Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Br J Clin Pharmacol ; 85(6): 1052-1062, 2019 06.
Article in English | MEDLINE | ID: mdl-30650219

ABSTRACT

The biological effects of the bisphosphonates (BPs) as inhibitors of calcification and bone resorption were first described in the late 1960s. In the 50 years that have elapsed since then, the BPs have become the leading drugs for the treatment of skeletal disorders characterized by increased bone resorption, including Paget's disease of bone, bone metastases, multiple myeloma, osteoporosis and several childhood inherited disorders. The discovery and development of the BPs as a major class of drugs for the treatment of bone diseases is a paradigm for the successful journey from "bench to bedside and back again". Several of the leading BPs achieved "blockbuster" status as branded drugs. However, these BPs have now come to the end of their patent life, making them highly affordable. The opportunity for new clinical applications for BPs also exists in other areas of medicine such as ageing, cardiovascular disease and radiation protection. Their use as inexpensive generic medicines is therefore likely to continue for many years to come. Fifty years of research into the pharmacology of bisphosphonates have led to a fairly good understanding about how these drugs work and how they can be used safely in patients with metabolic bone diseases. However, while we seemingly know much about these drugs, a number of key aspects related to BP distribution and action remain incompletely understood. This review summarizes the existing knowledge of the (pre)clinical and translational pharmacology of BPs, and highlights areas in which understanding is lacking.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Diseases, Metabolic/drug therapy , Bone Remodeling/drug effects , Diphosphonates/therapeutic use , Animals , Bone Density Conservation Agents/adverse effects , Bone Density Conservation Agents/pharmacokinetics , Bone Diseases, Metabolic/diagnosis , Bone Diseases, Metabolic/epidemiology , Bone Diseases, Metabolic/physiopathology , Diphosphonates/adverse effects , Diphosphonates/pharmacokinetics , Humans , Risk Factors , Treatment Outcome
2.
J Biol Chem ; 286(5): 3315-22, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21084289

ABSTRACT

We present here a study of a eukaryotic trans-prenylsynthase from the malaria pathogen Plasmodium vivax. Based on the results of biochemical assays and contrary to previous indications, this enzyme catalyzes the production of geranylgeranyl pyrophosphate (GGPP) rather than farnesyl pyrophosphate (FPP). Structural analysis shows that the product length is constrained by a hydrophobic cavity formed primarily by a set of residues from the same subunit as the product as well as at least one other from the dimeric partner. Furthermore, Plasmodium GGPP synthase (GGPPS) can bind nitrogen-containing bisphosphonates (N-BPs) strongly with the energetically favorable cooperation of three Mg(2+), resulting in inhibition by this class of compounds at IC(50) concentrations below 100 nM. In contrast, human and yeast GGPPSs do not accommodate a third magnesium atom in the same manner, resulting in their insusceptibility to N-BPs. This differentiation is in part attributable to a deviation in a conserved motif known as the second aspartate-rich motif: whereas the aspartates at the start and end of the five-residue motif in FFPP synthases and P. vivax GGPPSs both participate in the coordination of the third Mg(2+), an asparagine is featured as the last residue in human and yeast GGPPSs, resulting in a different manner of interaction with nitrogen-containing ligands.


Subject(s)
Geranylgeranyl-Diphosphate Geranylgeranyltransferase/chemistry , Plasmodium vivax/enzymology , Amino Acid Motifs , Amino Acid Sequence , Diphosphonates/metabolism , Diphosphonates/pharmacology , Enzyme Inhibitors , Geranylgeranyl-Diphosphate Geranylgeranyltransferase/antagonists & inhibitors , Humans , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Magnesium , Nitrogen , Polyisoprenyl Phosphates/biosynthesis , Yeasts
3.
Calcif Tissue Int ; 90(3): 202-10, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22249525

ABSTRACT

Differences in the binding affinities of bisphosphonates for bone mineral have been proposed to determine their localizations and duration of action within bone. The main objective of this study was to test the hypothesis that mineral binding affinity affects bisphosphonate distribution at the basic multicellular unit (BMU) level within both cortical and cancellous bone. To accomplish this objective, skeletally mature female rabbits (n = 8) were injected simultaneously with both low- and high-affinity bisphosphonate analogs bound to different fluorophores. Skeletal distribution was assessed in the rib, tibia, and vertebra using confocal microscopy. The staining intensity ratio between osteocytes contained within the cement line of newly formed rib osteons or within the reversal line of hemiosteons in vertebral trabeculae compared to osteocytes outside the cement/reversal line was greater for the high-affinity compared to the low-affinity compound. This indicates that the low-affinity compound distributes more equally across the cement/reversal line compared to a high-affinity compound, which concentrates mostly near surfaces. These data, from an animal model that undergoes intracortical remodeling similar to humans, demonstrate that the affinity of bisphosphonates for the bone determines the reach of the drugs in both cortical and cancellous bone.


Subject(s)
Bone Density Conservation Agents/pharmacokinetics , Bone Remodeling/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Diphosphonates/pharmacokinetics , Animals , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Bone Remodeling/physiology , Bone and Bones/cytology , Female , Haversian System/cytology , Haversian System/drug effects , Haversian System/metabolism , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/metabolism , Osteoporosis/drug therapy , Rabbits , Tissue Distribution/physiology
4.
Bone ; 42(5): 848-60, 2008 May.
Article in English | MEDLINE | ID: mdl-18325866

ABSTRACT

Bisphosphonates (BPs) target bone due to their high affinity for calcium ions. During osteoclastic resorption, these drugs are released from the acidified bone surface and taken up by osteoclasts, where they act by inhibiting the prenylation of small GTPases essential for osteoclast function. However, it remains unclear exactly how osteoclasts internalise BPs from bone and whether other cells in the bone microenvironment can also take up BPs from the bone surface. We have investigated this using a novel fluorescently-labelled alendronate analogue (FL-ALN), and by examining changes in protein prenylation following treatment of cells with risedronate (RIS). Confocal microscopic analysis showed that FL-ALN was efficiently internalised from solution or from the surface of dentine by resorbing osteoclasts into intracellular vesicles. Accordingly, unprenylated Rap1A accumulated to the same extent whether osteoclasts were cultured on RIS-coated dentine or with RIS in solution. By contrast, J774 macrophages internalised FL-ALN and RIS from solution, but took up comparatively little from dentine, due to their inability to resorb the mineral. Calvarial osteoblasts and MCF-7 tumour cells internalised even less FL-ALN and RIS, both from solution and from the surface of dentine. Accordingly, the viability of J774 and MCF-7 cells was drastically reduced when cultured with RIS in solution, but not when cultured on dentine pre-coated with RIS. However, when J774 macrophages were co-cultured with rabbit osteoclasts, J774 cells that were adjacent to resorbing osteoclasts frequently internalised more FL-ALN than J774 cells more distant from osteoclasts. This was possibly a result of increased availability of BP to these J774 cells due to transcytosis through osteoclasts, since FL-ALN partially co-localised with trancytosed, resorbed matrix protein within osteoclasts. In addition, J774 cells occupying resorption pits internalised more FL-ALN than those on unresorbed surfaces. These data demonstrate that osteoclasts are able to take up large amounts of BP, due to their ability to release the BP from the dentine surface during resorption. By contrast, non-resorbing cells take up only small amounts of BP that becomes available due to natural desorption from the dentine surface. However, BP uptake by non-resorbing cells can be increased when cultured in the presence of resorbing osteoclasts.


Subject(s)
Dentin/metabolism , Diphosphonates/metabolism , Macrophages/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Alendronate/metabolism , Animals , Bone Density Conservation Agents/metabolism , Bone Density Conservation Agents/pharmacokinetics , Bone Density Conservation Agents/pharmacology , Bone Resorption/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Diphosphonates/pharmacokinetics , Diphosphonates/pharmacology , Endocytosis/physiology , Etidronic Acid/analogs & derivatives , Etidronic Acid/metabolism , Etidronic Acid/pharmacokinetics , Etidronic Acid/pharmacology , Extracellular Matrix Proteins/metabolism , Macrophages/cytology , Mice , Microscopy, Fluorescence , Osteoblasts/cytology , Osteoclasts/cytology , Protein Prenylation/drug effects , Rabbits , Risedronic Acid , Skull/cytology , rap1 GTP-Binding Proteins/metabolism
5.
Bone ; 48(2): 259-66, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20920623

ABSTRACT

Nitrogen-containing bisphosphonates (N-BPs) are widely used to block bone destruction associated with bone metastasis because they are effective inhibitors of osteoclast-mediated bone resorption. More specifically, once internalized by osteoclasts, N-BPs block the activity of farnesyl pyrophosphate synthase (FPPS), a key enzyme in the mevalonate pathway. In addition to their antiresorptive activity, preclinical evidence shows that N-BPs have antiangiogenic properties. However, the exact reasons for which N-BPs inhibit angiogenesis remain largely unknown. Using different angiogenesis models, we examined here the effects of zoledronate, risedronate and three structural analogs of risedronate (NE-58025, NE-58051 and NE-10790) with lower potencies to inhibit FPPS activity. Risedronate and zoledronate were much more potent than NE-compounds at inhibiting both endothelial cell proliferation in vitro and vessel sprouting in the chicken egg chorioallantoic membrane (CAM) assay. In addition, only risedronate and zoledronate inhibited the revascularization of the prostate gland in testosterone-stimulated castrated rats. Moreover, as opposed to NE-compounds, risedronate and zoledronate induced intracellular accumulation of isopentenyl pyrophosphate (IPP) in endothelial cells by blocking the activity of the IPP-consuming enzyme FPPS. Thus, these results indicated that N-BPs inhibited angiogenesis in a FPPS-dependent manner. However, drug concentrations used to inhibit angiogenesis, both in vitro and in the CAM and prostate gland assays, were high. In contrast, a low concentration of risedronate (1 µM) was sufficient to inhibit blood vessel formation in the ex vivo rat aortic ring assay. Moreover, NE-58025 (which had a 7-fold lower potency than risedronate to inhibit FPPS activity) was as effective as risedronate to reduce angiogenesis in the rat aortic ring assay. In conclusion, our results suggest that low concentrations of N-BPs inhibit angiogenesis in a FPPS-independent manner, whereas higher drug concentrations were required to inhibit FPPS activity in vivo.


Subject(s)
Diphosphonates/pharmacology , Geranyltranstransferase/metabolism , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane , Diphosphonates/chemistry , Diphosphonates/therapeutic use , Endothelial Cells/cytology , Endothelial Cells/drug effects , Etidronic Acid/analogs & derivatives , Etidronic Acid/chemistry , Etidronic Acid/pharmacology , Etidronic Acid/therapeutic use , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Imidazoles/therapeutic use , Male , Neovascularization, Pathologic/drug therapy , Rats , Rats, Sprague-Dawley , Risedronic Acid , Zoledronic Acid
6.
Bone ; 49(1): 111-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21419243

ABSTRACT

The described ability of phosphonocarboxylate analogues of bisphosphonates (BPs) to inhibit Rab geranylgeranyl transferase (RGGT) is thought to be the mechanism underlying their cellular effects, including their ability to reduce macrophage cell viability and to inhibit osteoclast-mediated resorption. However, until now the possibility that at least some of the effects of these drugs may be mediated through other targets has not been excluded. Since RGGT is the most distal enzyme in the process of Rab prenylation, it has not proved possible to confirm the mechanism underlying the effects of these drugs by adding back downstream intermediates of the mevalonate pathway, the approach used to demonstrate that bisphosphonates act through this pathway. We now confirm that RGGT is the major pharmacological target of phosphonocarboxylates by using several alternative approaches. Firstly, analysis of several different phosphonocarboxylate drugs demonstrates a very good correlation between the ability of these drugs to inhibit RGGT with their ability to: (a) reduce macrophage cell viability; (b) induce apoptosis; and (c) induce vacuolation in rabbit osteoclasts. Secondly, we have found that cells from the gunmetal (gm/gm) mouse, which bear a homozygous mutation in RGGT that results in ~80% reduced activity of this enzyme compared to wild-type or heterozygous mice, are more sensitive to the effects of active phosphonocarboxylates (including reducing macrophage cell viability, inhibiting osteoclast formation and inhibiting fluid-phase endocytosis), confirming that these effects are mediated through inhibition of RGGT. In conclusion, these data demonstrate that all of the pharmacological effects of phosphonocarboxylates found thus far appear to be mediated through the specific inhibition of RGGT, highlighting the potential therapeutic value of this class of drugs.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Diphosphonates/metabolism , Diphosphonates/pharmacology , Alkyl and Aryl Transferases/metabolism , Animals , Apoptosis/drug effects , Cell Count , Cell Line , Cell Survival/drug effects , Diterpenes/pharmacology , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Heterozygote , Macrophages/drug effects , Macrophages/enzymology , Mice , Mice, Inbred Strains , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoclasts/drug effects , Osteoclasts/enzymology , Protein Prenylation/drug effects , Protein Transport/drug effects , Pyridines/pharmacology , Rabbits , Vacuoles/drug effects , Vacuoles/metabolism
7.
Ther Clin Risk Manag ; 6: 325-43, 2010 Jul 21.
Article in English | MEDLINE | ID: mdl-20668715

ABSTRACT

Bisphosphonates are the leading drugs for the treatment of osteoporosis. In randomized controlled trials (RCTs), alendronate, risedronate, and zoledronate have shown to reduce the risk of vertebral, nonvertebral, and hip fractures, whereas RCTs with ibandronate show antifracture efficacy at vertebral sites. Bisphosphonates are generally well tolerated and safe. Nevertheless, adverse events have been noted, and it is important to consider the strength of the evidence for causal relationships. Effects on the gastrointestinal tract and kidney function are well recognized, as are transient acute-phase reactions. Atrial fibrillation was first identified as a potential adverse event in a zoledronate trial, but subsequent trials and analyses failed to substantiate an association with bisphosphonates. Case reports have suggested a relationship between oral bisphosphonates and esophageal cancer, but this has not been demonstrated in epidemiologic studies. A possible association between bisphosphonate use and osteonecrosis of the jaw (ONJ) has also been suggested. However, the risk of ONJ in patients with osteoporosis appears to be very low, with no evidence from prospective RCTs of a causal association. There are reports of occasional occurrence of subtrochanteric or diaphyseal fractures in osteoporotic patients, but an association with bisphosphonate therapy is not substantiated by epidemiologic studies or prospective RCTs.

8.
J Biomed Mater Res A ; 85(4): 993-1000, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-17907244

ABSTRACT

Bisphosphonates (BPs), which display a high affinity for calcium phosphate surfaces, are able to selectively target bone mineral, where they are potent inhibitors of osteoclast-mediated bone resorption. The dissolution of synthetic hydroxyapatite (HAP) has been used previously as a model for BP effects on natural bone mineral. The present work examines the influence of BPs on carbonated apatite (CAP), which mimics natural bone more closely than does HAP. Constant composition dissolution experiments were performed at pH 5.50, physiological ionic strength (0.15M) and temperature (37 degrees C). Selected BPs were added at (0.5 x 10(-6)) to (50.0 x 10(-6))M, and adsorption affinity constants, K(L), were calculated from the kinetics data. The BPs showed concentration-dependent inhibition of CAP dissolution, with significant differences in rank order zoledronate > alendronate > risedronate. In contrast, for HAP dissolution at pH 5.50, the differences between the individual BPs were considerably smaller. The extent of CAP dissolution was also dependent on the relative undersaturation, sigma, and CAP dissolution rates increased with increasing carbonate content. These results demonstrate the importance of the presence of carbonate in mediating the dissolution of CAP, and the possible involvement of bone mineral carbonate in observed differences in bone affinities of BPs in clinical use.


Subject(s)
Apatites/metabolism , Diphosphonates/metabolism , Bone Substitutes/metabolism , Diphosphonates/chemistry , Hydrogen-Ion Concentration , Imidazoles/metabolism , Microscopy, Electron, Scanning , Spectroscopy, Fourier Transform Infrared , Temperature , Zoledronic Acid
9.
Chem Biol ; 15(12): 1296-306, 2008 Dec 22.
Article in English | MEDLINE | ID: mdl-19101474

ABSTRACT

Cryptosporidiosis is a neglected disease without a wholly effective drug. We present a study demonstrating nitrogen-containing bisphosphonates (N-BPs) to be capable of inhibiting Cryptosporidium parvum at low micromolar concentrations in infected MDCK cells. Predictably, the mechanism of action is based on inhibition of biosynthesis of isoprenoids but the target enzyme is unexpectedly a distinctive C. parvum enzyme dubbed nonspecific polyprenyl pyrophosphate synthase (CpNPPPS). This enzyme produces various isoprenoid products larger than FPP and is inhibited by N-BPs at subnanomolar concentrations. It is part of an isoprenoid pathway in Cryptosporidium distinctly different from other organisms. The proposed mechanism of action is corroborated by crystal structures of the enzyme with risedronate and zoledronate bound showing how this enzyme's unique chain length determinant region enables it to accommodate larger substrates and products. These results, combined with existing data on their clinical use, demonstrate that N-BPs are very promising anticryptosporidial drug candidates.


Subject(s)
Anti-Infective Agents/therapeutic use , Cryptosporidiosis/drug therapy , Cryptosporidium parvum/drug effects , Cryptosporidium parvum/enzymology , Dimethylallyltranstransferase/metabolism , Diphosphonates/therapeutic use , Animals , Cattle , Cells, Cultured , Chromatography, Liquid , Crystallography, X-Ray , Dimethylallyltranstransferase/antagonists & inhibitors , Diphosphonates/pharmacology , Fluorescent Antibody Technique , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Protein Prenylation
10.
J Pharmacol Exp Ther ; 322(1): 228-35, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17412884

ABSTRACT

Apomine, a 1,1-bisphosphonate-ester with antitumor activity, has previously been reported to strongly down-regulate 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoA reductase), the rate-limiting enzyme in the mevalonate pathway responsible for the prenylation of proteins. Here, we show that although apomine down-regulated HMG-CoA reductase protein levels in myeloma cells, it did not inhibit protein prenylation, and apomine-induced apoptosis could not be prevented by mevalonate, indicating that apomine cytotoxicity is independent from its effects on HMG-CoA reductase. Instead, apomine cytotoxicity was prevented by the addition of phosphatidylcholine, which is similar to the previously reported ability of phosphatidylcholine to overcome the cytotoxicity of farnesol, whereas phosphatidylcholine had no effect on down-regulation of HMG-CoA reductase by apomine. These findings raised the possibility that apomine, independent from its own cytotoxic effects, could enhance the antitumor effects of the competitive HMG-CoA reductase inhibitor lovastatin via down-regulating HMG-CoA reductase. Indeed, treatment with apomine in combination with lovastatin resulted in synergistic decreases in viable cell number and induction of apoptosis. At the concentrations used, apomine down-regulated HMG-CoA reductase protein levels without being cytotoxic. Accumulation of unprenylated Rap1A by lovastatin was enhanced in the presence of apomine. Furthermore, synergy was completely prevented by mevalonate, and apomine did not synergize with desoxolovastatin, which does not inhibit HMG-CoA reductase. We conclude that the synergistic drug interaction results from an enhancement by apomine of the effects of lovastatin, mediated by down-regulation of HMG-CoA reductase by apomine. Thus, these findings demonstrate a novel strategy for enhancing the antitumor effects of lovastatin.


Subject(s)
Antineoplastic Agents/pharmacology , Diphosphonates/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Multiple Myeloma/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Down-Regulation , Drug Synergism , Humans , Mevalonic Acid/metabolism , Mevalonic Acid/pharmacology , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Phosphatidylcholines/pharmacology , Protein Prenylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL