+ All Categories
Home > Documents > A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in...

A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in...

Date post: 07-Sep-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
9
A rat retinal damage model predicts for potential clinical visual disturbances induced by Hsp90 inhibitors Dan Zhou a, , Yuan Liu a , Josephine Ye a , Weiwen Ying a , Luisa Shin Ogawa a , Takayo Inoue a , Noriaki Tatsuta a , Yumiko Wada a , Keizo Koya a , Qin Huang b , Richard C. Bates a , Andrew J. Sonderfan a a Synta Pharmaceuticals Corp., 45 Hartwell Avenue, Lexington, MA 02421, USA b Department of Pathology and Laboratory Medicine, Veterans Affairs Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, USA abstract article info Article history: Received 11 June 2013 Revised 20 September 2013 Accepted 21 September 2013 Available online 1 October 2013 Keywords: Hsp90 inhibitors Visual impairment Retinal degeneration Drug safety assessment Predictive toxicology Rodent model In human trials certain heat shock protein 90 (Hsp90) inhibitors, including 17-DMAG and NVP-AUY922, have caused visual disorders indicative of retinal dysfunction; others such as 17-AAG and ganetespib have not. To understand these safety prole differences we evaluated histopathological changes and exposure proles of four Hsp90 inhibitors, with or without clinical reports of adverse ocular effects, using a rat retinal model. Retinal morphology, Hsp70 expression (a surrogate marker of Hsp90 inhibition), apoptotic induction and pharmacoki- netic drug exposure analysis were examined in rats treated with the ansamycins 17-DMAG and 17-AAG, or with the second-generation compounds NVP-AUY922 and ganetespib. Both 17-DMAG and NVP-AUY922 induced strong yet restricted retinal Hsp70 up-regulation and promoted marked photoreceptor cell death 24 h after the nal dose. In contrast, neither 17-AAG nor ganetespib elicited photoreceptor injury. When the relationship between drug distribution and photoreceptor degeneration was examined, 17-DMAG and NVP-AUY922 showed substantial retinal accumulation, with high retina/plasma (R/P) ratios and slow elimination rates, such that 51% of 17-DMAG and 65% of NVP-AUY922 present at 30 min post-injection were retained in the retina 6 h post-dose. For 17-AAG and ganetespib, retinal elimination was rapid (90% and 70% of drugs eliminated from the retina at 6 h, respectively) which correlated with lower R/P ratios. These ndings indicate that prolonged inhibition of Hsp90 activity in the eye results in photoreceptor cell death. Moreover, the results suggest that the retina/plasma exposure ratio and retinal elimination rate proles of Hsp90 inhibitors, irrespective of their chemical class, may predict for ocular toxicity potential. © 2013 The Authors. Published by Elsevier Inc. Introduction Heat shock protein 90 (Hsp90) is a ubiquitously expressed molecular chaperone required for the post-translational stability of its target substrates, known as client proteins, many of which are critical for cell growth, differentiation and survival (Taipale et al., 2010). In addition to playing an indispensable role in the normal homeostatic maintenance of organs and tissues, it is now recognized that the chaperone functions of Hsp90 can become subverted during tumorigenesis (Whitesell and Lindquist, 2005). In this setting Hsp90 can serve as a biochemical buffer to promote the structural and functional stability of a number of onco- genic signaling proteins causally implicated in human cancers (Trepel et al., 2010; Whitesell and Lindquist, 2005). Of note, pharmacologic inhibition of Hsp90 results in destabilization and targeted proteasomal destruction of these clients and a unique feature of Hsp90 blockade is that it provides a means to simultaneously inhibit multiple oncogenic pathways. For these reasons, Hsp90 has emerged as an attractive molec- ular target for the development of novel cancer therapeutics (Banerji, 2009; Kim et al., 2009). Ocular toxicities are a common side-effect of systemic chemothera- peutic drugs (al-Tweigeri et al., 1996) and have also emerged as an im- portant clinical concern for newer molecularly-targeted agents entering standard oncology practice (Renouf et al., 2012). The rst class of targeted Hsp90 compounds characterized was the benzoquinone ansamycins, which include the natural product geldanamycin and its derivatives 17-DMAG (17-dimethylaminoethylamino-17-demethoxygeldanamycin) and 17-AAG (17-allylamino-17-demethoxygeldanamycin) (Taldone et al., 2008). In recent years, an increasing number of synthetic small molecule inhibitors of Hsp90 have also been developed that are based on a diverse variety of chemical scaffolds, including resorcinol, purine and benzamide structures (Biamonte et al., 2010; Taldone et al., 2008). In human clinical trials some Hsp90 inhibitors, including 17- DMAG, have been associated with visual disorders including blurred vision, ashes, delayed light/dark accommodation, night blindness and photophobia (Kummar et al., 2010; Pacey et al., 2011; Samuel et al., 2010; Shapiro et al., 2010). These ocular adverse effects are often reversible though dose-limiting; moreover, the clinical evaluation Toxicology and Applied Pharmacology 273 (2013) 401409 Corresponding author. Fax: +1 781 274 8228. E-mail address: [email protected] (D. Zhou). 0041-008X © 2013 The Authors. Published by Elsevier Inc. http://dx.doi.org/10.1016/j.taap.2013.09.018 Contents lists available at ScienceDirect Toxicology and Applied Pharmacology journal homepage: www.elsevier.com/locate/ytaap Open access under CC BY-NC-ND license. Open access under CC BY-NC-ND license.
Transcript
Page 1: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

Toxicology and Applied Pharmacology 273 (2013) 401–409

Contents lists available at ScienceDirect

Toxicology and Applied Pharmacology

j ourna l homepage: www.e lsev ie r .com/ locate /ytaap

A rat retinal damage model predicts for potential clinical visualdisturbances induced by Hsp90 inhibitors

Dan Zhou a,⁎, Yuan Liu a, Josephine Ye a, Weiwen Ying a, Luisa Shin Ogawa a, Takayo Inoue a, Noriaki Tatsuta a,Yumiko Wada a, Keizo Koya a, Qin Huang b, Richard C. Bates a, Andrew J. Sonderfan a

a Synta Pharmaceuticals Corp., 45 Hartwell Avenue, Lexington, MA 02421, USAb Department of Pathology and Laboratory Medicine, Veterans Affairs Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, USA

⁎ Corresponding author. Fax: +1 781 274 8228.E-mail address: [email protected] (D. Zhou).

0041-008X © 2013 The Authors. Published by Elsevier Inchttp://dx.doi.org/10.1016/j.taap.2013.09.018

a b s t r a c t

a r t i c l e i n f o

Article history:Received 11 June 2013Revised 20 September 2013Accepted 21 September 2013Available online 1 October 2013

Keywords:Hsp90 inhibitorsVisual impairmentRetinal degenerationDrug safety assessmentPredictive toxicologyRodent model

In human trials certain heat shock protein 90 (Hsp90) inhibitors, including 17-DMAG and NVP-AUY922, havecaused visual disorders indicative of retinal dysfunction; others such as 17-AAG and ganetespib have not. Tounderstand these safety profile differences we evaluated histopathological changes and exposure profiles offour Hsp90 inhibitors, with or without clinical reports of adverse ocular effects, using a rat retinal model. Retinalmorphology, Hsp70 expression (a surrogate marker of Hsp90 inhibition), apoptotic induction and pharmacoki-netic drug exposure analysis were examined in rats treated with the ansamycins 17-DMAG and 17-AAG, or withthe second-generation compounds NVP-AUY922 and ganetespib. Both 17-DMAG and NVP-AUY922 inducedstrong yet restricted retinal Hsp70 up-regulation and promoted marked photoreceptor cell death 24 h afterthe final dose. In contrast, neither 17-AAG nor ganetespib elicited photoreceptor injury. When the relationshipbetween drug distribution and photoreceptor degenerationwas examined, 17-DMAG and NVP-AUY922 showedsubstantial retinal accumulation, with high retina/plasma (R/P) ratios and slow elimination rates, such that 51%of 17-DMAG and 65% of NVP-AUY922 present at 30 min post-injectionwere retained in the retina 6 h post-dose.For 17-AAG and ganetespib, retinal elimination was rapid (90% and 70% of drugs eliminated from the retina at6 h, respectively) which correlated with lower R/P ratios. These findings indicate that prolonged inhibition ofHsp90 activity in the eye results in photoreceptor cell death.Moreover, the results suggest that the retina/plasmaexposure ratio and retinal elimination rate profiles of Hsp90 inhibitors, irrespective of their chemical class,may predict for ocular toxicity potential.

© 2013 The Authors. Published by Elsevier Inc. Open access under CC BY-NC-ND license.

Introduction

Heat shock protein 90 (Hsp90) is a ubiquitously expressedmolecularchaperone required for the post-translational stability of its targetsubstrates, known as client proteins, many of which are critical for cellgrowth, differentiation and survival (Taipale et al., 2010). In addition toplaying an indispensable role in the normal homeostatic maintenanceof organs and tissues, it is now recognized that the chaperone functionsof Hsp90 can become subverted during tumorigenesis (Whitesell andLindquist, 2005). In this setting Hsp90 can serve as a biochemical bufferto promote the structural and functional stability of a number of onco-genic signaling proteins causally implicated in human cancers (Trepelet al., 2010; Whitesell and Lindquist, 2005). Of note, pharmacologicinhibition of Hsp90 results in destabilization and targeted proteasomaldestruction of these clients and a unique feature of Hsp90 blockade

. Open access under CC BY-NC-ND lice

is that it provides a means to simultaneously inhibit multiple oncogenicpathways. For these reasons, Hsp90 has emerged as an attractive molec-ular target for the development of novel cancer therapeutics (Banerji,2009; Kim et al., 2009).

Ocular toxicities are a common side-effect of systemic chemothera-peutic drugs (al-Tweigeri et al., 1996) and have also emerged as an im-portant clinical concern for newer molecularly-targeted agents enteringstandard oncology practice (Renouf et al., 2012). Thefirst class of targetedHsp90 compounds characterized was the benzoquinone ansamycins,which include the natural product geldanamycin and its derivatives17-DMAG (17-dimethylaminoethylamino-17-demethoxygeldanamycin)and 17-AAG (17-allylamino-17-demethoxygeldanamycin) (Taldoneet al., 2008). In recent years, an increasing number of synthetic smallmolecule inhibitors of Hsp90 have also been developed that are basedon a diverse variety of chemical scaffolds, including resorcinol, purineand benzamide structures (Biamonte et al., 2010; Taldone et al.,2008). In human clinical trials some Hsp90 inhibitors, including 17-DMAG, have been associated with visual disorders including blurredvision, flashes, delayed light/dark accommodation, night blindnessand photophobia (Kummar et al., 2010; Pacey et al., 2011; Samuelet al., 2010; Shapiro et al., 2010). These ocular adverse effects areoften reversible though dose-limiting; moreover, the clinical evaluation

nse.

Page 2: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

402 D. Zhou et al. / Toxicology and Applied Pharmacology 273 (2013) 401–409

of the second-generation aminobenzamide-based inhibitor, PF-04929113(SNX-5422), was recently discontinued by Pfizer based on significantdrug-related ocular toxicity in both preclinical and clinical studies(Rajan et al., 2011). The molecular mechanisms underlying such visualdisturbances remain undefined, although recent animal toxicology stud-ies have suggested that retinal dysfunction linked to photoreceptor de-generation and cell deathmaybe a contributing factor (Rajan et al., 2011).

Interestingly, a number of other Hsp90 inhibitors that have under-gone clinical evaluation, including 17-AAG and ganetespib (a secondgeneration resorcinolic compound), have not manifested the same de-gree of visual abnormalities seen for these other compounds (Banerjiet al., 2005; Cho et al., 2011; Demetri et al., 2011; Goldman et al.,2010; Ramalingam et al., 2008; Ramanathan et al., 2005, 2007; Solitet al., 2007;Wong et al., 2011). The reasons for the superior ocular safe-ty profile exhibited by 17-AAG and ganetespib are presently unknown.Here, histopathological changes and retinal drug distribution profilesof four Hsp90 inhibitors, with or without reported clinical visual distur-bances, were evaluated in a rodent model in order to understand theobserved differences in ocular toxicity profile among agents in thisclass. A characteristic feature of targeted Hsp90 inhibition is the induc-tion of heat shock protein 70 (Hsp70), a related molecular chaperonethat plays a key role in the chaperone complex machinery (Mayer andBukau, 2005). In this regard, Hsp70 upregulation is a commonly usedsurrogate biomarker for Hsp90 blockade (Whitesell et al., 2003) andwas used as a biological readout for Hsp90 inhibition in the rat retina.Results presented in this report suggest that Hsp90 plays a criticalrole in normal retinal function and suggest that aberrant inhibition ofHsp90 activity in the eye results in structural damage of the retina andphotoreceptor cell death. In addition, the data suggest that the retina/plasma exposure ratio and retinal elimination rate profiles of individualHsp90 inhibitor compounds, irrespective of their chemical class, repre-sent predictive factors for assessing compounds' potential for oculardamage.

Materials and methods

Hsp90 inhibitors. 17-DMAG and 17-AAGwere purchased from LC Labora-tories (Woburn, MA) and NVP-AUY922 from Selleck Chemicals (Houston,TX). Ganetespib was synthesized by Synta Pharmaceuticals Corp.

Animals, tissue processing and immunohistochemistry. All in vivo proce-dures were approved by the Synta Pharmaceuticals Corp. InstitutionalAnimal Care and Use Committee and carried out in strict accordancewith the Guide for Care and Use of Laboratory Animals of the NationalInstitutes of Health. Male Sprague Dawley (SD) and Long Evansrats (220 ± 60 g; Charles River Laboratories, Wilmington, MA) weremaintained on a 12 h light–dark cycle. Rats were dosed with either17-DMAG (20 mg/kg), 17-AAG (80 mg/kg), ganetespib (20 mg/kg),NVP-AUY922 (10 mg/kg) or vehicle (17-DMAG was formulated inD5W [5% dextrose in water] and the other three compounds in DRD[10% DMSO/14% Cremophor RH40/76%D5W]). All four inhibitors weredosed intravenously as this represents the clinical route of administra-tion for each. Animals were treated on a 2 consecutive day dosingschedule. Eyeswere harvested 24 h after the last dose, fixed inModifiedDavidson's solution for 24–48 h, and then washed with 70–90% etha-nol. Paraffin-embedded eye sections (5 μm) were subject to routineH&E staining, TUNEL staining using the ApopTag Peroxidase ISOLApoptosis Detection kit (Millipore, Billerica, MA), or immunohisto-chemistry with a rabbit polyclonal antibody directed against Hsp70(1:100 dilution; #4872, Cell Signaling Technology, Danvers, MA),according to manufacturers' protocols. Negative control staining wasperformed in the absence of primary antibody.

Histological assessment. Histology slides were reviewed in a blindfashion by two independent investigators followed by a board certifiedpathologist. TUNEL stained sections were scored semi-quantitatively as

follows: negative (−), ≤20 cells/overall section; minimal (±), 21–50 positive cells/overall section; mild (+), N50 positive cells in lessthan 30% of overall retinal section; moderate (++), N50 positive cellsin 30–60% of the overall retinal section; severe (+++), N50 positivecells in greater than 60% of the overall retinal section. Quantitativeimage analysis was performed on the Hsp70 immunostained sections.Three prominently stained retinal regions per rat were randomly select-ed from the peripheral area, proximal to the optical nerve, and withinthe intervening region. Images were captured under identical electronicexposure profiles and analyzed using Image-Pro Plus software (MediaCybernetics Inc., Rockville, MD) at 20× magnification, and measuredas the percentage of Hsp70 positive and negative areas from the innersegment of the photoreceptor layer to the ganglion cell layer. Theaverage %Hsp70-positive area/rat valueswere calculated and expressedas a fold change (median ± SD) compared to the vehicle group. Statis-tical significance was set at P b 0.05 using one-way ANOVA.

Retinal morphological evaluation. As an index of photoreceptor cellloss, a quantitative measurement of mean outer nuclear layer (ONL)thickness was performed according to previously published procedures(LaVail and Lawson, 1986;Williams andHowell, 1983). Ratswere treat-ed with vehicle or 17-DMAG at 20 mg/kg/day, every other day (q.o.d.)for 2 weeks. This dose schedule provided repeated exposure to theinhibitor, and was identified in pilot studies as the maximally tolerateddose. Twenty-four hours after the final dose, eyes were enucleated,fixed inModified Davidson's solution and embedded in paraffin. Retinaltissue was sectioned (4-μm thickness) along the vertical meridianthrough the optic nerve and stained with H&E. In each of the superiorand inferior hemispheres, the ONL thickness was measured quantita-tively using Image-Pro Plus software. The ONL thickness of each of thesuperior and inferior hemispheres was measured in nine sets of threemeasurements each (total of 27 measurements per hemisphere). Eachset was centered on adjacent 500 ± 50 μm segments of the retina, withthe first point of measurement taken approximately 170 ± 30 μm fromthe optic nerve head and subsequent sets located toward the periphery.Comparisons of ONL thickness were made between vehicle and 17-DMAG-treated animals using 6 rats per group.

Western blotting. Retinal tissue (including the retinal pigment epitheli-um layer) from both eyes was dissected using previously describedmethodology (Barres et al., 1988) and homogenized in a lysing matrixtube (MP Biosciences, Santa Ana, CA) containing lysis buffer (CellSignaling Technology). Lysates were clarified by centrifugationand equal amounts of protein resolved by SDS-PAGE before transferto nitrocellulose membranes (Invitrogen, Carlsbad, CA). Membraneswere blocked and then immunoblottedwith antibodies directed againstHsp70/72 (C92F3A-5; Enzo Life Sciences, Farmingdale, NY) or GAPDH(Santa Cruz Biotechnology, Inc., Santa Cruz, CA). The antibody–antigencomplex was visualized and quantitated using the Odyssey system(LI-COR, Lincoln, NE).

Pharmacokinetic analysis. 17-DMAG (20 mg/kg), 17-AAG (80 mg/kg),ganetespib (20 mg/kg) andNVP-AUY922 (10 mg/kg)were i.v. adminis-tered to male SD rats (n = 3). For consistency with the experimentalprotocol, animals were given two consecutive daily doses of inhib-itor prior to PK assessment. Following retro-orbital blood collection,plasma samples were protein precipitated and analyzed by liquidchromatography–tandem mass spectrometry (LC–MS/MS). Noncom-partmental pharmacokinetic analysis was performed using PhoenixWinNonlin ver. 6.3 (Pharsight, Mountain View, CA) for individualanimals and mean data reported for the group.

For the determination of plasma and retinal tissue drug concentra-tions, inhibitors were i.v. administered to rats (n = 3) as two consecu-tive daily doses and plasma and retina samples were collected at 0.5, 6and 18 h after the final dose for bioanalysis. To do this, animals wereeuthanized and retinal samples from each time point were pooled and

Page 3: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

403D. Zhou et al. / Toxicology and Applied Pharmacology 273 (2013) 401–409

processedwith the Covaris Cryoprep system (Woburn,MA) followed byhomogenization in phosphate buffered salinewith an IKA homogenizer.Blood was collected from the vena cava for plasma preparation. Plasmaand homogenized retinal samples were then extracted by protein pre-cipitation and analyzed by LC–MS/MS. A Phenomenex Kinetex 2.6 μC18 (30 × 2.1 mm) column was used with a run time of 3.5 min persample.

Statistical analysis. Data were expressed as means ± SD. Mean ormedian values were analyzed using one-way ANOVA with Dunn'sMethod for multiple comparisons versus the control groups. P valuesless than 0.05 were considered statistically significant.

Results

Chemical structure and clinical visual disturbance profile ofHsp90 inhibitors

Four small molecule Hsp90 inhibitors, all of which exhibit competi-tive binding for the ATP pocket at the N-terminus of Hsp90, were usedin this study and their chemical structures are presented in Fig. 1. 17-DMAG and 17-AAG both belong to the ansamycin class. Despite theirclose structural similarity, visual disturbances have been associatedwith 17-DMAG, but not 17-AAG, in human clinical trials (Kummaret al., 2010; Pacey et al., 2011). Ganetespib is a novel second genera-tion resorcinolic triazolone compound that is structurally distinctand more potent than the first-generation ansamycins and doesnot cause visual disturbances (Ying et al., 2012). NVP-AUY922 isanother resorcinol-based second generation inhibitor of Hsp90(Eccles et al., 2008) and, in contrast to ganetespib, ocular toxicitieshave recently emerged as an undesirable side-effect for this compound(Samuel et al., 2010).

Fig. 1. Chemical structures of the Hsp90 inhibitors 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), 17-allylamino-17-demethoxygeldanamycin(17-AAG), ganetespib and NVP-AUY922. CLogP values calculated for each compoundusing ChemBioDraw software (CambridgeSoft) are shown.

17-DMAG induced retinal degeneration involves the photoreceptor outernuclear layer in rats

Histologically, the retina is composed of multiple layers of neuronalcells. H&E staining of a normal retina from a vehicle-treated SpragueDawley (SD) rat revealed an orderly structural organization (Fig. 2A),within which the ganglion cell layer (GCL), the photoreceptor inner nu-clear layer (INL) and outer nuclear layer (ONL) (containing the rod andcone cells) could be readily delineated. First a repeat-dose experimentusing SD rats treated with i.v. administration of 17-DMAG (20 mg/kg)or vehicle (D5W) q.o.d. for twoweekswasperformed. This dosewasde-termined to be themaximally tolerated dose (MTD) for the drug, abovewhich animalmortalitywas observed.When retinalmorphologieswereinitially examined by H&E staining no discernible differences were seenin vehicle-treated animals; in contrast, 17-DMAG exposure resulted inmarked attenuation of the thickness of the ONL and prominent cellulardegenerative changes within this region, as evidenced by extensivenuclear condensation and pyknosis (data not shown). These changessuggested that photoreceptor degeneration resulted as a consequenceof exposure to the Hsp90 inhibitor. Therefore the mean ONL thicknessin vehicle- and 17-DMAG-treated SD rats was examined as a measureto evaluate the degree of photoreceptor cell loss. The ONL thicknesswas measured along the entire vertical meridian, and the mean thick-ness values (n = 6 rats) are plotted in Fig. 2B. Compared to the vehiclecontrol rats, the mean ONL thickness in 17-DMAG treated animalswas markedly reduced along the entire length of the retina; by 20–37% within the inferior hemisphere and by 20–62% within the superiorhemisphere.Within themore sensitive superior hemisphere, the reduc-tions in thickness were greatest toward the periphery compared to themore central regions (Fig. 2B).

Fig. 2C (upper panel) shows representative H&E images of retinasobtained from vehicle and 17-DMAG treated SD rats, correspondingto segment 9 of the superior hemisphere shown in Fig. 2B. The loss ofONL thickness was readily apparent (reduced from approximately9–10 rows of nuclei to 3–4 following 17-DMAG treatment), as was amajor disorganization and substantial loss of photoreceptor inner andouter segment membranes. TUNEL staining of adjacent tissue sections(Fig. 2C, lower panel) revealed the presence of apoptotic cells, confinedto the ONL, consistent with a model of selective photoreceptor cellloss occurring in response to 17-DMAG exposure.

In subsequent experiments, rats were treated with vehicle or20 mg/kg 17-DMAG for two consecutive days and retinas harvested24 h following the last dosing. TUNEL staining confirmed that the cellu-lar damage occurring within the ONL was due to apoptotic loss of pho-toreceptors (Fig. 2D, upper panel). Because SD rats lack a pigmentedepithelial cell layer, we performed a comparative study using LongEvans rats to determine whether the presence of this layer conferredany protection from Hsp90 inhibitor-induced cytotoxicity. As shownin Fig. 2D (lower panel), 17-DMAG exposure also resulted in extensiveapoptotic induction throughout, and restricted to, the ONL in theseanimals. The structural damage and widespread cell death observedwithin the retinal photoreceptor layer underscore the ocular toxicityseen in rats treated with 17-DMAG.

Up-regulation of the inducible heat shock protein 70 (Hsp70) following17-DMAG exposure accompanies apoptotic induction

Hsp90 is ubiquitously observed throughout all layers of the retina(Dean and Tytell, 2001) and no significant changes in total proteinlevels were seen by Western blot following 17-DMAG treatment(Supplementary Fig. 1). Therefore levels of the stress protein Hsp70were examined as a surrogate marker for Hsp90 inhibition, sinceHsp70 expression is induced following treatmentwithHsp90 inhibitors(Whitesell et al., 2003). As shown in Fig. 3A, Hsp70 was robustly up-regulated following 17-DMAG treatment, displaying strong cytoplasmicstaining in the inner segment and synaptic regions of the photoreceptor

Page 4: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

Fig. 2. 17-DMAG-induced ocular toxicity involves apoptotic cell death within the photoreceptor outer nuclear layer (ONL) in both Sprague Dawley (SD, albino) and Long Evans(LE, pigmented) rats. (A) H&E staining of a normal retina from a vehicle (D5W)-treated SD rat shows an orderly arrangement of photoreceptors, including the outer nuclear layer(ONL), inner nuclear layer (INL) and ganglion cell layer (GCL). Scale bar, 50 μm. (B) Loss of ONL thickness across the retina following 17-DMAG treatment. SD rats were treated withrepeated doses of vehicle (openboxes) or 20 mg/kg17-DMAGq.o.d. (closed boxes) for twoweeks and eyes harvested 24 h after thefinal dose. TheONL thicknesswithin eachof the superiorand inferior hemispheres was measured in nine sets of adjacent 500 ± 50 μm segments from the optic nerve (ON). Each data point represents the mean ± SD obtained from 6 rats.(C) Representative tissue sections taken from point 9 in the superior hemisphere as shown in (B). H&E staining is shown in the upper panel and TUNEL staining in the lower. Scale bar,50 μm. (D) TUNEL staining of retinal tissue harvested from SD (upper panels) and LE (lower panels) rats 24 h after 2 consecutive daily doses of 20 mg/kg 17-DMAG confirms the presenceof apoptotic photoreceptors within the ONL. Insets show apoptotic cells in more detail. Original magnification, 100×. Scale bar, 50 μm.

404 D. Zhou et al. / Toxicology and Applied Pharmacology 273 (2013) 401–409

cells. Hsp70 induction was restricted to the photoreceptor and ONLlayers — a distribution consistent with targeted Hsp90 inhibitionoccurring selectively within the cells of this region. Next the kinet-ics of Hsp70 regulation were determined by Western blot (Fig. 3B).Hsp70 protein levels were maximally induced 24 h following thelast 17-DMAG treatment (Day 1), remained elevated for at least3 days, and then returned toward the baseline level at Day 5. Thistemporal induction of Hsp70 was confirmed by immunostaining(Fig. 3C, upper panel). Interestingly, when concomitant TUNELstaining was performed we observed almost identical kineticsof apoptotic induction (Fig. 3C, lower panel). The relative levelsof Hsp70 protein expression are quantitated in Fig. 3D and apopto-tic scores shown in Fig. 3E. Taken together, these data show that17-DMAG treatment aberrantly inhibits Hsp90 activity in retinalphotoreceptor cells and this exposure promotes subsequent celldeath.

Comparison of retinal degeneration in SD rats treated with the Hsp90inhibitors 17-AAG, ganetespib and NVP-AUY922

Retinal damage profiles of 17-AAG, ganetespib and NVP-AUY922were examined. Rats were treated with 2-day consecutive dosingof vehicle (DRD), 17-AAG (80 mg/kg), ganetespib (20 mg/kg) or NVP-AUY922 (10 mg/kg) before retinal harvest 24 h later. The 17-AAGdose used was the maximally tolerated dose (MTD) for this drug inorder to provide a direct comparison with 17-DMAG in the rat modelsystem. Continuing development of these two ansamycin Hsp90 inhib-itors as cancer therapeutics has largely been halted; however bothof the second-generation compounds ganetespib and NVP-AUY922are currently undergoing clinical evaluation inmultiple trials and cancertypes. Therefore the doses of ganetespib and NVP-AUY922 used inthis study were selected based on equivalency to clinically-relevanthuman dosing levels. Similar to what was observed for 17-DMAG,

Page 5: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

Fig. 3.Hsp70 expression is up-regulated following 17-DMAG exposure and accompanies apoptotic induction. SD rats were treatedwith 2 consecutive daily doses of vehicle (D5W) or 17-DMAGand retinas harvested after 1, 3 and 5 days. (A) Representative image of Hsp70 immunoreactivity in the SD rat retina following exposure to 17-DMAG. Scale bar, 50 μm. (B) Kineticsof Hsp70 protein regulation after 17-DMAG treatment assessed byWestern blot. Each lane represents retinal tissue from an individual animal. Hsp70 expression was maximally induced1 day following the last dose, remained elevated at Day 3, and then returned to baseline levels at Day 5. GAPDH included as a loading control. (C) Time course of Hsp70 (upper panel) andapoptosis (lower panel) induction determined by immunohistochemistry and TUNEL staining, respectively. Maximal expression of both Hsp70 and apoptotic photoreceptor destructionwas seen 1 day following the last 17-DMAG dosing. Scale bars, 50 μm. (D) Quantification of Hsp70 expression for Days 1, 3 and 5. *P b 0.05; one-way ANOVA. (E) Quantification of theapoptotic index (average histology scores) for Days 1, 3 and 5. *P b 0.05.

405D. Zhou et al. / Toxicology and Applied Pharmacology 273 (2013) 401–409

NVP-AUY922 treatment induced extensive photoreceptor cell death.In stark contrast, and consistent with an absence of clinically reportedvisual changes, neither 17-AAG nor ganetespib produced any detectableapoptotic photoreceptor injury (Fig. 4A, Table 1). As predicted by thesefindings, only NVP-AUY922 treatment elicited robust up-regulationof Hsp70 within the retinal tissue (Fig. 4B).

Fig. 4.Retinal toxicity profiles of 17-AAG, ganetespib andNVP-AUY922. (A) TUNEL staining of SDor AUY922. Inset shows apoptotic induction within the ONL in response to NVP-AUY922 exposWestern blot from 3 individual animals following Hsp90 inhibitor treatment. GAPDH included

Comparative pharmacokinetics

The relationship between drug distribution profiles and photorecep-tor degeneration using pharmacokinetic (PK) analyses was examinedfor each Hsp90 inhibitor. The plasma concentration–time profilespresented in Fig. 5A were obtained after the i.v. administration of each

rat retinal tissue 24 h after 2 consecutive daily doses of vehicle (DRD), 17-AAG, ganetespibure in more detail. Scale bar, 50 μm. (B) Retinal Hsp70 protein expression determined byas a loading control.

Page 6: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

Table 1Comparison of Hsp90 inhibitor ocular toxicity in SD rats.

Compound Dosage(mg/kg/day ×2, i.v.)

IHC staining Western blot

Apoptosisa

(# affected/total animals) (scoreb)Hsp70 increase vs. controla (% ± SD)

Vehicle 0 19/20 (−), 1/20 (±) 0 ± 917-AAG 80 0/7 (−) 14 ± 7Ganetespib 20 0/7 (−) 34 ± 1217-DMAG 20 9/9 (+)–(+++) 122 ± 24⁎

NVP-AUY922 10 6/6 (±)–(+++) 117 ± 7⁎

a Evaluations performed 24 h following final dose.b Histological score for apoptosis (TUNEL staining): (−) negative, (±) minimal, (+) mild, (++) moderate, and (+++) severe.⁎ P b 0.05. Multiple comparisons versus control group (Dunn's Method).

406 D. Zhou et al. / Toxicology and Applied Pharmacology 273 (2013) 401–409

compound at the above doses in SD rats and themain PKparameters aresummarized in Table 2. Collectively the data show that inhibitor differ-ences in standard PK parameters did not correlate with the capacityto induce retinal effects. Notably, the peak concentrations of 17-DMAGand NVP-AUY922, both of which were retinal damage inducers, weresubstantially lower than those for 17-AAG and ganetespib (Table 2).Indeed, ganetespib showed an almost 25-fold higher Cmax value than17-DMAG when administered at the same 20 mg/kg dose (Fig. 5A)and, over time, was present at higher circulating concentrations thanNVP-AUY922. Reflecting its dosing level (80 mg/kg), 17-AAG showedthe highest initial plasma drug concentrations however the short half-life of the compound (T1/2, 2.8 h) promoted rapid clearance fromthe bloodstream (Fig. 5A). Because the extent of plasma protein binding

Fig. 5.Comparative pharmacokinetic analyses. (A)Meanplasma concentration–time profiles aftNVP-AUY922 were dosed at 20 mg/kg, 80 mg/kg, 20 mg/kg and 10 mg/kg, respectively. Data reistration of Hsp90 inhibitors to SD rats. Retinal samples were collected at 0.5, 6 and 18 h post-retina/plasma (R/P) drug concentration ratios and (D) retinal elimination rates over time. Plaskinetic analysis. Data are presented from pooled samples representing 3 animals/time point. ThNVP-AUY922 and 17-DMAG,whichproduced severe ocular effects, displayed thehighest R/P ratretinal damage. Retinal elimination profiles correlatedwith the degree of observed toxicity. 17-Arapidly eliminated from the retina compared to 17-DMAG and NVP-AUY922.

can influence the activity of a drug we additionally determinedthe unbound fraction of each compound, listed in Table 2. Interestingly,despite their different potentials for retinal degeneration, the tworesorcinolic compounds showed almost identicalmean unbound plasmafraction values (2.4% and 2.5% for NVP-AUY922 and ganetespib, respec-tively) suggesting that the availability of these two potent inhibitoryagents for on-target Hsp90 effects was comparable.

Retina/plasma exposure ratio and retinal elimination rate are linked tophotoreceptor toxicity induced by Hsp90 inhibitors

A comprehensive evaluation of the SD rat tissue distribution profilesfor each Hsp90 inhibitor measured at 0.5, 6 and 18 h post-dosing is

er the i.v. administration of Hsp90 inhibitors to SD rats. 17-DMAG, 17-AAG, ganetespib andpresent mean ± SD (n = 3). (B) Retinal concentration–time profiles after the i.v. admin-dose. Data are presented on a logarithmic base 2 scale. (C) Comparison of Hsp90 inhibitorma and retinal samples were collected at 0.5 and 6 h post-dose and subject to pharmaco-e bar graph inset in panel (C) denotes the relative R/P ratios at the initial 0.5 h time point.ioswhereas the lowerR/P ratios of ganetespib and 17-AAGwere associatedwith less risk ofAG and ganetespib, both ofwhich showed an absence of adverse ocular effects, weremore

Page 7: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

Table 2Pharmacokinetics of selected Hsp90 inhibitors in SD rats.

Compound 17-AAG 17-DMAG Ganetespib NVP-AUY922

Dose (mg/kg/day ×2) 80 20 20 10T1/2 (h) 2.8 NR 6.4 5.6Tmax (h) 0.083 0.083 0.083 0.083Cmax (μM) 487 4.48 110 6.19AUCt (μM·h) 619 7.43 36.1 2.63Mean unboundplasma fractiona (%Fu)

5.0 16.0 2.5 2.4

NR, not reportable due to insufficient characterization of the terminal phase.a Concentration tested 10 μM.

407D. Zhou et al. / Toxicology and Applied Pharmacology 273 (2013) 401–409

presented in Table 3. Drug concentrations following i.v. administrationof each compound were found to be generally higher in the retina thanin the plasma, confirming the ability of each compound to penetratethe blood–retina barrier, however the overall plasma concentrationalone did not predict for retinal exposure. When retinal concentrationswere plotted as a function of time (Fig. 5B) it was found that treatmentwith the two second generation compounds resulted in comparablelevels of drug at the 0.5 h time point, yet the retinotoxic NVP-AUY922displayed higher sustained concentrations up to 18 h compared toganetespib. Similarly, while dosing the ansamycin inhibitors at theirrespective MTDs resulted in greater overall retinal values initially, 17-DMAG showed comparatively higher tissue retention than 17-AAGover time (Fig. 5B). In order to more directly compare the distributionprofiles, we determined the retina/plasma (R/P) drug concentrationratio for each compound (Table 3, Fig. 5C), and this parameter wasfound to directly relate to the photoreceptor cell death observed above.17-DMAGandNVP-AUY922, both ofwhich showed severe ocular effects,had R/P ratios at 0.5 h of 6.8 and 4.5, respectively, whereas 17-AAG (R/Pratio of 0.14) and ganetespib (R/P ratio of 1.1) had much lower values(Fig. 5C, Table 3). The higher R/P ratios for NVP-AUY922 and 17-DMAGindicated that these compounds distributed more readily to the retina.Further, at the 6 h time point, the R/P ratios for 17-DMAG and NVP-AUY922 remained greater than those of 17-AAG and ganetespib (51and 288 vs. 1.8 and 28.9, respectively). Moreover, these accumulationprofiles were directly associated with the retinal elimination rate(Fig. 5D), as suggested by Fig. 5B. For 17-AAG, retinal elimination wasrapid — at 6 h post-dose, 90% of 17-AAG present at 30 min had beeneliminated from the retina, accounting for the low R/P ratio. Ganetespibexhibited a similar profile, with 70% retinal elimination of the drugover the same time period. Both 17-DMAG and NVP-AUY922 showedsubstantially slower rates of elimination from the retina at 6 h (Fig. 5D,Table 3). Even at 18 h, NVP-AUY922 showed only 43% clearance com-pared to 87% for ganetespib. Taken together, these data suggest thatboth the degree and duration of Hsp90 inhibitor drug exposure in theretina are linked to ocular toxicity potential.

Table 3Summary of plasma and retinal drug concentrations following Hsp90 inhibitor treatment.

Hsp90 inhibitor Dose(mg/kg)

Time(h)

Plasma(μM)

Retina(nmol/g)

R/P ratioa Retinalelimination

17-AAG 80 0.5 444 59.8 0.14 –

6 3.36 6.18 1.8 90%18 0.52 1.47 2.8 98%

Ganetespib 20 0.5 7.29 7.79 1.1 –

6 0.08 2.30 28.9 70%18 0.005 0.99 190 87%

17-DMAG 20 0.5 4.71 32 6.8 –

6 0.32 16.2 51 49%18 0.29 8.64 30 73%

NVP-AUY922 10 0.5 1.33 5.91 4.5 –

6 0.013 3.85 288 35%18 0.004 3.36 752 43%

a Assuming 1 g tissue equivalent to 1 mL.

Discussion

The advent of molecularly targeted therapies has ushered in a newera for cancer treatment. While these agents are generally consideredto be more tumor-selective and hence elicit fewer side effects than tra-ditional chemotherapeutic drugs, toxicities in normal tissues and organsare still commonly observed. Because of their relatively small mass andrich vascular supply, the eyes are particularly susceptible to insult fromsystemically administered cytotoxic agents (Peponis et al., 2010). Inaddition, their high metabolic rate and inability to regenerate followingdamage are also causative factors. Exposure to toxic compounds canmodify the physiology of retinal neurons and this in turn can leadto major, and in some cases irreversible, visual sensory loss (Renoufet al., 2012). To date, the pathogenic mechanisms of many retinotoxicdrugs have not been well characterized although it is reasonable tosuggest that on-target effects resulting from suppression of oncogenictargets and signaling pathways, which also exist within ocular tissues,play a significant role.

In this study, we initially evaluated histopathological changesinduced within the retina by four Hsp90 inhibitors, two from theprototypical ansamycin class (17-DMAG and 17-AAG) and two second-generation resorcinolic compounds (ganetespib and NVP-AUY922).Although not included in the present report, ocular adverse eventshave not been noted in clinical trials of the purine scaffold-based syn-thetic Hsp90 inhibitor, BIIB021 (Dickson et al., 2013; Elfiky et al., 2008).Consistent with their clinical visual symptom profiles, 17-DMAG andNVP-AUY922 each inducedmarked photoreceptor cell death, but similareffects were not produced by either 17-AAG or ganetespib treatment.Interestingly, the morphologic and structural changes observed in therat retina (degeneration of the photoreceptor segments and thinningof the ONL) are similar to what has been seen following retinal photicinjury (Shahinfar et al., 1991; Youssef et al., 2011), suggesting that phar-macological insult and light damage may manifest similar pathologicoutcomes in this tissue.Moreover, the identical retinal damage producedby two structurally distinct Hsp90 inhibitors (17-DMAG and NVP-AUY922) provides further evidence that retinal toxicity induced by thisgroup of agents is a drug target-dependent process.

In rat ocular tissue, Hsp90 is abundantly expressed within the retina(Dean and Tytell, 2001) suggesting that its chaperoning activities playessential roles in normal photoreceptor function. Our findings indicatethat prolonged or aberrant inhibition of Hsp90 activity in the retinaresults in photoreceptor death. Hsp90 controls the folding of numerouskey signaling molecules required to maintain normal cell function,including ubiquitously expressed apoptotic regulators such as AKT andits effector molecule glycogen synthase kinase 3 (Sato et al., 2000).Therefore, blockade of pro-survival signals could readily account forthe rapid onset of apoptotic loss observed following sustained 17-DMAGorNVP-AUY922 exposure. In addition, thehigh degree of special-ization exhibited by photoreceptor cells predicts for unique chaperonerequirements (Kosmaoglou et al., 2008). In this regard, photoreceptorcell-type specific Hsp90 client proteins may represent critical targetswhose function is may be profoundly impacted by Hsp90 inhibition.For example, aryl hydrocarbon receptor interacting protein-like 1(AIPL1) is expressed by cone and rod photoreceptor cells and plays acritical role in cellular viability (Kirschman et al., 2010). Mutationsin this gene underlie one of the most severe forms of inherited retinaldystrophy, Leber Congenital Amaurosis (LCA) (den Hollander et al.,2008; Sohocki et al., 2000). Notably, AIPL1 has been shown to interactwith Hsp90 (Hidalgo-de-Quintana et al., 2008) and is thought to repre-sent a retina-specific co-chaperone that is required for the assemblyof essential photoreceptor enzymes including the phototransductionprotein, phosphodiesterase (Kirschman et al., 2010; Kosmaoglou et al.,2008). Studies investigating the precise client proteins and signal cas-cades that are destabilized following inhibition of Hsp90 in this tissueare underway. It is also interesting to note that, despitemultiple reportsof Hsp90 inhibitors inducing cytotoxic effects in cultured retinal

Page 8: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

408 D. Zhou et al. / Toxicology and Applied Pharmacology 273 (2013) 401–409

pigment epithelium (RPE) cells in vitro (Kaarniranta et al., 2005;Wu et al., 2010; Yao et al., 2010), only low endogenous levels ofHsp90 protein are found in these cells in situ (Dean and Tytell, 2001)and we observed no apoptotic changes in the SD rat RPE followingHsp90 inhibitor treatment.

Importantly, this study provides the first evidence of a mechanisticbasis for different ocular toxicity potentials exhibited by small moleculeinhibitors of Hsp90. The similar toxicity profiles and biological re-sponses elicited by 17-DMAG and NVP-AUY922 indicated that it wasnot simply chemical class that dictated the capacity to induce retinaldegeneration. Indeed the lack of retinal damage seen with 17-AAGand ganetespib, also in accordance with their clinical safety profiles,supported this premise. Among the compounds studied, 17-DMAGand NVP-AUY922 contain salt-forming amine groups and are more hy-drophilic than either 17-AAG or ganetespib, though it is unlikely thatthis pharmaceutical property alone is sufficient to account for the differ-ent toxicities seen within this group of therapeutic agents. Instead,our pharmacokinetic examination of the relationship between retinaldrug distribution profiles and photoreceptor degeneration showedthat the retention characteristics of each agent appeared to be a predic-tive factor for retinal damage. All four compounds showed distributioninto the retinal tissue; however the potential for photoreceptor damagewas related to the two interconnected parameters of retina/plasmaratio and retinal elimination rate. Indeed, the higher initial R/P ratiosfor 17-DMAG and NVP-AUY922 indicated that both compounds distrib-uted more readily to the retinal compartment compared to the non-retinotoxic inhibitors and actual drug concentrations of each weresustained at higher levels over time. In addition, these characteristicswere sufficient to account for the robust upregulation of Hsp70 seen 1to 3 days following17-DMAGexposure. Hsp70 induction,while a usefulmarker of Hsp90 inhibition, also reflects the activation of an evolution-arily conserved cellular stress response wherein this chaperone mayexert cytoprotective activity and play a central role in the triage ofdamaged proteins following proteotoxic stress (Lanneau et al., 2010).Overall, while all inhibitors successfully penetrated the blood–retinabarrier, the degree and duration of retinal exposure differed evenamong compounds of the same class, and it was these characteris-tics that determined the extent of photoreceptor injury.

Studies of ocular drug reactions are best performed in species forwhich the information can be most relevantly applied to the clinicalsetting. The SD rat is commonly used for regulatory toxicity testingand the retinotoxic responses to individual Hsp90 inhibitors observedin this strain reflected the safety profiles seen in patients. Howeversince these albino animals lack melanin in the RPE or the uveal tract,an important consideration for this study was whether pigmentationof the eye could affect the outcome of Hsp90 inhibitor exposure, giventhat a large number of structurally and pharmacologically unrelateddrugs show significant affinity for binding to melanin. In this regard,while drug binding to melanin itself is not predictive of retinal toxicity(Leblanc et al., 1998), the presence of this biological pigment mayimpact local drug concentrations by promoting accumulation withinthe retinal compartment. As shown in Fig. 2, the pigmented LE rat strainremained susceptible to 17-DMAG-induced retinal damage — whichwas similarly restricted to the photoreceptor layer. Moreover, addition-al tissue distribution studies performed in LE rats also demonstratedhigher R/P ratios for the retinotoxic 17-DMAG and NVP-AUY922 inhib-itors compared to ganetespib and 17-AAG (D. Zhou, unpublished obser-vations). These data are consistent with the observations found in theSD rat and suggest that the presence of melanin in the pigment epithe-lium unlikely plays a major role in determining the relative retentionkinetics of these four compounds within the rat eye.

Finally, our findings validate the use of a rodentmodel to assess risksof visual disturbances and retinal dysfunction induced by targetedHsp90 inhibition. Evaluation of retinal pathology, heat shock proteinmodulation and profiles of retinal drug exposure in rats representeda relatively straightforward, sensitive and robust approach to detect

retinal cell death within 24 h after drug administration. This metho-dology provides a practical advantage over more sophisticated experi-mental approaches, such as electroretinography (ERG), which requiresspecialized equipment to assess clinical visual changes that take consid-erably longer times tomanifest andmonitor. Perhapsmost significantly,our results correlated with the adverse event profile in humans for eachof the agents examined, and suggest that this strategy may predict thepotential for retinal damage when applied to pre-clinical screening ofcandidate Hsp90 inhibitor compounds. Despite some species-relateddifferences (e.g. types and density of cone cells) rat and human eyesshare the same basic structure and function and rodent models havetraditionally proven useful for the investigation of ocular disease patho-genesis, response to therapies, and toxicology screening. It is importantto note, however, that many of the visual disorders seen in patientsare reversible — therefore the photoreceptor death observed in the ratretina likely does not account for all pathological processes occurringin the human eye as a consequence of aberrant Hsp90 inhibition.

In summary, Hsp90 client proteins play important roles in normalretinal function andprolongedHsp90 inhibition can lead to vision disor-ders such as those that have been seen in the clinical setting. Our find-ings reveal that the drug retina/plasma exposure ratio and eliminationrate profiles play crucial roles in ocular toxicity and may be used asindicators of Hsp90 inhibitor-induced damage in rats. Importantly, theretinotoxicity potential of each of the Hsp90 inhibitors defined in therodentmodelwas entirely consistentwith their observed clinical profilein humans. Based on these findings, it appears that adverse oculareffects may be successfully minimized by administration of Hsp90inhibitors with favorable drug properties that include, although arenot necessarily limited to, lower retina/plasma exposure ratios andfaster retinal elimination. In addition, improved understanding of path-ological mechanisms that result in retinal damage will have importantimplications for appropriate monitoring protocols designed to preventvisual impairment as a result of targeted Hsp90 therapies.

Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.taap.2013.09.018.

Conflict of interest statement

All authors with the exception of Qin Huang are current or formeremployees of Synta Pharmaceuticals Corporation.

Acknowledgments

We thank the following Synta colleagues: SuqinHe for assistancewiththeWestern blots; AmyGuan and Yilin Dong for technical assistancewiththe in vivo tissue distribution; Dinesh Chimmanamada for providingreagents; and David Proia for helpful and insightful discussions on themanuscript.

References

al-Tweigeri, T., Nabholtz, J.M., Mackey, J.R., 1996. Ocular toxicity and cancer chemotherapy.A review. Cancer 78, 1359–1373.

Banerji, U., 2009. Heat shock protein 90 as a drug target: some like it hot. Clin. Cancer Res.15, 9–14.

Banerji, U., O'Donnell, A., Scurr, M., Pacey, S., Stapleton, S., Asad, Y., Simmons, L., Maloney,A., Raynaud, F., Campbell, M., Walton, M., Lakhani, S., Kaye, S., Workman, P., Judson, I.,2005. Phase I pharmacokinetic and pharmacodynamic study of 17-allylamino, 17-demethoxygeldanamycin in patients with advanced malignancies. J. Clin. Oncol. 23,4152–4161.

Barres, B.A., Silverstein, B.E., Corey, D.P., Chun, L.L.Y., 1988. Immunological, morphological,and electrophysiological variations among retinal ganglion cells purified by panning.Neuron 1, 791–803.

Biamonte, M.A., VandeWater, R., Arndt, J.W., Scannevin, R.H., Perret, D., Lee, W.C., 2010.Heat shock protein 90: inhibitors in clinical trials. J. Med. Chem. 53, 3–17.

Cho, D.C., Heath, E.I., Cleary, J.M., Kwak, E.L., Gandhi, L., Lawrence, D.P., Zack, C., Teofilovici,F., Bradley, R., Karol, M.D., Shapiro, G., LoRusso, P., 2011. A phase I dose-escalationstudy of the Hsp90 inhibitor ganetespib (STA-9090) administered twice weekly topatients with solid tumors: updated report. J. Clin. Oncol. 29, A13051.

Dean, D.O., Tytell, M., 2001. Hsp25 and −90 immunoreactivity in the normal rat eye.Invest. Ophthalmol. Vis. Sci. 42, 3031–3040.

Page 9: A rat retinal damage model predicts for potential clinical visual ... · bances, were evaluated in a rodent model in order to understand the observed differences in ocular toxicity

409D. Zhou et al. / Toxicology and Applied Pharmacology 273 (2013) 401–409

Demetri, G.D., Heinrich, M.C., Chimelowski, B., Morgan, J.A., George, S., Bradley, R.,Blackman, R.K., Teofilovici, F., Fletcher, J.A., Tap, W., von Mehren, M., 2011. Anopen-label phase II study of the Hsp90 inhibitor ganetespib (STA-9090) in patients(pts) with metastatic and/or unresectable GIST. J. Clin. Oncol. 29, A10011.

den Hollander, A.I., Roepman, R., Koenekoop, R.K., Cremers, F.P., 2008. Leber congenitalamaurosis: genes, proteins and disease mechanisms. Prog. Retin. Eye Res. 27,391–419.

Dickson, M.A., Okuno, S.H., Keohan, M.L., Maki, R.G., D'Adamo, D.R., Akhurst, T.J.,Antonescu, C.R., Schwartz, G.K., 2013. Phase II study of the HSP90-inhibitor BIIB021in gastrointestinal stromal tumors. Ann. Oncol. 24, 252–257.

Eccles, S.A., Massey, A., Raynaud, F.I., Sharp, S.Y., Box, G., Valenti, M., Patterson, L., deHaven Brandon, A., Gowan, S., Boxall, F., Aherne, W., Rowlands, M., Hayes, A.,Martins, V., Urban, F., Boxall, K., Prodromou, C., Pearl, L., James, K., Matthews, T.P.,Cheung, K.M., Kalusa, A., Jones, K., McDonald, E., Barril, X., Brough, P.A., Cansfield,J.E., Dymock, B., Drysdale, M.J., Finch, H., Howes, R., Hubbard, R.E., Surgenor, A.,Webb, P., Wood, M., Wright, L., Workman, P., 2008. NVP-AUY922: a novel heatshock protein 90 inhibitor active against xenograft tumor growth, angiogenesis,and metastasis. Cancer Res. 68, 2850–2860.

Elfiky, A., Saif, M.W., Beeram, M., O'Brien, S., Lammanna, N., Castro, J.E., Woodworth, J.,Perea, R., Storgard, C., Von Hoff, D.D., 2008. BIIB021, an oral, synthetic non-ansamycin Hsp90 inhibitor: phase I experience. J. Clin. Oncol. 26, 2503.

Goldman, J.W., Raju, R.N., Gordon, G.A., Vukovic, V.M., Bradley, R., Rosen, L.S., 2010.A phase I dose-escalation study of the Hsp90 inhibitor STA-9090 administered onceweekly in patients with solid tumors. J. Clin. Oncol. 28, A2529.

Hidalgo-de-Quintana, J., Evans, R.J., Cheetham, M.E., van der Spuy, J., 2008. The Lebercongenital amaurosis protein AIPL1 functions as part of a chaperone heterocomplex.Invest. Ophthalmol. Vis. Sci. 49, 2878–2887.

Kaarniranta, K., Ryhanen, T., Karjalainen, H.M., Lammi, M.J., Suuronen, T., Huhtala, A.,Kontkanen, M., Terasvirta, M., Uusitalo, H., Salminen, A., 2005. Geldanamycinincreases 4-hydroxynonenal (HNE)-induced cell death in human retinal pigmentepithelial cells. Neurosci. Lett. 382, 185–190.

Kim, Y.S., Alarcon, S.V., Lee, S., Lee, M.J., Giaccone, G., Neckers, L., Trepel, J.B., 2009. Updateon Hsp90 inhibitors in clinical trial. Curr. Top. Med. Chem. 9, 1479–1492.

Kirschman, L.T., Kolandaivelu, S., Frederick, J.M., Dang, L., Goldberg, A.F., Baehr, W.,Ramamurthy, V., 2010. The Leber congenital amaurosis protein, AIPL1, is neededfor the viability and functioning of cone photoreceptor cells. Hum. Mol. Genet. 19,1076–1087.

Kosmaoglou, M., Schwarz, N., Bett, J.S., Cheetham, M.E., 2008. Molecular chaperones andphotoreceptor function. Prog. Retin. Eye Res. 27, 434–449.

Kummar, S., Gutierrez, M.E., Gardner, E.R., Chen, X., Figg, W.D., Zajac-Kaye, M., Chen, M.,Steinberg, S.M., Muir, C.A., Yancey, M.A., Horneffer, Y.R., Juwara, L., Melillo, G., Ivy, S.P.,Merino, M., Neckers, L., Steeg, P.S., Conley, B.A., Giaccone, G., Doroshow, J.H., Murgo,A.J., 2010. Phase I trial of 17-dimethylaminoethylamino-17-demethoxygeldanamycin(17-DMAG), a heat shock protein inhibitor, administered twice weekly in patientswith advanced malignancies. Eur. J. Cancer 46, 340–347.

Lanneau, D., Wettstein, G., Bonniaud, P., Garrido, C., 2010. Heat shock proteins:cell protection through protein triage. ScientificWorldJournal 10, 1543–1552.

LaVail, M.M., Lawson, N.R., 1986. Development of a congenic strain of pigmented andalbino rats for light damage studies. Exp. Eye Res. 43, 867–869.

Leblanc, B., Jezequel, S., Davies, T., Hanton, G., Taradach, C., 1998. Binding of drugs to eyemelanin is not predictive of ocular toxicity. Regul. Toxicol. Pharmacol. 28, 124–132.

Mayer, M.P., Bukau, B., 2005. Hsp70 chaperones: cellular functions and molecularmechanism. Cell. Mol. Life Sci. 62, 670–684.

Pacey, S., Wilson, R.H., Walton, M., Eatock, M.M., Hardcastle, A., Zetterlund, A., Arkenau,H.T., Moreno-Farre, J., Banerji, U., Roels, B., Peachey, H., Aherne, W., de Bono, J.S.,Raynaud, F., Workman, P., Judson, I., 2011. A phase I study of the heat shock protein90 inhibitor alvespimycin (17-DMAG) given intravenously to patients with advancedsolid tumors. Clin. Cancer Res. 17, 1561–1570.

Peponis, V., Kyttaris, V.C., Chalkiadakis, S.E., Bonovas, S., Sitaras, N.M., 2010. Ocular sideeffects of anti-rheumatic medications: what a rheumatologist should know. Lupus19, 675–682.

Rajan, A., Kelly, R.J., Trepel, J.B., Kim, Y.S., Alarcon, S.V., Kummar, S., Gutierrez, M., Crandon,S., Zein,W.M., Jain, L., Mannargudi, B., Figg,W.D., Houk, B.E., Shnaidman, M., Brega, N.,Giaccone, G., 2011. A phase I study of PF-04929113 (SNX-5422), an orally bio-available heat shock protein 90 inhibitor, in patients with refractory solid tumormalignancies and lymphomas. Clin. Cancer Res. 17, 6831–6839.

Ramalingam, S.S., Egorin, M.J., Ramanathan, R.K., Remick, S.C., Sikorski, R.P., Lagattuta, T.F.,Chatta, G.S., Friedland, D.M., Stoller, R.G., Potter, D.M., Ivy, S.P., Belani, C.P., 2008. Aphase I study of 17-allylamino-17-demethoxygeldanamycin combined with paclitaxelin patients with advanced solid malignancies. Clin. Cancer Res. 14, 3456–3461.

Ramanathan, R.K., Trump, D.L., Eiseman, J.L., Belani, C.P., Agarwala, S.S., Zuhowski, E.G.,Lan, J., Potter, D.M., Ivy, S.P., Ramalingam, S., Brufsky, A.M., Wong, M.K., Tutchko, S.,Egorin, M.J., 2005. Phase I pharmacokinetic-pharmacodynamic study of 17-(allylamino)-17-demethoxygeldanamycin (17AAG, NSC 330507), a novel inhibitorof heat shock protein 90, in patients with refractory advanced cancers. Clin. CancerRes. 11, 3385–3391.

Ramanathan, R.K., Egorin, M.J., Eiseman, J.L., Ramalingam, S., Friedland, D.,Agarwala, S.S., Ivy, S.P., Potter, D.M., Chatta, G., Zuhowski, E.G., Stoller, R.G.,Naret, C., Guo, J., Belani, C.P., 2007. Phase I and pharmacodynamic study of 17-(allylamino)-17-demethoxygeldanamycin in adult patients with refractory advancedcancers. Clin. Cancer Res. 13, 1769–1774.

Renouf, D.J., Velazquez-Martin, J.P., Simpson, R., Siu, L.L., Bedard, P.L., 2012. Ocular toxicityof targeted therapies. J. Clin. Oncol. 30, 3277–3286.

Samuel, T.A., Sessa, C., Britten, C., Milligan, K.S., Mita, M.M., Banerji, U., Pluard, T.J., Stiegler,P., Quadt, C., Shapiro, G., 2010. AUY922, a novel HSP90 inhibitor: final results of afirst-in-human study in patients with advanced solid malignancies. J. Clin. Oncol. 28(15 s), 2528.

Sato, S., Fujita, N., Tsuruo, T., 2000. Modulation of Akt kinase activity by binding to Hsp90.Proc. Natl. Acad. Sci. U. S. A. 97, 10832–10837.

Shahinfar, S., Edward, D.P., Tso, M.O., 1991. A pathologic study of photoreceptor cell deathin retinal photic injury. Curr. Eye Res. 10, 47–59.

Shapiro, G., Kwak, E.L., Dezube, B.J., Lawrence, D.P., Cleary, J.M., Lewis, S., Squires, M., Lock,V., Lyons, J.F., Yule, M., 2010. Phase I pharmacokinetic and pharmacodynamic study ofthe heat shock protein 90 inhibitor AT13387 in patients with refractory solid tumors.J. Clin. Oncol. 28 (15 s), 3069.

Sohocki, M.M., Perrault, I., Leroy, B.P., Payne, A.M., Dharmaraj, S., Bhattacharya, S.S.,Kaplan, J., Maumenee, I.H., Koenekoop, R., Meire, F.M., Birch, D.G., Heckenlively, J.R.,Daiger, S.P., 2000. Prevalence of AIPL1 mutations in inherited retinal degenerativedisease. Mol. Genet. Metab. 70, 142–150.

Solit, D.B., Ivy, S.P., Kopil, C., Sikorski, R., Morris, M.J., Slovin, S.F., Kelly, W.K., DeLaCruz, A.,Curley, T., Heller, G., Larson, S., Schwartz, L., Egorin, M.J., Rosen, N., Scher, H.I., 2007.Phase I trial of 17-allylamino-17-demethoxygeldanamycin in patients with advancedcancer. Clin. Cancer Res. 13, 1775–1782.

Taipale, M., Jarosz, D.F., Lindquist, S., 2010. HSP90 at the hub of protein homeostasis:emerging mechanistic insights. Nat. Rev. Mol. Cell Biol. 11, 515–528.

Taldone, T., Gozman, A., Maharaj, R., Chiosis, G., 2008. Targeting Hsp90: small-moleculeinhibitors and their clinical development. Curr. Opin. Pharmacol. 8, 370–374.

Trepel, J., Mollapour, M., Giaccone, G., Neckers, L., 2010. Targeting the dynamic HSP90complex in cancer. Nat. Rev. Cancer 10, 537–549.

Whitesell, L., Lindquist, S.L., 2005. HSP90 and the chaperoning of cancer. Nat. Rev. Cancer5, 761–772.

Whitesell, L., Bagatell, R., Falsey, R., 2003. The stress response: implications for the clinicaldevelopment of hsp90 inhibitors. Curr. Cancer Drug Targets 3, 349–358.

Williams, T.P., Howell, W.L., 1983. Action spectrum of retinal light-damage in albino rats.Invest. Ophthalmol. Vis. Sci. 24, 285–287.

Wong, K., Koczywas, M., Goldman, J.W., Paschold, E.H., Horn, R.L., Blackman, R.K.,Teofilovici, F., Shapiro, G., Socinski, M.A., 2011. An open-label phase II study of theHsp90 inhibitor ganetespib (STA-9090) as monotherapy in patients with advancednon-small cell lung cancer (NSCLC). J. Clin. Oncol. 29, A7500.

Wu, W.C., Wu, M.H., Chang, Y.C., Hsieh, M.C., Wu, H.J., Cheng, K.C., Lai, Y.H., Kao, Y.H.,2010. Geldanamycin and its analog induce cytotoxicity in cultured human retinalpigment epithelial cells. Exp. Eye Res. 91, 211–219.

Yao, J.Q., Liu, Q.H., Chen, X., Yang, Q., Xu, Z.Y., Hu, F., Wang, L., Li, J.M., 2010. Hsp90inhibitor 17-allylamino-17-demethoxygeldanamycin inhibits the proliferation ofARPE-19 cells. J. Biomed. Sci. 17, 30.

Ying, W., Du, Z., Sun, L., Foley, K.P., Proia, D.A., Blackman, R.K., Zhou, D., Inoue, T., Tatsuta,N., Sang, J., Ye, S., Acquaviva, J., Ogawa, L.S., Wada, Y., Barsoum, J., Koya, K., 2012.Ganetespib, a unique triazolone-containing Hsp90 inhibitor, exhibits potent anti-tumor activity and a superior safety profile for cancer therapy. Mol. Cancer Ther.11, 475–484.

Youssef, P.N., Sheibani, N., Albert, D.M., 2011. Retinal light toxicity. Eye (Lond.) 25,1–14.


Recommended