Next Article in Journal
ELK1/MTOR/S6K1 Pathway Contributes to Acquired Resistance to Gefitinib in Non-Small Cell Lung Cancer
Previous Article in Journal
Effects of Nutraceutical Compositions Containing Rhizoma Gastrodiae or Lipoic Acid in an In Vitro Induced Neuropathic Pain Model
Previous Article in Special Issue
Sustained Effectiveness and Safety of Therapeutic miR-10a/b in Alleviating Diabetes and Gastrointestinal Dysmotility without Inducing Cancer or Inflammation in Murine Liver and Colon
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Statins—Their Role in Bone Tissue Metabolism and Local Applications with Different Carriers

by
Marcin Mateusz Granat
1,
Joanna Eifler-Zydel
2 and
Joanna Kolmas
2,*
1
Department of Clinical and Experimental Pharmacology, Faculty of Medicine, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
2
Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(4), 2378; https://doi.org/10.3390/ijms25042378
Submission received: 4 January 2024 / Revised: 13 February 2024 / Accepted: 15 February 2024 / Published: 17 February 2024
(This article belongs to the Special Issue Feature Papers in Molecular Endocrinology and Metabolism)

Abstract

:
Statins, widely prescribed for lipid disorders, primarily target 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase competitively and reversibly, resulting in reduced low-density lipoprotein cholesterol (LDL-C). This mechanism proves effective in lowering the risk of lipid-related diseases such as ischemic cerebrovascular and coronary artery diseases. Beyond their established use, statins are under scrutiny for potential applications in treating bone diseases. The focus of research centers mainly on simvastatin, a lipophilic statin demonstrating efficacy in preventing osteoporosis and aiding in fracture and bone defect healing. Notably, these effects manifest at elevated doses (20 mg/kg/day) of statins, posing challenges for systematic administration due to their limited bone affinity. Current investigations explore intraosseous statin delivery facilitated by specialized carriers. This paper outlines various carrier types, characterizing their structures and underscoring various statins’ potential as local treatments for bone diseases.

1. Introduction

Statins, originally derived from mold fungi, constitute a class of medications widely employed in treating lipid disorders, which stand as a primary risk factor for atherosclerosis and related conditions like coronary artery disease, ischemic cerebrovascular disease, and peripheral vascular diseases [1,2,3]. These compounds function as competitive, reversible inhibitors of the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, a key catalyst in the early stages of hepatic cholesterol biosynthesis. Approximately two thirds of the body’s cholesterol is synthesized de novo in the liver, with the transformation mediated by HMG-CoA reductase serving as the rate-limiting step in this process [4].
Inhibiting this enzyme’s activity diminishes cellular cholesterol levels, prompting the upregulation of low-density lipoprotein (LDL) receptors on the cell surface and enhancing the cellular uptake of LDL from the bloodstream. The consequential reduction in plasma LDL stands as the primary therapeutic effect of statins [4,5]. Additionally, these drugs exhibit the capability to modestly decrease triglyceride levels, elevate high-density lipoprotein (HDL) concentration, and reduce lipid susceptibility to oxidation [6]. Moreover, their cardioprotective effects may extend beyond lipid profile modulation, possibly involving increased endothelial nitric oxide synthesis, leading to improved vascular endothelial function [7].
Statins might mitigate cardiovascular event risk by stabilizing atherosclerotic plaques, inhibiting metalloproteinase secretion by macrophages, and impeding extracellular matrix breakdown, thus fortifying plaque fibrous membranes [8,9]. This drug class also influences the coagulation system, diminishing platelet aggregation and promoting fibrinolysis [6,10]. Statins may further modulate arterial wall cell numbers by restricting smooth muscle cell proliferation and enhancing apoptosis [11]. Notably, they exhibit antioxidant, immunomodulatory, and anti-inflammatory effects, addressing the emerging emphasis on chronic inflammation’s role in atherosclerosis development [12,13]. Their pleiotropic effects and generally favorable patient tolerance make statins a cornerstone in cardiovascular disease treatment (see Figure 1).
Commonly utilized statins include mevastatin, lovastatin, and pravastatin (natural statins); simvastatin (a semi-synthetic statin); and fluvastatin, rosuvastatin, pitavastatin, cerivastatin, and atorvastatin (synthetic statins) [14,15]. Their chemical formulas are depicted in Figure 2. Notably, variations in their chemical structures impact lipophilicity, hydrophilicity, and subsequent absorption, distribution, metabolism, and excretion [14,16].
The molecule of each statin consists of three main parts: an HMG-CoA analogue, a complex ring structure responsible for binding the statin to HMG-CoA reductase, and a side chain structure determining solubility. Statins vary greatly in solubility due to the presence/absence of polar moieties in their predominantly hydrophobic backbones. Lipophilic statins can easily penetrate deeper into cell membranes and enter cells through passive diffusion, and are therefore widely distributed in various tissues. They are metabolized by cytochrome P450 (CYP) enzymes upon binding to the membrane. In turn, hydrophilic statins remain bound to the polar surface of the membrane and require the transport of proteins into the cell, show greater hepatoselectivity and lower potential for uptake by peripheral cells than lipophilic statins, and are mostly eliminated unmetabolized.
Upon oral administration, statins undergo substantial hepatic first-pass effects, limiting their systemic bioavailability to 5–30% of the administered dose due to high hepatic uptake. In plasma, nearly 95% of statins and their metabolites are protein-bound [14,16]. Some, like simvastatin, are administered as inactive prodrugs requiring hydrolysis catalyzed by esterases and peroxidases for therapeutic effect. Despite generally good tolerability, serious adverse effects, especially at higher doses, may occur. Serving as substrates for CYP3A4 (cytochrome P450 family 3 subfamily A member 4) isoenzyme and OATP-2 (organic anion transporting polypeptide-2), statins can engage in clinically significant interactions with other drugs affecting these proteins.
Common adverse effects of statin therapy include elevated liver transaminases, with hepatotoxicity and nephrotoxicity as potential risks. Some rare but severe adverse effects are myopathy and rhabdomyolysis, with the risk directly proportional to statin concentration in plasma [17,18].
Currently, research explores statin use beyond cardiac diseases related to lipid disorders, including potential applications in bone diseases, injuries, and tissue defects [19,20].
Our study focuses on investigating the effects of statins on bone tissue metabolism, considering different carriers for drug application.
It should be noted that statins, like most drugs, are typically administered orally. However, the oral administration of statins presents drawbacks, such as first-pass metabolism in the liver and degradation in the gastrointestinal tract, resulting in limited bioavailability. Another concern is the occurrence of adverse effects such as myopathy, kidney and liver damage, and rhabdomyolysis. Hence, alternative routes of statin administration are under investigation, particularly for potential use in bone diseases where prolonged action directly within the bone tissue is required.
We conducted a comprehensive search on databases such as PubMed, Embase, IEEE Explorer, and Google Scholar, accessing them on 27 December 2023, due to their leading role in scientific research and English-language content. The search phrases included “statin bone tissue”, “statin bone metabolism”, “statin bone application”, “statin bone”, “statin osteoporosis”, and “statin biomaterials”. The publications considered spanned from 1999 to 2023, complemented by insights from pharmacology and pharmaceutical chemistry books, contributing approximately 15% of the overall information.

2. Statins—Their Role in Bone Tissue Metabolism

As early as the late 20th century, observations indicated that statins can stimulate osteogenesis and offer therapeutic benefits to patients with osteoporosis [21,22,23,24,25]. Subsequent research has unveiled their potential as robust modulators of bone healing and the inflammation process, demonstrating their capacity to stimulate osteoinduction and angiogenesis—key phenomena for bone reconstruction [26,27]. They also exhibit the ability to limit the loss of bone tissue by inhibiting both osteoclastogenesis and the apoptosis of osteoblasts [28,29,30]. Therefore, the beneficial effect of statins on bone tissue metabolism results from their action through various mechanisms, as confirmed in numerous animal models of osteoporosis and mechanical bone damage [31,32,33,34,35]. Lipophilic statins, particularly simvastatin, lovastatin, and atorvastatin, appear to be the most effective in preventing osteoporosis and supporting the healing of bone fractures and defects [22,23,36].
Several mechanisms have been proposed to explain the anabolic effect of statins on bone tissue (see Figure 3). Firstly, these drugs can stimulate the formation of new tissue by increasing the expression of a gene for bone morphogenetic proteins (BMPs), specific growth factors regulating osteoblast and chondroblast proliferation and differentiation [21,26,37,38]. Statins increase BMP-2 expression by modulating small GTP-binding proteins, particularly the Ras and Rho proteins [37]. It is important to note that only lipophilic statins can activate the promoter of this gene. Another suggested mechanism related to the primary target point of statins involves blocking HMG-CoA reductase, inhibiting the conversion of HMG-CoA to mevalonate, and indirectly limiting the biosynthesis of its derivatives, such as farnesyl pyrophosphate or geranylgeranyl pyrophosphate [22,36,37,38,39]. These derivatives adversely affect bone tissue metabolism. For example, farnesyl diphosphate can activate the glucocorticoid receptor, potentially increasing bone resorption and reducing osteoblast proliferation. Geranylgeranyl pyrophosphate likely also negatively affects osteogenesis [21,22]. By affecting the TGFβ/Smad3 pathway, statins protect osteoblasts against apoptosis. TGFβ (transforming growth factor beta) activates Smad3, promoting the formation of new bone tissue by stimulating the synthesis of extracellular matrix proteins, intensifying mineralization, and increasing alkaline phosphatase activity—a marker of early osteoblast differentiation (see Figure 3) [28,30].
Studies have shown that bone marrow stem cells in the presence of statins exhibit increased activity of this protein, with the drug released from the carrier stimulating the mineralization of the extracellular matrix [40,41,42,43]. The loss of Smad3 results in the apoptosis of osteoblasts and osteocytes, leading to decreased bone mass. By enhancing its expression in these cells, statins may contribute to their protection. These drugs can also modulate osteoclastogenesis by inhibiting it through the OPG/RANKL/RANK pathway [40]. RANKL (receptor activator of NF-κB ligand), RANK (receptor activator of NF-κB), and osteoprotegerin (OPG) play key roles in this process. OPG reduces the proliferation of osteoclasts, while RANK, belonging to the tumor necrosis factor receptor (TNFR) superfamily, enhances osteoclastogenesis. RANKL, being a membrane protein on the surface of osteoblasts, acts as a key factor for osteoclast function by blocking their apoptosis [26]. Mice lacking RANKL or RANK developed significant osteoporosis due to the complete absence of osteoclasts and the consequent lack of bone resorption and remodeling. In turn, OPG-deficient mice showed osteoporosis resulting from an increased number of osteoclasts. The balance of this signaling pathway is crucial for proper, balanced bone remodeling, and statins can modulate it.
In studies on a mouse osteocyte cell line, these drugs have been shown to increase the expression of the OPG gene and decrease the expression of the RANKL gene [22,24]. It has also been proven that the number of osteoclasts decreases with increasing statin concentration. Studies also suggest that statins may promote neovascularization because they increase gene expression for vascular endothelial growth factor (VEGF) and fibroblast growth factor 2 (FGF-2). The processes of osteogenesis and angiogenesis are closely related, and coordinated vascularization is of key importance in the process of bone tissue reconstruction. It not only provides oxygen and nutrients to the developing tissue, but also allows the transport of mesenchymal stem cells from the bone marrow, further supporting bone regeneration [44,45,46,47,48,49].
The ability of statins to simultaneously stimulate these two processes makes them even more promising potential drugs in the therapy of bone damage and defects [50,51]. Unfortunately, systemically administered statins have a low affinity for bone tissue, making the systemic use of these drugs in attempts to treat bone diseases potentially inconclusive as to their effectiveness [51,52]. Additionally, as mentioned previously, most statins undergo a significant first-pass effect, further limiting their bioavailability in the bones. Therefore, orally administered statins in clinical doses required for lipid-lowering therapy are not able to exert a strong positive effect on the anabolism and catabolism of bone tissue [51,52,53]. Achieving these effects would require administering drugs at much higher doses, significantly increasing the risk of systemic adverse effects. Therefore, the topical application of statins directly to the bone tissue may be an alternative option that allows for bypassing the hepatic metabolism of these drugs, increasing their bioavailability, and reducing the dose. Other resulting benefits include reducing the risk of systemic adverse effects, as well as eliminating the need for repeated administration of the drug.

3. Statin Local Delivery Methods and Carriers

The local administration of statins in the case of bone tissue disorders seems justified for several reasons. Firstly, bone tissue is highly vascularized, and systemic treatment requires the use of large doses of therapeutic substances to achieve an adequate concentration at the target site [54,55]. Therefore, the direct administration of statins to bone tissue allows for a reduction in dosage compared to systemic administration [56]. Additionally, the carrier for intraosseous drug delivery can provide a matrix for the infiltration of mesenchymal cells and often serves as a filling material for bone defects [57,58]. It is essential to emphasize that the optimal carrier should exhibit appropriate degradation rates to ensure the proper growth of newly formed bone tissue while preventing the formation of fibrous tissue and fibrous encapsulation of the carrier. The local administration of statins can effectively reduce the risk of adverse effects, primarily myopathy characterized by muscle pain and weakness, the elevation of creatine kinase (CK) levels in the serum, and, in extreme cases, rhabdomyolysis, which is a life-threatening condition [57,58].
It is worth noting that our study did not focus on the specific doses of statins used in individual research. The results regarding statin dosage in the treatment of bone tissue diseases remain inconclusive. Depending on the dose, these drugs can exhibit either positive or negative effects on bone health. For instance, high doses of simvastatin (20 mg/kg bw/day) have been shown to stimulate new bone tissue formation, while low doses (1 mg/kg bw/day) may inhibit reconstruction and increase bone resorption [59]. Despite promising results, the practical applications of statins in treating bone diseases require further research, including optimizing dosage and exploring effective methods of application.
Table 1 presents the literature review results regarding carriers for statin delivery into the bone tissue. The data indicate that statin delivery systems used mainly simvastatin; in a few cases, lovastatin [60,61,62]; pitavastatin [63]; rosuvastatin [64,65,66]; and fluvastatin [67,68,69,70,71].
Carriers were equipped with statins in various ways. The simplest method involves impregnating bone scaffolds, but this has the drawback of rapid drug dissolution in the bloodstream, leading to reduced bioavailability. Alternatively, spray techniques and ultrasonic dispersion can coat statins onto bone scaffolds, allowing for only low-dose application. Other approaches involve micro- and nanospheres, chemical compound combinations, polymerization, and blending with biomaterials, each with its advantages and limitations.
Based on their chemical composition, carriers can be categorized into those made of inorganic materials, natural polymers, synthetic polymers, and polymer/inorganic material composites. The choice of carrier materials is dictated by the structure and properties of bone tissue, composed of an organic matrix (mainly collagen type I fibers) and an inorganic phase, primarily biological apatite, a nanocrystalline calcium phosphate. Inorganic carriers offer high compatibility, bioactivity, and bioresorbability, but may lack sufficient mechanical strength, especially when serving as bone defect fillers. On the other hand, polymers, despite high biocompatibility, may lack the necessary hardness. Therefore, composite carriers seem to be the optimal solution, providing adequate mechanical properties, biodegradation, and controlled statin release at the target site.
In the case of inorganic carriers, calcium phosphates are primarily utilized due to their similarity to biological apatite. Among the most commonly used calcium phosphates, hydroxyapatite and crystalline calcium orthophosphates (in both low- and high-temperature forms; respectively, β and α) can be mentioned, with α-TCP being a compound that is much more soluble than β-TCP and hydroxyapatite. α-TCP has been employed in the form of bone cement, a material that solidifies in reaction with water to form a hard filling for bone defects. β-TCP has also been used in the form of cement, as well as a biphasic material with hydroxyapatite, thereby increasing the material’s bioresorbability and bioactivity. In the study [72], unconventional hydroxyapatite in the form of nanofibers was used, while in [73], together with simvastatin, it served as a coating for a titanium implant. In this case, hydroxyapatite was used as a factor for improving osteointegration and as a carrier for delivering a statin. The studies [74,75] focus on the use of a scaffold of calcium sulfate loaded with simvastatin. Research conducted using inorganic materials was carried out in both in vitro conditions (primarily on BMSCs cells) and in vivo animal models (rats, mice, and rabbits). In all the presented studies, the simultaneous administration of the inorganic material with statin resulted in the increased formation of new bone tissue, increased density of the created tissue, and improvement in bone tissue regeneration, which were observed between the 3rd and 12th week of the experiment.
Significantly more research has focused on the utilization of both natural and synthetic polymers, as well as composite materials, as potential carriers for statins. This emphasis likely stems from the inherent drawbacks of inorganic materials (primarily calcium phosphates), notably, their brittleness and, thus, insufficient mechanical strength, which render them unsuitable for areas subjected to high stresses. Polymers are characterized by high flexibility, whereas calcium phosphate/polymer composites combine the hardness of inorganic materials with the elasticity of polymers, ensuring excellent mechanical properties akin to those of bone tissue, which is also a natural composite.
Among the natural polymers utilized for statin carriers, chitosan has been predominant, with collagen or prepared atelocollagen being alternative materials. Gelatin has also been employed in hydrogel form. Concerning synthetic polymers, a wide array have been utilized, including both biodegradable (e.g., PLA, PLLA, PCL, PLGA) and non-biodegradable (PUR, PEEK) polymers. The composite materials described in the available literature consist of hydroxyapatite or beta-TCP combined with polymers such as poly(e-caprolactone), poly(glycerol sebacate), gelatin-nanofibrillar cellulose, PCL, and PLGA. Polymer and composite materials are prepared in various forms, including porous scaffolds, microspheres, nanomicelles, fibers, nanoparticles, and coatings on titanium surfaces. The available literature predominantly comprises in vivo studies on rats and rabbits, which consistently indicate increased mineralization, bone tissue formation, and improvements in bone parameters in each case.
A significant aspect investigated under laboratory conditions is the release of statins from previously obtained carriers. Various release profiles have been observed depending on the chemical composition of the carrier, its form, and its preparation method. Generally, the constructed carriers are designed to release statins gradually over an extended period (from 50–90% within 7 days) [76,77,78]. In the study by [78], a typical microporous structure of calcium phosphate foam was employed, which proved to be a suitable carrier for pitavastatin: the “burst release” effect was relatively minimal, followed by the gradual release of the statin over 72 h. Various approaches have been applied to prolong the release time. For instance, delayed release was achieved in carriers derived from biomimetic beta-TCP by coating it with an additional layer of apatite [63]. This resulted in a matrix that released 20% less simvastatin over 7 days. In the study by [79], significantly prolonged release of simvastatin was obtained from PCL microspheres contained within a collagen coating covering PET polymer (constructing artificial ligaments). Over 14 days, slightly over 50% of the drug was released, and approximately 75% within nearly 40 days.
Table 1. The effects of various statins combined with different types of carriers on bone.
Table 1. The effects of various statins combined with different types of carriers on bone.
Statin (Dose)CarrierModel of Study/Duration of TreatmentFindingsReference
Inorganic materials
Simvastatin
(0.25 and 0.5 mg)
α-TCP (α-tricalcium phosphate)In vivo/69 healthy Wistar adult rats; 8 weeksBone regeneration in rat calvarial defects was noticed[80]
Simvastatin
(0.1 mg)
α-TCP (α-tricalcium phosphate)In vivo/72 healthy Wistar rats; 8 weeksStimulation of bone regeneration occurred[81]
Simvastatin
(6% concentration)
Apatite cementsIn vivo/18 ovariectomized rats; 3 weeksBone mineral density increased[82]
Simvastatin
(4 mg/mL)
β-TCP (β-tricalcium phosphate)In vitro/drug release in simulation
body fluid solution; 7 days
Controlled release of the drug with a reduction of approximately 25% compared to control samples was observed [76]
Simvastatin
(0.1 mg)
β-TCP (β-tricalcium phosphate)In vivo/72 healthy Wistar rats; 8 weeksStimulation of bone regeneration occurred[81]
Simvastatin
(0.1, 0.9, and 1.7 mg)
β-TCP (β-tricalcium phosphate)In vivo/162 healthy male Sprague Dawley rats; 6 weeksDecreased mineral apposition was observed, and after 26 weeks, increased fibrous area fraction, β-TCP area fraction, and particle size and number were noticed[83]
Simvastatin
(0.1 mg)
Calcium phosphateIn vivo/15 healthy female Wistar rats; 8 weeksBone-like tissue was formed[84]
Simvastatin
(1, 5, and 10% concentrations)
Calcium phosphate cement (an equimolar mixture
of tetracalcium phosphate and dicalcium phosphate
anhydrous)
In vivo/40 healthy New Zealand white rabbits; 4 weeksNew bone formation was observed[85]
Simvastatin
(0.5 and 0.25 mg/g cement)
Calcium
phosphate cement
In vitro/Saos-2 cells; 7 daysPromotion of bone formation was noticed[86]
Simvastatin
(0.1, 0.25 and 0.5 mg/g cement)
Calcium phosphate cement (β-tricalcium phosphate and monocalcium
phosphate anhydrous in molar ratio of 1:1)
In vitro/bone marrow
macrophages isolated from mice; 12 days
Inhibition of osteoclastic differentiation was observed[87]
Simvastatin
(0.5 mg)
Calcium sulphateIn vivo/18 healthy New Zealand white rabbits; 8 weeksAn area of newly formed bone was noticed[74]
Simvastatin
(1 mg)
Calcium sulphateIn vivo/45 healthy male Wistar rats; 8 weeksStimulation of bone regeneration was observed[75]
Simvastatin
(0.125 mg)
HydroxyapatiteIn vivo/12 healthy New Zealand
white rabbits; 8 weeks
Increased bone volume was noticed[72]
Simvastatin
(0.45 mg)
HydroxyapatiteIn vivo/20 adult Japanese white rabbits; 8 weeksNew bone formation was observed[73]
Simvastatin
(0.1 mg)
HydroxyapatiteIn vivo/72 healthy Wistar rats; 8 weeksStimulation of bone regeneration occurred[88]
Simvastatin
(10 mM)
Hydroxyapatite-coated titaniumIn vitro/bone mesenchymal stem cells (BMSCs); 14 days
In vivo/48 adult male Sprague Dawley rats; 6 weeks
In vitro: Enhanced osteogenesis and osteointegration occurred
In vivo: Maximum forces of the Sim-Low and Sim-High groups were significantly higher
than those of the Control and HA groups
[88]
Simvastatin
(0.01 and 0.001 g/L)
Mesoporous titania thin filmsIn vitro/MC3T3-E1 pre-osteoblasts cells; 21 daysIncubation the formation of a complex network of pre-collagen filaments was observed[89]
Simvastatin
(10 mM)
NanohydroxyapatiteIn vivo/36 ovariectomized Sprague Dawley rats; 12 weeksNew bone formation around implant surfaces was noticed[90]
Simvastatin
(5 mg/kg)
Titanium implantsIn vivo/54 ovariectomized Sprague Dawley rats; 84 daysThe bone healing process was observed[91]
Simvastatin
(50 μg/implant)
Titanium Kirschner wires coated with PDLLA (poly(D,L-lactide)) and PUR (polyurethane)In vivo/200 female Sprague Dawley rats; 6 weeksImproved fracture healing was present[92]
Natural and synthetic polymers and composites
Simvastatin
(5, 10, and 20 mg/15 g solutions)
3D—PGHS (as-fabricated 3D fibrous
scaffolds of poly (ε-caprolactone) poly (glycerol-sebacate) hydroxyapatite nanoparticles)
In vitro/human
mesenchymal stem cells (hMSCs) and human umbilical vein endothelial cells (HUVECs); 7 days
Osteogenic differentiation and migration as well as tube formation occurred[93]
Simvastatin
(10 mg/mL)
ALN-CD (alendronate—β-cyclodextrin) conjugateIn vivo/44 healthy female Sprague Dawley rats; 4 weeksThe study stated that ALN-CD conjugates not only act as tissue-specific carriers but preserve new bone formation[94]
Simvastatin
(2.5 mg/mL dissolved in 0.2 mL water)
ACS (atelocollagen sponge)In vivo/20 adult male Japanese white rabbits; 12 weeksNew bone formation was observed[95]
Pitavastatin
(0.1 μM)
β-cyclodextrin-grafted chitosan and gelatinIn vivo/40 specific-pathogen-free male Sprague Dawley rats; 4 weeksBone formation was observed[63]
Lovastatin
(1.2 mg/layer)
β-TCP/PCL (β-tricalcium phosphate/polycaprolactone) microchips and PCL nanofiber membranesIn vivo/24 ovariectomized New Zealand rabbits; 12 weeksBone parameters significantly improved[60]
Simvastatin
(0.5 μM)
BPPD (bis(PLGA-phe-PEG)-qDETA)In vitro/bone marrow mesenchymal stem cells (BMSCs); 6 daysPromotion of osteogenesis in BMSCs was observed[96]
Simvastatin
(4 mg)
ChitosanIn vitro/BMSC culture; 14 days
In vivo/6 healthy ovariectomized rats; 8 weeks
In vitro: A positive effect on cell proliferation was noticed
In vivo: The bone regeneration process was observed
[97]
Simvastatin
(0.25 mg)
ChitosanIn vivo/21 healthy Sprague Dawley rats; 8 weeksNo significant difference between the
control and experimental
groups was found
[98]
Simvastatin
(0.05 mg)
ChitosanIn vitro/human bone marrow
mesenchymal stem cells (hbMMSCs); 14 days
Chitosan scaffold is a bioactive compatible material with regenerative
potential for hBMMSCs
[99]
Simvastatin
(5 mg/0.5 mL)
ChitosanIn vivo/12 healthy male albino New Zealand rabbits; 6 weeksThe process of bone regeneration was noticed[100]
Simvastatin
(2.5 mg/mL)
Collagen graftIn vivo/9 healthy New Zealand white rabbits; 14 daysAn osteoinductive effect was noticed[101]
Simvastatin
(2.5 mg/mL)
Collagen matrixIn vivo/14 healthy New Zealand white rabbits; 14 daysNew bone formation was observed[102]
Rosuvastain
(0.1, 0.5, and 2.5 mg/mL)
Collagen spongesIn vivo/18 healthy New Zealand white female rabbits; 4 weeksStimulation of bone formation occurred[64]
Simvastatin
(1% concentration)
Gel (composed of polymer 2% HPMC K100M and 20% poloxamer 407)In vivo/72 healthy Sprague Dawley rats; 56 daysBone regeneration was observed[103]
Simvastatin
(250 μg)
Gelatin hydrogelIn vivo/60 healthy virgin female Sprague Dawley rats; 8 weeksAcceleration of fracture healing was observed[104]
Fluvastatin
(1 mM)
Gelatin hydrogelIn vivo/60 healthy male Sprague Dawley rats; 4 weeksInduced osteogenesis in rat calvarial bone was observed[67]
Simvastatin
(2.5 mg/mL dissolved in 0.2 mL water)
Gelatin hydrogelIn vivo/20 adult male Japanese white rabbits; 12 weeksNew bone formation was observed[95]
Simvastatin
(125 μg)
Gelatin hydrogelIn vivo/42 healthy mature mature Japanese rabbits; 8 weeksPromotion of tendon–bone healing at an early stage via angiogenesis and osteogenesis occurred but did not affect the biomechanical property in the long term[105]
Simvastatin
(0.5 μM)
GNTS (gelatin-nanofibrillar
cellulose- β tricalcium phosphate)
In vivo/30 healthy male Sprague Dawley rats; 8 weeksNewly formed bone structures were noticed [106]
Simvastatin
(100 nM)
Methylated β-cyclodextrinsIn vitro/MC3T3-E1 cells; 14 daysALP production and the expression of bone sialoprotein and osteocalcin were noticed[107]
Simvastatin
(2.2 mg)
Methylcellulose gel and PLA
(polylactide membrane)
In vivo/56 healthy female ICR Swiss mice; 44 daysAn increase in bone thickness was observed[108]
Simvastatin
(0.5 mg)
NLC (nanostructured lipid carrier)In vivo/20 healthy rabbits; 4 weeksEnhanced bone formation was observed[109]
Simvastatin
(20 μg)
PCL (poly (ε-caprolactone))In vivo/90 healthy Wistar albino rats; 6 monthsAn increase in bone mineralization was noticed[110]
Simvastatin
(100 μg/mL)
PCL (poly (ε-caprolactone)) and collagenIn vitro/primary human umbilical vein endothelial cells (pHUVECs); 21 daysEnhanced osteogenic differentiation was noticed[111]
Simvastatin
(2.2 mg)
PCL (poly (ε-caprolactone)) fibrous sheets and structured nanofibers with a gelatin shellIn vivo/24 healthy male New Zealand white rabbits; 12 weeksGood cell viability and effective osteoinductive and barrier properties were observed[112]
Simvastatin
(5% concentration)
PCL (poly (ε-caprolactone)) nanofibers loaded with polyaniline-coated titanium oxide nanoparticles (TiO2/PANI)In vitro/MC3T3-E1 osteoblast cells; 14 daysProfound cell proliferation was observed[113]
Simvastatin
(dose not stated)
PCL-HA (poly(ε-caprolactone- hydroxyapatite)) microspheresIn vitro/bone marrow mesenchymal stromal cells (BMSCs); 21 days
In vivo/3 healthy Sprague Dawley rats; 8 weeks
Osteogenic differentiation of BMSCs was noticed in vitro.
Promotion of vascular network and functional bone formation was observed in vivo
[114]
Simvastatin
(5% concentration)
PCL-HA (poly(ε-caprolactone-hydroxyapatite)) composite coated
on biodegradable Mg alloy nanofibers
In vitro/MC3T3 mouse
osteoblast cell line; 7 days
An increase in bone regeneration and control of its degradation occurred[115]
Simvastatin
(from 2.5 × 10−6 to 2.5 × 10−10 M)
PECL (poly (ethylene
glycol))-poly(ε-caprolactone))
In vitro/human osteoblast-like MG-63 cells; 7 daysOsteoblast differentiation and mineralization were observed[116]
Fluvastatin
(0.01 and 0.1 μM)
PEGDM (poly (ethylene glycol) dimethacrylate)In vitro/human mesenchymal stem cell (hMSC); 14 daysAn increase in hMSC CBFA1, ALP, and COL I gene expression was noticed, which indicated an effect on osteogenic differentiation[68]
Simvastatin
(0.5 mg)
PEG-PLA (polyethylene glycol- polylactic acid) polymeric
nanomicelles
In vivo/6 healthy New Zealand white rabbits; 4 weeksOsteoblasts and new capillaries around the trabecular bone were found[117]
Simvastatin
(0.28 and 0.31 μg/mg)
PEG-PLGA (poly (ethylene glycol))-block-poly(lactic-co-glycolic acid)In vivo/6 healthy ovariectomized Sprague Dawley rats; 12 weeksA bone formation effect was present[118]
Simvastatin
(2 mg/mL)
PEEK (polyetheretherketone)bio-compositeIn vitro/MC3T3-E1 pre-osteoblasts; 14 daysOsteogenic differentiation was observed[79]
Simvastatin
(1 mg/mL)
PET (polyethylene terephthalate)In vivo/36 healthy New Zealand white rabbits; 8 weeksBone healing was observed[119]
Fluvastatin
(75 μg)
PGA (propylene glycol alginate)In vivo/60 healthy female Wistar rats; 2 weeksAn increase in bone volume was noticed[69]
Fluvastatin
(75 μg)
PGA (propylene glycol alginate)In vivo/48 healthy female Wistar rats; 4 weeksAn increase in bone–implant contact and mineralized bone volume was observed[70]
Lovastatin
(1 mg/mL)
PGA-PEG (poly(glycolide)-poly(ethylene glycol))In vitro/mice; 7 daysThe study showed that the maximum tolerated dose in mice can be increased[61]
Simvastatin
(5 mg)
PLA (polylactic acid)In vivo/16 healthy New Zealand white rabbits; 12 weeksHigh-density spots were observed and the margins of the defects were more irregular[120]
Simvastatin
(4mg/g PLG)
PLG (poly(lactide-co-glycolide))In vitro/rat bone marrow cells; 10 daysBone cell mineralization was observed[121]
Fluvastatin
(0.5 and 1 mg/kg)
PLGA (poly (lactic-co-glycolic acid))In vivo/40 healthy Sprague Dawley rats; 4 weeksMore bone trabeculae were observed[71]
Simvastatin
(1 mg)
PLGA (poly (lactic-co-glycolic
acid))
In vitro/human osteoblastic cell line (hFOB); 11 daysOsteoblastic differentiation was observed[122]
Simvastatin
(0.6% concentration)
PLGA (poly (lactic-co-glycolic acid)) coated around titaniumIn vitro/human gingival fibroblasts (HGFs) and stem cells from human exfoliated deciduous teeth (SHEDs); 7 daysHigh cell viability was observed[123]
Simvastatin
(20 mg/kg)
PLGA (poly (lactic-co-glycolic acid))-encapsulated hydroxyapatiteIn vivo/24 healthy female Wistar rats; 45 daysSignificant improvement in the bone surface was observed[124]
Simvastatin
(2, 5, and 8% concentrations)
PLGA (poly (lactide-co-glycolide)) microspheres using the electrospraying methodIn vitro/human MG-63
osteoblast cells; 7 days
Good biocompatibility of the electrosprayed PLGA microspheres was observed, which increased in the presence of a statin[125]
Simvastatin
(5% concentration)
PLGA (poly (lactic-co-glycolic acid)) microspheres loaded into hydrogel-loaded BCP (biphasic calcium phosphate)In vitro/MC3T3-E1 pre-osteoblast cells; 7 daysBone remodeling gene and protein expression were observed[126]
Simvastatin
(3 mg of simvastatin/PLGA)
PLGA (poly (lactic-co-glycolic
acid)) with a rapidly absorbable calcium sulfate
In vivo/60 healthy male Sprague Dawley rats; 12 weeksOsteogenic and angiogenic activity and bone healing process increased[127]
Simvastatin
(0.5 μM)
PLGA-PEG (poly (lactic acid-co-glycolic acid)-polyethylene glycol))In vitro/BMSCs; 6 daysImprovement in bone healing was observed[96]
Simvastatin
(1 mg)
PLLA (poly-L-lactide)In vivo/29 healthy male Sprague Dawley rats; 8 weeksNew bone formation and increased bone mineral density were observed[128]
Simvastatin
(~ 120 mg/kg/day)
Polyethylene particlesIn vivo/21 healthy female and male C57BL/J6 mice; 14 daysNew bone formation was noticed[129]
Simvastatin
(2.2 mg)
Poly(ethylene glycol)-block-poly(simvastatin)In vivo/144 healthy male Sprague Dawley rats; 8 weeksA significant osteogenic effect was noticed[130]
Simvastatin
(0.5 mM)
Poly (N-isopropylacrylamide)
Brush-modified mesoporous hydroxyapatite
In vivo/20 ovariectomized Wistar rats; 6 weeksPromotion of osteogenesis was observed[131]
Lovastatin
(200 μg/g of foam)
Polyurethane (PUR)In vivo/6 healthy male Sprague Dawley rats; 4 weeksAn increase in the density of the newly formed bone was observed[62]
Simvastatin
(5 mg/mL)
Polyurethane nanofibersIn vivo/32 healthy adult male
Wistar rats; 4 weeks
Induction of bone healing was noticed[132]
Rosuvastatin
(5 mg/mL)
PVA-SF (polyvinyl alcohol–silk fibroin) core-shell nanofibersIn vitro/Human adipose-derived
stem cells
(hADSCs); 21 days
Improved cell proliferation and osteogenic differentiation occurred[65]
Simvastatin
(2 mg)
SIM-DOME (methylcellulose gel under
a polylactic acid dome membrane)
In vivo/44 healthy mature female Sprague Dawley rats; 24 daysNew bone formation was observed[94]
Simvastatin
(0.5–1 μM)
Poly(l-lactide-co-glycolide)In vivo/4 healthy male C57/BL/6 J mice; 12 weeksNew bone formation was observed[133]
Organic non-polymer materials
Simvastatin
(10 mg)
Gelfoam soaked with normal salineIn vivo/50 humans; 12 weeksAn increase in bone density occurred[134]
Simvastatin
(0.1 and 1 mg)
Hyaluronic acid (HA)
hydrogels
In vivo/12 healthy male New Zealand rabbits; 8 weeksA significant influence on osteogenesis was observed[135]
Simvastatin
(0.01, 0.1, and 1 μM)
Injectable tissue-engineered bone (ITB)In vitro/human adipose-derived stromal cells (hADSCs); 14 days
In vivo/26 healthy BALB/C homozygous nude mice; 4 weeks
Osteoblastic differentiation in vitro and bone formation in vivo were observed[136]
Rosuvastatin
(3 mg/mL)
SF (silk fibroin) nanofibersIn vitro/Human adipose-derived
stem cells
(hADSCs); 21 days
Osteogenic gene differentiation was observed[66]

4. Conclusions

The treatment of large bone defects and effective bone tissue regeneration remain challenges in orthopedics and dental surgery. Constant efforts are made to identify both bone substitute materials with appropriate physicochemical, mechanical, and biological properties, as well as factors that would expedite the process of new bone tissue formation and the development of blood vessels within it.
Therefore, the discovery that statins, drugs that have long been used to lower cholesterol levels in the blood, exhibit strong proangiogenic and osteogenic effects has generated significant interest in their potential use as supportive factors in the treatment of serious bone disorders. Our literature review has demonstrated that statins, acting pleiotropically through various mechanisms, attenuate osteoblast apoptosis, influence the reduction in osteoclast differentiation, and stimulate bone tissue formation, among other effects, by promoting osteoblast differentiation.
Of course, like most drugs, statins exhibit numerous adverse effects, which can be particularly hazardous when high doses are required for the therapy of bone disorders. Hence, much research is focused on developing local delivery systems for statins to bone tissue, which, in addition to minimizing adverse effects, would improve bioavailability and allow for a reduction in drug dosage.
The results of this review suggest that carriers based on inorganic materials (primarily various calcium phosphates and calcium sulfate), polymeric materials (especially chitosan, collagen poly-e-caprolactone, and poly(lactide-co-glycolide)), and inorganic–organic composite materials are capable of delivering statins locally to bone tissue, releasing them over time, and significantly enhancing bone tissue regeneration compared to mere filling. The findings from these studies are promising and highlight the need for further investigation. It is worth noting that only one human study has been published so far; thus, clinical trials should proceed.
While there are numerous works in the literature dedicated to the issue of bone carriers for statins, there are no articles focusing on the combination of different antiresorptive and osteogenic factors with statins. Future research could be dedicated to examining the impact of the simultaneous administration of statins and other factors stimulating bone tissue regeneration (e.g., magnesium ions, strontium, or silicates). These could be introduced in the form of substituted calcium phosphate. Future studies should also focus on determining the appropriate statin dosage under in vivo conditions to ensure therapy effectiveness and safety. It is worth noting that the majority of studies only address one statin, simvastatin. Therefore, further research should also include other representatives of this group.

Author Contributions

M.M.G. wrote the manuscript, prepared the table with the literature, and supervised the manuscript and the figures; J.E.-Z. wrote the manuscript; J.K. conceived the idea, wrote and supervised the manuscript, prepared the figures, and supervised the table with the literature. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Stancu, C.; Sima, A. Statins: Mechanism of action and effects. J. Cell Mol. Med. 2001, 5, 378–387. [Google Scholar] [CrossRef]
  2. Maron, D.J.; Fazio, S.; Linton, M.F. Current perspectives on statins. Circulation 2000, 101, 207–213. [Google Scholar] [CrossRef] [PubMed]
  3. Egom, E.E.; Hafeez, H. Biochemistry of Statins. Adv. Clin. Chem. 2016, 73, 127–168. [Google Scholar] [CrossRef] [PubMed]
  4. Hunninghake, D.B. HMG-CoA reductase inhibitors. Curr. Opin. Lipidol. 1992, 3, 22–28. [Google Scholar] [CrossRef]
  5. Watson, K.E.; Fonarow, G.C. The past, present, and future of statin therapy. Rev. Cardiovasc. Med. 2005, 6, 129–139. [Google Scholar]
  6. Puccetti, L.; Pasqui, A.L.; Auteri, A.; Bruni, F. Mechanisms for antiplatelet action of statins. Curr. Drug Targets Cardiovasc. Haematol. Disord. 2005, 5, 121–126. [Google Scholar] [CrossRef] [PubMed]
  7. Mohammadkhani, N.; Gharbi, S.; Rajani, H.F.; Farzaneh, A.; Mahjoob, G.; Hoseinsalari, A.; Korsching, E. Statins: Complex outcomes but increasingly helpful treatment options for patients. Eur. J. Pharmacol. 2019, 863, 172704. [Google Scholar] [CrossRef] [PubMed]
  8. Lahera, V.; Goicoechea, M.; de Vinuesa, S.G.; Miana, M.; de las Heras, N.; Cachofeiro, V.; Luño, J. Endothelial dysfunction, oxidative stress and inflammation in atherosclerosis: Beneficial effects of statins. Curr. Med. Chem. 2007, 14, 243–248. [Google Scholar] [CrossRef]
  9. Vaughan, C.J.; Gotto, A.M.; Basson, C.T. The evolving role of statins in the management of atherosclerosis. J. Am. Coll. Cardiol. 2000, 35, 1–10. [Google Scholar] [CrossRef]
  10. Schachter, M. Chemical, pharmacokinetic and pharmacodynamic properties of statins: An update. Fundam. Clin. Pharmacol. 2005, 19, 117–125. [Google Scholar] [CrossRef]
  11. Bellosta, S.; Ferri, N.; Bernini, F.; Paoletti, R.; Corsini, A. Non-lipid-related effects of statins. Ann. Med. 2000, 32, 164–176. [Google Scholar] [CrossRef]
  12. Athyros, V.G.; Kakafika, A.I.; Tziomalos, K.; Karagiannis, A.; Mikhailidis, D.P. Pleiotropic effects of statins—Clinical evidence. Curr. Pharm. Des. 2009, 15, 479–489. [Google Scholar] [CrossRef]
  13. Rossini, E.; Biscetti, F.; Rando, M.M.; Nardella, E.; Cecchini, A.L.; Nicolazzi, M.A.; Covino, M.; Gasbarrini, A.; Massetti, M.; Flex, A. Statins in High Cardiovascular Risk Patients: Do Comorbidities and Characteristics Matter? Int. J. Mol. Sci. 2022, 23, 9326. [Google Scholar] [CrossRef] [PubMed]
  14. Sirtori, C.R. The pharmacology of statins. Pharmacol. Res. 2014, 88, 3–11. [Google Scholar] [CrossRef] [PubMed]
  15. Koch, C.G. Statin therapy. Curr. Pharm. Des. 2012, 18, 6284–6290. [Google Scholar] [CrossRef] [PubMed]
  16. Climent, E.; Benaiges, D.; Pedro-Botet, J. Hydrophilic or lipophilic statins? Front. Cardiovasc. Med. 2021, 8, 687585. [Google Scholar] [CrossRef]
  17. Waters, D.D. What the statin trials have taught us. Am. J. Cardiol. 2006, 98, 129–134. [Google Scholar] [CrossRef]
  18. McKenney, J.M. Pharmacologic characteristics of statins. Clin Cardiol. 2003, 26 (Suppl. 3), 32–38. [Google Scholar] [CrossRef] [PubMed]
  19. Corsini, A.; Bellosta, S.; Baetta, R.; Fumagalli, R.; Bernini, F. New insights into the pharmacodynamics and pharmacokinetic properties of statins. Pharmacol. Ther. 1999, 84, 413–428. [Google Scholar] [CrossRef] [PubMed]
  20. Dagli-Hernandez, C.; Zhou, Y.; Lauschke, V.M.; Genvigir, F.D.V.; Hirata, T.D.C.; Hirata, M.H.; Hirata, R.D.C. Pharmacogenomics of statins: Lipid response and other outcomes in Brazilian cohorts. Pharmacol Rep. 2022, 74, 47–66. [Google Scholar] [CrossRef]
  21. Yaturu, S. Skeletal effects of statins. Endocr. Pract. 2003, 9, 315–320. [Google Scholar] [CrossRef] [PubMed]
  22. Sharif, P.S.; Abdollahi, M. A systematic review on the relation between use of statins and osteoporosis. Int. J. Pharmacol. 2011, 7, 180–188. [Google Scholar] [CrossRef]
  23. Cruz, A.C.; Gruber, B.L. Statins and osteoporosis: Can these lipid-lowering drugs also bolster bones? Clevel. Clin. J. Med. 2002, 69, 277–288. [Google Scholar] [CrossRef]
  24. An, T.; Hao, J.; Sun, S.; Li, R.; Yang, M.; Cheng, G.; Zhou, M. Efficacy of statins for osteoporosis: A systematic review and meta-analysis. Osteoporos. Int. 2017, 28, 47–57. [Google Scholar] [CrossRef] [PubMed]
  25. Tsartsalis, A.N.; Dokos, C.; Kaiafa, D.G.; Tsartsalis, D.N.; Kattamis, A.; Hatzitolios, A.I.; Savopoulos, C.G. Statins, bone formations and osteoporosis: Hope or hype? Hormones 2012, 11, 126–139. [Google Scholar] [CrossRef]
  26. Wang, C.Y.; Liao, J.K. Current advances in statin treatment: From molecular mechanisms to clinical practice. Arch. Med. Sci. 2007, 4A, 91–96. [Google Scholar]
  27. Oryan, A.; Kamali, A.; Moshiri, A. Potential mechanisms and applications of statins on osteogenesis: Current modalities, conflicts and future directions. J. Controll. Rel. 2015, 215, 12–24. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, P.Y.; Sun, J.S.; Tsuang, Y.H.; Chen, M.H.; Weng, P.W.; Lin, F.H. Simvastatin promotes osteoblast viability and differentiation via Ras/Smad/Erk/ BMP-2 signalling pathway. Nutr. Res. 2010, 30, 191–199. [Google Scholar] [CrossRef]
  29. Montagnani, A.; Gonnelli, S.; Cepollaro, C.; Pacini, S.; Campagna, M.S.; Franci, M.B.; Lucani, B.; Gennari, C. Effect of simvastatin treatment on bone mineral density and bone turnover in hypercholesterolemic postmenopausal women: A 1-year longitudinal study. Bone 2003, 32, 427–433. [Google Scholar] [CrossRef]
  30. Kaji, H.; Naito, J.; Inoue, Y.; Sowa, H.; Sugimoto, T.; Chihara, K. Statin suppresses apoptosis in osteoblastic cells: Role of transforming growth factor–beta-Smad3 pathway. Horm. Metab. Res. 2008, 40, 746–751. [Google Scholar] [CrossRef]
  31. Moshiri, A.; Sharifi, A.M.; Oryan, A. Role of simvastatin on fracture healing and osteoporosis: A systematic review on in vivo investigation. Clin. Exp. Pharmacol. Physiol. 2016, 43, 659–684. [Google Scholar] [CrossRef] [PubMed]
  32. Mundy, G.R. Statins and their potential for osteoporosis. Bone 2001, 29, 495–497. [Google Scholar] [CrossRef] [PubMed]
  33. Tahamtan, S.; Shirban, F.; Baghernija, M.; Johnston, T.P.; Sahebkar, A. The effects of statins on dental and oral health: A review of preclinical and clinical studies. J. Transl. Med. 2020, 18, 155. [Google Scholar] [CrossRef] [PubMed]
  34. Shahrezaee, M.; Oryan, A.; Bastami, F.; Hosseinpour, S.; Shahrezaee, M.H.; Kamali, A. Comparative impact of systemic delivery of atorvastatin, simvastatin, and lovastatin on bone mineral density of the ovariectomized rats. Endocrine 2018, 60, 138–150. [Google Scholar] [CrossRef] [PubMed]
  35. Zhou, H.; Xie, Y.; Baloch, Z.; Shi, Q.; Huo, Q.; Ma, T. The effect of atorvastatin, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor (HMG-CoA), on the prevention of osteoporosis in ovariectomized rabbits. J. Bone Miner. Metab. 2017, 35, 245–254. [Google Scholar] [CrossRef]
  36. Antonenko, A.; Leahy, A.; Babenko, M.; Lyons, D. Low-dose hydrophilic statins are the preferred agents for females at risk of osteoporosis. Bone Rep. 2022, 16, 101152. [Google Scholar] [CrossRef]
  37. Jadhav, S.B.; Jain, G.K. Statins and osteoporosis: A new role for old drugs. J. Pharm. Pharmacol. 2006, 58, 3–18. [Google Scholar] [CrossRef]
  38. Shah, S.R.; Werlang, C.A.; Kasper, F.K.; Mikos, A.G. Novel applications of statins for bone regenerations. Nat. Sci. Rev. 2015, 2, 85–99. [Google Scholar] [CrossRef]
  39. Hong, W.; Wei, Z.; Qiu, Z.; Li, Z.; Fu, C.; Ye, Z.; Xu, X. Atorvastatin promotes bone formation in aged apoE–/– mice through the Sirt1–Runx2 axis. J. Orthop. Surg. Res. 2020, 15, 303. [Google Scholar] [CrossRef]
  40. Mundy, G.; Garrett, R.; Harris, S.; Chan, J.; Chen, D.; Rossini, G.; Boyce, B.; Zhao, M.; Gutierrez, G. Stimulation of bone formation in vitro and in rodents by statins. Science 1999, 286, 1946–1949. [Google Scholar] [CrossRef]
  41. Lee, W.S.; Lee, E.G.; Sung, M.S.; Choi, Y.J.; Yoo, W.H. Atorvastatin inhibits osteoclast differentiation by suppressing NF-Κb and MAPK signaling during IL-1 β-induced osteoclastogenesis. Korean J. Intern. Med. 2018, 33, 397–406. [Google Scholar] [CrossRef]
  42. Tan, J.; Yang, N.; Fu, X.; Cui, Y.; Guo, Q.; Ma, T.; Yin, X.; Leng, H.; Song, C. Single-dose local simvastatin injection improves implant fixation via increased angiogenesis and bone formation in an ovariectomized rat model. Med. Sci. Monit. 2015, 21, 1428–1439. [Google Scholar]
  43. Sabandal, M.M.I.; Schäfer, E.; Imper, J.; Jung, S.; Kleinheinz, J.; Sielker, S. Simvastatin Induces In Vitro Mineralization Effects of Primary Human Odontoblast-like Cells. Materials. 2020, 3, 4679. [Google Scholar] [CrossRef]
  44. Von Stechow, D.; Fish, S.; Yahalom, D.; Bab, I.; Chorev, M.; Müller, R.; Alexander, J.M. Does simvastatin stimulate bone formation in vivo? BMC Musculoskelet. Disord. 2003, 28, 8. [Google Scholar] [CrossRef]
  45. Kabra, S.; Thosar, N.R.; Malviya, N.S. Exploring the Synergistic Effect of Simvastatin in Oral Health Applications: A Literature Review. Cureus 2023, 15, e44411. [Google Scholar] [CrossRef]
  46. Goes, P.; Lima, A.P.; Melo, I.M.; Rêgo, R.O.; Lima, V. Effect of atorvastatin in radiographic density on alveolar bone loss in Wistar rats. Braz. Dent. J. 2010, 21, 193–198. [Google Scholar] [CrossRef] [PubMed]
  47. El-Nabarawi, N.; El-Wakd, M.; Salem, M. Atorvastatin, a double weapon in osteoporosis treatment: An experimental and clinical study. Drug Des. Dev. Ther. 2017, 2, 1383–1391. [Google Scholar] [CrossRef] [PubMed]
  48. Qadir, F.; Alam, S.M.; Zehra, T.; Mehmood, A.; Siddiqi, A.Q. Role of Pitavastatin in prevention of osteopenic changes in ovariectomized rats. J. Coll. Physicians Surg. Pak. 2016, 26, 41–45. [Google Scholar] [PubMed]
  49. Sorial, A.K.; Anjum, S.A.; Cook, M.J.; Board, T.N.; O’Neil, T.W. Statins: Bone biology and revision arthroplasty: Review of clinical and experimental evidence. Ther. Adv. Muscoskel. Dis. 2020, 12, 1759720X20966229. [Google Scholar]
  50. Tang, Q.O.; Tran, G.T.; Gamie, Z.; Graham, S.; Tsialogiannis, E.; Tsiridis, E.; Linder, T.; Tsiridis, E. Statins: Under investigation for increasing bone mineral density and augmenting fracture healing. Expert Opin. Investig. Drugs 2008, 17, 1435–1463. [Google Scholar] [CrossRef]
  51. Leutner, M.; Butylina, M.; Matzhold, C.; Klimek, P.; Cuhaj, C.; Bellach, L.; Baumgartner-Parzer, S.; Reiter, B.; Preindl, K.; Kautzky, A.; et al. Simvastatin therapy in higher dosage deteriorates bone quality: Consistent evidence from population-wide patient data and interventional mouse studies. Biomed. Pharmacother. 2023, 158, 114089. [Google Scholar] [CrossRef]
  52. Uzzan, B.; Cohen, R.; Nicolas, P.; Cucherat, M.; Perret, G.Y. Effects of statins on bone mineral density: A meta-analysis of clinical studies. Bone 2007, 40, 1581–1587. [Google Scholar] [CrossRef]
  53. Lin, T.K.; Chou, P.; Lin, C.H.; Hung, Y.J.; Jong, G.P. Long-term effect of statins on the risk of new-onset osteoporosis: A nationwide population-based cohort study. PLoS ONE 2018, 13, e0196713. [Google Scholar] [CrossRef] [PubMed]
  54. Watson, E.C.; Adams, R.H. Biology of bone: The vasculature of the skeletal system. Cold Spring Harb. Perspect. Med. 2018, 8, a031559. [Google Scholar] [CrossRef]
  55. Yin, S.; Zhang, W.; Zhang, Z.; Jiang, X. Recent advances in scaffold design and material for vascularized tissue-engineered bone regeneration. Adv. Healthc. Mater. 2019, 8, 1801433. [Google Scholar] [CrossRef]
  56. Elavarasu, S.; Shutanthiran, T.K.; Naveen, D. Statins: A new era in local drug delivery. J. Pharm. Bioallied. Sci. 2012, 4, 248–251. [Google Scholar] [CrossRef]
  57. Jin, H.; Ji, Y.; Cui, Y.; Xu, L.; Liu, H.; Wang, J. Simvastatin-Incorporated Drug Delivery Systems for Bone Regeneration. ACS Biomater. Sci. Eng. 2021, 7, 2177–2191. [Google Scholar] [CrossRef]
  58. Anupama Devi, V.K.; Ray, S.; Arora, U.; Mitra, S.; Sionkowska, A.; Jaiswal, A.K. Dual drug delivery platforms for bone tissue engineering. Front. Bioeng. Biotechnol. 2022, 10, 969843. [Google Scholar]
  59. Kheirallah, M.; Almeshaly, H. Simvastatin, dosage and delivery system for supporting bone regeneration, an update review. J. Oral Maxillofac. Surg. Med. Pathol. 2016, 28, 205–209. [Google Scholar] [CrossRef]
  60. Liu, X.; Li, T.; Wang, F.; Sun, F.; Hu, J.; Ye, X.; Wang, D.; Yang, X. Controlling sustained statins release in multi-layered composite scaffolds for healing of osteoporotic bone defects. Biomater Adv. 2022, 137, 212838. [Google Scholar] [CrossRef]
  61. Moore, T.L.; Schreurs, A.S.; Morrison, R.A.; Jelen, E.K.; Loo, J.; Globus, R.K.; Alexis, F. Polymer-Coated Hydroxyapatite Nanoparticles for the Delivery of Statins. Nanomed. Nanotechnol. 2014, 5, 5. [Google Scholar] [CrossRef]
  62. Yoshii, T.; Hafeman, A.E.; Nyman, J.S.; Esparza, J.M.; Shinomiya, K.; Spengler, D.M.; Mundy, G.R.; Gutierrez, G.E.; Guelcher, S.A. A sustained release of lovastatin from biodegradable, elastomeric polyurethane scaffolds for enhanced bone regeneration. Tissue Eng. A 2010, 16, 2369–2379. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, W.; Xie, G.; Lu, Y.; Wang, J.; Feng, B.; Wang, Q.; Xu, K.; Bao, J. An improved osseointegration of metal implants by pitavastatin loaded multilayer films with osteogenic and angiogenic properties. Biomaterials 2022, 280, 121260. [Google Scholar] [CrossRef] [PubMed]
  64. Monjo, M.; Rubert, M.; Wohlfahrt, J.C.; Rønold, H.J.; Ellingsen, J.E.; Lyngstadaas, S.P. In vivo performance of absorbable collagen sponges with rosuvastatin in critical-size cortical bone defects. Acta Biomater. 2010, 6, 1405–1412. [Google Scholar] [CrossRef] [PubMed]
  65. Kalani, M.M.; Babak, N.; Rahimi, A. Electrospun coresheath poly (vinyl alcohol)/silk fibroin nanofibers with rosuvastatin release functionality for enhancing osteogenesis of human adipose-derived stem cells. Mater. Sci. Eng. 2019, 99, 129–139. [Google Scholar] [CrossRef] [PubMed]
  66. Kalani, M.M.; Nourmohammadi, J.; Negahdari, B. Osteogenic potential of rosuvastatin immobilized on silk fibroin nanofibers using argon plasma treatment. Biomed. Mater. 2019, 14, 025002. [Google Scholar] [CrossRef] [PubMed]
  67. Tanabe, K.; Nomoto, H.; Okumori, N.; Miura, T.; Yoshinari, M. Osteogenic effect of fluvastatin combined with biodegradable gelatin-hydrogel. Dent. Mater. J. 2012, 31, 489–493. [Google Scholar] [CrossRef]
  68. Benoit, D.S.; Nuttelman, C.R.; Collins, S.D.; Anseth, K.S. Synthesis and characterization of a fluvastatin-releasing hydrogel delivery system to modulate hMSC differentiation and function for bone regeneration. Biomaterials 2006, 27, 6102–6110. [Google Scholar] [CrossRef]
  69. Moriyama, Y.; Ayukawa, Y.; Ogino, Y.; Atsuta, I.; Koyano, K. Topical application of statin affects bone healing around implants. Clin. Oral Implant. Res. 2008, 19, 600–605. [Google Scholar] [CrossRef]
  70. Moriyama, Y.; Ayukawa, Y.; Ogino, Y.; Atsuta, I.; Todo, M.; Takao, Y.; Koyano, K. Local application of fluvastatin improves peri-implant bone quantity and mechanical properties: A rodent study. Acta Biomater. 2010, 6, 1610–1618. [Google Scholar] [CrossRef]
  71. Masuzaki, T.; Ayukawa, Y.; Moriyama, Y.; Jinno, Y.; Atsuta, I.; Ogino, Y.; Koyano, K. The effect of a single remote injection of statin-impregnated poly (lactic-co-glycolic acid) microspheres on osteogenesis around titanium implants in rat tibia. Biomaterials 2010, 31, 3327–3334. [Google Scholar] [CrossRef]
  72. Chalisserry, E.P.; Nam, S.Y.; Anil, S. Simvastatin Loaded Nano Hydroxyapatite in Bone Regeneration: A Study in the Rabbit Femoral Condyle. Curr. Drug Deliv. 2019, 16, 530–537. [Google Scholar] [CrossRef] [PubMed]
  73. Gao, S.; Shiota, M.; Fujii, M.; Chen, K.; Shimogishi, M.; Sato, M.; Kasugai. Combination of simvastatin and hydroxyapatite fiber induce bone augmentation. Open J. Regen. Medicine. 2013, 02, 53–60. [Google Scholar] [CrossRef]
  74. Huang, X.; Huang, Z.; Li, W. Highly efficient release of simvastatin from simvastatin-loaded calcium sulphate scaffolds enhances segmental bone regeneration in rabbits. Mol. Med. Rep. 2014, 9, 2152–2158. [Google Scholar] [CrossRef]
  75. Nyan, M.; Sato, D.; Oda, M.; Machida, T.; Kobayashi, H.; Nakamura, T.; Kasugai, S. Bone formation with the combination of simvastatin and calcium sulfate in critical-sized rat calvarial defect. J. Pharmacol. Sci. 2007, 104, 384–386. [Google Scholar] [CrossRef]
  76. Chou, J.; Ito, T.; Bishop, D.; Otsuka, M.; Ben-Nissan, B.; Milthorpe, B. Controlled release of simvastatin from biomimetic β-TCP drug delivery system. PLoS ONE 2013, 8, e54676. [Google Scholar] [CrossRef]
  77. Laskus-Zakrzewska, A.; Kazimierczak, P.; Kolmas, J. Porous Composite granules with potential function on bone substitute and simvastatin releasing system: A preliminary study. Materials 2021, 14, 5068. [Google Scholar] [CrossRef] [PubMed]
  78. Khurana, K.; Guillem-Marti, J.; Soldera, F.; Mücklich, F.; Canal, C.; Ginebra, M.-P. Injectable calcium phosphate foams for the delivery of Pitavastatin as osteogenic and angiogenic agent. J. Biomed. Mater. Res. 2019, 108, 760–777. [Google Scholar] [CrossRef]
  79. Deng, L.J.; Wu, Y.L.; He, X.H.; Xie, K.N.; Xie, L.; Deng, Y. Simvastatin delivery on PEEK for bioactivity and osteogenesis enhancements. J. Biomater. Sci. Polym. Ed. 2018, 29, 2237–2251. [Google Scholar] [CrossRef] [PubMed]
  80. Nyan, M.; Sato, D.; Kihara, H.; Machida, T.; Ohya, K.; Kasugai, S. Effects of the combination with alpha-tricalcium phosphate and simvastatin on bone regeneration. Clin. Oral Implant. Res. 2009, 20, 280–287. [Google Scholar] [CrossRef]
  81. Rojbani, H.; Nyan, M.; Ohya, K.; Kasugai, S. Evaluation of the osteoconductivity of α-tricalcium phosphate, β-tricalcium phosphate, and hydroxyapatite combined with or without simvastatin in rat calvarial defect. J. Biomed. Mater. Res. A 2011, 98, 488–498. [Google Scholar] [CrossRef]
  82. Hamada, H.; Ohshima, H.; Otsuka, M. Dissolution medium responsive simvastatin release from biodegradable apatite cements and the therapeutic effect in osteoporosis rats. J. Appl. Biomater. Funct. Mater. 2012, 10, 22–28. [Google Scholar]
  83. Ma, B.; Clarke, S.A.; Brooks, R.A.; Rushton, N. The effect of simvastatin on bone formation and ceramic resorption in a peri-implant defect model. Acta Biomater. 2008, 4, 149–155. [Google Scholar] [CrossRef]
  84. de Santana, W.M.; de Sousa, D.N.; Ferreira, V.M.; Duarte, W.R. Simvastatin and biphasic calcium phosphate affects bone formation in critical-sized rat calvarial defects. Acta Cir. Bras. 2016, 31, 300–307. [Google Scholar] [CrossRef]
  85. Yin, H.; Li, Y.G.; Si, M.; Li, J.M. Simvastatin-loaded macroporous calcium phosphate cement: Preparation, in vitro characterization, and evaluation of in vivo performance. J. Biomed. Mater. Res. A 2012, 100, 2991–3000. [Google Scholar] [CrossRef]
  86. Montazerolghaem, M.; Engqvist, H.; Karlsson Ott, M. Sustained release of simvastatin from premixed injectable calcium phosphate cement. J. Biomed. Mater. Res. A 2014, 102, 340–347. [Google Scholar] [CrossRef]
  87. Montazerolghaem, M.; Rasmusson, A.; Melhus, H.; Engqvist, H.; Karlsson Ott, M. Simvastatin-doped pre-mixed calcium phosphate cement inhibits osteoclast differentiation and resorption. J. Mater. Sci. Mater. Med. 2016, 27, 83. [Google Scholar] [CrossRef]
  88. Sun, T.; Huang, J.; Zhang, W.; Zheng, X.; Wang, H.; Liu, J.; Leng, H.; Yuan, W.; Song, C. Simvastatin-hydroxyapatite coatings prevent biofilm formation and improve bone formation in implant-associated infections. Bioact. Mater. 2022, 13, 44–56. [Google Scholar] [CrossRef]
  89. López-Álvarez, M.; López-Puente, V.; Rodríguez-Valencia, C.; Angelomé, P.C.; Liz-Marzán, L.M.; Serra, J.; Pastoriza-Santos, I.; González, P. Osteogenic effects of simvastatin-loaded mesoporous titania thin films. Biomed. Mater. 2018, 13, 025017. [Google Scholar] [CrossRef] [PubMed]
  90. Fang, W.; Zhao, S.; He, F.; Liu, L.; Yang, G. Influence of simvastatin-loaded implants on osseointegration in an ovariectomized animal model. Biomed. Res. Int. 2015, 2015, 831504. [Google Scholar] [CrossRef] [PubMed]
  91. Du, Z.; Chen, J.; Yan, F.; Xiao, Y. Effects of Simvastatin on bone healing around titanium implants in osteoporotic rats. Clin. Oral Implant. Res. 2009, 20, 145–150. [Google Scholar] [CrossRef] [PubMed]
  92. Pauly, S.; Luttosch, F.; Morawski, M.; Haas, N.P.; Schmidmaier, G.; Wildemann, B. Simvastatin locally applied from a biodegradable coating of osteosynthetic implants improves fracture healing comparable to BMP-2 application. Bone 2009, 45, 505–511. [Google Scholar] [CrossRef]
  93. Rezk, A.I.; Kim, J.Y.; Kim, B.S.; Park, C.H.; Kim, C.S. De novo dual functional 3D scaffold using computational simulation with controlled drug release. J. Colloid Interface Sci. 2022, 625, 12–23. [Google Scholar] [CrossRef] [PubMed]
  94. Lee, Y.; Liu, X.; Nawshad, A.; Marx, D.B.; Wang, D.; Reinhardt, R.A. Role of prostaglandin pathway and alendronate-based carriers to enhance statin-induced bone. Mol. Pharm. 2011, 8, 1035–1042. [Google Scholar] [CrossRef]
  95. Mukozawa, A.; Ueki, K.; Marukawa, K.; Okabe, K.; Moroi, A.; Nakagawa, K. Bone healing of critical-sized nasal defects in rabbits by statins in two different carriers. Clin. Oral Implant. Res. 2011, 22, 1327–1335. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, C.Z.; Fu, Y.C.; Jian, S.C.; Wang, Y.H.; Liu, P.L.; Ho, M.L.; Wang, C.K. Synthesis and characterization of cationic polymeric nanoparticles as simvastatin carriers for enhancing the osteogenesis of bone marrow mesenchymal stem cells. J. Colloid Interface Sci. 2014, 432, 190–199. [Google Scholar] [CrossRef]
  97. Xue, Y.; Wu, M.; Liu, Z.; Song, J.; Luo, S.; Li, H.; Li, Y.; Jin, L.; Guan, B.; Lin, M.; et al. In vitro and in vivo evaluation of chitosan scaffolds combined with simvastatin-loaded nanoparticles for guided bone regeneration. J. Mater. Sci. Mater. Med. 2019, 30, 47. [Google Scholar] [CrossRef]
  98. Ghadri, N.; Anderson, K.M.; Adatrow, P.; Stein, S.H.; Su, H.J.; Garcia-Godoy, F.; Karydis, A.; Bumgardner, J.D. Evaluation of Bone Regeneration of Simvastatin Loaded Chitosan Nanofiber Membranes in Rodent Calvarial Defects. J. Biomater. Nanobiotechnol. 2018, 9, 210–231. [Google Scholar] [CrossRef]
  99. Alsawah, G.M.; Al-Obaida, M.I.; Al-Madi, E.M. Effect of a Simvastatin-Impregnated Chitosan Scaffold on Cell Growth and Osteoblastic Differentiation. Appl. Sci. 2021, 11, 5346. [Google Scholar] [CrossRef]
  100. Delan, W.K.; Zakaria, M.; Elsaadany, B.; ElMeshad, A.N.; Mamdouh, W.; Fares, A.R. Formulation of simvastatin chitosan nanoparticles for controlled delivery in bone regeneration: Optimization using Box-Behnken design, stability and in vivo study. Int J. Pharm. 2020, 577, 119038. [Google Scholar] [CrossRef]
  101. Wong, R.W.; Rabie, A.B. Statin collagen grafts used to repair defects in the parietal bone of rabbits. Br. J. Oral Maxillofac. Surg. 2013, 41, 244–248. [Google Scholar] [CrossRef]
  102. Wong, R.W.; Rabie, A.B. Histologic and ultrastructural study on statin graft in rabbit skulls. J. Oral Maxillofac. Surg. 2005, 63, 1515–1521. [Google Scholar] [CrossRef] [PubMed]
  103. Ruan, H.; Yu, Y.; Guo, X.; Jiang, Q.; Luo, Y. The possibility of healing alveolar bone defects with simvastatin thermosensitive gel: In vitro/in vivo evaluation. Drug Des. Devel. Ther. 2018, 12, 1997–2003. [Google Scholar] [CrossRef] [PubMed]
  104. Fukui, T.; Ii, M.; Shoji, T.; Matsumoto, T.; Mifune, Y.; Kawakami, Y.; Akimaru, H.; Kawamoto, A.; Kuroda, T.; Saito, T.; et al. The therapeutic effect of local administration of low-dose simvastatin-conjugated gelatin hydrogel for fracture healing. J. Bone Miner. Res. 2012, 27, 1118–1131. [Google Scholar] [CrossRef]
  105. Oka, S.; Matsumoto, T.; Kubo, S.; Matsushita, T.; Sasaki, H.; Nishizawa, Y.; Matsuzaki, T.; Saito, T.; Nishida, K.; Tabata, Y.; et al. Local administration of low-dose simvastatin-conjugated gelatin hydrogel for tendon-bone healing in anterior cruciate ligament reconstruction. Tissue Eng. Part A 2013, 19, 1233–1243. [Google Scholar] [CrossRef] [PubMed]
  106. Sukul, M.; Min, Y.K.; Lee, S.Y.; Lee, B.T. Osteogenic potential of simvastatin loaded gelatin-nanofibrillar cellulose-β tricalcium phosphate hydrogel scaffold in critical-sized rat calvarial defect. Eur. Polym. J. 2015, 73, 308–323. [Google Scholar] [CrossRef]
  107. Terauchi, M.; Inada, T.; Tonegawa, A.; Tamura, A.; Yamaguchi, S.; Harada, K.; Yui, N. Supramolecular inclusion complexation of simvastatin withmethylated β-cyclodextrins for promoting osteogenic differentiation. Int. J. Biol. Macromol. 2016, 93, 1492–1498. [Google Scholar] [CrossRef]
  108. Thylin, M.R.; McConnell, J.C.; Schmid, M.J.; Reckling, R.R.; Ojha, J.; Bhattacharyya, I.; Marx, D.B.; Reinhardt, R.A. Effects of simvastatin gels on murine calvarial bone. J. Periodontol. 2002, 73, 1141–1148. [Google Scholar] [CrossRef]
  109. Yue, X.; Niu, M.; Zhang, T.; Wang, C.; Wang, Z.; Wu, W.; Zhang, Q.; Lai, C.; Zhou, L. In vivo evaluation of a simvastatin-loaded nanostructured lipid carrier for bone tissue regeneration. Nanotechnology 2016, 27, 115708. [Google Scholar] [CrossRef]
  110. Pişkin, E.; Işoğlu, I.A.; Bölgen, N.; Vargel, I.; Griffiths, S.; Cavuşoğlu, T.; Korkusuz, P.; Güzel, E.; Cartmell, S. In vivo performance of simvastatin-loaded electrospun spiral-wound polycaprolactone scaffolds in reconstruction of cranial bone defects in the rat model. J. Biomed. Mater. Res. A 2009, 90, 1137–1151. [Google Scholar] [CrossRef]
  111. Saberi, A.; Kouhjani, M.; Mohammadi, M.; Hosta-Rigau, L. Novel scaffold platforms for simultaneous induction osteogenesis and angiogenesis in bone tissue engineering: A cutting-edge approach. J. Nanobiotechnol. 2023, 21, 351. [Google Scholar] [CrossRef]
  112. Yu, D.; Huang, C.; Jiang, C.; Zhu, H. Features of a simvastatin-loaded multi-layered co-electrospun barrier membrane for guided bone regeneration. Exp. Ther. Med. 2021, 22, 713. [Google Scholar] [CrossRef]
  113. Rezk, A.I.; Bhattarai, D.P.; Park, J.; Park, C.H.; Kim, C.S. Polyaniline-coated titanium oxide nanoparticles and simvastatin-loaded poly(ε-caprolactone) composite nanofibers scaffold for bone tissue regeneration application. Colloids Surf. B Biointerfaces 2020, 192, 111007. [Google Scholar] [CrossRef]
  114. Li, G.; Li, Y.; Zhang, X.; Gao, P.; Xia, X.; Xiao, S.; Wen, J.; Guo, T.; Yang, W.; Li, J. Strontium and simvastatin dual loaded hydroxyapatite microsphere reinforced poly(ε-caprolactone) scaffolds promote vascularized bone regeneration. J. Mater. Chem. B 2023, 11, 1115–1130. [Google Scholar] [CrossRef] [PubMed]
  115. Rezk, A.I.; Mousa, H.M.; Lee, J.; Park, C.H.; Kim, C.S. Composite PCL/HA/simvastatin electrospun nanofiber coating on biodegradable Mg alloy for orthopedic implant application. J. Coat. Technol. Res. 2019, 16, 477–489. [Google Scholar] [CrossRef]
  116. Liu, X.; Li, X.; Zhou, L.; Li, S.; Sun, J.; Wang, Z.; Gao, Y.; Jiang, Y.; Lu, H.; Wang, Q.; et al. Effects of simvastatin-loaded polymeric micelles on human osteoblast-like MG-63 cells. Colloids Surf. B Biointerfaces 2013, 102, 420–427. [Google Scholar] [CrossRef]
  117. Feng, X.; Yue, X.; Niu, M. Simvastatin-Loaded Nanomicelles Enhance the Osteogenic Effect of Simvastatin. J. Nanomaterials. 2020, 2020, 1072765. [Google Scholar] [CrossRef]
  118. Lin, C.W.; Lee, C.Y.; Lin, S.Y.; Kang, L.; Fu, Y.C.; Chen, C.H.; Wang, C.K. Bone-Targeting Nanoparticles of a Dendritic (Aspartic acid)3-Functionalized PEG-PLGA Biopolymer Encapsulating Simvastatin for the Treatment of Osteoporosis in Rat Models. Int. J. Mol. Sci. 2022, 23, 10530. [Google Scholar] [CrossRef]
  119. Zhang, P.; Han, F.; Li, Y.; Chen, J.; Chen, T.; Zhi, Y.; Jiang, J.; Lin, C.; Chen, S.; Zhao, P. Local delivery of controlled-release simvastatin to improve the biocompatibility of polyethylene terephthalate artificial ligaments for reconstruction of the anterior cruciate ligament. Int. J. Nanomed. 2016, 11, 465–478. [Google Scholar] [CrossRef] [PubMed]
  120. Yueyi, C.; Xiaoguang, H.; Jingying, W.; Quansheng, S.; Jie, T.; Xin, F.; Yingsheng, X.; Chunli, S. Calvarial defect healing by recruitment of autogenous osteogenic stem cells using locally applied simvastatin. Biomaterials 2013, 34, 9373–9380. [Google Scholar] [CrossRef]
  121. Whang, K.; Grageda, E.; Khan, A.; McDonald, J.; Lawton, M.; Satsangi, N. A novel osteotropic biomaterial OG-PLG: In vitro efficacy. J. Biomed. Mater. Res. A 2005, 74, 247–253. [Google Scholar] [CrossRef]
  122. Gentile, P.; Nandagiri, V.K.; Daly, J.; Chiono, V.; Mattu, C.; Tonda-Turo, C.; Ciardelli, G.; Ramtoola, Z. Localised controlled release of simvastatin from porous chitosan-gelatin scaffolds engrafted with simvastatin loaded PLGA-microparticles for bone tissue engineering application. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 59, 249–257. [Google Scholar] [CrossRef]
  123. Littuma, G.J.S.; Sordi, M.B.; Borges Curtarelli, R.; Aragonês, Á.; da Cruz, A.C.C.; Magini, R.S. Titanium coated with poly(lactic-co-glycolic) acid incorporating simvastatin: Biofunctionalization of dental prosthetic abutments. J. Periodontal. Res. 2020, 55, 116–124. [Google Scholar] [CrossRef] [PubMed]
  124. Kaur, M.; Nagpal, M.; Grewal, A.K.; Chauhan, S.; Dora, C.P.; Singh, T.G. Molecular Complex of HSIM-loaded Polymeric Nanoparticles: Potential Carriers in Osteoporosis. Curr. Drug Targets 2023, 24, 1066–1078. [Google Scholar] [CrossRef] [PubMed]
  125. Nath, S.D.; Son, S.; Sadiasa, A.; Min, Y.K.; Lee, B.T. Preparation and characterization of PLGA microspheres by the electrospraying method for delivering simvastatin for bone regeneration. Int. J. Pharm. 2013, 443, 87–94. [Google Scholar] [CrossRef]
  126. Nath, S.D.; Linh, N.T.; Sadiasa, A.; Lee, B.T. Encapsulation of simvastatin in PLGA microspheres loaded into hydrogel loaded BCP porous spongy scaffold as a controlled drug delivery system for bone tissue regeneration. J. Biomater. Appl. 2014, 28, 1151–1163. [Google Scholar] [CrossRef] [PubMed]
  127. Fu, Y.C.; Wang, Y.H.; Chen, C.H.; Wang, C.K.; Wang, G.J.; Ho, M.L. Combination of calcium sulfate and simvastatin-controlled release microspheres enhances bone repair in critical-sized rat calvarial bone defects. Int. J. Nanomed. 2015, 10, 7231–7240. [Google Scholar]
  128. Yan, M.; Ni, J.; Shen, H.; Song, D.; Ding, M.; Huang, J. Local controlled release of simvastatin and PDGF from core/shell microspheres promotes bone regeneration in vivo. RSC Adv. 2017, 7, 19621–19629. [Google Scholar] [CrossRef]
  129. von Knoch, F.; Wedemeyer, C.; Heckelei, A.; Saxler, G.; Hilken, G.; Brankamp, J.; Sterner, T.; Landgraeber, S.; Henschke, F.; Löer, F.; et al. Promotion of bone formation by simvastatin in polyethylene particle-induced osteolysis. Biomaterials 2005, 26, 5783–5789. [Google Scholar] [CrossRef]
  130. Venkatesan, N.; Liyanage, A.D.T.; Castro-Núñez, J.; Asafo-Adjei, T.; Cunningham, L.L.; Dziubla, T.D.; Puleo, D.A. Biodegradable polymerized simvastatin stimulates bone formation. Acta Biomater. 2019, 93, 192–199. [Google Scholar] [CrossRef]
  131. Wu, T.; Sun, J.; Tan, L.; Yan, Q.; Li, L.; Chen, L.; Liu, X.; Bin, S. Enhanced osteogenesis and therapy of osteoporosis using simvastatin loaded hybrid system. Bioact. Mater. 2020, 5, 348–357. [Google Scholar] [CrossRef]
  132. Hajializade, M.; Moghtadaei, M.; Mirzaei, A.; Abdollahi Kordkandi, S.; Babaheidarian, P.; Pazoki-Toroudi, H.; Yeganeh, A. Significant effect of simvastatin and/or ezetimibe-loaded nanofibers on the healing of femoral defect: An experimental study. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 111, 110861. [Google Scholar] [CrossRef] [PubMed]
  133. Wadagaki, R. Osteogenic induction of bone marrow-derived stromal cells on simvastatin-releasing, biodegradable, nano-to microscale fiber scaffolds. Ann. Biomed. Eng. 2011, 39, 1872–1881. [Google Scholar] [CrossRef]
  134. Harsha, G.; Madhavi, S.; Arthi, S.; Haritha, S. Evaluation of efficacy of simvastatin in bone regeneration following local application in third molar extraction socket: A randomized control trial. Natl. J. Maxillofac Surg. 2023, 14, 286–293. [Google Scholar] [CrossRef] [PubMed]
  135. Bae, M.S.; Yang, D.H.; Lee, J.B.; Heo, D.N.; Kwon, Y.D.; Youn, I.C.; Choi, K.; Hong, J.H.; Kim, G.T.; Choi, Y.S.; et al. Photo-cured hyaluronic acid-based hydrogels containing simvastatin as a bone tissue regeneration scaffold. Biomaterials 2011, 32, 8161–8171. [Google Scholar] [CrossRef]
  136. Zhou, Y.; Ni, Y.; Liu, Y.; Zeng, B.; Xu, Y.; Ge, W. The role of simvastatin in the osteogenesis of injectable tissue-engineered bone based on human adipose-derived stromal cells and platelet-rich plasma. Biomaterials 2010, 31, 5325–5335. [Google Scholar] [CrossRef]
Figure 1. Main and pleiotropic effects of statins (down arrow—decrease, up arrow—increase).
Figure 1. Main and pleiotropic effects of statins (down arrow—decrease, up arrow—increase).
Ijms 25 02378 g001
Figure 2. Chemical structure of statins (L—lipophilic statin; H—hydrophilic statin).
Figure 2. Chemical structure of statins (L—lipophilic statin; H—hydrophilic statin).
Ijms 25 02378 g002
Figure 3. Mechanism of statins’ actions on bone metabolism (Abbreviations: BMP-2—bone morphogenetic protein 2; ER—endoplasmic reticulum; HMG-CoA—3-hydroxy-3-methyl-glutaryl-coenzyme A; OPG—osteoprotegerin; PI-3-K—phosphatidylinositol 3-kinase; PP—pyrophosphate; RANK—receptor activator of NF-κB; RANKL—receptor activator of NF-κB ligand; Smad3—SMAD family member 3; TGFβ—transforming growth factor beta; VEGF—vascular endothelial growth factor). Red down arrow—decrease, red up arrow—increase.
Figure 3. Mechanism of statins’ actions on bone metabolism (Abbreviations: BMP-2—bone morphogenetic protein 2; ER—endoplasmic reticulum; HMG-CoA—3-hydroxy-3-methyl-glutaryl-coenzyme A; OPG—osteoprotegerin; PI-3-K—phosphatidylinositol 3-kinase; PP—pyrophosphate; RANK—receptor activator of NF-κB; RANKL—receptor activator of NF-κB ligand; Smad3—SMAD family member 3; TGFβ—transforming growth factor beta; VEGF—vascular endothelial growth factor). Red down arrow—decrease, red up arrow—increase.
Ijms 25 02378 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Granat, M.M.; Eifler-Zydel, J.; Kolmas, J. Statins—Their Role in Bone Tissue Metabolism and Local Applications with Different Carriers. Int. J. Mol. Sci. 2024, 25, 2378. https://doi.org/10.3390/ijms25042378

AMA Style

Granat MM, Eifler-Zydel J, Kolmas J. Statins—Their Role in Bone Tissue Metabolism and Local Applications with Different Carriers. International Journal of Molecular Sciences. 2024; 25(4):2378. https://doi.org/10.3390/ijms25042378

Chicago/Turabian Style

Granat, Marcin Mateusz, Joanna Eifler-Zydel, and Joanna Kolmas. 2024. "Statins—Their Role in Bone Tissue Metabolism and Local Applications with Different Carriers" International Journal of Molecular Sciences 25, no. 4: 2378. https://doi.org/10.3390/ijms25042378

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop