1
|
Dillenburg M, Peterson CD, Dolot R, Ligori K, Kitto KF, Wilcox GL, Fairbanks CA, Wagner CR. HINT1 Inhibitors as Selective Modulators of MOR-NMDAR Cross-Regulation and Non-Opioid Analgesia. ACS Chem Neurosci 2025; 16:604-618. [PMID: 39913175 DOI: 10.1021/acschemneuro.4c00564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2025] Open
Abstract
Human histidine triad nucleotide-binding protein 1 (HINT1) has recently become a protein of interest due to its involvement in several CNS processes, including neuroplasticity and the development of several neuropsychiatric disorders. Crucially, HINT1 behaves as a mediator for cross-regulation of the mu-opioid receptor (MOR) and N-methyl-d-aspartate receptor (NMDAR). Active site inhibition of HINT1 using small-molecule inhibitors has been demonstrated to have a significant impact on this cross-regulatory relationship in vivo. Herein, we describe the development of a series of ethenoadenosine HINT1 inhibitors to further evaluate the effect of HINT1 inhibition on morphine's blockade of NMDA-evoked behaviors, the development of acute endomorphin-2 tolerance, and analgesia. X-ray crystallographic analysis and HINT1 binding experiments demonstrate that modifications to the inhibitor nucleobase greatly impact the inhibitor binding interactions with HINT1. Our results reveal a complex structure-activity relationship for HINT1 inhibitors, in which minor modifications to the ethenoadenosine scaffold resulted in dramatic changes to their activity in these assays modeling MOR-NMDAR interaction. Specifically, we observed the ability of HINT1 inhibitors to selectively affect individual pathways of MOR-NMDAR crosstalk. Furthermore, we observed that a carbamate ethenoadenosine inhibitor of HINT1 can induce analgesia while not affecting opioid tolerance. Additionally, although past studies have indicated that the loss of HINT1 expression can result in the downregulation of p53, we have shown that the inhibition of HINT1 has no effect on either the expression of HINT1 or p53. These studies highlight the critical role of HINT1 in MOR-NMDAR crosstalk and demonstrate the intriguing potential of using HINT1 active-site inhibitors as tools to probe its role in these biochemical pathways and its potential as a novel pain target.
Collapse
Affiliation(s)
- Maxwell Dillenburg
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Cristina D Peterson
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rafal Dolot
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| | - Kostana Ligori
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kelley F Kitto
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - George L Wilcox
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carolyn A Fairbanks
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carston R Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
2
|
Jiang L, Pan T, Lv Q, Yuan W, Liu X, Qu X, Luo D, Wan S, Cui S. Novel ProTide prodrugs of 5-fluoro-2'-deoxyuridine for the treatment of liver cancer. Eur J Med Chem 2023; 260:115763. [PMID: 37659196 DOI: 10.1016/j.ejmech.2023.115763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/09/2022] [Revised: 04/08/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2023]
Abstract
ProTide prodrug technology has emerged as a promising way for the development of anti-viral and anti-tumor drugs, whereas, there are fewer applications for the treatment of liver cancer. Herein, a series of distinct 3'-ester ProTide prodrugs of 5-fluoro-2'-deoxyuridine (FdUR) were synthesized and evaluated for their anti-liver cancer activity. The most efficient prodrug 11b reached a sub-micromolar activity (IC50 = 0.42 ± 0.13 μM) against HepG2 and over 100-fold and 200-fold improvements compared to 5-FU, respectively. 11b also demonstrated favorable selectivity towards normal liver cells L-02 (IC50 > 100 μM). In vitro metabolic stability studies revealed that 11b is stable in the plasma and could be activated rapidly in the liver, which supported that 11b is liver-targeted. Importantly, to more accurately evaluate the anti-HCC activity of 11b, the liver orthotopic model was built and 11b significantly suppressed tumor growth (TGI = 75.5%) at a dose of 60 mg/kg/2d in vivo without obvious toxicity. Overall, these promising results indicated that 11b could serve as a safe and effective prodrug of 5-FU nucleoside for liver cancer therapy.
Collapse
Affiliation(s)
- Leilei Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
| | - Ting Pan
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Qin Lv
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
| | - Wenmin Yuan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
| | - Xiaochun Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China
| | - Xianjun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dongdong Luo
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China.
| | - Shengbiao Wan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266071, China.
| | - Shuxiang Cui
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
3
|
Siekierska I, Lukaszewicz M, Worch R, Jankowska-Anyszka M, Piecyk K. Application of Phosphoramidate ProTide Technology for the Synthesis of 5'-mRNA Cap Analogs Modified on the Exocyclic Amine Group. ChemMedChem 2023; 18:e202200490. [PMID: 36658701 DOI: 10.1002/cmdc.202200490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/09/2022] [Revised: 12/18/2022] [Indexed: 01/21/2023]
Abstract
Aryloxy triester phosphoramidate methodology, commonly known as ProTide technology, is one of the most widely used prodrug approaches applied to therapeutic nucleosides. This approach has been used extensively by the pharmaceutical industry and researchers in medicinal chemistry. Herein we report our adaptation of this effective method for the synthesis of bioactive 5'-mRNA cap analogues as inhibitors for targeting cap-dependent translation. The synthesis was performed in two main stages: preparation of N2-modified guanosine analogues and their subsequent transformation into prodrugs using phenylethoxy-l-alaninyl phosphorochloridate. The prepared pro-nucleotide cap analogues were tested for their capacity in enzymatic activation, inhibitory properties in a rabbit reticulocyte lysate system, and passive membrane translocation properties.
Collapse
Affiliation(s)
- Izabela Siekierska
- Faculty of Chemistry, University of Warsaw, 1 Pasteur St., 02-093, Warsaw, Poland
| | - Maciej Lukaszewicz
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-089, Warsaw, Poland
| | - Remigiusz Worch
- Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093, Warsaw, Poland
| | | | - Karolina Piecyk
- Faculty of Chemistry, University of Warsaw, 1 Pasteur St., 02-093, Warsaw, Poland
| |
Collapse
|
4
|
Roy B, Navarro V, Peyrottes S. Prodrugs of Nucleoside 5'-Monophosphate Analogues: Overview of the Recent Literature Concerning their Synthesis and Applications. Curr Med Chem 2023; 30:1256-1303. [PMID: 36093825 DOI: 10.2174/0929867329666220909122820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/20/2022] [Revised: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 11/22/2022]
Abstract
Nucleoside analogues are widely used as anti-infectious and antitumoral agents. However, their clinical use may face limitations associated with their physicochemical properties, pharmacokinetic parameters, and/or their peculiar mechanisms of action. Indeed, once inside the cells, nucleoside analogues require to be metabolized into their corresponding (poly-)phosphorylated derivatives, mediated by cellular and/or viral kinases, in order to interfere with nucleic acid biosynthesis. Within this activation process, the first-phosphorylation step is often the limiting one and to overcome this limitation, numerous prodrug approaches have been proposed. Herein, we will focus on recent literature data (from 2015 and onwards) related to new prodrug strategies, the development of original synthetic approaches and novel applications of nucleotide prodrugs (namely pronucleotides) leading to the intracellular delivery of 5'-monophosphate nucleoside analogues.
Collapse
Affiliation(s)
- Béatrice Roy
- Team Nucleosides & Phosphorylated Effectors, Institute for Biomolecules Max Mousseron (IBMM), University of Montpellier, Route de Mende, 34293 Montpellier, France
| | - Valentin Navarro
- Team Nucleosides & Phosphorylated Effectors, Institute for Biomolecules Max Mousseron (IBMM), University of Montpellier, Route de Mende, 34293 Montpellier, France
| | - Suzanne Peyrottes
- Team Nucleosides & Phosphorylated Effectors, Institute for Biomolecules Max Mousseron (IBMM), University of Montpellier, Route de Mende, 34293 Montpellier, France
| |
Collapse
|
5
|
Jacobson BA, Ahmad Z, Chen S, Waldusky G, Dillenburg M, Stoian E, Cambron DA, Patel AJ, Patel MR, Wagner CR, Kratzke RA. 4Ei-10 interdiction of oncogenic cap-mediated translation as therapy for non-small cell lung cancer. Invest New Drugs 2021; 39:636-643. [PMID: 33230623 DOI: 10.1007/s10637-020-01036-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/06/2020] [Accepted: 11/18/2020] [Indexed: 11/28/2022]
Abstract
In order to suppress 5' cap-mediated translation a highly available inhibitor of the interaction between the 5' mRNA cap and the eIF4E complex has been developed. 4Ei-10 is a member of the class of ProTide compounds and has elevated membrane permeability and is a strong active chemical antagonist for eIF4E. Once taken up by cells it is converted by anchimeric activation of the lipophilic 2-(methylthio) ethyl protecting group and after that Hint1 P-N bond cleavage to N7-(p-chlorophenoxyethyl) guanosine 5'-monophosphate (7-Cl-Ph-Ethyl-GMP). Using this powerful interaction, it has been demonstrated that 4Ei-10 inhibits non-small cell lung cancer (NSCLC) cell growth. In addition, treatment of NSCLC cells with 4Ei-10 results in suppression of translation and diminished expression of a cohort of cellular proteins important to maintaining the malignant phenotype and resisting apoptosis such as Bcl-2, survivin, and ornithine decarboxylase (ODC). Finally, as a result of targeting the translation of anti-apoptotic proteins, NSCLC cells are synergized to be more sensitive to the existing anti-neoplastic treatment gemcitabine currently used in NSCLC therapy.
Collapse
Affiliation(s)
- Blake A Jacobson
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Zeeshan Ahmad
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Maxwell Dillenburg
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Anil J Patel
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Manish R Patel
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Carston R Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Robert A Kratzke
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
- Division of Heme-Onc-Transplant, University of Minnesota Medical School, MMC 480, 420 Delaware St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
6
|
Strom A, Shah R, Wagner CR. "Switching On" Enzyme Substrate Specificity Analysis with a Fluorescent Competitive Inhibitor. Biochemistry 2021; 60:440-450. [PMID: 33513008 DOI: 10.1021/acs.biochem.0c00954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Abstract
Enzymatically driven change to the spectroscopic properties of a chemical substrate or product has been a linchpin in the development of continuous enzyme kinetics assays. These assays inherently necessitate substrates or products that naturally comply with the constraints of the spectroscopic technique being used, or they require structural changes to the molecules involved to make them observable. Here we demonstrate a new analytical kinetics approach with enzyme histidine triad nucleotide binding protein 1 (HINT1) that allows us to extract both useful kcat values and a rank-ordered list of substrate specificities without the need to track substrates or products directly. Instead, this is accomplished indirectly using a "switch on" competitive inhibitor that fluoresces maximally only when bound to the HINT1 enzyme active site. Kinetic information is extracted from the duration of the diminished fluorescence when the monitorable inhibitor-bound enzyme is challenged with saturating concentrations of a nonfluorescent substrate. We refer to the loss of fluorescence, while the substrate competes for the fluorescent probe in the active site, as the substrate's residence transit time (RTT). The ability to assess kcat values and substrate specificity by monitoring the RTTs for a set of substrates with a competitive "switch on" inhibitor should be broadly applicable to other enzymatic reactions in which the "switch on" inhibitor has sufficient binding affinity over the enzymatic product.
Collapse
Affiliation(s)
- Alexander Strom
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rachit Shah
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carston R Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
7
|
Schwarz DC, Williams SK, Dillenburg M, Wagner CR, Gestwicki JE. A Phosphoramidate Strategy Enables Membrane Permeability of a Non-nucleotide Inhibitor of the Prolyl Isomerase Pin1. ACS Med Chem Lett 2020; 11:1704-1710. [PMID: 32944137 PMCID: PMC7488286 DOI: 10.1021/acsmedchemlett.0c00170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/05/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022] Open
Abstract
The membrane permeability of nucleotide-based drugs, such as sofosbuvir (Sovaldi), requires installation of phosphate-caging groups. One strategy, termed "ProTide", masks the anionic phosphate through an N-linked amino ester and an O-linked aromatic phospho-ester, such that release of the active drug requires consecutive enzymatic liberation by an esterase and then a phosphoramidase, such as Hint1. Because Hint1 is known to be selective for nucleotides, it was not clear if the ProTide approach could be deployed for non-nucleotides. Here, we demonstrate that caging of a phosphate-containing inhibitor of the prolyl isomerase Pin1 increases its permeability. Moreover, this compound was processed by both esterase and phosphoramidase activity, releasing the active molecule to bind and inhibit Pin1 in cells. Thus, Hint1 appears to recognize a broader set of substrates than previously appreciated. It seems possible that other potent, but impermeable, phosphate-containing inhibitors might likewise benefit from this approach.
Collapse
Affiliation(s)
- Daniel
M. C. Schwarz
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
| | - Sarah K. Williams
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
| | - Maxwell Dillenburg
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carston R. Wagner
- Department
of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jason E. Gestwicki
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
8
|
Itumoh EJ, Data S, Leitao EM. Opening up the Toolbox: Synthesis and Mechanisms of Phosphoramidates. Molecules 2020; 25:E3684. [PMID: 32823507 PMCID: PMC7463754 DOI: 10.3390/molecules25163684] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/03/2020] [Revised: 08/05/2020] [Accepted: 08/11/2020] [Indexed: 11/25/2022] Open
Abstract
This review covers the main synthetic routes to and the corresponding mechanisms of phosphoramidate formation. The synthetic routes can be separated into six categories: salt elimination, oxidative cross-coupling, azide, reduction, hydrophosphinylation, and phosphoramidate-aldehyde-dienophile (PAD). Examples of some important compounds synthesized through these routes are provided. As an important class of organophosphorus compounds, the applications of phosphoramidate compounds, are also briefly introduced.
Collapse
Affiliation(s)
- Emeka J. Itumoh
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand; (E.J.I.); (S.D.)
- Department of Industrial Chemistry, Ebonyi State University, Abakaliki 480001, Ebonyi State, Nigeria
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Shailja Data
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand; (E.J.I.); (S.D.)
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Erin M. Leitao
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand; (E.J.I.); (S.D.)
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| |
Collapse
|
9
|
Matos de Souza MR, Cunha MS, Okon A, Monteiro FLL, Campanati L, Wagner CR, da Costa LJ. In Vitro and In Vivo Characterization of the Anti-Zika Virus Activity of ProTides of 2'-C-β-Methylguanosine. ACS Infect Dis 2020; 6:1650-1658. [PMID: 32525653 DOI: 10.1021/acsinfecdis.0c00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Abstract
The ProTide approach has emerged as a powerful tool to improve the intracellular delivery of nucleotide analogs with antiviral and anticancer activity. Here, we characterized the anti-ZIKV (ZIKV, Zika virus) activity of two ProTides of 2'-C-β-methylguanosine. ProTide UMN-1001 is a 2'-C-β-methylguanosine tryptamine phosphoramidate monoester, and ProTide UMN-1002 is a 2-(methylthio)-ethyl-2'-C-β-methylguanosine tryptamine phosphoramidate diester. UMN-1002 undergoes stepwise intracellular activation to the corresponding nucleotide monophosphate followed by P-N bond cleavage by intracellular histidine triad nucleotide binding protein 1 (Hint1). UMN-1001 is activated by Hint1 but is less cell-permeable than UMN-1002. UMN-1001 and UMN-1002 were found to be more potent than 2'-C-β-methylguanosine against ZIKV in human-derived microvascular endothelial and neuroblastoma cells and in reducing ZIKV RNA replication. Studies with a newborn mouse model of ZIKV infection demonstrated that, while treatment with 2'-C-β-methylguanosine and UMN-1001 was lethal, treatment with UMN-1002 was nontoxic and significantly reduced ZIKV infection. Our data suggests that anchimeric activated ProTides of 2'-C-β-methyl nucleosides should be further investigated for their potential as anti-ZIKV therapeutics.
Collapse
Affiliation(s)
| | | | - Aniekan Okon
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | | | - Carston R. Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | |
Collapse
|
10
|
Strom A, Tong CL, Wagner CR. Histidine triad nucleotide-binding proteins HINT1 and HINT2 share similar substrate specificities and little affinity for the signaling dinucleotide Ap4A. FEBS Lett 2020; 594:1497-1505. [PMID: 31990367 DOI: 10.1002/1873-3468.13745] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/28/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 11/07/2022]
Abstract
Human histidine triad nucleotide-binding protein 2 (hHINT2) is an important player in human mitochondrial bioenergetics, but little is known about its catalytic capabilities or its nucleotide phosphoramidate prodrug (proTide)-activating activity akin to the cytosolic isozyme hHINT1. Here, a similar substrate specificity profile (kcat /Km ) for model phosphoramidate substrates was found for hHINT2 but with higher kcat and Km values when compared with hHINT1. A broader pH range for maximum catalytic activity was determined for hHINT2 (pK1 = 6.76 ± 0.16, pK2 = 8.41 ± 0.07). In addition, the known hHINT1-microphthalmia-inducing transcription factor-regulating molecule Ap4 A was found to have no detectable binding to HINT1 nor HINT2 by isothermal titration calorimetry. These results demonstrate that despite differences in their sequence and localization, HINT1 and HINT2 have similar nucleotide substrate specificities, which should be considered in future proTide design and in studies of their natural function.
Collapse
Affiliation(s)
- Alexander Strom
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Cher Ling Tong
- Department of Biochemistry Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Carston R Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
11
|
Abstract
Despite successful vaccination programs and effective treatments for some viral infections, humans are still losing the battle with viruses. Persisting human pandemics, emerging and re-emerging viruses, and evolution of drug-resistant strains impose continuous search for new antiviral drugs. A combination of detailed information about the molecular organization of viruses and progress in molecular biology and computer technologies has enabled rational antivirals design. Initial step in establishing efficacy of new antivirals is based on simple methods assessing inhibition of the intended target. We provide here an overview of biochemical and cell-based assays evaluating the activity of inhibitors of clinically important viruses.
Collapse
Affiliation(s)
- Michaela Rumlová
- Department of Biotechnology, University of Chemistry and Technology, Prague 166 28, Czech Republic.
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague 166 28, Czech Republic.
| |
Collapse
|
12
|
Liang D, Ge D, Lv Y, Huang W, Wang B, Li W. Silver-Catalyzed Radical Arylphosphorylation of Unactivated Alkenes: Synthesis of 3-Phosphonoalkyl Indolines. J Org Chem 2018; 83:4681-4691. [DOI: 10.1021/acs.joc.8b00450] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/03/2023]
Affiliation(s)
- Deqiang Liang
- Department of Chemistry, Kunming University, Kunming 650214, China
- Yunnan Engineering Technology Research Center for Plastic Films, Kunming 650214, China
| | - Dandan Ge
- Department of Chemistry, Kunming University, Kunming 650214, China
| | - Yanping Lv
- Department of Chemistry, Kunming University, Kunming 650214, China
| | - Wenzhong Huang
- Yunnan Engineering Technology Research Center for Plastic Films, Kunming 650214, China
| | - Baoling Wang
- Yunnan Engineering Technology Research Center for Plastic Films, Kunming 650214, China
| | - Weili Li
- Yunnan Engineering Technology Research Center for Plastic Films, Kunming 650214, China
| |
Collapse
|
13
|
Maize KM, Shah R, Strom A, Kumarapperuma S, Zhou A, Wagner CR, Finzel BC. A Crystal Structure Based Guide to the Design of Human Histidine Triad Nucleotide Binding Protein 1 (hHint1) Activated ProTides. Mol Pharm 2017; 14:3987-3997. [PMID: 28968488 DOI: 10.1021/acs.molpharmaceut.7b00664] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/08/2023]
Abstract
Nucleotide analogues that incorporate a metabolically labile nucleoside phosphoramidate (a ProTide) have found utility as prodrugs. In humans, ProTides can be cleaved by human histidine triad nucleotide binding protein 1 (hHint1) to expose the nucleotide monophosphate. Activation by this route circumvents highly selective nucleoside kinases that limit the use of nucleosides as prodrugs. To better understand the diversity of potential substrates of hHint1, we created and studied a series of phosphoramidate nucleosides. Using a combination of enzyme kinetics, X-ray crystallography, and isothermal titration calorimetry with both wild-type and inactive mutant enzymes, we have been able to explore the energetics of substrate binding and establish a structural basis for catalytic efficiency. Diverse nucleobases are well tolerated, but portions of the ribose are needed to position substrates for catalysis. Beneficial characteristics of the amine leaving group are also revealed. Structural principles revealed by these results may be exploited to tune the rate of substrate hydrolysis to strategically alter the intracellular release of the product nucleoside monophosphate from the ProTide.
Collapse
Affiliation(s)
- Kimberly M Maize
- Department of Medicinal Chemistry, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Rachit Shah
- Department of Medicinal Chemistry, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Alex Strom
- Department of Medicinal Chemistry, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Sidath Kumarapperuma
- Department of Medicinal Chemistry, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Andrew Zhou
- Department of Medicinal Chemistry, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Carston R Wagner
- Department of Medicinal Chemistry, University of Minnesota , Minneapolis, Minnesota 55455, United States
| | - Barry C Finzel
- Department of Medicinal Chemistry, University of Minnesota , Minneapolis, Minnesota 55455, United States
| |
Collapse
|