1
|
Xu Q, Sharif M, James E, Dismorr JO, Tucker JHR, Willcox BE, Mehellou Y. Phosphonodiamidate prodrugs of phosphoantigens (ProPAgens) exhibit potent Vγ9/Vδ2 T cell activation and eradication of cancer cells. RSC Med Chem 2024; 15:2462-2473. [PMID: 39026632 PMCID: PMC11253855 DOI: 10.1039/d4md00208c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/30/2024] [Indexed: 07/20/2024] Open
Abstract
The phosphoantigen (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) is an established activator of Vγ9/Vδ2 T cells and stimulates downstream effector functions including cytotoxicity and cytokine production. In order to improve its drug-like properties, we herein report the design, synthesis, serum stability, in vitro metabolism, and biological evaluation of a new class of symmetrical phosphonodiamidate prodrugs of methylene and difluoromethylene monophosphonate derivatives of HMBPP. These prodrugs, termed phosphonodiamidate ProPAgens, were synthesized in good yields, exhibited excellent serum stability (>7 h), and their in vitro metabolism was shown to be initiated by carboxypeptidase Y. These phosphonodiamidate ProPAgens triggered potent activation of Vγ9/Vδ2 T cells, which translated into efficient Vγ9/Vδ2 T cell-mediated eradication of bladder cancer cells in vitro. Together, these findings showcase the potential of these phosphonodiamidate ProPAgens as Vγ9/Vδ2 T cell modulators that could be further developed as novel cancer immunotherapeutic agents.
Collapse
Affiliation(s)
- Qin Xu
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University Cardiff CF10 3NB UK
| | - Maria Sharif
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham B15 2TT UK
- Cancer Immunology and Immunotherapy Centre, University of Birmingham Birmingham B15 2TT UK
| | - Edward James
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University Cardiff CF10 3NB UK
| | - Jack O Dismorr
- School of Chemistry, University of Birmingham Birmingham B15 2TT UK
| | - James H R Tucker
- School of Chemistry, University of Birmingham Birmingham B15 2TT UK
| | - Benjamin E Willcox
- Institute of Immunology and Immunotherapy, University of Birmingham Birmingham B15 2TT UK
- Cancer Immunology and Immunotherapy Centre, University of Birmingham Birmingham B15 2TT UK
| | - Youcef Mehellou
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University Cardiff CF10 3NB UK
- Medicines Discovery Institute, Cardiff University Cardiff CF10 3AT UK
| |
Collapse
|
2
|
Yan VC, Pham CD, Ballato ES, Yang KL, Arthur K, Khadka S, Barekatain Y, Shrestha P, Tran T, Poral AH, Washington M, Raghavan S, Czako B, Pisaneschi F, Lin YH, Satani N, Hammoudi N, Ackroyd JJ, Georgiou DK, Millward SW, Muller FL. Prodrugs of a 1-Hydroxy-2-oxopiperidin-3-yl Phosphonate Enolase Inhibitor for the Treatment of ENO1-Deleted Cancers. J Med Chem 2022; 65:13813-13832. [PMID: 36251833 PMCID: PMC9620261 DOI: 10.1021/acs.jmedchem.2c01039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancers harboring homozygous deletion of the glycolytic enzyme enolase 1 (ENO1) are selectively vulnerable to inhibition of the paralogous isoform, enolase 2 (ENO2). A previous work described the sustained tumor regression activities of a substrate-competitive phosphonate inhibitor of ENO2, 1-hydroxy-2-oxopiperidin-3-yl phosphonate (HEX) (5), and its bis-pivaloyoxymethyl prodrug, POMHEX (6), in an ENO1-deleted intracranial orthotopic xenograft model of glioblastoma [Nature Metabolism 2020, 2, 1423-1426]. Due to poor pharmacokinetics of bis-ester prodrugs, this study was undertaken to identify potential non-esterase prodrugs for further development. Whereas phosphonoamidate esters were efficiently bioactivated in ENO1-deleted glioma cells, McGuigan prodrugs were not. Other strategies, including cycloSal and lipid prodrugs of 5, exhibited low micromolar IC50 values in ENO1-deleted glioma cells and improved stability in human serum over 6. The activity of select prodrugs was also probed using the NCI-60 cell line screen, supporting its use to examine the relationship between prodrugs and cell line-dependent bioactivation.
Collapse
Affiliation(s)
- Victoria C. Yan
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States,. Twitter: @victoriacyanide
| | - Cong-Dat Pham
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Elliot S. Ballato
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Kristine L. Yang
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Kenisha Arthur
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Sunada Khadka
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States,Department
of Cancer Biology, University of Texas MD
Anderson Cancer Center, Houston, Texas 77054, United States
| | - Yasaman Barekatain
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States,Department
of Cancer Biology, University of Texas MD
Anderson Cancer Center, Houston, Texas 77054, United States
| | - Prakriti Shrestha
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Theresa Tran
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Anton H. Poral
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Mykia Washington
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Sudhir Raghavan
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Barbara Czako
- Institute
of Applied Cancer Science, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Federica Pisaneschi
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Yu-Hsi Lin
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Nikunj Satani
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Naima Hammoudi
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Jeffrey J. Ackroyd
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Dimitra K. Georgiou
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Steven W. Millward
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Florian L. Muller
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| |
Collapse
|
3
|
Abstract
Remdesivir (RDV; GS-5734, Veklury), the first FDA-approved antiviral to treat COVID-19, is a single-diastereomer monophosphoramidate prodrug of an adenosine analogue. RDV is taken up in the target cells and metabolized in multiple steps to form the active nucleoside triphosphate (TP) (GS-443902), which, in turn, acts as a potent and selective inhibitor of multiple viral RNA polymerases. In this report, we profiled the key enzymes involved in the RDV metabolic pathway with multiple parallel approaches: (i) bioinformatic analysis of nucleoside/nucleotide metabolic enzyme mRNA expression using public human tissue and lung single-cell bulk mRNA sequence (RNA-seq) data sets, (ii) protein and mRNA quantification of enzymes in human lung tissue and primary lung cells, (iii) biochemical studies on the catalytic rate of key enzymes, (iv) effects of specific enzyme inhibitors on the GS-443902 formation, and (v) the effects of these inhibitors on RDV antiviral activity against SARS-CoV-2 in cell culture. Our data collectively demonstrated that carboxylesterase 1 (CES1) and cathepsin A (CatA) are enzymes involved in hydrolyzing RDV to its alanine intermediate MetX, which is further hydrolyzed to the monophosphate form by histidine triad nucleotide-binding protein 1 (HINT1). The monophosphate is then consecutively phosphorylated to diphosphate and triphosphate by cellular phosphotransferases. Our data support the hypothesis that the unique properties of RDV prodrug not only allow lung-specific accumulation critical for the treatment of respiratory viral infection such as COVID-19 but also enable efficient intracellular metabolism of RDV and its MetX to monophosphate and successive phosphorylation to form the active TP in disease-relevant cells.
Collapse
|
4
|
Lášek T, Dobiáš J, Buděšínský M, Kozák J, Lapuníková B, Rosenberg I, Birkuš G, Páv O. Synthesis of phosphonate derivatives of 2'-deoxy-2'-fluorotetradialdose d-nucleosides and tetradialdose d-nucleosides. Tetrahedron 2021; 89:132159. [PMID: 33879930 PMCID: PMC8049856 DOI: 10.1016/j.tet.2021.132159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/02/2021] [Accepted: 04/09/2021] [Indexed: 11/24/2022]
Abstract
Analogs of nucleosides and nucleotides represent a promising pool of potential therapeutics. This work describes a new synthetic route leading to 2'-deoxy-2'-fluorotetradialdose D-nucleoside phosphonates. Moreover, a new universal synthetic route leading to tetradialdose d-nucleosides bearing purine nucleobases is also described. All new compounds were tested as triphosphate analogs for inhibitory potency against a variety of viral polymerases. The fluorinated nucleosides were transformed to phosphoramidate prodrugs and evaluated in cell cultures against various viruses including influenza and SARS-CoV-2.
Collapse
Affiliation(s)
- Tomáš Lášek
- IOCB Prague, Flemingovo Nám. 2, 160 00, Prague, Czech Republic.,UCT Prague, Technická 5, 166 28, Prague, Czech Republic
| | - Juraj Dobiáš
- IOCB Prague, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | | | - Jaroslav Kozák
- IOCB Prague, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | | | - Ivan Rosenberg
- IOCB Prague, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | - Gabriel Birkuš
- IOCB Prague, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | - Ondřej Páv
- IOCB Prague, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| |
Collapse
|
5
|
Li J, Liu S, Shi J, Wang X, Xue Y, Zhu HJ. Tissue-Specific Proteomics Analysis of Anti-COVID-19 Nucleoside and Nucleotide Prodrug-Activating Enzymes Provides Insights into the Optimization of Prodrug Design and Pharmacotherapy Strategy. ACS Pharmacol Transl Sci 2021; 4:870-887. [PMID: 33855276 PMCID: PMC8033752 DOI: 10.1021/acsptsci.1c00016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Indexed: 12/16/2022]
Abstract
Nucleoside and nucleotide analogs are an essential class of antivirals for COVID-19 treatment. Several nucleoside/nucleotide analogs have shown promising effects against SARS-CoV-2 in vitro; however, their in vivo efficacy is limited. Nucleoside/nucleotide analogs are often formed as ester prodrugs to improve pharmacokinetics (PK) performance. After entering cells, the prodrugs undergo several enzymatic metabolism steps to form the active metabolite triphosphate nucleoside (TP-Nuc); prodrug activation is therefore associated with the abundance and catalytic activity of the corresponding activating enzymes. Having the activation of nucleoside/nucleotide prodrugs occur at the target site of action, such as the lung, is critical for anti-SARS-CoV-2 efficacy. Herein, we conducted an absolute quantitative proteomics study to determine the expression of relevant activating enzymes in human organs related to the PK and antiviral efficacy of nucleoside/nucleotide prodrugs, including the lung, liver, intestine, and kidney. The protein levels of prodrug-activating enzymes differed significantly among the tissues. Using catalytic activity values reported previously for individual enzymes, we calculated prodrug activation profiles in these tissues. The prodrugs evaluated in this study include nine McGuigan phosphoramidate prodrugs, two cyclic monophosphate prodrugs, two l-valyl ester prodrugs, and one octanoate prodrug. Our analysis showed that most orally administered nucleoside/nucleotide prodrugs were primarily activated in the liver, suggesting that parenteral delivery routes such as inhalation and intravenous infusion could be better options when these antiviral prodrugs are used to treat COVID-19. The results also indicated that the l-valyl ester prodrug design can plausibly improve drug bioavailability and enhance effects against SARS-CoV-2 intestinal infections. This study further revealed that an octanoate prodrug could provide a long-acting antiviral effect targeting SARS-CoV-2 infections in the lung. Finally, our molecular docking analysis suggested several prodrug forms of favipiravir and GS-441524 that are likely to exhibit favorable PK features over existing prodrug forms. In sum, this study revealed the activation mechanisms of various nucleoside/nucleotide prodrugs relevant to COVID-19 treatment in different organs and shed light on the development of more effective anti-COVID-19 prodrugs.
Collapse
Affiliation(s)
- Jiapeng Li
- Department
of Clinical Pharmacy, University of Michigan
College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, Michigan 48109, United States
| | - Shuhan Liu
- Department
of Clinical Pharmacy, University of Michigan
College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, Michigan 48109, United States
- Department
of Pharmaceutical Sciences, University of
Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
| | - Jian Shi
- Department
of Clinical Pharmacy, University of Michigan
College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, Michigan 48109, United States
| | - Xinwen Wang
- Department
of Pharmaceutical Sciences, Northeast Ohio
Medical University College of Pharmacy, Rootstown, Ohio 44272, United States
| | - Yanling Xue
- Department
of Clinical Pharmacy, University of Michigan
College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, Michigan 48109, United States
| | - Hao-Jie Zhu
- Department
of Clinical Pharmacy, University of Michigan
College of Pharmacy, 428 Church Street, Room 4565 NUB, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
6
|
Hassan AY, El-Sebaey SA, El Deeb MA, Elzoghbi MS. Potential antiviral and anticancer effect of imidazoles and bridgehead imidazoles generated by HPV-Induced cervical carcinomas via reactivating the P53/ pRb pathway and inhibition of CA IX. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
7
|
Groaz E, De Jonghe S. Overview of Biologically Active Nucleoside Phosphonates. Front Chem 2021; 8:616863. [PMID: 33490040 PMCID: PMC7821050 DOI: 10.3389/fchem.2020.616863] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/30/2020] [Indexed: 12/25/2022] Open
Abstract
The use of the phosphonate motif featuring a carbon-phosphorous bond as bioisosteric replacement of the labile P–O bond is widely recognized as an attractive structural concept in different areas of medicinal chemistry, since it addresses the very fundamental principles of enzymatic stability and minimized metabolic activation. This review discusses the most influential successes in drug design with special emphasis on nucleoside phosphonates and their prodrugs as antiviral and cancer treatment agents. A description of structurally related analogs able to interfere with the transmission of other infectious diseases caused by pathogens like bacteria and parasites will then follow. Finally, molecules acting as agonists/antagonists of P2X and P2Y receptors along with nucleotidase inhibitors will also be covered. This review aims to guide readers through the fundamentals of nucleoside phosphonate therapeutics in order to inspire the future design of molecules to target infections that are refractory to currently available therapeutic options.
Collapse
Affiliation(s)
- Elisabetta Groaz
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Steven De Jonghe
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Wiemer AJ. Metabolic Efficacy of Phosphate Prodrugs and the Remdesivir Paradigm. ACS Pharmacol Transl Sci 2020; 3:613-626. [PMID: 32821882 PMCID: PMC7409933 DOI: 10.1021/acsptsci.0c00076] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Indexed: 02/08/2023]
Abstract
![]()
Drugs that contain phosphates (and
phosphonates or phosphinates)
have intrinsic absorption issues and are therefore often delivered
in prodrug forms to promote their uptake. Effective prodrug forms
distribute their payload to the site of the intended target and release
it efficiently with minimal byproduct toxicity. The ability to balance
unwanted payload release during transit with desired release at the
site of action is critical to prodrug efficacy. Despite decades of
research on prodrug forms, choosing the ideal prodrug form remains
a challenge which is often solved empirically. The recent emergency
use authorization of the antiviral remdesivir for COVID-19 exemplifies
a new approach for delivery of phosphate prodrugs by parenteral dosing,
which minimizes payload release during transit and maximizes tissue
payload distribution. This review focuses on the role of metabolic
activation in efficacy during oral and parenteral dosing of phosphate,
phosphonate, and phosphinate prodrugs. Through examining prior structure–activity
studies on prodrug forms and the choices that led to development of
remdesivir and other clinical drugs and drug candidates, a better
understanding of their ability to distribute to the planned site of
action, such as the liver, plasma, PBMCs, or peripheral tissues, can
be gained. The structure–activity relationships described here
will facilitate the rational design of future prodrugs.
Collapse
Affiliation(s)
- Andrew J Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, United States.,Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
9
|
Rautio J, Kärkkäinen J, Sloan KB. Prodrugs – Recent approvals and a glimpse of the pipeline. Eur J Pharm Sci 2017; 109:146-161. [DOI: 10.1016/j.ejps.2017.08.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 01/12/2023]
|
10
|
Abstract
The ProTide technology is a prodrug approach developed for the efficient intracellular delivery of nucleoside analogue monophosphates and monophosphonates. In this approach, the hydroxyls of the monophosphate or monophosphonate groups are masked by an aromatic group and an amino acid ester moiety, which are enzymatically cleaved-off inside cells to release the free nucleoside monophosphate and monophosphonate species. Structurally, this represents the current end-point of an extensive medicinal chemistry endeavor that spans almost three decades. It started from the masking of nucleoside monophosphate and monophosphonate groups by simple alkyl groups and evolved into the sophisticated ProTide system as known today. This technology has been extensively employed in drug discovery, and it has already led to the discovery of two FDA-approved (antiviral) ProTides. In this work, we will review the development of the ProTide technology, its application in drug discovery, and its role in the improvement of drug delivery and efficacy.
Collapse
Affiliation(s)
- Youcef Mehellou
- School of Pharmacy and Pharmaceutical Sciences , Cardiff University , Redwood Building , Cardiff CF10 3NB , U.K
| | - Hardeep S Rattan
- School of Pharmacy, College of Medical and Dental Sciences , University of Birmingham , Edgbaston , Birmingham B15 2TT , U.K
| | - Jan Balzarini
- Laboratory of Virology and Chemotherapy , Rega Institute for Medical Research , Herestraat 49 , 3000 Leuven , Belgium
| |
Collapse
|
11
|
Rebbapragada I, Birkus G, Perry J, Xing W, Kwon H, Pflanz S. Molecular Determinants of GS-9620-Dependent TLR7 Activation. PLoS One 2016; 11:e0146835. [PMID: 26784926 PMCID: PMC4718629 DOI: 10.1371/journal.pone.0146835] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/22/2015] [Indexed: 01/04/2023] Open
Abstract
GS-9620 is an orally administered agonist of Toll-like receptor (TLR)7 currently being evaluated in clinical studies for the treatment of chronic HBV and HIV patients. GS-9620 has shown antiviral efficacy in preclinical models of chronic hepadnavirus infection in woodchuck as well as chimpanzee. However, the molecular determinants of GS-9620-dependent activation of TLR7 are not well defined. The studies presented here elucidate GS-9620 subcellular distribution and characterize its molecular interactions with human TLR7 using structure-guided mutational analysis. Based on our results we present a molecular model of TLR7 bound to GS-9620. We also determine that several coding SNPs had no effect on GS-9620-dependent TLR7 activation. In addition, our studies provide evidence that TLR7 exists in a ligand-independent oligomeric state and that, TLR7 activation by GS-9620 is likely associated with compound-induced conformational changes. Finally, we demonstrate that activation of NF-κB and Akt pathways in primary plasmacytoid dendritic cells occur as immediate downstream cellular responses to GS-9620 stimulation. The data presented here further our understanding of the molecular parameters governing TLR7 activation by GS-9620, and more generally by nucleos/tide-related ligands.
Collapse
Affiliation(s)
| | - Gabriel Birkus
- Department of Biology, Gilead Sciences Inc., Foster City, California, USA
| | - Jason Perry
- Department of Structural Chemistry Gilead Sciences Inc., Foster City, California, USA
| | - Weimei Xing
- Department of Biology, Gilead Sciences Inc., Foster City, California, USA
| | - HyockJoo Kwon
- Department of Biology, Gilead Sciences Inc., Foster City, California, USA
| | - Stefan Pflanz
- Department of Biology, Gilead Sciences Inc., Foster City, California, USA
- * E-mail:
| |
Collapse
|
12
|
Intracellular Activation of Tenofovir Alafenamide and the Effect of Viral and Host Protease Inhibitors. Antimicrob Agents Chemother 2015; 60:316-22. [PMID: 26503655 DOI: 10.1128/aac.01834-15] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/19/2015] [Indexed: 12/22/2022] Open
Abstract
Tenofovir alafenamide fumarate (TAF) is an oral phosphonoamidate prodrug of the HIV reverse transcriptase nucleotide inhibitor tenofovir (TFV). Previous studies suggested a principal role for the lysosomal serine protease cathepsin A (CatA) in the intracellular activation of TAF. Here we further investigated the role of CatA and other human hydrolases in the metabolism of TAF. Overexpression of CatA or liver carboxylesterase 1 (Ces1) in HEK293T cells increased intracellular TAF hydrolysis 2- and 5-fold, respectively. Knockdown of CatA expression with RNA interference (RNAi) in HeLa cells reduced intracellular TAF metabolism 5-fold. Additionally, the anti-HIV activity and the rate of CatA hydrolysis showed good correlation within a large set of TFV phosphonoamidate prodrugs. The covalent hepatitis C virus (HCV) protease inhibitors (PIs) telaprevir and boceprevir potently inhibited CatA-mediated TAF activation (50% inhibitory concentration [IC50] = 0.27 and 0.16 μM, respectively) in vitro and also reduced its anti-HIV activity in primary human CD4(+) T lymphocytes (21- and 3-fold, respectively) at pharmacologically relevant concentrations. In contrast, there was no inhibition of CatA or any significant effect on anti-HIV activity of TAF observed with cobicistat, noncovalent HIV and HCV PIs, or various prescribed inhibitors of host serine proteases. Collectively, these studies confirm that CatA plays a pivotal role in the intracellular metabolism of TAF, whereas the liver esterase Ces1 likely contributes to the hepatic activation of TAF. Moreover, this work demonstrates that a wide range of viral and host PIs, with the exception of telaprevir and boceprevir, do not interfere with the antiretroviral activity of TAF.
Collapse
|
13
|
Kubota K, Inaba SI, Nakano R, Watanabe M, Sakurai H, Fukushima Y, Ichikawa K, Takahashi T, Izumi T, Shinagawa A. Identification of activating enzymes of a novel FBPase inhibitor prodrug, CS-917. Pharmacol Res Perspect 2015; 3:e00138. [PMID: 26171222 PMCID: PMC4492754 DOI: 10.1002/prp2.138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/19/2015] [Accepted: 02/23/2015] [Indexed: 11/30/2022] Open
Abstract
CS-917 (MB06322) is a selective small compound inhibitor of fructose 1,6-bisphosphatase (FBPase), which is expected to be a novel drug for the treatment of type 2 diabetes by inhibiting gluconeogenesis. CS-917 is a bisamidate prodrug and activation of CS-917 requires a two-step enzyme catalyzed reaction. The first-step enzyme, esterase, catalyzes the conversion of CS-917 into the intermediate form (R-134450) and the second-step enzyme, phosphoramidase, catalyzes the conversion of R-134450 into the active form (R-125338). In this study, we biochemically purified the CS-917 esterase activity in monkey small intestine and liver. We identified cathepsin A (CTSA) and elastase 3B (ELA3B) as CS-917 esterases in the small intestine by mass spectrometry, whereas we found CTSA and carboxylesterase 1 (CES1) in monkey liver. We also purified R-134450 phosphoramidase activity in monkey liver and identified sphingomyelin phosphodiesterase, acid-like 3A (SMPADL3A), as an R-134450 phosphoramidase, which has not been reported to have any enzyme activity. Recombinant human CTSA, ELA3B, and CES1 showed CS-917 esterase activity and recombinant human SMPDL3A showed R-134450 phosphoramidase activity, which confirmed the identification of those enzymes. Identification of metabolic enzymes responsible for the activation process is the requisite first step to understanding the activation process, pharmacodynamics and pharmacokinetics of CS-917 at the molecular level. This is the first identification of a phosphoramidase other than histidine triad nucleotide-binding protein (HINT) family enzymes and SMPDL3A might generally contribute to activation of the other bisamidate prodrugs.
Collapse
Affiliation(s)
- Kazuishi Kubota
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. Tokyo, Japan
| | - Shin-Ichi Inaba
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. Tokyo, Japan
| | - Rika Nakano
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. Tokyo, Japan
| | - Mihoko Watanabe
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. Tokyo, Japan
| | - Hidetaka Sakurai
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. Tokyo, Japan
| | - Yumiko Fukushima
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. Tokyo, Japan
| | - Kimihisa Ichikawa
- Biologics Technology Research Laboratories, Daiichi Sankyo Co., Ltd. Tokyo, Japan
| | - Tohru Takahashi
- New Modality Research Laboratories, Daiichi Sankyo Co., Ltd. Tokyo, Japan
| | - Takashi Izumi
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. Tokyo, Japan
| | - Akira Shinagawa
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd. Tokyo, Japan
| |
Collapse
|
14
|
Abstract
A substantial portion of metabolism involves transformation of phosphate esters, including pathways leading to nucleotides and oligonucleotides, carbohydrates, isoprenoids and steroids, and phosphorylated proteins. Because the natural substrates bear one or more negative charges, drugs that target these enzymes generally must be charged as well, but small charged molecules can have difficulty traversing the cell membrane by means other than endocytosis. The resulting dichotomy has stimulated a great deal of effort to develop effective prodrugs, compounds that carry little or no charge to enable them to transit biological membranes, but able to release the parent drug once inside the target cell. This chapter presents recent studies on advances in prodrug forms, along with representative examples of their application to marketed and developmental drugs.
Collapse
Affiliation(s)
- Andrew J Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, 06269, USA
| | | |
Collapse
|
15
|
Andrei G, Topalis D, De Schutter T, Snoeck R. Insights into the mechanism of action of cidofovir and other acyclic nucleoside phosphonates against polyoma- and papillomaviruses and non-viral induced neoplasia. Antiviral Res 2014; 114:21-46. [PMID: 25446403 DOI: 10.1016/j.antiviral.2014.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 09/22/2014] [Accepted: 10/21/2014] [Indexed: 12/30/2022]
Abstract
Acyclic nucleoside phosphonates (ANPs) are well-known for their antiviral properties, three of them being approved for the treatment of human immunodeficiency virus infection (tenofovir), chronic hepatitis B (tenofovir and adefovir) or human cytomegalovirus retinitis (cidofovir). In addition, cidofovir is mostly used off-label for the treatment of infections caused by several DNA viruses other than cytomegalovirus, including papilloma- and polyomaviruses, which do not encode their own DNA polymerases. There is considerable interest in understanding why cidofovir is effective against these small DNA tumor viruses. Considering that papilloma- and polyomaviruses cause diseases associated either with productive infection (characterized by high production of infectious virus) or transformation (where only a limited number of viral proteins are expressed without synthesis of viral particles), it can be envisaged that cidofovir may act as antiviral and/or antiproliferative agent. The aim of this review is to discuss the advances in recent years in understanding the mode of action of ANPs as antiproliferative agents, given the fact that current data suggest that their use can be extended to the treatment of non-viral related malignancies.
Collapse
Affiliation(s)
- G Andrei
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Belgium.
| | - D Topalis
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Belgium
| | - T De Schutter
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Belgium
| | - R Snoeck
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Belgium
| |
Collapse
|
16
|
Abstract
Galactosialidosis is a human lysosomal storage disease caused by deficiency in the multifunctional lysosomal protease cathepsin A (also known as protective protein/cathepsin A, PPCA, catA, HPP, and CTSA; EC 3.4.16.5). Previous structural work on the inactive precursor human cathepsin A (zymogen) led to a two-stage model for activation, where proteolysis of a 1.6-kDa excision peptide is followed by a conformational change in a blocking peptide occluding the active site. Here we present evidence for an alternate model of activation of human cathepsin A, needing only cleavage of a 3.3-kDa excision peptide to yield full enzymatic activity, with no conformational change required. We present x-ray crystallographic, mass spectrometric, amino acid sequencing, enzymatic, and cellular data to support the cleavage-only activation model. The results clarify a longstanding question about the mechanism of cathepsin A activation and point to new avenues for the design of mechanism-based inhibitors of the enzyme.
Collapse
Affiliation(s)
- Nilima Kolli
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003; Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Scott C Garman
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003; Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003.
| |
Collapse
|
17
|
Amidate prodrugs of 9-[2-(phosphonomethoxy)ethyl]adenine as inhibitors of adenylate cyclase toxin from Bordetella pertussis. Antimicrob Agents Chemother 2013; 58:664-71. [PMID: 24145524 DOI: 10.1128/aac.01685-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Adenylate cyclase toxin (ACT) is the key virulence factor of Bordetella pertussis that facilitates its invasion into the mammalian body. 9-[2-(Phosphonomethoxy)ethyl]adenine diphosphate (PMEApp), the active metabolite of the antiviral drug bis(POM)PMEA (adefovir dipivoxil), has been shown to inhibit ACT. The objective of this study was to evaluate six novel amidate prodrugs of PMEA, both phenyloxy phosphonamidates and phosphonodiamidates, for their ability to inhibit ACT activity in the J774A.1 macrophage cell line. The two phenyloxy phosphonamidate prodrugs exhibited greater inhibitory activity (50% inhibitory concentration [IC50] = 22 and 46 nM) than the phosphonodiamidates (IC50 = 84 to 3,960 nM). The inhibitory activity of the prodrugs correlated with their lipophilicity and the degree of their hydrolysis into free PMEA in J774A.1 cells. Although the prodrugs did not inhibit ACT as effectively as bis(POM)PMEA (IC50 = 6 nM), they were significantly less cytotoxic. Moreover, they all reduced apoptotic effects of ACT and prevented an ACT-induced elevation of intracellular [Ca(2+)]i. The amidate prodrugs were less susceptible to degradation in Caco-2 cells compared to bis(POM)PMEA, while they exerted good transepithelial permeability in this assay. As a consequence, a large amount of intact amidate prodrug is expected to be available to target macrophages in vivo. This feature makes nontoxic amidate prodrugs attractive candidates for further investigation as novel antimicrobial agents.
Collapse
|
18
|
Keough DT, Špaček P, Hocková D, Tichý T, Vrbková S, Slavětínská L, Janeba Z, Naesens L, Edstein MD, Chavchich M, Wang TH, de Jersey J, Guddat LW. Acyclic Nucleoside Phosphonates Containing a Second Phosphonate Group Are Potent Inhibitors of 6-Oxopurine Phosphoribosyltransferases and Have Antimalarial Activity. J Med Chem 2013; 56:2513-26. [DOI: 10.1021/jm301893b] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Dianne T. Keough
- The School of Chemistry and
Molecular Biosciences, The University of Queensland, Brisbane 4072, Queensland, Australia
| | - Petr Špaček
- Institute of Organic Chemistry
and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. Flemingovo nám. 2, CZ-166 10 Prague 6, Czech Republic
| | - Dana Hocková
- Institute of Organic Chemistry
and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. Flemingovo nám. 2, CZ-166 10 Prague 6, Czech Republic
| | - Tomáš Tichý
- Institute of Organic Chemistry
and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. Flemingovo nám. 2, CZ-166 10 Prague 6, Czech Republic
| | - Silvie Vrbková
- Institute of Organic Chemistry
and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. Flemingovo nám. 2, CZ-166 10 Prague 6, Czech Republic
| | - Lenka Slavětínská
- Institute of Organic Chemistry
and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. Flemingovo nám. 2, CZ-166 10 Prague 6, Czech Republic
| | - Zlatko Janeba
- Institute of Organic Chemistry
and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. Flemingovo nám. 2, CZ-166 10 Prague 6, Czech Republic
| | - Lieve Naesens
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat
10, B-3000 Leuven, Belgium
| | - Michael D. Edstein
- Australian Army Malaria Institute, Enoggera, Brisbane, Queensland 4051,
Australia
| | - Marina Chavchich
- Australian Army Malaria Institute, Enoggera, Brisbane, Queensland 4051,
Australia
| | - Tzu-Hsuan Wang
- The School of Chemistry and
Molecular Biosciences, The University of Queensland, Brisbane 4072, Queensland, Australia
| | - John de Jersey
- The School of Chemistry and
Molecular Biosciences, The University of Queensland, Brisbane 4072, Queensland, Australia
| | - Luke W. Guddat
- The School of Chemistry and
Molecular Biosciences, The University of Queensland, Brisbane 4072, Queensland, Australia
| |
Collapse
|
19
|
Inhibition of CatA: an emerging strategy for the treatment of heart failure. Future Med Chem 2013; 5:399-409. [DOI: 10.4155/fmc.13.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The lysosomal serine carboxypeptidase CatA has a very important and well-known structural function as well as a, so far, less explored catalytic function. A complete loss of the CatA protein results in the lysosomal storage disease galactosialidosis caused by intralysosomal degradation of β-galactosidase and neuraminidase 1. However, mice with a catalytically inactive CatA enzyme show no signs of this disease. This observation establishes a clear distinction between structural and catalytic functions of the CatA enzyme. Recently, several classes of orally bioavailable synthetic inhibitors of CatA have been identified. Pharmacological studies in rodents indicate a remarkable influence of CatA inhibition on cardiovascular disease progression and identify CatA as a promising novel target for the treatment of heart failure.
Collapse
|
20
|
|
21
|
Meneghesso S, Vanderlinden E, Stevaert A, McGuigan C, Balzarini J, Naesens L. Synthesis and biological evaluation of pyrimidine nucleoside monophosphate prodrugs targeted against influenza virus. Antiviral Res 2012; 94:35-43. [DOI: 10.1016/j.antiviral.2012.01.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 12/24/2011] [Accepted: 01/18/2012] [Indexed: 11/16/2022]
|