IF in JCR  CiteScore Rank  About IJO  Current Issue  Featured Articles  Articles In Press  Recent Accepted
International Journal of Ophthalmology

2017; 10(9): 1361-1369


·Basic Research·



 

The ocular toxicity and pharmacokinetics of simvastatin following intravitreal injection in mice

  

Dennis Y. Tse1,2, Seong Jae Kim3, Inyoung Chung3, Feng He4, Theodore G. Wensel4, Samuel M. Wu1

 

1Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA

2School of Optometry, the Hong Kong Polytechnic University, Hong Kong, China

3Department of Ophthalmology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea

4Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA

Co-first authors: Dennis Y. Tse, Seong Jae Kim and Inyoung Chung

Correspondence to: Inyoung Chung. Department of Ophthalmology, College of Medicine, Gyeongsang National University, Jinju 52727, Korea. inyoung@gnu.ac.kr

 

Received: 2016-09-07        Accepted: 2017-04-21

 

Abstract

AIM: To investigate the retinal toxicity and pharmacokinetics of simvastatin intravitreally injected into mice.

METHODS: Forty-eight 6-8-week-old C57BL/6J mice were used in this study. Simvastatin was intravitreally injected into the right eye of each mouse; the left eye was injected with vehicle and was used as a control. Bilateral dark-adapted electroretinography (ERG) was performed 1 and 7d following injection. Histology was examined using a combination of light, fluorescence and electron microscopy. High-performance liquid chromatography (HPLC) was used to determine the decay in the retinal simvastatin concentration.

RESULTS: ERG revealed no significant changes in the simvastatin-injected eyes compared to control. Histologic studies showed normal retinal morphology in eyes injected with simvastatin up to a final vitreal concentration of 200 μmol/L. No significant changes in the number of photoreceptors, bipolar cells or ganglion cells were found. The retinal simvastatin concentration decayed exponentially, with a half-life of 1.92-2.41h.

CONCLUSION: Intravitreal injection of up to 200 μmol/L simvastatin produced no signs of adverse effects in the mouse retina. Simvastatin reaches the retina shortly after intravitreal injectionand has a short half-life.

KEYWORDS: simvastatin; retina; electroretinography; high-performance liquid chromatography; electron microscopy; intravitreal injection

DOI:10.18240/ijo.2017.09.05

 

Citation: Tse DY, Kim SJ, Chung I, He F, Wensel TG, Wu SM. The ocular toxicity and pharmacokinetics of simvastatin following intravitreal injection in mice. Int J Ophthalmol  2017;10(9):1361-1369

 

INTRODUCTION

Statins, potent inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, have widely been prescribed for the treatment of hyperlipidemia since 1987[1] and have recently been demonstrated to possess an expanding spectrum of activities, including anti-inflammatory, anti-angiogenic, anti-oxidant[2], immunosuppressive[3] and neuroprotective[4-6] effects. Statins have been reported to be beneficial to patients with a variety of ocular diseases such as diabetic retinopathy[7], age-related macular degeneration (AMD)[8] and retinal ischemia and to potentially be useful in glaucoma[9-12].

Simvastatin, the semisynthetic statin tested in the present study, is one of the most potent of all the identified statins; simvastatin inhibits the diabetes-induced increases in vascular endothelial growth factor (VEGF) expression, protects the blood-retinal barrier and suppresses the progression of streptozotocin-induced diabetic retinopathy in rats[13-15]. A recent study has shown that simvastatin activated the anti-oxidative defense protein HO-1 in cultured human retinal pigment epithelial (RPE) cells[16] and that this activity contributed to its cytoprotective effect against AMD. Considering the potential adverse side effects of intravitreally-injected steroids [e.g. intraocular pressure (IOP) elevation, cataract] and anti-VEGF drugs (e.g. intolerance, cost, and unknown deleterious cumulative systemic effects) commonly used in the treatment in AMD or diabetic retinopathy, simvastatin might be a potential alternative drug candidate as several reports have suggested that simvastatin inhibits the progression of these two diseases[17-20]. It has also been shown that simvastatin enhanced retinal ganglion cell (RGC) survival and protected visual functions in rodent models of acute retinal ischemia/reperfusion injury[21-23] and optic nerve lesioning[24]. Simvastatin inhibits the Rho/Rho-kinase pathway and thus might have therapeutic potential in the prevention of cicatrical contraction of proliferative membranes in vivo study. And thus, simvastatin might provide a new strategy for the treatment and prevention of the development of proliferative vitreoretinal diseases[25].

In most of the literature, statins were administered orally or injected intraperitoneally. From a clinical perspective, some ocular conditions would be better managed if the intraocular statin concentration was increased more efficiently via direct intravitreal injection; such conditions include when ocular conditions are acute or when systemic side effects should be avoided[26-27]. From a research perspective, intravitreal injection is superior to systemic delivery routes because the other eye can serve as an internal control in certain experimental settings[28].

Our objective was to study the retinal toxicity of simvastatin to mice using electroretinography (ERG) and electron microscopy. These results are pivotal in guiding future laboratory and clinical studies on the dosage and delivery of simvastatin. This study is particularly relevant to mouse research models, as such disease models are widely available and are easily combined with genetic manipulations or surgical interventions[29]. The long-term goal of this line of investigation is to utilize mouse models to investigate the potential neuroprotective effects of simvastatin against avariety of retinal diseases such as glaucoma and AMD[30-32].

MATERIALS AND METHODS

Animal Preparations  Forty-eight 6-8 weeks old C57BL/6J mice obtained from the Jackson Laboratory (Bar Harbor, ME, USA) were used for the experiments. The animals were treated in accordance with the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research and with the animal welfare guidelines of the IACUC of Baylor College of Medicine.

Intravitreal Injection  Because vitreal simvastatin concentrations of 5 and 15 μmol/L have been shown to be effective and did not produce sign of toxicity in previous animal studies[24-25], we selected higher vitreal simvastatin concentrations of 50 and 200 μmol/L for this retinal toxicity study to provide at least 10× margin over the previous studies.

Simvastatin (#S6196, Sigma-Aldrich Corp. St. Louis, MO, USA) was converted to its active form according to the manufacturer’s instructions[33], diluted in sterile buffered balanced salt solutions (pH 7.5; Alcon Laboratories, Inc., Fort Worth, TX, USA), and then filtered using a 0.22-μm PVDF filter (Millipore, Billerica, MA, USA). Aliquots were stored at -20 before use. The mice were first anesthetized by intraperitoneally injecting weight-based doses of ketamine (95 mg/mL) and xylazine (5 mg/mL). Then, a single drop of 0.5% proparacaine hydrochloride (Alcon Laboratories, Inc., Fort Worth, TX, USA) and 1% tropicamide were applied to both eyes. One microliter of 0.5 or 2.0 mmol/L simvastatin was injected into the vitreous of the right eye under a stereomicroscope using a Nanofil syringe fitted with a 33-gauge beveled needle (WPI, Sarasota, FL, USA). One microliter of an equivalent vehicle solution was injected into the left eye as a control. Assuming that the injected 1 μL of solution was diluted into the 10 μL of vitreous fluid in the eye[34-35], the vitreal concentrations of simvastatin were 50 and 200 μmol/L for the injected 0.5 and 2.0 mmol/L solutions, respectively. The needle was inserted behind the limbus through the pars plana at an oblique angle to avoid damaging the crystallized lens. To prevent the injected solution from escaping the eye when the needle was withdrawn, the needle tip was held in the eye for 30s after the injection to facilitate mixing.

Electroretinography Recordings  In vivo scotopic ERG was recorded bilaterally from the mice 1 and 7d after intravitreal injection. A pre-injection measurement was performed on a different cohort of mice as a reference. Prior to each ERG assessment, the mice were allowed to adapt to the dark for at least 2.5h. Under dim redlight, the mice were prepared for ERG testing as described previously[30]. The signals were amplified using a Grass P122 amplifier and were band-pass filtered from 0.1 to 1000 Hz (Grass Instruments, West Warwick, RI, USA). The data were acquired using a National Instruments data acquisition board (USB-6216, National Instruments, TX, USA) at a sampling rate of 10 kHz. The traces were analyzed using custom codes written in MATLAB (MathWorks, Natick, MA, USA)[30].

The flashes used for scotopic b-wave measurements were generated using cyan light-emitting diodes calibrated with a photometer (ILT1700, International Light, MA, USA) and were converted to the unit photoisomerizations/rod, where 1 scot cd m2=581 photoisomerizations/rod/s[30]. To remove oscillatory potentials before fitting, the scotopic b-wave was digitally filtered using the filtfilt function in MATLAB (low-pass filter; Fc=60 Hz). The positive and negative scotopic threshold responses (STRs) were measured under stimuli of various intensities as described previously[30]. The cone ERG recording was performed according to a paired-flash method using xenon flashes[36]. An initial conditioning flash saturated both rods and cones 2s before a probe flash. The ERG signal recorded by the probe flash is attributed to responses driven by the cones because cones recover faster than rods. In this manuscript, the stimulus intensities are presented on a log scale, where 1E+0=1 photoisomerizations/rod. All statistical analyses were performed using paired t-tests in SPSS version 20 (IBM).

Pharmacokinetic Analysis via High-performance Liquid Chromatography  The ocular concentration of simvastatin in the current study was measured using the retinal tissue instead of the vitreous humor, which is commonly used in rabbit pharmacokinetic models[37-39]. This experimental approach is advantageous because the retina itself is typically the target of treatment.

The injected eyes were enucleated at 1, 3, 6, 12, 24 or 48h to measure the retinal concentration of the drug. For each of the above time points, at least three microcentrifuge tube samples were analyzed. Each sample contained four whole retinae extracted from four eyes. First, 100 μL of phosphate buffered saline (PBS) at 4 was added to the sample before the samples were homogenized using amotorized mortar (#47747-370, VWR International LLC, Radnor, PA, USA) for a total of 6min. Then, 900 μL of acetonitrile at 4 was added to the sample. The mixture was subsequently vortexed for 2min and then ultra-centrifuged at 68 000× g for 20min at 4. The supernatant was transferred to auto-sampler vials and then concentrated to a volume of 50-80 µL using a SpeedVac. The resulting samples contained 50% acetonitrile and 0.1% (v/v) trifluoroacetic acid (TFA).

The simvastatin standards (Sigma-Aldrich Corp., St. Louis, MO, USA) were dissolved in 100 μL of 50% acetonitrile containing 0.1% (v/v) TFA and were vortexed for 1min. All samples were filtered through a 0.45 μm Acrodisc® 3 CR PTFE filter to remove insoluble particles before injection into a C18 high-performance liquid chromatography (HPLC) column (Grace Davison Discovery Sciences, 4.6×250 mm, 5-micron, Vydac 218TP C18).

The samples were analyzed using Shimadzu gradient HPLC (Shimadzu Co., Kyoto, Japan) in a system of 0.1% aqueous TFA (buffer A) versus 0.1% TFA in acetonitrile (buffer B) at a flow rate of 1 mL/min. A 100 μL volume of each sample was injected into the C18 column, which was pre-equilibrated with 50% buffer B. Simvastatin was eluted with a two-step gradient of acetonitrile: 50% acetonitrile containing 0.1% TFA for 10min followed by 80% acetonitrile containing 0.1% TFA for an additional 10min. Simvastatin was monitored by measuring the absorbance at 238 nm using a Shimadzu SPD-M20A UV/VIS photodiode array detector (Shimadzu Co.) interfaced to a computer running Shimadzu LCsolution software. The standard curve, which was created using eight different concentrations of the simvastatin standard, was linear from 2 to 640 pmol (correlation coefficient 0.99988). The detection limit using this method was estimated to be approximately 1.5 pmol (signal-to-noise ratio greater than 2).

Pharmacokinetic data were then analyzed using Phoenix WinNonlin software version 5.3 (Certera, St. Louis, MO, USA). The following equation was used for the one-compartment model:

where C(t) denotes the quantity of simvastatin at time t, Vd denotes the volume of distribution, and K(h-1) denotes the elimination rate constant. In addition, data were analyzed using a non-compartmental model for reference.

Retinal Histology  On the 7th day following injection, anesthetized mice were euthanatized via cervical dislocation, and their eyes were enucleated after ERG recording was conducted. For histologic studies, a large full-thickness incision was made in the cornea, and the anterior segment was removed. Then, the eye cups were fixed in 3% glutaraldehyde in phosphate buffer. The tissue was then washed in 1 mol/L sodium phosphate buffer, pH 7.3, and post-fixed in 1% osmium tetroxide for 1h at room temperature. The dehydrated tissue was infiltrated with acetone and Poly/Bed 812 plastic resin. The tissue was then embedded in plastic block molds with 100% Poly/Bed 812. Sections (1 μm thick) were generated on an ultramicrotome, placed on glass slides and stained with toluidine blue for light microscopy. The areas of interest were trimmed, and 80-nm-thick ultra-thin sections were sliced using a Leica Ultracut R Ultramicrotome, mounted on 100-mesh copper grids, and stained with 2% uranyl acetate and Reynold’s lead citrate. The specimens were imaged using a Zeiss CEM 902 electron microscope to study the retinal ultrastructure. For immunohistochemistry (IHC) and cell counting studies, the eyes were carefully dissected to isolate whole retinae, which were then incubated in 4% paraformaldehyde (Electron Microscopy Science, Fort Washington, PA, USA) in Dulbecco's phosphate-buffered saline (DPBS, pH7.4, Invitrogen, LaJolla, CA, USA) at room temperature for 45min for fixation. An indirect antibody method was adopted for IHC. First, retinae were sliced into vertical 40-μm-thick sections using a microtome (Vibratome; Leica Microsystems, Bannockburn, IL, USA) and then blocked with 10% donkey serum (Jackson ImmunoResearch, West Grove, PA, USA) in TBS [DPBS containing 0.5% Triton X-100 (Sigma) and 0.1% sodium azide (Sigma), pH 7.2] at 4 overnight to reduce nonspecific labeling. The free-floating sections were incubated in primary antibodies diluted in TBS containing 3% donkey serum at 4 for 4d. Controls lacking primary antibodies were also processed. After several rinses, the sections were transferred and incubated in a TBS solution containing 3% normal donkey serum and donkey-hosted secondary antibodies conjugated with Cy3 (1:200, Jackson ImmunoResearch) or Alexa Fluor 488 (1:200 dilution, Molecular Probes, Eugene, OR) at 4 overnight. A fluorescent nuclear dye, TO-PRO3 (1:3000 dilution, Molecular Probes, Eugene, OR, Cat. No. T3605), was applied together with the secondary antibodies.

After rinsing several times, the sections were mounted with Vectashield medium (Vector Laboratories, Burlingame, CA, USA), cover slipped and, finally, observed under a confocal laser scanning microscope (LSM 510; Zeiss, Thornwood, NY, USA). Images were acquired using Zeiss LSM software and a 40× or a 63× oil-immersion objective. Adobe Photoshop CS5 (Adobe Systems, San Jose, CA, USA) was used to crop images and to apply uniform brightness and contrast adjustments.

In the present study, rod bipolar cells were immuno-labeled with a mouse antibody against PKCα (1:250 dilution; BD Transduction Labs, San Jose, CA, USA, Cat. No. 610107)[40]. Photoreceptor nuclei in the outer retina and ganglion cell nuclei in the inner retina were stained using the fluorescent nuclear dye TO-PRO3[41]. Cone cell bodies were immuno-labeled with a rabbit antibody against GNAT2 (1:100 dilution; Santa Cruz Biotechnology, Santa Cruz, CA, USA, Cat. No. SC-390)[42-43]. The dimensions were measured using built-in tools in Zeiss LSM software.

To determine whether injection of 200 μmol/L simvastatin produced any toxicity to the retina and caused any retinal cell loss, the thicknesses of the two retinal nuclear layers were gauged, and the numbers of cone soma, rod bipolar cells and ganglion cells were systematically counted in the two groups. Five mice, each of which received injections of 200 μmol/L simvastatin into one eye and vehicle solution into the other eye, were sacrificed. Three sections from each eye were imaged, for a sample size of 15 per group; the number of cells in each 200-μm region were counted, and the dimensions were measured.


RESULTS

Electroretinography  Changes in the amplitudes of the scotopic a-wave and b-wave against an increasing stimulus intensity measured 1 and 7d after injection of 50 or 200 μmol/L simvastatin are shown in Figure 1. As shown in panels A-D, eyes injected with either 50 or 200 μmol/L simvastatin showed no significant differences in the a- or b-waves at 9 stimulus intensities 1 or 7d after injection on ERG compared to the control eyes. Baseline a-wave and b-wave responses from another 7 pairs of eyes before injection are shown in panel E for reference.

Dennis Y. Tse1

Figure 1 Changes in the amplitudes of the scotopic a-wave and b-wave against an increasing stimulus intensity measured after injection of simvastatin  A: Day 1 after injection of 50 μmol/L simvastatin; B: Day 7 after injection of 50 μmol/L simvastatin; C: Day 1 after injection of 200 μmol/L simvastatin; D: Day 7 after injection of 200 μmol/L simvastatin; E: Pre-injection.

 

To test whether simvastatin produced any toxicity to the retinal cone pathway, we employed paired flash ERG recordings to isolate the cone-driven responses from the rod-driven responses. As shown in Figure 2, there were no significant differences in the cone a-wave or b-wave between the simvastatin-injected eyes and the vehicle-injected control eyes based on ERG recordings performed 1 and 7d after injection (7 pairs of eyes). Figure 3 shows the changes in the positive STRs (pSTRs) and the negative STRs (nSTRs) in response to increasing stimulus intensities 1 and 7d after injection of 50 or 200 μmol/L simvastatin. There were no significant differences in the mean pSTR or nSTR between simvastatin-injected eyes and control eyes for either concentration at each time point. Given that the pSTR and the nSTR represent inner retinal signaling from third-order neurons such as RGCs and AII amacrine cells[44-46], our results suggest that a vitreal simvastatin concentration of 200 μmol/L or less does not alter the electrophysiological functions of ganglion cells.

Dennis Y. Tse2

Figure 2 The differences in the cone a-wave or b-wave between the simvastatin-injected eyes and the vehicle-injected control eyes based on ERG recordings  A: Pre-injection; B: Day 1 after injection of 200 μmol/L simvastatin; C: Day 7 after injection of 200 μmol/L simvastatin.

Dennis Y. Tse3

Figure 3  Changes in the positive STRs (pSTRs) and the negative STRs (nSTRs) in response to increasing stimulus intensities after injection of simvastatin  A: Day 1 after injection of 50 μmol/L simvastatin; B: Day 7 after injection of 50 μmol/L simvastatin; C: Day 1 after injection of 200 μmol/L simvastatin; D: Day 7 after injection of 200 μmol/L simvastatin; E: Pre-injection.

 

Pharmacokinetic Analysis  The peak simvastatin concentration was calculated according to appropriate standard curves using LCsolution software. These standard curves, created using eight different concentrations of simvastatin, were linear from 0 to 640 pmol (correlation coefficient >0.99988). The detection limit was below 2 pmol.

Simvastatin was detectable in retinal samples collected 1h following the injection. Samples collected at the subsequent post-injection time points showed decreasing concentrations of simvastatin. These findings suggest that intravitreally injected simvastatin arrives at the retina shortly after injection; additionally, simvastatin reached its maximum concentration within 1h. The change in the mean simvastatin concentration in the retina over time as measured using HPLC was fitted using a one-compartment model (Figure 4). The half-lives of simvastatin for injected two concentrations of 200 and 50 µmol/L were found to be 1.92 and 2.41h, respectively (Table 1). Projected Cmax were 506.37 and 75.23 mol, respectively. Mean residence time were 2.77 and 3.48h, respectively.

Dennis Y. Tse4

Figure 4  The change in the mean simvastatin concentration in the retina over time as measured using HPLC was fitted using a one-compartment model  A, B: Injection of 200 μmol/L simvastatin; C, D: Injection of 50 μmol/L simvastatin.

Table 1 Pharmacokinetic parameters of simvastatin in retina


NCA: Non-compartmental analysis; 1CM: 1-compartment modeling; Cmax: Observed/predicted max quantity; Tmax: Time to Cmax; AUC: Area under the curve; MRT: Mean residence time; CL: Clearance. Mean wet weight of mouse retina=3.3 mg; data taken from Cerani et al[47].

 

Retinal Histology  Gross examination of eye specimens revealed no evidence of retinal tearing, retinal detachment, or retinal hemorrhage or any signs of infection in any of the simvastatin-injected or control eyes. The mean thicknesses of the outer nuclear layer (ONL) were 62.5±0.9 and 63.0±1.3 μm in the simvastatin and control groups. The ONL thicknesses in terms of cell number (rods and cones) were 11.7±0.2 and 11.9±0.2 in the simvastatin and control groups. The mean numbers of cone soma were 14.5±1.7 and 15.4±1.0 per 200-μm horizontal ONL section in the simvastatin and control groups. Similarly, the mean numbers of rod bipolar cells (RBCs) in the inner nuclear layer (INL) were 22.9±0.7 and 22.7±0.5 per 200 μm in the simvastatin and control groups. The mean thicknesses of the INL were 43.4±1.3 and 44.0±1.8 μm in the simvastatin and control groups. Finally, the numbers of ganglion cells per 200-μm horizontal ganglion cell layer section were 23±0.6 and 23.1±0.4 in the simvastatin and control groups (Figure 5). There was no significant difference (paired t-test, P>0.05) in the measured layer thicknesses or in the numbers of counted neurons between the 200 μmol/L simvastatin-injected group and the control group (Table 2). These findings suggested that intravitreally injecting 200 μmol/L simvastatin did not induce a loss of retinal neurons.

Dennis Y. Tse5

Figure 5 The cellular profiles of the Simvastatin-injected group with those of the control group  A: A bar chart comparing the cellular profiles of the Simvastatin-injected group with those of the control group. Data points and error bars represent the mean values and standard errors, respectively. B: A representative fluorescent micrograph of a retina processed by staining with the nuclear dye TO-PRO3 (blue) and the antibody GNAT2 (green). A higher-than-normal exposure was used for the green channel to optimally show the cone soma and to facilitate cell counting. Ganglion cell soma were stained in blue in the inner retina. C: A representative fluorescent micrograph of a retina processed by staining with the antibody PKCα (red) and the nuclear dye TO-PRO3 (blue).

Table 2 Comparing the retinal cellular profile of simvastatin injected eyes and vehicle injected control eyes


Low magnification histologic examination via light microscopy conducted 7d after injection revealed normal neurosensory retinae. No signs of inflammation- or toxicity-induced changes in any retinal layer were found. There were no observable differences in layer thicknesses or cell morphologies between the simvastatin-injected retinae and the control retinae (Figure 6).

Dennis Y. Tse6

Figure 6 Light micrographs of vertical retinal sections of mouse eyes processed 7d after intravitreal injection of (A) the control vehicle solution or (B) 2.0 mmol/L Simvastatin  GCL: Ganglion cell layer; IPL: Inner plexiform layer; INL: Inner nuclear layer; OPL: Outer plexiform layer; ONL: Outer nuclear layer; OS: Outer segment of photoreceptors.

 

High magnification histologic examination of individual retinal layers via electron microscopy revealed very similar ultrastructure and cell morphologies between the simvastatin-injected retinae and the control retinae (Figure 7). No abnormalities in photoreceptors, bipolar cells or ganglion cells were identified in the simvastatin-injected eyes 7d after injection.

Dennis Y. Tse7

Figure 7 Electron micrographs of vertical retinal sections of mouse eyes processed 7d after intravitreal injection of (left column) the control vehicle solution or (right column) 200 μmol/L Simvastatin  A, B: Ganglion cells; C, D: Inner nuclear layer; E, F: Photoreceptor inner segment; G, H: Photoreceptor outer segment and retinal pigment epithelium.

 

DISCUSSION

Simvastatin has been suggested to have a pleiotropic protective effect against a variety of ocular diseases, including diabetic retinopathy, AMD, retinal ischemia and glaucoma. More recently, its protective effect against glaucomatous neuronal damage has been hypothesized to be related to its anti-inflammatory properties[11]. However, the ocular toxicity and pharmacokinetics of simvastatin had not been adequately studied previously. In the present study, we have shown that intravitreal injection of up to 200 μmol/L simvastatin produced no apparent adverse effects in the mouse retina based on results from functional tests via ERG and histologic examinations via fluorescence and electron microscopy.

ERG is a useful tool for evaluating retinal functions both experimentally and clinically. In the present study, ERG was performed using a comprehensive range of stimuli to assess the functions of a comprehensive range of retinal neurons. Depending on the stimulus intensity, the a-wave and the b-wave are generated via the rod pathway, the cone pathway or a combination of the two. The scotopic a-wave and b-wave data suggest that simvastatin up to a vitreal concentration of 200 μmol/L does not influence the functions of rod photoreceptors or rod-driven bipolar cells. There were no significant differences in the cone a-wave or b-wave between the simvastatin-injected eyes and the control eyes. This observation indicated that a vitreal simvastatin concentration of 200 μmol/L did not produce any signs of toxicity to the retinal cone pathway. The pSTR and the nSTR are the most sensitive components of the ERG, and our results suggest that a vitreal simvastatin concentration of 200 μmol/L or less does not alter the electrophysiological functions of ganglion cells.

Traditionally, statins are administered systemically via oral ingestion, subcutaneous injection, or intraperitoneal injection. In contrast, delivery via intravitreal injection has not been commonly used but has the advantage of enabling the application of a higher dose intraocularly and monocularly within a short interval. Intravitreal injection is also likely to be more efficient when the therapeutic target site is located at the proximal retina, where drugs administered systemically moving from the choroid must overcome diffusion barriers to reach the retina. One successful application of simvastatin was that intravitreally administered 15 μmol/L simvastatin had no apparent adverse effects in rabbits and prevented the progression of induced proliferative vitreoretinopathy[25].

We found that the ocular half-life of simvastatin in the retina following intravitreal administration was 1.92-2.41h; this half-life is relatively short compared with that of other drugs administered via intravitreal injection. For example, the half-lives of dexamethasone and triamcinolone were previously reported to be 3.48h and 1.57d, respectively[37-38]. We cannot directly compare those findings to the present results because the previous studies performed measurements on the rabbit eye. We did not find any related data using mice. In the present study, the HPLC data suggested that intravitreally injected simvastatin rapidly reached the retina and had a short retinal half-life. Our findings indicate that a sustained release mechanism such as an intravitreal implant[48] would be needed to extend the application of simvastatin to the treatment of chronic vitreal-retinal diseases.

Since the calculated half-life is about 2h and simvastatin is mostly cleared in 12h, the 1d time point is appropriate to evaluate acute toxicity and the 1wk time point was designed to evaluate delayed damages, for example, those resulted from inflammation or secondary degeneration. Our results have further shown that administering up to 200 μmol/L simvastatin via intravitreal injection produced no apparent adverse effects on the retinal ultrastructure of mouse eyes, as the retinae showed no signs of anomalies one week after injection. Therefore, a vitreal simvastatin concentration between 50 and 200 μmol/L would be a reasonable starting point for future therapeutic trials using mouse models of acute ocular diseases.

Though the weakest point of this study is that the volume of the mouse vitreous is much smaller than that of the human vitreous, the present study is the first to assess the toxicity of simvastatin in mice, which represent a very useful model animal because of their potential for genetic modification and the wide availability of mouse models of disease. This study represents an important step in the exploration of the full potential benefits of simvastatin to patients with ocular disorders, including diabetic retinopathy, AMD, retinal ischemia, glaucoma and proliferative vitreoretinopathy. Intravitreal injection of simvastatin is a highly efficient route of delivery, but the half-life of intravitreally injected simvastatin is relatively short. To extend its application to the treatment of chronic ocular disorders, a slow-release drug delivery system or vehicle might be necessary to sustain a therapeutic simvastatin dosage in the retina.


ACKNOWLEDGEMENTS

We thank Zhuo Yang for help with animal husbandry and retina extraction, Ralph Nichols for help with electron microscopy, and Roy Jacoby for carefully reading the manuscript.

Foundations: Supported by the National Institute of Health under Award Number R01 EY004446 & R01 EY019908, NIH Vision Core EY02520, the Retina Research Foundation (Houston), Research to Prevent Blindness Inc., and Hong Kong Polytechnic University grants G-UA7J and G-YBQT.

Conflicts of Interest: Tse DY, None; Kim SJ, None; Chung I, None; He F, None; Wensel TG, None; Wu SM, None.


REFERENCES

1 Molgaard J, von Schenck H, Olsson AG. Effects of simvastatin on plasma lipid, lipoprotein and apolipoprotein concentrations in hypercholesterolaemia. Eur Heart J 1988;9(5):541-551. [CrossRef] [PubMed]

2 Cimino M, Gelosa P, Gianella A, Nobili E, Tremoli E, Sironi L. Statins: multiple mechanisms of action in the ischemic brain. Neuroscientist 2007;13(3):208-213. [CrossRef] [PubMed]

3 Kwak B, Mulhaupt F, Veillard N, Pelli G, Mach F. The HMG-CoA reductase inhibitor simvastatin inhibits IFN-gamma induced MHC class II expression in human vascular endothelial cells. Swiss Med Wkly 2001;131(3-4):41-46. [PubMed]

4 Zacco A, Togo J, Spence K, Ellis A, Lloyd D, Furlong S, Piser T. 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors protect cortical neurons from excitotoxicity. J Neurosci 2003;23(35):11104-11111. [PubMed]

5 Vaughan CJ, Delanty N. Neuroprotective properties of statins in cerebral ischemia and stroke. Stroke 1999;30(9):1969-1973. [CrossRef] [PubMed]

6 Menge T, von Büdingen HC, Zamvil SS, Hartung HP, Kieseier BC, Stüve O. Statins for treatment of CNS diseases. Status report from research and clinical practice. Nervenarzt 2005;76(4):426-437. [CrossRef] [PubMed]

7 Gordon B, Chang S, Kavanagh M, Berrocal M, Yannuzzi L, Robertson C, Drexler A. The effects of lipid lowering on diabetic retinopathy. Am J Ophthalmol 1991;112(4):385-391. [CrossRef]

8 Tan JSL, Mitchell P, Rochtchina E, Wang JJ. Statins and the long-term risk of incident age-related macular degeneration: the blue mountains eye study. Am J Ophthalmol 2007;143(4):685-687. [CrossRef] [PubMed]

9 Stein JD, Newman-Casey PA, Talwar N, Nan B, Richards JE, Musch DC. The relationship between statin use and open-angle glaucoma. Ophthalmology 2012;119(10):2074-2081. [CrossRef] [PMC free article] [PubMed]

10 Leung DY, Li FC, Kwong YY, Tham CC, Chi SC, Lam DS. Simvastatin and disease stabilization in normal tension glaucoma: a cohort study. Ophthalmology 2010;117(3):471-476. [CrossRef] [PubMed]

11 Soto I, Howell GR. The complex role of neuroinflammation in glaucoma. Cold Spring Harb Perspect Med 2014;4(8). pii:017269. [CrossRef] [PMC free article] [PubMed]

12 Davaro F, Forde SD, Garfield M, Jiang Z, Halmen K, Tamburro ND, Kurt-Jones E, Fitzgerald KA, Golenbock DT, Wang D. 3-Hydroxyl-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor (statin)-induced 28-kDa interleukin-1beta interferes with mature IL-1beta signaling. J Biol Chem 2014;289(23):16214-16222. [CrossRef] [PMC free article] [PubMed]

13 Miyahara S, Kiryu J, Yamashiro K, Miyamoto K, Hirose F, Tamura H, Katsuta H, Nishijima K, Tsujikawa A, Honda Y. Simvastatin inhibits leukocyte accumulation and vascular permeability in the retinas of rats with streptozotocin-induced diabetes. Am J Pathol 2004;164(5):1697-1706. [CrossRef]

14 Zhang W, Yan H. Simvastatin increases circulating endothelial progenitor cells and reduces the formation and progression of diabetic retinopathy in rats. Exp Eye Res 2012;105:1-8. [CrossRef] [PubMed]

15 Al-Shabrawey M, Bartoli M, El-Remessy AB, Ma G, Matragoon S, Lemtalsi T, Caldwell RW, Caldwell RB. Role of NADPH oxidase and Stat3 in statin-mediated protection against diabetic retinopathy. Invest Ophthalmol Vis Sci 2008;49(7):3231-3238. [CrossRef] [PMC free article] [PubMed]

16 Kim KJ, Kim KS, Kim NR, Chin HS. Effects of simvastatin on the expression of heme oxygenase-1 in human RPE cells. Invest Ophthalmol Vis Sci 2012;53(10):6456-6464. [CrossRef] [PubMed]

17 James ER. The etiology of steroid cataract. J Ocul Pharmacol Ther 2007;23(5):403-420. [CrossRef] [PubMed]

18 Binder S. Loss of reactivity in intravitreal anti-VEGF therapy: tachyphylaxis or tolerance? Br J Ophthalmol 2012;96(1):1-2. [CrossRef] [PubMed]

19 Dibas A, Yorio T. Glucocorticoid therapy and ocular hypertension. Eur J Pharmacol 2016;787:57-71. [CrossRef] [PubMed]

20 Scott AW, Bressler SB. Long-term follow-up of vascular endothelial growth factor inhibitor therapy for neovascular age-related macular degeneration. Curr Opin Ophthalmol 2013;24(3):190-196. [CrossRef] [PubMed]

21 Schmeer C, Gamez A, Tausch S, Witte OW, Isenmann S. Statins modulate heat shock protein expression and enhance retinal ganglion cell survival after transient retinal ischemia/reperfusion in vivo. Invest Ophthalmol Vis Sci 2008;49(11):4971-4981. [CrossRef] [PubMed]

22 Krempler K, Schmeer CW, Isenmann S, Witte OW, Löwel S. Simvastatin improves retinal ganglion cell survival and spatial vision after acute retinal ischemia/reperfusion in mice. Invest Ophthalmol Vis Sci 2011;52(5):2606-2618. [CrossRef] [PubMed]

23 Ko ML, Chen CF, Peng PH, Peng YH. Simvastatin upregulates Bcl-2 expression and protects retinal neurons from early ischemia/reperfusion injury in the rat retina. Exp Eye Res 2011;93(5):580-585. [CrossRef] [PubMed]

24 Kretz A, Schmeer C, Tausch S, Isenmann S. Simvastatin promotes heat shock protein 27 expression and Akt activation in the rat retina and protects axotomized retinal ganglion cells in vivo. Neurobiol Dis 2006;21(2):421-430. [CrossRef] [PubMed]

25 Kawahara S, Hata Y, Kita T, Arita R, Miura M, Nakao S, Mochizuki Y, Enaida H, Kagimoto T, Goto Y, Hafezi-Moghadam A, Ishibashi T. Potent inhibition of cicatricial contraction in proliferative vitreoretinal diseases by statins. Diabetes 2008;57(10):2784-2793. [CrossRef] [PMC free article] [PubMed]

26 Sasaki H, Yamamura K, Mukai T, Nishida K, Nakamura J, Nakashima M, Ichikawa M. Enhancement of ocular drug penetration. Crit Rev Ther Drug Carrier Syst 1999;16(1):85-146. [CrossRef]

27 Lee JE, Lim DW, Park HJ, Shin JH, Lee SM, Oum BS. Intraocular toxicity and pharmacokinetics of candesartan in a rabbit model. Invest Ophthalmol Vis Sci 2011;52(6):2924-2929. [CrossRef] [PubMed]

28 Chiu K, Chang RC, So KF. Intravitreous injection for establishing ocular diseases model. J Vis Exp 2007;8:313. [CrossRef]

29 Levkovitch-Verbin H. Animal models of optic nerve diseases. Eye (Lond) 2004;18(11):1066-1074. [CrossRef] [PubMed]

30 Frankfort BJ, Khan AK, Tse DY, Chung I, Pang JJ, Yang Z, Gross RL, Wu SM. Elevated intraocular pressure causes inner retinal dysfunction before cell loss in a mouse model of experimental glaucoma. Invest Ophthalmol Vis Sci 2013;54(1):762-770. [CrossRef] [PMC free article] [PubMed]

31 Reichstein D, Ren L, Filippopoulos T, Mittag T, Danias J. Apoptotic retinal ganglion cell death in the DBA/2 mouse model of glaucoma. Exp Eye Res 2007;84(1):13-21. [CrossRef] [PubMed]

32 Barathi VA, Yeo SW, Guymer RH, Wong TY, Luu CD. Effects of simvastatin on retinal structure and function of a high-fat atherogenic mouse model of thickened Bruch's membrane. Invest Ophthalmol Vis Sci 2014;55(1):460-468. [CrossRef] [PubMed]

33 Sadeghi MM, Collinge M, Pardi R, Bender JR. Simvastatin modulates cytokine-mediated endothelial cell adhesion molecule induction: involvement of an inhibitory g protein. J Immunol 2000;165(5):2712-2718. [CrossRef]

34 Remtulla S, Hallett PE. A schematic eye for the mouse, and comparisons with the rat. Vision Res 1985;25(1):21-31. [CrossRef]

35 Lebrun-Julien F, Bertrand MJ, De Backer O, Stellwagen D, Morales CR, Di Polo A, Barker PA. ProNGF induces TNFalpha-dependent death of retinal ganglion cells through a p75NTR non-cell-autonomous signaling pathway. Proc Natl Acad Sci U S A 2010;107(8):3817-3822. [CrossRef] [PMC free article] [PubMed]

36 Pennesi ME, Howes KA, Baehr W, Wu SM. Guanylate cyclase-activating protein (GCAP) 1 rescues cone recovery kinetics in GCAP1/GCAP2 knockout mice. Proc Natl Acad Sci U S A 2003;100(11):6783-6788. [CrossRef] [PMC free article] [PubMed]

37 Abd-El-Barr MM, Albini TA, Carvounis PE, He F, Manzano RP, Chevez-Barrios P, Wensel TG, Wu SM, Holz ER. Safety and pharmokinetics of triamcinolone hexacetonide in rabbit eyes. J Ocul Pharmacol Ther 2008;24(2):197-205. [CrossRef] [PubMed]

38 Kwak HW, D'Amico DJ. Evaluation of the retinal toxicity and pharmacokinetics of dexamethasone after intravitreal injection. Arch Ophthalmol 1992;110(2):259-266. [CrossRef]

39 Chin HS, Park TS, Moon YS, Oh JH. Difference in clearance of intravitreal triamcinolone acetonide between vitrectomized and nonvitrectomized eyes. Retina 2005;25(5):556-560. [CrossRef]

40 Zhang J, Yang Z, Wu SM. Development of cholinergic amacrine cells is visual activity-dependent in the postnatal mouse retina. J Comp Neurol 2005;484(3):331-343. [CrossRef] [PubMed]

41 Vekslin S, Ben-Yosef T. Spatiotemporal expression pattern of ceramide kinase-like in the mouse retina. Mol Vis 2010;16:2539-2549. [PMC free article] [PubMed]

42 Chang B, Dacey MS, Hawes NL, Hitchcock PF, Milam AH, Atmaca-Sonmez P, Nusinowitz S, Heckenlively JR. Cone photoreceptor function loss-3, a novel mouse model of achromatopsia due to a mutation in Gnat2. Invest Ophthalmol Vis Sci 2006;47(11):5017-5021. [CrossRef] [PubMed]

43 Kostic C, Crippa SV, Pignat V, Bemelmans AP, Samardzija M, Grimm C, Wenzel A, Arsenijevic Y. Gene therapy regenerates protein expression in cone photoreceptors in Rpe65(R91W/R91W) mice. PLoS One 2011;6(2):e16588. [CrossRef] [PMC free article] [PubMed]

44 Saszik SM, Robson JG, Frishman LJ. The scotopic threshold response of the dark-adapted electroretinogram of the mouse. J Physiol (Lond) 2002;543(Pt 3):899-916. [CrossRef] [PMC free article] [PubMed]

45 Abd-El-Barr MM, Pennesi ME, Saszik SM, Barrow AJ, Lem J, Bramblett DE, Paul DL, Frishman LJ, Wu SM. Genetic dissection of rod and cone pathways in the dark-adapted mouse retina. J Neurophysiol 2009;102(3):1945-1955. [CrossRef] [PMC free article] [PubMed]

46 Moshiri A, Gonzalez E, Tagawa K, Maeda H, Wang M, Frishman LJ, Wang SW. Near complete loss of retinal ganglion cells in the math5/brn3b double knockout elicits severe reductions of other cell types during retinal development. Dev Biol 2008;316(2):214-227. [CrossRef] [PMC free article] [PubMed]

47 Cerani A, Tetreault N, Menard C, Lapalme E, Patel C, Sitaras N, Beaudoin F, Leboeuf D, De Guire V, Binet F, Dejda A, Rezende FA, Miloudi K, Sapieha P. Neuron-derived semaphorin 3A is an early inducer of vascular permeability in diabetic retinopathy via neuropilin-1. Cell Metab 2013;18(4):505-518. [CrossRef] [PubMed]

48 Chang-Lin JE, Attar M, Acheampong AA, Robinson MR, Whitcup SM, Kuppermann BD, Welty D. Pharmacokinetics and pharmacodynamics of a sustained-release dexamethasone intravitreal implant. Invest Ophthalmol Vis Sci 2011;52(1):80-86. [CrossRef] [PubMed]

--------------------------------------------------------------------------------------------------------------------------------
All rights reserved by Press of International Journal of Ophthalmology (IJO PRESS)

www.ijo.cn