, O’Malley S, Stevens RC. Structural characterization of three novel hydroxamate-based zinc chelating inhibitors of the Clostridium botulinum serotype A neurotoxin light chain metalloprotease reveals a compact binding site resulting from 60/70 loop flexibility. Biochemistry 2011; 50:4019-28. [PMID: 21434688 PMCID: PMC3092028 DOI: 10.1021/bi2001483] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neurotoxins synthesized by Clostridium botulinum bacteria (BoNT), the etiological agent of human botulism, are extremely toxic proteins making them high-risk agents for bioterrorism. Small molecule inhibitor development has been focused on the light chain zinc-dependent metalloprotease domain of the neurotoxin, an effort that has been hampered by its relatively flexible active site. Developed in concert with structure--activity relationship studies, the X-ray crystal structures of the complex of BoNT serotype A light chain (BoNT/A LC) with three different micromolar-potency hydroxamate-based inhibitors are reported here. Comparison with an unliganded BoNT/A LC structure reveals significant changes in the active site as a result of binding by the unique inhibitor scaffolds. The 60/70 loop at the opening of the active site pocket undergoes the largest conformational change, presumably through an induced-fit mechanism, resulting in the most compact catalytic pocket observed in all known BoNT/A LC structures.
Collapse
Affiliation(s)
- Aaron A. Thompson
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Guan-Sheng Jiao
- Department of Chemistry, PanThera Biopharma LLC, Aiea, HI 96701
| | - Seongjin Kim
- Department of Chemistry, PanThera Biopharma LLC, Aiea, HI 96701
| | - April Thai
- Department of Chemistry, PanThera Biopharma LLC, Aiea, HI 96701
| | | | | | - Alan T. Johnson
- Department of Chemistry, PanThera Biopharma LLC, Aiea, HI 96701
| | - Gye Won Han
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Sean O’Malley
- Department of Chemistry, PanThera Biopharma LLC, Aiea, HI 96701
| | - Raymond C. Stevens
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037,Address correspondence to: The Scripps Research Institute, 10550 North Torrey Pines Rd., GAC-1200, La Jolla, CA 92037. Tel.: 858-784-9416; Fax: 858-784-9483;
| |
Collapse
|
17
|
García-Urdiales E, Alfonso I, Gotor V. Update 1 of: Enantioselective Enzymatic Desymmetrizations in Organic Synthesis. Chem Rev 2011; 111:PR110-80. [DOI: 10.1021/cr100330u] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Eduardo García-Urdiales
- Departamento de Química
Orgánica e Inorgánica, Facultad de Química, Universidad
de Oviedo, Julián Clavería, 8, 33006 Oviedo, Spain,
and
| | - Ignacio Alfonso
- Departamento de Química Biológica
y Modelización Molecular, Instituto de Química Avanzada
de Cataluña (IQAC, CSIC), Jordi Girona, 18-26, 08034, Barcelona,
Spain
| | - Vicente Gotor
- Departamento de Química
Orgánica e Inorgánica, Facultad de Química, Universidad
de Oviedo, Julián Clavería, 8, 33006 Oviedo, Spain,
and
| |
Collapse
|
18
|
Cardellina JH, Vieira RC, Eccard V, Skerry J, Montgomery V, Campbell Y, Roxas-Duncan V, Leister W, LeClair CA, Maloney DJ, Padula D, Pescitelli G, Khavrutskii I, Hu X, Wallqvist A, Smith LA. Separation of Betti Reaction Product Enantiomers: Absolute Configuration and Inhibition of Botulinum Neurotoxin A. ACS Med Chem Lett 2011; 2:396-401. [PMID: 22102940 DOI: 10.1021/ml200028z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The racemic product of the Betti reaction of 5-chloro-8-hydroxyquinoline, benzaldehyde and 2-aminopyridine was separated by chiral HPLC to determine which enantiomer inhibited botulinum neurotoxin serotype A. When the enantiomers unexpectedly proved to have comparable activity, the absolute structures of (+)-(R)-1 and (-)-(S)-1 were determined by comparison of calculated and observed circular dichroism spectra. Molecular modeling studies were undertaken in an effort to understand the observed bioactivity and revealed different ensembles of binding modes, with roughly equal binding energies, for the two enantiomers.
Collapse
Affiliation(s)
- John H. Cardellina
- Division of Integrated Toxicology, U.S. Army Medical Research Institute for Infectious Diseases, Frederick, Maryland, United States
| | - Rebecca C. Vieira
- Division of Integrated Toxicology, U.S. Army Medical Research Institute for Infectious Diseases, Frederick, Maryland, United States
| | - Vanessa Eccard
- Division of Integrated Toxicology, U.S. Army Medical Research Institute for Infectious Diseases, Frederick, Maryland, United States
| | - Janet Skerry
- Division of Integrated Toxicology, U.S. Army Medical Research Institute for Infectious Diseases, Frederick, Maryland, United States
| | - Vicki Montgomery
- Division of Integrated Toxicology, U.S. Army Medical Research Institute for Infectious Diseases, Frederick, Maryland, United States
| | - Yvette Campbell
- Division of Integrated Toxicology, U.S. Army Medical Research Institute for Infectious Diseases, Frederick, Maryland, United States
| | - Virginia Roxas-Duncan
- Division of Integrated Toxicology, U.S. Army Medical Research Institute for Infectious Diseases, Frederick, Maryland, United States
| | - William Leister
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370, Bethesda, Maryland, United States
| | - Christopher A. LeClair
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370, Bethesda, Maryland, United States
| | - David J. Maloney
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH, 9800 Medical Center Drive, MSC 3370, Bethesda, Maryland, United States
| | - Daniele Padula
- Department of Chemistry, University of Pisa, Pisa, Italy
| | | | - Ilja Khavrutskii
- Biotechnology High Performance Computer Software Application Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Frederick, Maryland, United States
| | - Xin Hu
- Biotechnology High Performance Computer Software Application Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Frederick, Maryland, United States
| | - Anders Wallqvist
- Biotechnology High Performance Computer Software Application Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Frederick, Maryland, United States
| | - Leonard A. Smith
- Office of Chief Scientist, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States
| |
Collapse
|
19
|
Šilhár P, Alakurtti S, Čapková K, Xiaochuan F, Shoemaker CB, Yli-Kauhaluoma J, Janda KD. Synthesis and evaluation of library of betulin derivatives against the botulinum neurotoxin A protease. Bioorg Med Chem Lett 2011; 21:2229-31. [PMID: 21421315 DOI: 10.1016/j.bmcl.2011.02.115] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 02/25/2011] [Accepted: 02/28/2011] [Indexed: 10/18/2022]
Abstract
Botulinum neurotoxins (BoNTs) are the most toxic proteins currently known. Current treatments for botulinum poisoning are all protein based with a limited window of opportunity. Inhibition of the BoNT light chain protease (LC) has emerged as a new therapeutic strategy for the treatment of botulism as it may provide an effective post-exposure remedy. As such, a small library of 40 betulin derivatives was synthesized and screened against the light chain of BoNT serotype A (LC/A); five positive hits (IC(50) <100 μM) were uncovered. Detailed evaluation of inhibition mechanism of three most active compounds revealed a competitive model, with sub-micromolar K(i) value for the best inhibitor (7). Unfortunately, an in vitro cell-based assay did not show any protection of rat cerebellar neurons against BoNT/A intoxication by 7.
Collapse
Affiliation(s)
- Peter Šilhár
- Department of Chemistry, Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Salzameda NT, Eubanks LM, Zakhari JS, Tsuchikama K, DeNunzio NJ, Allen KN, Hixon MS, Janda KD. A cross-over inhibitor of the botulinum neurotoxin light chain B: a natural product implicating an exosite mechanism of action. Chem Commun (Camb) 2011; 47:1713-5. [PMID: 21203627 DOI: 10.1039/c0cc04078a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Clostridium botulinum produces the most lethal toxins known to man, as such they are high risk terrorist threats, and alarmingly there is no approved therapeutic. We report the first cross-over small molecule inhibitor of these neurotoxins and propose a mechanism by which it may impart its inhibitory activity.
Collapse
Affiliation(s)
- Nicholas T Salzameda
- Department of Chemistry, and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Li B, Peet NP, Butler MM, Burnett JC, Moir DT, Bowlin TL. Small molecule inhibitors as countermeasures for botulinum neurotoxin intoxication. Molecules 2010; 16:202-20. [PMID: 21193845 PMCID: PMC6259422 DOI: 10.3390/molecules16010202] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 12/20/2010] [Accepted: 12/29/2010] [Indexed: 11/18/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are the most potent of known toxins and are listed as category A biothreat agents by the U.S. CDC. The BoNT-mediated proteolysis of SNARE proteins inhibits the exocytosis of acetylcholine into neuromuscular junctions, leading to life-threatening flaccid paralysis. Currently, the only therapy for BoNT intoxication (which results in the disease state botulism) includes experimental preventative antibodies and long-term supportive care. Therefore, there is an urgent need to identify and develop inhibitors that will serve as both prophylactic agents and post-exposure ‘rescue’ therapeutics. This review focuses on recent progress to discover and develop small molecule inhibitors as therapeutic countermeasures for BoNT intoxication.
Collapse
Affiliation(s)
- Bing Li
- Microbiotix, Inc., One Innovation Drive, Worcester, MA 01605, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-508-757-2800; Fax: +1-508-757-1999
| | - Norton P. Peet
- Microbiotix, Inc., One Innovation Drive, Worcester, MA 01605, USA
| | | | - James C. Burnett
- Target, Structure-Based Drug Discovery Group, SAIC-Frederick, Inc., National Cancer Institute at Frederick, 1050 Boyles Street, Frederick, MD 21702, USA; E-Mail: (J.C.B.)
| | - Donald T. Moir
- Microbiotix, Inc., One Innovation Drive, Worcester, MA 01605, USA
| | - Terry L. Bowlin
- Microbiotix, Inc., One Innovation Drive, Worcester, MA 01605, USA
| |
Collapse
|
22
|
Eubanks LM, Šilhár P, Salzameda NT, Zakhari JS, Xiaochuan F, Barbieri JT, Shoemaker CB, Hixon MS, Janda KD. Identification of a Natural Product Antagonist against the Botulinum Neurotoxin Light Chain Protease. ACS Med Chem Lett 2010; 1:268-272. [PMID: 20959871 DOI: 10.1021/ml100074s] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are the etiological agents responsible for botulism, a disease characterized by peripheral neuromuscular blockade and a characteristic flaccid paralysis of humans. BoNTs are the most lethal known poisons affecting humans and has been recognized as a potential bioterrorist threat. Current treatments for botulinum poisoning are predominately prophylactic in nature relying on passive immunization with antitoxins. Inhibition of the BoNT light chain metalloprotease (LC) has emerged as a new therapeutic strategy for the treatment of botulism that may provide an effective post-exposure remedy. A high-throughput screening effort against the light chain of BoNT serotype A (LC/A) was conducted with the John Hopkins Clinical Compound Library comprised of over 1,500 existing drugs. Lomofungin, a natural product first isolated in the late 1960's, was identified as an inhibitor of LC/A, displaying classical noncompetitive inhibition kinetics with a K(i) of 6.7 ± 0.7 µM. Inhibitor combination studies reveal that lomofungin binding is nonmutually exclusive (synergistic). The inhibition profile of lomofungin has been delineated by the use of both an active site inhibitor, 2,4-dichlorocinnamic hydroxamate, and a noncompetitive inhibitor d-chicoric acid; the mechanistic implications of these observations are discussed. Lastly, cellular efficacy was investigated using a rat primary cell model which demonstrated that lomofungin can protect against SNAP-25 cleavage, the intracellular protein target of LC/A.
Collapse
Affiliation(s)
- Lisa M. Eubanks
- Departments of Chemistry and Immunology
- The Skaggs Institute for Chemical Biology
- The Worm Institute for Research and Medicine
| | - Peter Šilhár
- Departments of Chemistry and Immunology
- The Skaggs Institute for Chemical Biology
| | | | - Joseph S. Zakhari
- Departments of Chemistry and Immunology
- The Skaggs Institute for Chemical Biology
| | - Feng Xiaochuan
- Department of Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, 200 Westboro Road, North Grafton, Massachusetts 01536
| | - Joseph T. Barbieri
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226
| | - Charles B. Shoemaker
- Department of Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, 200 Westboro Road, North Grafton, Massachusetts 01536
| | - Mark S. Hixon
- Departments of Chemistry and Immunology
- The Skaggs Institute for Chemical Biology
- The Worm Institute for Research and Medicine
| | - Kim D. Janda
- Departments of Chemistry and Immunology
- The Skaggs Institute for Chemical Biology
- The Worm Institute for Research and Medicine
| |
Collapse
|