1
|
Salari S, Lee H, Tsantrizos YS, Park J. Inhibition of human mevalonate kinase by allosteric inhibitors of farnesyl pyrophosphate synthase. FEBS Open Bio 2024; 14:1320-1339. [PMID: 38923323 PMCID: PMC11301271 DOI: 10.1002/2211-5463.13853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Mevalonate kinase is a key regulator of the mevalonate pathway, subject to feedback inhibition by the downstream metabolite farnesyl pyrophosphate. In this study, we validated the hypothesis that monophosphonate compounds mimicking farnesyl pyrophosphate can inhibit mevalonate kinase. Exploring compounds originally synthesized as allosteric inhibitors of farnesyl pyrophosphate synthase, we discovered mevalonate kinase inhibitors with nanomolar activity. Kinetic characterization of the two most potent inhibitors demonstrated Ki values of 3.1 and 22 nm. Structural comparison suggested features of these inhibitors likely responsible for their potency. Our findings introduce the first class of nanomolar inhibitors of human mevalonate kinase, opening avenues for future research. These compounds might prove useful as molecular tools to study mevalonate pathway regulation and evaluate mevalonate kinase as a potential therapeutic target.
Collapse
Affiliation(s)
- Saman Salari
- Department of BiochemistryMemorial University of NewfoundlandSt. John'sCanada
| | - Hiu‐Fung Lee
- Department of ChemistryMcGill UniversityMontrealCanada
| | | | - Jaeok Park
- Department of BiochemistryMemorial University of NewfoundlandSt. John'sCanada
| |
Collapse
|
2
|
Bekić M, Vasiljević M, Stojanović D, Kokol V, Mihajlović D, Vučević D, Uskoković P, Čolić M, Tomić S. Phosphonate-Modified Cellulose Nanocrystals Potentiate the Th1 Polarising Capacity of Monocyte-Derived Dendritic Cells via GABA-B Receptor. Int J Nanomedicine 2022; 17:3191-3216. [PMID: 35909813 PMCID: PMC9329576 DOI: 10.2147/ijn.s362038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/26/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Phosphonates, like 3-AminoPropylphosphonic Acid (ApA), possess a great potential for the therapy of bone tumours, and their delivery via cellulose nanocrystals (CNCs) seems a promising approach for their increased efficacy in target tissues. However, the immunological effects of CNC-phosphonates have not been investigated thoroughly. The main aim was to examine how the modification of CNCs with phosphonate affects their immunomodulatory properties in human cells. Methods Wood-based native (n) CNCs were modified via oxidation (ox-CNCs) and subsequent conjugation with ApA (ApA-CNCs). CNCs were characterised by atomic force microscopy (AFM) and nanoindentation. Cytotoxicity and immunomodulatory potential of CNCs were investigated in cultures of human peripheral blood mononuclear cells (PBMCs) and monocyte-derived dendritic cells (MoDCs)/T cells co-cultures by monitoring phenotype, cytokines production, allostimulatory and Th/Treg polarisation capacity. Results AFM showed an increase in CNCs' thickens, elasticity modulus and hardness during the modification with ApA. When applied at non-toxic doses, nCNCs showed a tolerogenic potential upon internalisation by MoDCs, as judged by their increased capacity to up-regulate tolerogenic markers and induce regulatory T cells (Treg), especially when present during the differentiation of MoDCs. In contrast, ox- and ApA-CNCs induced oxidative stress and autophagy in MoDCs, which correlated with their stimulatory effect on the maturation of MoDCs, but also inhibition of MoDCs differentiation. ApA-CNC-treated MoDCs displayed the highest allostimulatory and Th1/CTL polarising activity in co-cultures with T cells. These effects of ApA-CNCs were mediated via GABA-B receptor-induced lowering of cAMP levels in MoDCs, and they could be blocked by GABA-B receptor inhibitor. Moreover, the Th1 polarising and allostimulatory capacity of MoDCs differentiated with ApA-CNC were largely preserved upon the maturation of MoDCs, whereas nCNC- and ox-CNC-differentiated MoDCs displayed an increased tolerogenic potential. Conclusion The delivery of ApA via CNCs induces potent DC-mediated Th1 polarisation, which could be beneficial in their potential application in tumour therapy.
Collapse
Affiliation(s)
- Marina Bekić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Miloš Vasiljević
- Center for Biomedical Sciences, Medical Faculty Foča, University of East Sarajevo, Foča, Bosnia and Herzegovina
| | - Dušica Stojanović
- Department for Construction and Special Materials, Faculty for Technology and Metallurgy, University in Belgrade, Belgrade, Serbia
| | - Vanja Kokol
- Department of Textile Materials and Design, Faculty of Mechanical Engineering, University of Maribor, Maribor, Slovenia
| | - Dušan Mihajlović
- Center for Biomedical Sciences, Medical Faculty Foča, University of East Sarajevo, Foča, Bosnia and Herzegovina
| | - Dragana Vučević
- Center for Biomedical Sciences, Medical Faculty Foča, University of East Sarajevo, Foča, Bosnia and Herzegovina
| | - Petar Uskoković
- Department for Construction and Special Materials, Faculty for Technology and Metallurgy, University in Belgrade, Belgrade, Serbia
| | - Miodrag Čolić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia.,Center for Biomedical Sciences, Medical Faculty Foča, University of East Sarajevo, Foča, Bosnia and Herzegovina.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Sergej Tomić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
Lee HF, Lacbay CM, Boutin R, Matralis AN, Park J, Waller DD, Guan TL, Sebag M, Tsantrizos YS. Synthesis and Evaluation of Structurally Diverse C-2-Substituted Thienopyrimidine-Based Inhibitors of the Human Geranylgeranyl Pyrophosphate Synthase. J Med Chem 2022; 65:2471-2496. [PMID: 35077178 DOI: 10.1021/acs.jmedchem.1c01913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Novel analogues of C-2-substituted thienopyrimidine-based bisphosphonates (C2-ThP-BPs) are described that are potent inhibitors of the human geranylgeranyl pyrophosphate synthase (hGGPPS). Members of this class of compounds induce target-selective apoptosis of multiple myeloma (MM) cells and exhibit antimyeloma activity in vivo. A key structural element of these inhibitors is a linker moiety that connects their (((2-phenylthieno[2,3-d]pyrimidin-4-yl)amino)methylene)bisphosphonic acid core to various side chains. The structural diversity of this linker moiety, as well as the side chains attached to it, was investigated and found to significantly impact the toxicity of these compounds in MM cells. The most potent inhibitor identified was evaluated in mouse and rat for liver toxicity and systemic exposure, respectively, providing further optimism for the potential value of such compounds as human therapeutics.
Collapse
Affiliation(s)
- Hiu-Fung Lee
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Cyrus M Lacbay
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Rebecca Boutin
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Alexios N Matralis
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Jaeok Park
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Daniel D Waller
- Department of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
- Division of Hematology, McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Tian Lai Guan
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Michael Sebag
- Department of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
- Division of Hematology, McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Youla S Tsantrizos
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
4
|
Zhou X, Cornel EJ, Fan Z, He S, Du J. Bone-Targeting Polymer Vesicles for Effective Therapy of Osteoporosis. NANO LETTERS 2021; 21:7998-8007. [PMID: 34529430 DOI: 10.1021/acs.nanolett.1c02150] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
With the aging of the population, postmenopausal osteoporosis becomes increasingly widespread and severe as fractures caused by osteoporosis may lead to permanent disabilities and even death. Inspired by extracellular vesicles that participate in bone remodeling, we present a biomimicking polymer vesicle for bone-targeted β-estradiol (E2) delivery. This vesicle is self-assembled from a poly(ε-caprolactone)28-block-poly[(l-glutamic acid)7-stat-(l-glutamic acid-alendronic acid)4] (PCL28-b-P[Glu7-stat-(Glu-ADA)4]) diblock copolymer. The alendronic acid (ADA) on the coronas endows the polymer vesicles with a high bone affinity and acts synergistically with E2 to achieve an enhanced therapeutic effect. As confirmed with ovariectomized osteoporosis rat models, bone loss was significantly reversed as the recovery rates of total BMD (bone mineral density) and trabecular BMD were 70.4% and 99.3%, respectively. Overall, this work provides fresh insight into the treatment of osteoporosis.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Erik Jan Cornel
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Zhen Fan
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Shisheng He
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Jianzhong Du
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| |
Collapse
|
5
|
Calcium Chelidonate: Semi-Synthesis, Crystallography, and Osteoinductive Activity In Vitro and In Vivo. Pharmaceuticals (Basel) 2021; 14:ph14060579. [PMID: 34204329 PMCID: PMC8235635 DOI: 10.3390/ph14060579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
Calcium chelidonate [Ca(ChA)(H2O)3]n was obtained by semi-synthesis using natural chelidonic acid. The structure of the molecular complex was determined by X-ray diffraction analysis. The asymmetric unit of [Ca(ChA)(H2O)3]n includes chelidonic acid coordinated through three oxygen atoms, and three water ligands. The oxygen atoms of acid and oxygen atoms of water from each asymmetric unit are also coordinated to the calcium of another one, forming an infinite linear complex. Calcium geometry is close to the trigonal dodecahedron (D2d). The intra-complex hydrogen bonds additionally stabilize the linear species, which are parallel to the axis. In turn the linear species are packed into the 3D structure through mutual intercomplex hydrogen bonds. The osteogenic activity of the semi-synthetic CaChA was studied in vitro on 21-day hAMMSC culture and in vivo in mice using ectopic (subcutaneous) implantation of CaP-coated Ti plates saturated in vitro with syngeneic bone marrow. The enhanced extracellular matrix ECM mineralization in vitro and ectopic bone tissue formation in situ occurred while a water solution of calcium chelidonate at a dose of 10 mg/kg was used. The test substance promotes human adipose-derived multipotent mesenchymal stromal/stem cells (hAMMSCs), as well as mouse MSCs to differentiate into osteoblasts in vitro and in vivo, respectively. Calcium chelidonate is non-toxic and can stimulate osteoinductive processes.
Collapse
|
6
|
Roy S, Mukherjee P, Das PK, Ghosh PR, Datta P, Kundu B, Nandi SK. Local delivery systems of morphogens/biomolecules in orthopedic surgical challenges. MATERIALS TODAY COMMUNICATIONS 2021; 27:102424. [DOI: 10.1016/j.mtcomm.2021.102424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Park J, Pandya VR, Ezekiel SJ, Berghuis AM. Phosphonate and Bisphosphonate Inhibitors of Farnesyl Pyrophosphate Synthases: A Structure-Guided Perspective. Front Chem 2021; 8:612728. [PMID: 33490038 PMCID: PMC7815940 DOI: 10.3389/fchem.2020.612728] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Phosphonates and bisphosphonates have proven their pharmacological utility as inhibitors of enzymes that metabolize phosphate and pyrophosphate substrates. The blockbuster class of drugs nitrogen-containing bisphosphonates represent one of the best-known examples. Widely used to treat bone-resorption disorders, these drugs work by inhibiting the enzyme farnesyl pyrophosphate synthase. Playing a key role in the isoprenoid biosynthetic pathway, this enzyme is also a potential anticancer target. Here, we provide a comprehensive overview of the research efforts to identify new inhibitors of farnesyl pyrophosphate synthase for various therapeutic applications. While the majority of these efforts have been directed against the human enzyme, some have been targeted on its homologs from other organisms, such as protozoan parasites and insects. Our particular focus is on the structures of the target enzymes and how the structural information has guided the drug discovery efforts.
Collapse
Affiliation(s)
- Jaeok Park
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Vishal R Pandya
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Sean J Ezekiel
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | | |
Collapse
|
8
|
Manaswiyoungkul P, de Araujo ED, Gunning PT. Targeting prenylation inhibition through the mevalonate pathway. RSC Med Chem 2020; 11:51-71. [PMID: 33479604 PMCID: PMC7485146 DOI: 10.1039/c9md00442d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/10/2019] [Indexed: 12/13/2022] Open
Abstract
Protein prenylation is a critical mediator in several diseases including cancer and acquired immunodeficiency syndrome (AIDS). Therapeutic intervention has focused primarily on directly targeting the prenyltransferase enzymes, FTase and GGTase I and II. To date, several drugs have advanced to clinical trials and while promising, they have yet to gain approval in a medical setting due to off-target effects and compensatory mechanisms activated by the body which results in drug resistance. While the development of dual inhibitors has mitigated undesirable side effects, potency remains sub-optimal for clinical development. An alternative approach involves antagonizing the upstream mevalonate pathway enzymes, FPPS and GGPPS, which mediate prenylation as well as cholesterol synthesis. The development of these inhibitors presents novel opportunities for dual inhibition of cancer-driven prenylation as well as cholesterol accumulation. Herein, we highlight progress towards the development of inhibitors against the prenylation machinery.
Collapse
Affiliation(s)
- Pimyupa Manaswiyoungkul
- Department of Chemistry , University of Toronto , 80 St. George Street , Toronto , Ontario M5S 3H6 , Canada
| | - Elvin D de Araujo
- Department of Chemical and Physical Sciences , University of Toronto Mississauga , 3359 Mississauga Rd N. , Mississauga , Ontario L5L 1C6 , Canada .
| | - Patrick T Gunning
- Department of Chemical and Physical Sciences , University of Toronto Mississauga , 3359 Mississauga Rd N. , Mississauga , Ontario L5L 1C6 , Canada .
- Department of Chemistry , University of Toronto , 80 St. George Street , Toronto , Ontario M5S 3H6 , Canada
| |
Collapse
|
9
|
Liu Q, Miao Y, Wang X, Lv G, Peng Y, Li K, Li M, Qiu L, Lin J. Structure-based virtual screening and biological evaluation of novel non-bisphosphonate farnesyl pyrophosphate synthase inhibitors. Eur J Med Chem 2020; 186:111905. [DOI: 10.1016/j.ejmech.2019.111905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/14/2019] [Accepted: 11/20/2019] [Indexed: 02/09/2023]
|
10
|
Targeting heme-oxidized soluble guanylate cyclase to promote osteoblast function. Drug Discov Today 2019; 25:422-429. [PMID: 31846712 DOI: 10.1016/j.drudis.2019.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/25/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022]
Abstract
The enzyme soluble guanylate cyclase (sGC) plays an essential part in the nitric oxide (NO) signaling pathway by binding to the prosthetic heme group; thereby catalyzing the synthesis of cyclic guanosine monophosphate (cGMP)-dependent protein kinases. Impaired NO-sGC-cGMP signaling could lead to osteoblast apoptosis by mechanisms involving the oxidative-stress-induced shift of the redox state of the reduced heme to oxidized sGC, leading to diminished heme binding to the enzyme and rendering the sGC unresponsive to NO. Targeting oxidized sGC to enhance cGMP production could restore proliferation and differentiation of osteoblasts into osteocytes. Here, the potential role of sGC activators of an oxidized or heme-free sGC as a target for promoting osteoblast function is reviewed and strategies for delivering drugs to bone are identified.
Collapse
|
11
|
Farrell KB, Karpeisky A, Thamm DH, Zinnen S. Bisphosphonate conjugation for bone specific drug targeting. Bone Rep 2018; 9:47-60. [PMID: 29992180 PMCID: PMC6037665 DOI: 10.1016/j.bonr.2018.06.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/26/2022] Open
Abstract
Bones provide essential functions and are sites of unique biochemistry and specialized cells, but can also be sites of disease. The treatment of bone disorders and neoplasia has presented difficulties in the past, and improved delivery of drugs to bone remains an important goal for achieving effective treatments. Drug targeting strategies have improved drug localization to bone by taking advantage of the high mineral concentration unique to the bone hydroxyapatite matrix, as well as tissue-specific cell types. The bisphosphonate molecule class binds specifically to hydroxyapatite and inhibits osteoclast resorption of bone, providing direct treatment for degenerative bone disorders, and as emerging evidence suggests, cancer. These bone-binding molecules also provide the opportunity to deliver other drugs specifically to bone by bisphosphonate conjugation. Bisphosphonate bone-targeted therapies have been successful in treatment of osteoporosis, primary and metastatic neoplasms of the bone, and other bone disorders, as well as refining bone imaging. In this review, we focus upon the use of bisphosphonate conjugates with antineoplastic agents, and overview bisphosphonate based imaging agents, nanoparticles, and other drugs. We also discuss linker design potential and the current state of bisphosphonate conjugate research progress. Ongoing investigations continue to expand the possibilities for bone-targeted therapeutics and for extending their reach into clinical practice.
Collapse
Affiliation(s)
- Kristen B Farrell
- MBC Pharma Inc., 12635 East Montview Blvd., Aurora, CO 80045-0100, United States of America
| | - Alexander Karpeisky
- MBC Pharma Inc., 12635 East Montview Blvd., Aurora, CO 80045-0100, United States of America
| | - Douglas H Thamm
- Flint Animal Cancer Center, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523-1620, United States of America
| | - Shawn Zinnen
- MBC Pharma Inc., 12635 East Montview Blvd., Aurora, CO 80045-0100, United States of America
| |
Collapse
|
12
|
Cawthray J, Wasan E, Wasan K. Bone-seeking agents for the treatment of bone disorders. Drug Deliv Transl Res 2018; 7:466-481. [PMID: 28589453 DOI: 10.1007/s13346-017-0394-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The targeting and delivery of therapeutic and diagnostic agents to bone tissue presents both a challenge and opportunity. Osteoporosis, Paget's disease, cancer, and bone metastases are all skeletal diseases whose treatment would benefit from new targeted therapeutic strategies. Osteoporosis, in particular, is a very prevalent disease, affecting over one in three women and one in five men in Canada alone with the cost to the healthcare system estimated at over $2.3 billion in 2010. Bone tissue is often considered a rigid structure when in reality there is a process of continuous remodeling that takes place via complex endocrine-regulated cell signaling pathways in addition to the signaling pathways unique to bone tissue. It is these specific boneremodeling processes that provide unique targeting opportunities but also present a number of challenges.
Collapse
Affiliation(s)
- Jacqueline Cawthray
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Ellen Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kishor Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
13
|
Young RN, Grynpas MD. Targeting therapeutics to bone by conjugation with bisphosphonates. Curr Opin Pharmacol 2018; 40:87-94. [PMID: 29626715 DOI: 10.1016/j.coph.2018.03.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/20/2018] [Indexed: 12/24/2022]
Abstract
Bisphosphonates target and bind avidly to the mineral (hydroxyapatite) found in bone. This targeting ability has been exploited to design and prepare bisphosphonate conjugate prodrugs to deliver a wide variety of drug molecules selectively to bones. It is important that conjugates be stable in the blood stream and that conjugate that is not taken up by bone is eliminated rapidly. The prodrugs should release active drug at a rate appropriate so as to provide efficacy. Radiolabelling is the best method to quantify and evaluate pharmacokinetics, tissue distribution, bone uptake and release of the active drug(s). Recent reports have described bisphosphonate conjugates derived from the antiresorptive drug, alendronic acid and anabolic prostanoid drugs that effectively deliver prostaglandins and prostaglandin EP4 receptor agonists to bone and show enhanced anabolic efficacy and tolerability compared to the drugs alone. These conjugate drugs can be dosed infrequently (weekly or bimonthly) whereas the free drugs must be dosed daily.
Collapse
Affiliation(s)
- Robert N Young
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada.
| | - Marc D Grynpas
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, and University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Alter C, Neumann B, Stammler H, Hoge B. Bis(diethylamino)pentafluorophenylphosphane as Valuable Precursor for the Design of Tetrafluorophenylphosphanes, Tetrafluorophenylphosphinic and ‐phosphonic Acids. Eur J Inorg Chem 2018. [DOI: 10.1002/ejic.201701402] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Christian Alter
- Fakultät für Chemie Centrum für Molekulare Materialien Universität Bielefeld Universitätsstraße 25 33615 Bielefeld Germany
| | - Beate Neumann
- Fakultät für Chemie Centrum für Molekulare Materialien Universität Bielefeld Universitätsstraße 25 33615 Bielefeld Germany
| | - Hans‐Georg Stammler
- Fakultät für Chemie Centrum für Molekulare Materialien Universität Bielefeld Universitätsstraße 25 33615 Bielefeld Germany
| | - Berthold Hoge
- Fakultät für Chemie Centrum für Molekulare Materialien Universität Bielefeld Universitätsstraße 25 33615 Bielefeld Germany
| |
Collapse
|
15
|
Park J, Zielinski M, Magder A, Tsantrizos YS, Berghuis AM. Human farnesyl pyrophosphate synthase is allosterically inhibited by its own product. Nat Commun 2017; 8:14132. [PMID: 28098152 PMCID: PMC5253651 DOI: 10.1038/ncomms14132] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/02/2016] [Indexed: 12/13/2022] Open
Abstract
Farnesyl pyrophosphate synthase (FPPS) is an enzyme of the mevalonate pathway and a well-established therapeutic target. Recent research has focused around a newly identified druggable pocket near the enzyme's active site. Pharmacological exploitation of this pocket is deemed promising; however, its natural biological function, if any, is yet unknown. Here we report that the product of FPPS, farnesyl pyrophosphate (FPP), can bind to this pocket and lock the enzyme in an inactive state. The Kd for this binding is 5–6 μM, within a catalytically relevant range. These results indicate that FPPS activity is sensitive to the product concentration. Kinetic analysis shows that the enzyme is inhibited through FPP accumulation. Having a specific physiological effector, FPPS is a bona fide allosteric enzyme. This allostery offers an exquisite mechanism for controlling prenyl pyrophosphate levels in vivo and thus contributes an additional layer of regulation to the mevalonate pathway. Farnesyl pyrophosphate (FPP) is a key building block for the synthesis of many lipids. Here the authors determine the crystal structure of farnesyl pyrophosphate synthase (FPPS) with its bound product and use kinetic measurements to show that FPP is an allosteric effector of the enzyme.
Collapse
Affiliation(s)
- Jaeok Park
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 0B1
| | - Michal Zielinski
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 0B1
| | - Alexandr Magder
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 0B1
| | - Youla S Tsantrizos
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 0B1.,Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A 0B8
| | - Albert M Berghuis
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 0B1
| |
Collapse
|
16
|
Abstract
Osteoporosis, which is characterized by resorption of bone exceeding formation, remains a significant human health concern, and the impact of this condition will only increase with the "graying" of the worldwide population. This review focuses on current and emerging approaches for delivering therapeutic agents to restore bone remodeling homeostasis. Well-known antiresorptive and anabolic agents, such as estrogen, estrogen analogs, bisphosphonates, calcitonin, and parathyroid hormone, along with newer modulators and antibodies, are primarily administered orally, intravenously, or subcutaneously. Although these treatments can be effective, continuing problems include patient noncompliance and adverse systemic or remote-site effects. Controlled drug delivery via polymeric, targeted, and active release systems extends drug half-life by shielding against premature degradation and improves bioavailability while also providing prolonged, sustained, or intermittent release at therapeutic doses to more effectively treat osteoporosis and associated fracture risk.
Collapse
Affiliation(s)
- T A Asafo-Adjei
- Department of Biomedical Engineering, University of Kentucky, 522A Robotics and Manufacturing Building, Lexington, KY, 40506-0108, USA
| | - A J Chen
- Department of Biomedical Engineering, University of Kentucky, 522A Robotics and Manufacturing Building, Lexington, KY, 40506-0108, USA
| | - A Najarzadeh
- Department of Biomedical Engineering, University of Kentucky, 522A Robotics and Manufacturing Building, Lexington, KY, 40506-0108, USA
| | - D A Puleo
- Department of Biomedical Engineering, University of Kentucky, 522A Robotics and Manufacturing Building, Lexington, KY, 40506-0108, USA.
| |
Collapse
|
17
|
Rodriguez JB, Falcone BN, Szajnman SH. Approaches for Designing new Potent Inhibitors of Farnesyl Pyrophosphate Synthase. Expert Opin Drug Discov 2016; 11:307-20. [DOI: 10.1517/17460441.2016.1143814] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Marzinzik AL, Amstutz R, Bold G, Bourgier E, Cotesta S, Glickman JF, Götte M, Henry C, Lehmann S, Hartwieg JCD, Ofner S, Pellé X, Roddy TP, Rondeau JM, Stauffer F, Stout SJ, Widmer A, Zimmermann J, Zoller T, Jahnke W. Discovery of Novel Allosteric Non-Bisphosphonate Inhibitors of Farnesyl Pyrophosphate Synthase by Integrated Lead Finding. ChemMedChem 2015; 10:1884-91. [PMID: 26381451 DOI: 10.1002/cmdc.201500338] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Indexed: 12/27/2022]
Abstract
Farnesyl pyrophosphate synthase (FPPS) is an established target for the treatment of bone diseases, but also shows promise as an anticancer and anti-infective drug target. Currently available anti-FPPS drugs are active-site-directed bisphosphonate inhibitors, the peculiar pharmacological profile of which is inadequate for therapeutic indications beyond bone diseases. The recent discovery of an allosteric binding site has paved the way toward the development of novel non-bisphosphonate FPPS inhibitors with broader therapeutic potential, notably as immunomodulators in oncology. Herein we report the discovery, by an integrated lead finding approach, of two new chemical classes of allosteric FPPS inhibitors that belong to the salicylic acid and quinoline chemotypes. We present their synthesis, biochemical and cellular activities, structure-activity relationships, and provide X-ray structures of several representative FPPS complexes. These novel allosteric FPPS inhibitors are devoid of any affinity for bone mineral and could serve as leads to evaluate their potential in none-bone diseases.
Collapse
Affiliation(s)
| | - René Amstutz
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland.,Conim AG, Oberwiler Kirchweg 4c, 6300, Zug, Switzerland
| | - Guido Bold
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | | | - Simona Cotesta
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | - J Fraser Glickman
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland.,High Throughput and Spectroscopy Resource Center, Rockefeller University, New York, NY, 10065, USA
| | - Marjo Götte
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | - Christelle Henry
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | - Sylvie Lehmann
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | | | - Silvio Ofner
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | - Xavier Pellé
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | - Thomas P Roddy
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland.,Agios, Cambridge, MA, 02139-4169, USA
| | | | - Frédéric Stauffer
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | - Steven J Stout
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland.,Merck Research Laboratories, 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Armin Widmer
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | - Johann Zimmermann
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland.,Polyphor, Hegenheimermattweg 125, 4123, Allschwil, Switzerland
| | - Thomas Zoller
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland
| | - Wolfgang Jahnke
- Novartis Institutes for BioMedical Research, Basel, 4002, Switzerland.
| |
Collapse
|