Scholarly article on topic 'A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee'

A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee Academic research paper on "Biological sciences"

CC BY-NC-ND
0
0
Share paper
Academic journal
Osteoarthritis and Cartilage
OECD Field of science
Keywords
{Osteoarthritis / "Wnt signaling" / Chondrocyte / ACLT / DMOAD / "Small molecule"}

Abstract of research paper on Biological sciences, author of scientific article — V. Deshmukh, H. Hu, C. Barroga, C. Bossard, S. KC, et al.

Summary Objectives Osteoarthritis (OA) is a degenerative disease characterized by loss of cartilage and increased subchondral bone within synovial joints. Wnt signaling affects the pathogenesis of OA as this pathway modulates both the differentiation of osteoblasts and chondrocytes, and production of catabolic proteases. A novel small-molecule Wnt pathway inhibitor, SM04690, was evaluated in a series of in vitro and in vivo animal studies to determine its effects on chondrogenesis, cartilage protection and synovial-lined joint pathology. Design A high-throughput screen was performed using a cell-based reporter assay for Wnt pathway activity to develop a small molecule designated SM04690. Its properties were evaluated in bone-marrow-derived human mesenchymal stem cells (hMSCs) to assess chondrocyte differentiation and effects on cartilage catabolism by immunocytochemistry and gene expression, and glycosaminoglycan breakdown. In vivo effects of SM04690 on Wnt signaling, cartilage regeneration and protection were measured using biochemical and histopathological techniques in a rodent acute cruciate ligament tear and partial medial meniscectomy (ACLT + pMMx) OA model. Results SM04690 induced hMSC differentiation into mature, functional chondrocytes and decreased cartilage catabolic marker levels compared to vehicle. A single SM04690 intra-articular (IA) injection was efficacious in a rodent OA model, with increased cartilage thickness, evidence for cartilage regeneration, and protection from cartilage catabolism observed, resulting in significantly improved Osteoarthritis Research Society International (OARSI) histology scores and biomarkers, compared to vehicle. Conclusions SM04690 induced chondrogenesis and appeared to inhibit joint destruction in a rat OA model, and is a candidate for a potential disease modifying therapy for OA.

Academic research paper on topic "A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee"

Accepted Manuscript

A Small Molecule Inhibitor of the Wnt Pathway (SM04690) As a Potential Disease Modifying Agent for the Treatment of Osteoarthritis of the Knee

Vishal Deshmukh, PhD, Haide Hu, PhD, Charlene Barroga, PhD, Carine Bossard, PhD, Sunil KC, PhD, Luis Dellamary, BS, Joshua Stewart, BS, Kevin Chiu, BS, Maureen Ibanez, MS, Melinda Pedraza, BS, Tim Seo, MS, Long Do, PhD, Shawn Cho, MS, Joseph Cahiwat, BS, Betty Tam, PhD, Jeyanesh R.S. Tambiah, MBChB, John Hood, PhD, Nancy E. Lane, MD, Yusuf Yazici, MD

PII: S1063-4584(17)31167-6

DOI: 10.1016/j.joca.2017.08.015

Reference: YJOCA 4078

To appear in: Osteoarthritis and Cartilage

Received Date: Revised Date: Accepted Date:

2 January 2017 18 July 2017 30 August 2017

Osteoarthritis

Please cite this article as: Deshmukh V, Hu H, Barroga C, Bossard C, KC S, Dellamary L, Stewart J, Chiu K, Ibanez M, Pedraza M, Seo T, Do L, Cho S, Cahiwat J, Tam B, Tambiah JRS, Hood J, Lane NE, Yazici Y, A Small Molecule Inhibitor of the Wnt Pathway (SM04690) As a Potential Disease Modifying Agent for the Treatment of Osteoarthritis of the Knee, Osteoarthritis and Cartilage (2017), doi: 10.1016/ j.joca.2017.08.015.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1 Title Page

2 A Small Molecule Inhibitor of the Wnt Pathway (SM04690) As a Potential Disease

3 Modifying Agent for the Treatment of Osteoarthritis of the Knee

5 Running title: Wnt pathway inhibitor as potential DMOAD for OA

7 Authors: Vishal Deshmukh1, Haide Hu1, Charlene Barroga1, Carine Bossard1, Sunil

8 KC1, Luis Dellamary1, Joshua Stewart1, Kevin Chiu1, Maureen Ibanez1, Melinda

9 Pedraza1, Tim Seo1, Long Do1, Shawn Cho1, Joseph Cahiwat1, Betty Tam1, Jeyanesh

10 R.S. Tambiah1, John Hood1, Nancy E. Lane2 , Yusuf Yazici1 +

11 +corresponding author

13 Affiliations: 1 Samumed, LLC, San Diego, CA, USA

14 2 University of California, Davis, CA, USA

16 Author Emails: Vishal Deshmukh, PhD (vishal@samumed.com), Haide Hu, PhD

17 (huyong21@gmail.com), Charlene Barroga, PhD (charlene@samumed.com), Sunil KC,

18 PhD (sunil@samumed.com), Luis Dellamary, BS (luis@samumed.com), Joshua

19 Stewart, BS (josh@samumed.com), Kevin Chiu, BS (kevin@samumed.com), Carine

20 Bossard, PhD (carine@samumed.com), Maureen Ibanez, MS

21 (maureen@samumed.com), Melinda Pedraza, BS (melinda@samumed.com), Tim Seo,

22 MS (tim@samumed.com), Long Do, PhD (long@samumed.com), Shawn Cho, MS

23 (shawn@samumed.com), Joseph Cahiwat, BS (joec@samumed.com), Betty Tam, PhD

24 (betty@samumed.com), Jeymi Tambiah, MBChB (jeymi@samumed.com), John Hood,

25 PhD (john@impactbiosciences.com), Nancy E. Lane, MD (nelane@ucdavis.edu), Yusuf

26 Yazici, MD (yusuf@samumed.com)

28 + Main corresponding author: Yusuf Yazici, MD, Samumed LLC. 9381 Judicial Drive,

29 Suite 160, San Diego, CA 92121; Tel: 858-926-2962; yusuf@samumed.com

46 Abstract

48 Objectives

50 Osteoarthritis (OA) is a degenerative disease characterized by loss of cartilage and

51 increased subchondral bone within synovial joints. Wnt signaling affects the

52 pathogenesis of OA as this pathway modulates both the differentiation of osteoblasts

53 and chondrocytes, and production of catabolic proteases. A novel small molecule Wnt

54 pathway inhibitor, SM04690, was evaluated in a series of in vitro and in vivo animal

55 studies to determine its effects on chondrogenesis, cartilage protection and synovial-

56 lined joint pathology.

58 Design

60 A high-throughput screen was performed using a cell-based reporter assay for Wnt

61 pathway activity to develop a small molecule designated SM04690. Its properties were

62 evaluated in bone marrow derived human mesenchymal stem cells (hMSCs) to assess

63 chondrocyte differentiation and effects on cartilage catabolism by immunocytochemistry

64 and gene expression, and glycosaminoglycan breakdown. In vivo effects of SM04690

65 on Wnt signaling, cartilage regeneration and protection were measured using

66 biochemical and histopathological techniques in a rodent acute cruciate ligament tear

67 and partial medial meniscectomy (ACLT+pMMx) OA model.

69 Results

71 SM04690 induced hMSC differentiation into mature, functional chondrocytes and

72 decreased cartilage catabolic marker levels compared to vehicle. A single SM04690

73 intra-articular (IA) injection was efficacious in a rodent OA model, with increased

74 cartilage thickness, evidence for cartilage regeneration, and protection from cartilage

75 catabolism observed, resulting in significantly improved Osteoarthritis Research Society

76 International (OARSI) histology scores and biomarkers, compared to vehicle.

78 Conclusions

80 SM04690 induced chondrogenesis and appeared to inhibit joint destruction in a rat OA

81 model, and is a candidate for a potential disease modifying therapy for OA.

83 Keywords: Osteoarthritis, Wnt signaling, chondrocyte, ACLT, DMOAD, small molecule

85 INTRODUCTION

87 Osteoarthritis affects an estimated 27 million adults in the U.S. and is a leading cause of

88 disability worldwide1. The disease is characterized by breakdown of articular cartilage

89 and growth of subchondral bone causing pain, decreased mobility, limitation of function

90 and failure of synovial joints2. The only current therapeutic options for OA are

91 symptomatic pain management and surgical intervention3, 4 There is therefore an

92 unmet need for disease modifying OA drugs (DMOADs). Mesenchymal stem cells

93 (MSCs) in synovium and subchondral bone are capable of differentiation into cartilage

94 forming chondrocytes, bone forming osteoblasts, or adipocytes5. Compared to healthy

95 synovium, synovium of subjects with meniscal injury and early OA is enriched in stem

96 cells6-8, suggesting the failure to regenerate articular cartilage may not be due to

97 insufficient stem cell supply, but rather their inappropriate differentiation when

98 attempting to restore healthy cartilage homeostasis.

100 Wnt proteins interact with stem and differentiated cells to orchestrate organogenesis,

101 cell differentiation, morphogenesis and tissue remodeling. In adults, the role of the Wnt

102 pathway in tissue formation is extended to homeostatic control through the tightly

103 regulated differentiation of resident stem cells to replenish and repair adult tissues9, 10

104 Activation of Wnt signaling is initiated by Wnt proteins binding to their cognate

105 receptors, and is modulated by endogenous antagonists such as Dickkopf, Wnt-

106 inhibitory signaling protein, secreted Frizzled related protein and Cereberus. In the

107 canonical Wnt/p-catenin-signaling pathway, binding of Wnt proteins to cell surface

108 receptors leads to stabilized p-catenin levels by inhibition of p-catenin phosphorylation

109 and proteasome-mediated degradation. Stabilized p-catenin translocates to the nucleus

110 and interacts with the T-cell factor/lymphoid enhancer factor (TCF/LEF) family of

111 transcription factors to activate Wnt target genes9, 10 Wnt signaling is subject to

112 complex modulation at multiple levels, resulting in signaling acting more as rheostats,

113 than binary switches in most tissues. Modulating this pathway is an attractive

114 therapeutic approach for regenerative medicine, however success has been limited due

115 to lack of potent, safe therapeutic agents11.

117 Recently, increased understanding of cartilage growth mechanisms during OA

118 development and aging have identified new molecular mechanisms and targets.

119 Amongst these, Wnt signaling has been shown to play a critical role in OA

120 pathogenesis, particularly cartilage and subchondral bone remodeling12. A single

121 nucleotide polymorphism analysis demonstrated an association of both proximal femur

122 bone shape and risk of hip OA with an Arg324Gly substitution mutation in FrzB, a Wnt

123 antagonist, implicating the Wnt pathway as a potential regulator of the disease13, 14 At a

124 molecular level, the balance of Wnt signaling maintains osteoblast and chondrocyte

125 lineage fates and their homeostasis. Increased mechanotransductive force increases

126 Wnt signaling in the joint, which results in osteoblast formation and release of proteases

127 that remodel articular cartilage into an osteoconductive matrix, while decreased Wnt

128 signaling stimulates chondrogenesis12. Wnt signaling has also been implicated in the

129 induction of protease production, especially matrix metalloproteinases (MMP1, MMP-3

130 and MMP-13) by chondrocytes and synovial tissue in response to mechanical stress

131 and pro-inflammatory cytokines15. In the context of a joint with abnormal loading, this

132 has the potential to lead to a progressive disease such as OA, with Wnt playing a

133 central role in its pathogenesis.

135 In this study, SM04690, a small molecule Wnt pathway inhibitor being developed as a

136 potential DMOAD, was evaluated for its effects on chondrocyte differentiation, cartilage

137 regeneration and protection, and prevention of joint destruction in a preclinical model of

138 knee OA.

140 METHODS

142 Cell culture and differentiation

143 SW480 cells (ATCC) cultured in Dulbecco's modified Eagle's medium (DMEM,

144 ThermoFisher, Carlsbad, CA) with 10% fetal bovine serum (FBS) were transduced with

145 the TCF/LEF-luciferase lentivirus (Qiagen, Germantown, MD) and stable clones were

146 selected using puromycin. Primary bone marrow derived hMSCs (Lonza Inc., Basel,

147 Switzerland) were grown in MSCGM™ mesenchymal stem cell growth medium (Lonza),

148 and used between passages 2 and 6.

150 High-throughput screening

151 A chemical library was screened in a cellular TCF/LEF reporter-based assay using

152 SW480 cells. Compounds were transferred into screening plates using ECHO 550

153 (Labcyte, San Jose, CA). SW480 cells were plated (3000/well) in DMEM with 1% FBS.

154 After 48 hrs, BrightGlo (Promega, Madison, WI) luminescence was measured using

155 Envision (Perkin Elmer, Waltham, MA). For chondrogenic differentiation, hMSCs were

156 plated (20,000/well) in incomplete chondrocyte differentiation medium (iCDM; Lonza)

157 and treated with SM04690 or compounds- FH535, IWR-1, ICG001, iCRT14, KY02111,

158 and, CX-4945 (Sigma-Aldrich) or Transforming Growth Factor p3 (TGFp3; 20ng/ml,

159 Peprotech Inc., Rocky Hill, NJ). On Day 5, cells were fixed, stained with 1 pg/mL

160 Rhodamine B and imaged using Cellomics CellInsight (ThermoFisher).

162 Chondrocyte Differentiation

163 For chondrocyte differentiation, hMSCs were plated in either 48-well plates

164 (40,000/well) or dispensed into 15ml conical tubes (150,000/tube) in iCDM and treated

165 with SM04690 in DMSO or TGFp3 (20ng/ml). Cells were incubated for 21 days, with

166 media changes every 5 days, fixed and immunostained with specific antibodies or with

167 Alcian Blue (1% in acetic acid, pH 2.5) or Safranin O (0.1% aqueous solution). Cells

168 were imaged using EVOS FL Microscope (ThermoFisher). Gene expression was

169 measured by quantitative real time PCR (qRT-PCR) using SYBR Green based, gene

170 specific primers.

171 Primary calvarial cells were isolated from mouse E13.5 embryos using collagenase

172 (Sigma-Aldrich, St. Louis, MI) and plated in poly-L-lysine coated plates. After 5 days

173 cells were treated with DMSO or SM04690. On Day 21, cells were fixed and stained

174 with Alcian Blue.

176 Cartilage Matrix Degradation- Glycosaminoglycan (GAG) and Nitric Oxide (NO)

177 measurement

178 hMSCs were differentiated into chondrocytes using TGFp3 for 21 days followed by

179 treatment with either TNFa (20ng/ml) + Oncostatin M (10ng/ml) or IL1 p (10ng/ml) and

180 SM04690 for 72hrs. Chondrocytes were digested with papain (Sigma). GAG content

181 was measured using the dimethylmethylene blue (DMMB) kit (Chondrex, Redmond,

182 WA) and absorbance at 535nm was measured using Cytation 3 (Biotek, Winooski, VT).

183 NO was measured using Greiss reagent kit (Promega).

185 Pharmacokinetics

186 Following single IA SM04690 injection into Sprague-Dawley (SD) rats (12 weeks old,

187 male), knee joints were collected, flash frozen and stored at -70°C. Bone and cartilage

188 samples were isolated from tibias, homogenized and SM04690 extracted with acidified

189 organic solvent. Extracts were analyzed using Phenomenex PFP column and HPLC

190 gradient method in tandem to a triple quadrupole mass spectrometer (API Triple Quad

191 3000) with a Turboionspray source for detection in positive ion mode.

193 Surgery-induced OA model

194 All animal studies were performed in accordance with approved Samumed, LLC Animal

195 Committee protocols. Male SD rats were housed at Samumed LLC, and provided food

196 and water ad libitum. At 10 weeks postnatal age, 24 rats were subjected to severing of

197 the anterior cruciate, medial collateral and medial meniscotibial ligaments

198 (ACLT+pMMx). One-week post-surgery, all rats were randomized and given IA

199 SM04690 (0.3pg in 50pl) or vehicle (n=12 rats/group). Thirteen weeks post-surgery,

200 knee joints were isolated, fixed in 10% formalin, decalcified and embedded in paraffin

201 blocks. Frontal sections (5 pm thick, 3 sections to ensure redundancy) from different

202 levels, each 100 pm apart, were obtained and stained with Safranin O/Fast Green. At

203 least 12 sections were obtained from each rat and imaged using a light microscope

204 (EVOS FL, Life Technologies).

206 OARSI Scoring, Cartilage Protection and Regeneration

207 Histological evaluation was performed by 2 blinded observers. Images were scored

208 based on the OARSI cartilage histopathology scoring system by evaluating damage

209 based on grade (depth of progression into cartilage) and stage (extent of joint

210 involvement)16. Briefly, femurs and tibias were evaluated separately, and each assigned

211 a score for cartilage damage grade on a scale of 0 to 6 (0 - intact surface; 6 - extensive

212 deformation) and cartilage damage stage on a scale from 0 to 4 (0 - normal joint; 4 -

213 >50% involvement). Total score was the product of Grade x Stage (0 representing

214 normal joint; 24 representing severe OA)16. Twelve sections were scored per rat and no

215 rats were excluded. Following scoring, the study was unblinded and the 4 sections from

216 each rat with lowest scores (representing least cartilage damage) were excluded from

217 further analysis. Mean OARSI score for each rat was generated by averaging the

218 scores from the 2 blinded observers. Additionally, histological evaluation was repeated

219 by 4 independent blinded observers based on a modified objective quantitative

220 histomorphometrical OARSI scoring system17. Safranin O staining intensity and

221 cartilage thickness were measured using ImageJ18.

222 Quantification of articular chondrocytes was performed using immunohistochemistry for

223 doublecortin (Dcx), followed by imaging (Keyence, confocal mode), and analysis using

224 Cellomics (ThermoFisher). Levels of circulating biomarkers were measured by ELISA

225 on weeks 3, 4, 6.

226 In a second study, 24 rats (male, 10-weeks old) were subjected to the ACLT+pMMx

227 procedure. One-week post-surgery all rats were randomized and given an IA injection of

228 SM04690 (0.3pg) or vehicle (n=12 rats/group). On week 5 after surgery, cartilage was

229 isolated from the rats for gene expression measurement of chondrocyte markers and

230 proteases.

232 Statistical analysis

233 Statistical analysis was performed using Prism 7 (Graphpad Inc, La Jolla, CA). EC5o

234 values were obtained using sigmoidal dose-response curve-fitting. For those outcomes

235 in which an underlying normal distribution was assumed, an independent, two-tailed t-

236 test was utilized for two group comparisons, and for more than two groups one-way

237 ANOVA was utilized. OARSI scores, for which the normal distribution was not assumed,

238 were evaluated using Mann-Whitney U test. Data were represented as Mean ± 95%

239 confidence interval (CI) or Mean ± standard deviation (SD) as noted in the figure

240 legends, and significance values listed. Significance was set at p<0.05. *p<0.05,

241 **p<0.01, ***p<0.001

243 RESULTS

245 Wnt Pathway Modulation- Cellular High-Throughput Screening Identified SM04690 as a

246 Potent and Selective Inhibitor of Wnt Signaling.

248 Small-molecule inhibitors of Wnt signaling were identified using a high-throughput

249 TCF/LEF-reporter assay in SW480 colon cancer cells bearing a mutation in the APC

250 protein19, which leads to constitutively active canonical Wnt signaling. Hits were

251 counter-screened against SW480 cells expressing a SV40-driven luciferase reporter, to

252 eliminate compounds that acted non-specifically. Based on the elucidated structure

253 activity relationship from these compounds, iterative medicinal chemistry efforts

254 generated SM04690 (EC50= 19.5 nM) which did not affect the SV40 luciferase reporter

255 (Figure 1a). The activity of SM04690 was then compared with known Wnt pathway

256 inhibitors such as FH53520, IWR-121, ICG00121, iCRT1422, KY0211123, and, CX-494524.

257 SM04690 was found to be ~150-500-fold more potent than the other compounds

258 (Supplementary Figure 1a, b). Inhibition of Wnt signaling was confirmed with SM04690

259 treatment in SW480 cells by qRT-PCR, and Western blot for p-catenin and its target

260 proteins (Figure 1b, c). Bone-marrow-derived hMSCs (CD29+, CD44+ CD166+ CD105+

261 CD45-), treated with SM04690, showed a dose-dependent decrease in the expression

262 of Wnt pathway genes (ASCL1, LEF1, TCF7L2, TCF7, C-MYC and AXIN2), as

263 measured by qRT-PCR (Figure 1d) and Western Blot (Figure 1e). SM04690 also

264 inhibited the expression of AXIN2, TCF7 and LEF1 in hMSCs (Supplementary Figure

265 1c) and AXIN2 and LGR5 in IEC6 (intestinal stem cells sensitive to Wnt activation;

266 Supplementary Figure 1d) when the Wnt pathway was selectively activated using either

267 Wnt3a or a GSK3p inhibitor, CHIR-99021. The inhibition of Wnt target genes by

268 SM04690 (30nM) was comparable to the activity of CX-494524 (10|jM) and better than

269 KY02111 (10|M)23. SM04690 had minimal effects on the non-canonical Wnt pathway

270 and the BMP pathway (Supplementary Figures 2, 3). Changes in the expression of

271 genes in these pathways (e.g., decreased expression of DAAM1, LEF1, RHOA, WNT4,

272 BMP4 and increased expression of DKK1, NLK1, WNT16, MAPK13, SMAD7) correlated

7 25 28

273 with Wnt pathway inhibition and the induction of chondrogenic differentiation7

275 Cartilage Regeneration- Wnt Pathway Inhibitor SM04690 Induced Chondrocyte

276 Differentiation in vitro.

278 Chondrogenesis occurs as a result of MSC and progenitor cell differentiation. To assess

279 SM04690 effects on early chondrogenesis, hMSCs were treated for 5 days, and

280 Rhodamine B stained chondrogenic nodules29 were measured as an indicator of early

281 cell condensation phenotype associated with chondrogenesis induction (Figure 2a, b).

282 SM04690 promoted aggregation of hMSCs in a dose-dependent manner (EC5o = 10nM,

283 Supplementary Figure 4a), increasing Rhodamine B stained colonies >40-fold

284 (p<0.0001), compared to DMSO treated control. TGFp330, was used as a positive

285 control. hMSCs treated with SM04690 for 3 days demonstrated significantly increased

286 (p=0.0006) induction of Sox931 by immunocytochemistry; Figure 2c, d) and Western blot

287 (Supplementary Figure 4b), compared to DMSO, indicating an early chondrocyte

288 lineage phenotype.

290 hMSCs treated with SM04690 in high density three dimensional, 21-day pellet culture

291 conditions efficiently differentiated into mature chondrocytes, as evidenced by larger

292 pellets with increased Safranin O staining relative to DMSO treated control (Figure 2e).

293 Additionally, hMSCs treated with SM04690 for 21 days under monolayer conditions

294 demonstrated chondrocyte differentiation, with the presence of mature chondrocyte

295 specific proteins and cartilage matrix components (TIMP1, Type II collagen, aggrecan,

296 Alcian Blue, Safranin O, Rhodamine B, Toluidine Blue and CD44)30-32 as compared to

297 DMSO treated cells (Figure 2f). Treatment with SM04690 also induced significant

298 increases in the sulfated GAG content of differentiated chondrocytes32 (Figure 2g)

299 compared to DMSO control (p=0.023).

301 Following SM04690 treatment, qRT-PCR analysis of differentiated cells further

302 confirmed increased expression of chondrocyte associated genes, including SOX9,

303 cartilage oligomeric matrix protein (COMP), aggrecan, COL2A1, TGFp1, tissue inhibitor

304 of metalloproteinase 1 (TIMP1), CD44, and Type 10 collagen (COL10A1) (Figure 3a),

305 as compared to DMSO treatment, with expression levels greater than those induced by

306 CX-4945 (10pM; p<0.05 for all genes except COL2A1) and KY02111 (10pM; p<0.05)

307 (Supplementary Figure 5a). SM04690 treatment decreased expression of genes

308 associated with osteoblast differentiation (BGLAP [osteocalcin], ALPL [Alkaline

309 phosphatase], BMP4, RUNX2)29, 33 and tendon or ligament differentiation (COL1A1)

310 (p<0.01, Figure 3b) with levels comparable to CX-4945 (10|M) and KY02111 (10|M)

311 treatment (Supplementary Figure 5b). While increased C0L10A1 expression was

312 observed in vitro, a corresponding increase was not observed in the in vivo model

313 treated with SM04690 (Supplementary Figure 9a), suggesting increased C0L10A1

314 expression may be a cell culture system result and not indicative of hypertrophic

315 differentiation34 induced by SM04690 treatment.

317 Further, the ability of SM04690 to induce differentiation of mouse progenitor cells into

318 chondrocytes was evaluated. SM04690 treatment for 21 days induced chondrocyte

319 differentiation in ATDC5 cells (mouse chondrogenic cell line35), as measured by

320 Safranin O and Alcian Blue staining (Figure 3c). Additionally, primary calvarial cells

321 isolated from E13.5 mouse embryos36 and cultured in the presence of SM04690

322 (100nM) for 21 days showed a marked increase in the number and size of Alcian Blue

323 stained chondrocytes, increasing the area of field covered with chondrocytes to >30%

324 compared to <3% in the DMSO treated control (Figure 3d, e).

326 Since SM04690 inhibited Wnt signaling, several commercially available Wnt inhibitors

327 were tested in the early chondrogenic aggregation and chondrocyte differentiation

328 assays. In line with their activities in the TCF/LEF reporter assay (Supplementary Figure

329 1a, b), several Wnt pathway inhibitors were ~50-500 fold less potent than SM04690

330 (Supplementary Figure 6a, b). SM04690 also induced ~3-10-fold more Alcian Blue and

331 Safranin O stained chondrocyte colonies, at ~200-fold lower EC50 as compared to CX-

332 4945, KY02111 and ICG-001 (Supplementary Figure 6c-h).

334 Cartilage Protection- Wnt Pathway Inhibitor SM04690 Protected Chondrocytes from

335 Catabolic Breakdown in vitro.

337 In addition to regeneration of cartilage, long-term disease modification in OA requires

338 inhibited degradation of newly formed and existing cartilage. Therefore, the effects of

339 SM04690 on chondrocytes and hMSCs and cartilage catabolism under

340 pathophysiological OA-like conditions were evaluated. Chondrocytes and hMSCs were

341 treated either with a combination of TNF-a (20ng/ml) and oncostatin M (10ng/ml), or

342 IL1p (10ng/ml) to mimic cytokine-induced cartilage degeneration during OA progression.

343 When treated with these cytokines, cultured chondrocytes and hMSCs upregulated the

344 expression of MMP-1, MMP-3, MMP-13 and IHH37 as measured by qRT-PCR (Figure

345 4a, b, Supplementary Figure 7a-d), increased extracellular secreted GAG29, as

346 measured by DMMB reaction (Figure 4c), and released increased amounts of NO29

347 (Figure 4d), as measured by the Greiss reaction, compared to untreated chondrocytes.

348 Treatment with SM04690 inhibited cytokine-induced expression of matrix degrading

349 enzymes (MMP-1, MMP-3, MMP-13, IHH) (Figure 4a, b, Supplementary Figure 7a-d),

350 with potency similar or improved (p<0.05) compared to CX-4945 (10 pM) and KY02111

351 (10 pM). Consistent with this effect, SM04690 treatment also significantly decreased

352 breakdown and release of GAGs (Figure 4c) and NO (Figure 4d) compared to controls

353 (p<0.05).

355 Pharmacokinetics- IA SM04690 Demonstrated Sustained Local and Minimal Systemic

356 Exposure

358 Pharmacokinetic evaluation of a single IA SM04690 injection in rats demonstrated knee

359 joint residence time >180 days (Supplementary Figure 8a). Systemic exposure of drug

360 was below quantifiable plasma levels (lower limit of quantification= 10nM) and no

361 obvious adverse effects (weight loss, significant swelling, signs of pain or distress) were

362 observed in the treated rats.

364 Cartilage Regeneration- IA SM04690 Promoted Cartilage Growth and Improved Joint

365 Health in a Rat Model of Knee Osteoarthritis.

367 The ACLT+pMMx rat OA model causes severe joint destabilization, leading to OA-like

368 disease with cartilage degradation within 1-2 weeks16, 38, 39 The efficacy of SM04690

369 was evaluated in the ACLT+pMMx rat knee OA model, following a single IA injection of

370 SM04690 (0.3pg) or vehicle control. Histological examination of the joints 12 weeks post

371 vehicle injection (13 weeks post-surgery) showed loss of smooth cartilage articular

372 surface and decreased intercondylar space between the femur and tibia in these rats

373 (Figure 5a). In contrast, SM04690 treated animals showed increased articular cartilage

374 and smooth cartilage surface as indicated by significantly increased Safranin O-Fast

375 Green staining intensity (p<0.0001; Supplementary Figure 8c) and cartilage thickness

376 measurements (p=0.0042; Supplementary Figure 8d). Blinded OARSI histopathology

377 analysis16 of the most severely affected joint areas, revealed a significant decrease

378 (p=0.008) in treatment group scores (Figure 5b) compared to vehicle controls.

379 Additionally, blinded scoring performed using a modified objective quantitative

380 histomorphometrical OARSI scoring method17 also showed a significant decrease

381 (p=0.0005) in the OARSI score with SM04690 treatment (Supplementary Figure 8b),

382 indicating improvement in joint morphology.

384 Serum was evaluated during the 12-week post- treatment period for N-terminal

385 propeptide of collagen IIA (PIIANP), a biomarker of cartilage matrix synthesis40. Levels

386 of PIIANP in serum were significantly increased (p=0.0186) at 3 weeks post-treatment in

387 rats treated with SM04690 as compared to vehicle control and remained slightly

388 elevated even at 5 weeks post-treatment (p>0.05; Figure 5c).

390 Additionally, the onset of chondrogenic differentiation in the SM04690 treated animals

391 was confirmed by increased gene expression of Col2a1, aggrecan and COMP in the

392 cartilage as compared to vehicle treatment (Figure 5d). However, no significant changes

393 were observed in the expression levels of Col10a, a chondrocyte hypertrophy marker,

394 (Supplementary Figure 9a) after 4 weeks from treatment with SM04690, as compared to

395 vehicle. Further, quantification of chondrocytes in the superficial zone of the articular

396 cartilage at 12- weeks post injection showed a significant increase (p=0.001) in the

397 number of Doublecortin (Dcx) positive chondrocytes41 in SM04690 treated rats

398 compared to vehicle (Figure 5e, f).

400 Cartilage Protection- Single IA Injection of SM04690 Decreased Cartilage Breakdown in

401 a Rat Model of Knee Osteoarthritis.

403 Local regulation of matrix degradation markers has previously been demonstrated by a

404 significant increase in mRNA levels of ADAMTS4 and MMP-13 in animal models of

405 OA38, 42 MMP-1, MMP-3, MMP-13 and ADAMTS5 were highly expressed in the articular

406 cartilage of ACL-transected, vehicle-treated animals. In SM04690 treated rats,

407 significant (p<0.05) reductions of protease levels (approximately 2-3-fold) were

408 observed at 4 weeks post injection as compared to vehicle-treated animals (Figure 5g).

409 Further, circulating COMP levels, which correlate with OA disease severity43, were

410 slightly reduced in SM04690-treated rats as compared to control rats at 2 weeks post-

411 treatment, and significantly reduced (p=0.0064) at 5 weeks post-treatment (Figure 5h).

413 Wnt Pathway Modulation- SM04690 Inhibited the Wnt Pathway in a Rat Model of Knee

414 Osteoarthritis.

416 The expression of Wnt pathway genes in the cartilage from ACLT+pMMx rats was

417 evaluated using a qRT-PCR panel comprising 84 Wnt pathway genes. Decreased

418 expression (p<0.05) of several Wnt pathway genes (e.g. GSK3ß, Dvl1, Wnt3a, TCF7,

419 Axin 2, p-catenin), and upregulation in Wnt inhibitory genes (e.g. DKK1, WIF1) was

420 observed in the cartilage of animals treated with SM04690 as compared to vehicle

421 controls (Supplementary Figure 9b-d). Finally, decreased expression and nuclear

422 localization of p-catenin was observed in articular chondrocytes in SM04690- treated

423 rats as compared to vehicle-treated rats (Supplementary Figure 10a-c).

425 DISCUSSION

427 Wnt signaling has been shown to be a pivotal pathway in OA that modulates bone and

428 chondrocyte lineage specification, protease production and joint homeostasis. SM04690

429 is being developed as a novel small molecule inhibitor of the Wnt pathway. In this study,

430 we used several in vitro assays for Wnt signaling, chondrogenesis and cartilage

431 catabolism to test the effects of SM04690 on these processes. In vitro, SM04690 was

432 ~50-500-fold more potent than published Wnt inhibitors across multiple cellular assays.

433 Functional inhibition of Wnt signaling in cartilage in the rat ACLT+pMMx model

434 demonstrated by both gene expression and inhibited p-catenin nuclear localization,

435 provided evidence for the ability of SM04690 to modulate the Wnt pathway in vivo, in

436 the context of OA.

438 The appearance of Rhodamine B staining with SM04690- treated hMSCs and mouse

439 stem cells in vitro indicated significant and dose-dependent induction of early

440 chondrocyte condensation, followed by differentiation into mature chondrocytes that

441 expressed both Type II collagen and Aggrecan that are required for the specialized

442 extracellular matrix rich in highly sulfated GAGs necessary to provide elastic

443 biomechanical properties for motion and weight bearing30. In the ACLT+pMMx model,

444 SM04690 treatment led to increased cartilage as demonstrated by Safranin O staining,

445 GAG measurement and serum biomarker PIIANP. An increased number of Dcx positive

446 articular chondrocytes as well as increased expression of chondrogenic genes in the

447 cartilage, as compared to vehicle controls, were consistent with induction of

448 chondrocyte differentiation in SM04690- treated rats. These data demonstrated the

449 ability of SM04690 to potentially regenerate functional chondrocytes and hence restore

450 cartilage in vivo.

452 Enzymatic tissue degradation plays a significant role in the progression of OA and

453 expression of proteases is increased in articular cartilage in response to joint injury38, 39

454 SM04690 inhibited the expression of catabolic enzymes in cytokine stimulated

455 chondrocytes and hMSCs in vitro, as well as in the ACLT+pMMx animals, compared to

456 vehicle controls, thus demonstrating specific control and regulation of catabolic

457 enzymes in the context of OA. SM04690 also decreased cytokine induced GAG

458 breakdown and NO production in vitro, and levels of circulating COMP in vivo, together

459 providing strong evidence that SM04690 prevented matrix breakdown by proteases

460 under the pathological conditions of OA. While additional studies are needed to

461 evaluate SM04690 effects on protease production and matrix catabolism in primary OA

462 patient-derived chondrocytes, these data suggested that SM04690 may have potential

463 cartilage protective effects.

465 Changes in gene expression, biomarkers and restored cartilage measured in the rat

466 ACLT+pMMx model experiments by histological OARSI scores indicated improvements

467 in joint morphology and attenuated OA phenotype with SM04690 treatment, despite the

468 ongoing absence of joint ligaments responsible for inducing the model. The observed

469 efficacy may be a result of a combination of mechanisms, including MSC differentiation

470 into chondrocytes, and cartilage protective effects of SM04690, however, the relative

471 contribution of each is unknown. In vivo pharmacokinetic data supported the use of a

472 single SM04690 IA injection for this study and the demonstrated efficacy of the

473 compound in human cells in vitro supported the rationale to pursue clinical development

474 of SM04690. In a phase 1 first-in-human clinical trial (N=61), SM04690 appeared safe

475 and well-tolerated (in press).

477 In the search for an effective treatment for OA, development of DMOADs has focused

478 on either slowing down the progression of the disease by targeting either matrix-

479 degrading enzymes (using MMP/ADAMTS inhibitors)42 or inflammatory mediators such

480 as IL1 p and TNFa44. Expression of factors such as FGF-18 and TGF-p to promote

481 chondrocyte proliferation and maturation, or stem cell based transplants have been

482 tested, but challenges remain with translation of these techniques into the clinic45, 46

483 Targeting the resident stem cell population in the articular region with an effective

484 therapy remains a promising approach29, 46.The potential to stimulate cartilage repair via

485 chondrocyte differentiation and reduce cartilage destroying proteases, could lead to a

486 beneficial treatment effect with SM04690. These data support the further development

487 of SM04690 as an injectable small molecule potential DMOAD for the treatment of knee

488 OA.

491 Author contributions:

493 Conception and design (J.H., C.Ba., S.K, H.H., V.D., L.D., J.S., N.L and Y.Y.)

494 Acquisition, analysis and interpretation of data (V.D., H.H., C.Ba., S.K., L.D., J.S., K.C.,

495 C.Bo, M.I., M.P., T.S., J.C., L.D., S.C., B.T., J.T,J.H., N.L. and Y.Y.)

496 Drafting the article (V.D., J.H. N.L and Y.Y.)

497 Final approval of the article (V.D., H.H., C.Ba., S.K., L.D., J.S., K.C., C.Bo, M.I., M.P.,

498 T.S., J.C., L.D., S.C., B.T., J.T., J.H., N.L. and Y.Y.)

500 Financial support:

502 Financial support for this study and its publication was provided by Samumed, LLC.

504 Conflict of Interest:

506 Samumed LLC salary and equity- V.D., C.Ba., S.K., L.D., J.S., K.C., C.Bo., M.I., M.P.,

507 T.S., J.C., L.D., S.C., B.T., J.T., and Y.Y.

508 Samumed LLC equity- H.H. and J.H.

509 Samumed consultant- N.L.

511 Acknowledgements:

513 We thank Sarah Kennedy, PhD, David Herman, PhD, Timothy Phalen, PhD, Benoit

514 Melchior, PhD and Hutch Humphreys for critical revision of the manuscript and

515 Christopher Swearingen, PhD for assistance with statistical analysis.

533 REFERENCES

535 1. Lawrence RC, Felson DT, Helmick CG, Arnold LM, Choi H, Deyo RA, et al.

536 Estimates of the prevalence of arthritis and other rheumatic conditions in the

537 United States. Part II. Arthritis Rheum 2008;58:26-35. doi:10.1002/art.23176.

538 2. Howell DS. Pathogenesis of osteoarthritis. Am J Med 1986;80:24-8.

539 3. McAlindon TE, Bannuru RR, Sullivan MC, Arden NK, Berenbaum F, Bierma-

540 Zeinstra SM, et al. OARSI guidelines for the non-surgical management of knee

541 osteoarthritis. Osteoarthritis Cartilage 2014;22:363-88.

542 doi:10.1016/j.joca.2014.01.003.

543 4. Katz JN, Earp BE, Gomoll AH. Surgical management of Osteoarthritis. Arthritis

544 Care Res 2010;62:1220-8. doi:10.1002/acr.20231.Surgical.

545 5. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al.

546 Multilineage potential of adult human mesenchymal stem cells. Science (80- )

547 1999;284:143-7. doi:10.1126/science.284.5411.143.

548 6. Jones EA, Crawford A, English A, Henshaw K, Mundy J, Corscadden D, et al.

549 Synovial fluid mesenchymal stem cells in health and early osteoarthritis: Detection

550 and functional evaluation at the single-cell level. Arthritis Rheum 2008;58:1731-

551 40. doi:10.1002/art.23485.

552 7. Sekiya I, Ojima M, Suzuki S, Yamaga M, Horie M, Koga H, et al. Human

553 mesenchymal stem cells in synovial fluid increase in the knee with degenerated

554 cartilage and osteoarthritis. J Orthop Res 2012;30:943-49. doi:10.1002/jor.22029.

555 8. Matsukura Y, Muneta T, Tsuji K, Koga H, Sekiya I. Mesenchymal Stem Cells in

556 Synovial Fluid Increase After Meniscus Injury. Clin Orthop Relat Res

557 2014;472:1357-64. doi:10.1007/s11999-013-3418-4.

558 9. Clevers H. Wnt/beta-Catenin Signaling in Development and Disease. Cell

559 2006;127:469-80. doi:10.1016/j.cell.2006.10.018.

560 10. Clevers H, Loh KM, Nusse R. Stem cell signaling. An integral program for tissue

561 renewal and regeneration: Wnt signaling and stem cell control. Science

562 2014;346:1248012. doi:10.1126/science.1248012.

563 11. Kahn M. Can we safely target the WNT pathway? Nat Rev Drug Discov

564 201 4;13:513-32. doi:10.1038/nrd4233.

565 12. Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin signaling in

566 mesenchymal progenitors controls osteoblast and chondrocyte differentiation

567 during vertebrate skeletogenesis. Dev Cell 2005;8:739-50.

568 doi:10.1016/j.devcel.2005.03.016.

569 13. Lane NE, Lian K, Nevitt MC, Zmuda JM, Lui L, Li J, et al. Frizzled-related protein

570 variants are risk factors for hip osteoarthritis. Arthritis Rheum 2006;54:1246-54.

571 doi: 10.1002/art.21673.

572 14. Baker-LePain JC, Lynch JA, Parimi N, McCulloch CE, Nevitt MC, Corr M, et al.

573 Variant alleles of the Wnt antagonist FRZB are determinants of hip shape and

574 modify the relationship between hip shape and osteoarthritis. Arthritis Rheum

575 201 2;64:1457-65. doi:10.1002/art.34526.

576 15. Ge X, Ma X, Meng J, Zhang C, Ma K, Zhou C. Role of Wnt-5A in interleukin-

577 1 beta-induced matrix metalloproteinase expression in rabbit temporomandibular

578 joint condylar chondrocytes. Arthritis Rheum 2009;60:2714-22.

579 doi: 10.1002/art.24779.

580 16. Pritzker KPH, Gay S, Jimenez SA, Ostergaard K, Pelletier JP, Revell K, et al.

581 Osteoarthritis cartilage histopathology: Grading and staging. Osteoarthritis

582 Cartilage. 2006;14:13-29. doi:10.1016/j.joca.2005.07.014.

583 17. Glasson SS, Chambers MG, Van Den Berg WB, Little CB. The OARSI

584 histopathology initiative - recommendations for histological assessments of

585 osteoarthritis in the mouse. Osteoarthritis Cartilage 2010;18:S17-S23.

586 doi: 10.1016/j.joca.2010.05.025.

587 18. Pastoureau P, Leduc S, Chomel A, De Ceuninck F. Quantitative assessment of

588 articular cartilage and subchondral bone histology in the meniscectomized guinea

589 pig model of osteoarthritis. Osteoarthritis Cartilage. 2003; 11:412-23.

590 http://www.ncbi.nlm.nih.gov/pubmed/12801481.

591 19. Yang J, Zhang W, Evans PM, Chen X, He X, Liu C. Adenomatous polyposis coli

592 (APC) differentially regulates beta-catenin phosphorylation and ubiquitination in

593 colon cancer cells. J Biol Chem 2006;281:17751-7. doi:10.1074/jbc.M600831200.

594 20. Handeli S, Simon JA. A small-molecule inhibitor of Tcf/ beta-catenin signaling

595 down-regulates PPAR and PPAR activities. Mol Cancer Ther 2008;7:521-9.

596 doi: 10.1158/1535-7163. MCT-07-2063.

597 21. Voronkov A, Krauss S. Wnt/beta-Catenin Signaling and Small Molecule Inhibitors.

598 Curr Pharm Des 2013;19:634-4. doi:10.2174/138161213804581837.

599 22. Gonsalves FC, Klein K, Carson BB, Katz S, Ekas LA, Evans S, et al. An RNAi-

600 based chemical genetic screen identifies three small-molecule inhibitors of the

601 Wnt/wingless signaling pathway. Proc Natl Acad Sci 2011; 108:5954-63.

602 doi: 10.1073/pnas. 1017496108.

603 23. Minami I, Yamada K, Otsuji TG, Yamamoto T, Shen Y, Otsuka S, et al. A Small

604 Molecule that Promotes Cardiac Differentiation of Human Pluripotent Stem Cells

605 under Defined, Cytokine- and Xeno-free Conditions. Cell Rep 2012;2:1448-60.

606 doi: 10.1016/j.celrep.2012.09.015.

607 24. Kim J, Hwan Kim S. CK2 Inhibitor CX-4945 Blocks TGF-ß1-Induced Epithelial-to-

608 Mesenchymal Transition in A549 Human Lung Adenocarcinoma Cells. Katoh M,

609 ed. PLoS One 2013;8:e74342. doi:10.1371/journal.pone.0074342.

610 25. Woods A, Beier F. RhoA/ROCK Signaling Regulates Chondrogenesis in a

611 Context-dependent Manner. J Biol Chem 2006;281: 13134-40.

612 doi: 10.1074/jbc.M509433200.

613 26. Shu B, Zhang M, Xie R, Wang M, Jin H, Hou W, et al. BMP2, but not BMP4, is

614 crucial for chondrocyte proliferation and maturation during endochondral bone

615 development. J Cell Sci 2011;124:3428-40. doi:10.1242/jcs.083659.

616 27. Zanotti S, Canalis E. Nemo-like kinase inhibits osteoblastogenesis by suppressing

617 bone morphogenetic protein and WNT canonical signaling. J Cell Biochem

618 2012;113:449-56. doi:10.1002/jcb.23365.

619 28. Stanton L-A, Underhill TM, Beier F. MAP kinases in chondrocyte differentiation.

620 Dev Biol 2003;263:165-75. http://www.ncbi.nlm.nih.gov/pubmed/14597193.

621 29. Johnson K, Zhu S, Tremblay MS, Payette JN, Wang J, Bouchez LC, et al. A Stem

622 Cell-Based Approach to Cartilage Repair. Science (80- ) 2012;336:717-21.

623 doi:10.1126/science.1215157.

624 30. Dahlin RL, Ni M, Meretoja V V., Kasper FK, Mikos AG. TGF-ß3-induced

625 chondrogenesis in co-cultures of chondrocytes and mesenchymal stem cells on

626 biodegradable scaffolds. Biomaterials 2014;35:123-32.

627 doi: 10.1016/j.biomaterials.2013.09.086.

628 31. Leung VYL, Gao B, Leung KKH, Melhado IG, Wynn SL, Au TYK, et al. SOX9

629 governs differentiation stage-specific gene expression in growth plate

630 chondrocytes via direct concomitant transactivation and repression. PLoS Genet

631 2011;7:e1002356. doi:10.1371/journal.pgen.1002356.

632 32. Yoon HJ, Kim SB, Somaiya D, Noh MJ, Choi K-B, Lim C-L, et al. Type II collagen

633 and glycosaminoglycan expression induction in primary human chondrocyte by

634 TGF-ß1. BMC Musculoskelet Disord 2015;16:141. doi:10.1186/s12891-015-0599-

635 x.

636 33. Phimphilai M, Zhao Z, Boules H, Roca H, Franceschi RT. BMP Signaling Is

637 Required for RUNX2-Dependent Induction of the Osteoblast Phenotype. J Bone

638 Miner Res 2006;21:637-46. doi:10.1359/jbmr.060109.

639 34. Zheng Q, Zhou G, Morello R, Chen Y, Garcia-Rojas X, Lee B. Type X collagen

640 gene regulation by Runx2 contributes directly to its hypertrophic chondrocyte-

641 specific expression in vivo. J Cell Biol 2003;162:833-42.

642 doi: 10.1083/jcb.200211089.

643 35. Altaf FM, Hering TM, Kazmi NH, Yoo JU, Johnstone B. Ascorbate-enhanced

644 chondrogenesis of ATDC5 cells. Eur Cells Mater 2006;12:64-9. doi:vol012a08

645 [pi i].

646 36. Aberg T, Rice R, Rice D, Thesleff I, Waltimo-Siren J. Chondrogenic potential of

647 mouse calvarial mesenchyme. J Histochem Cytochem 2005;53:53-663.

648 doi: 10.1369/jhc.4A6518.2005.

649 37. Murphy G, Knauper V, Atkinson S, Butler G, English W, Hutton M, et al. Matrix

650 metalloproteinases in arthritic disease. Arthritis Res 2002;4(Suppl 3):S39.

651 doi:10.1186/ar572.

652 38. Hayami T, Pickarski M, Zhuo Y, Wesolowski GA, Rodan GA, Duong LT.

653 Characterization of articular cartilage and subchondral bone changes in the rat

654 anterior cruciate ligament transection and meniscectomized models of

655 osteoarthritis. Bone 2006;38:234-43. doi:10.1016/j.bone.2005.08.007.

656 39. Hamilton CB, Pest MA, Pitelka V, Ratneswaran A, Beier F, Chesworth BM.

657 Weight-bearing asymmetry and vertical activity differences in a rat model of post-

658 traumatic knee osteoarthritis. Osteoarthritis Cartilage. 2015;23:1178-85.

659 doi: 10.1016/j.joca.2015.03.001.

660 40. Rousseau J-C, Zhu Y, Miossec P, Vignon E, Sandell LJ, Garnero P, et al. Serum

661 levels of type IIA procollagen amino terminal propeptide (PIIANP) are decreased

662 in patients with knee osteoarthritis and rheumatoid arthritis. Osteoarthritis V

663 Cartilage 2004;12:440-7. doi:10.1016/j.joca.2004.02.004.

664 41. Zhang Y, Ryan JA, Di Cesare PE, Liu J, Walsh CA, You Z. Doublecortin is

665 expressed in articular chondrocytes. Biochem Biophys Res Commun

666 2007;363:694-700. doi:10.1016/j.bbrc.2007.09.030.

667 42. Li N-G, Shi Z-H, Tang Y-P, Wang Z-J, Song S-L, Qian L-H, et al. New hope for

668 the treatment of osteoarthritis through selective inhibition of MMP-13. Curr Med

669 Chem 2011;18:977-1001. http://www.ncbi.nlm.nih.gov/pubmed/21254976.

670 43. Verma P, Dalal K. Serum cartilage oligomeric matrix protein (COMP) in knee

671 osteoarthritis: a novel diagnostic and prognostic biomarker. J Orthop Res

672 2013;31:999-1006. doi:10.1002/jor.22324.

673 44. Calich ALG, Domiciano DS, Fuller R. Osteoarthritis: can anti-cytokine therapy

674 play a role in treatment? Clin Rheumatol 2010;29:451-5. doi:10.1007/s10067-009-

675 1 352-3.

676 45. Chen B, Qin J, Wang H, Magdalou J, Chen L. Effects of adenovirus-mediated

677 bFGF, IL-1 Ra and IGF-1 gene transfer on human osteoarthritic chondrocytes and

678 osteoarthritis in rabbits. Exp Mol Med 2010;42:684.

679 doi:10.3858/emm.2010.42.10.067.

680 46. Sierra R, Wyles C, Houdek M, Behfar A. Mesenchymal stem cell therapy for

681 osteoarthritis: current perspectives. Stem Cells Cloning Adv Appl 2015:117.

682 doi: 10.2147/SCCAA.S68073.

1 Figure 1

2 SM04690 was a potent and specific inhibitor of Wnt signaling

3 SW480 cells and hMSCs treated with SM04690 or DMSO for 48hrs (a) Dose response

4 of TCF/LEF promoter driven- or SV40 promoter driven- luciferase reporters in SW480

5 cells (n=4, Mean ± 95% CI). (b) Expression of genes in the Wnt signaling pathway in

6 SW480 cells as measured by qRT-PCR. Fold change relative to DMSO (n=3, Mean ±

7 95% CI, Ascll ***p<0.0001, LEF1 ***p=0.0001, TCF7L2 **p=0.0038, TCF7 ***p=0.0005,

8 c-myc ***p=0.0003, Axin2 *p=0.014, t-test). (c) Expression of proteins in the Wnt

9 signaling pathway in SW480 cells as measured by Western blot. (d) Expression of

10 genes in the Wnt signaling pathway in hMSCs as measured by qRT-PCR. Fold change

11 relative to DMSO (n=3, Mean ± 95% CI, Ascl1 **p=0.002, LEF1 *p=0.017, TCF7L2

12 **p=0.004, TCF7 **p=0.0053, c-myc ***p<0.0001, Axin2 ***p=0.0008, t-test). (e)

13 Expression of proteins in the Wnt signaling pathway in hMSCs as measured by Western

14 blot.

22 Supplementary Figure 1

23 SM04690 was a potent and specific inhibitor of Wnt signaling

24 (a, b) SW480 cells and hMSCs treated with SM04690 or various Wnt pathway inhibitors

25 or DMSO for 48hrs. (a) Dose response of TCF/LEF promoter driven- luciferase reporter

26 in SW480 cells (n=4, Mean ± 95% CI). (b) EC50 values for Wnt inhibitors in (a). (c, d)

27 Gene expression of Wnt pathway genes in (c) hMSCs and (d) IEC6 cells, stimulated

28 either with Wnt3a (200 ng/ml) or CHIR99021 (4 pM) and treated with SM04690 (30nM)

29 or CX-4945 (3pM) or KY02111 (10pM) for 16hrs. Fold change relative to unstimulated

30 control (n=4, Mean ± 95% CI, *p<0.05, ***p<0.01, ***p<0.001, one-way ANOVA).

42 SM04690 was a potent and specific inhibitor of Wnt signaling

43 (a, b) Expression of genes in the non-canonical Wnt pathway in hMSCs following

44 treatment with DMSO or SM04690 (30nM) or CX-4945 (10 pM) or KY02111 (10pM) for

45 48hrs in the (a) absence or (b) presence of Wnt5a (100ng/ml), measured by qRT-PCR.

46 Expression represented as ddCt relative to DMSO (n=4, Mean ± 95% CI, *p<0.05,

47 **p<0.01, one-way ANOVA).

60 SM04690 was a potent and specific inhibitor of Wnt signaling

61 Expression of genes in the BMP pathway in hMSCs following treatment with DMSO or

62 SM04690 (30nM, 100nM) for 6hrs, 40hrs or 72hrs measured by qRT-PCR. Expression

63 represented as ddCt relative to DMSO (n=4, Mean ± 95% CI, *p<0.05, **p<0.01,

64 ***p<0.001, t-test vs DMSO at each timepoint).

77 Figure 2

78 SM04690 induced chondrocyte differentiation from hMSCs

79 (a) hMSCs treated with SM04690 (30nM) or DMSO or TGFp3 (20ng/ml) for 5 days and

80 stained with Rhodamine B (scale bars, 200 pm). (b) Quantification of the number of

81 Rhodamine B stained colonies in (a) (n=6, Mean ± 95% CI, ***p<0.0001, t-test). (c)

82 hMSCs treated with SM04690 (30nM) or DMSO or TGFp3 (20ng/ml) for 3 days and

83 stained for Sox9 (scale bars, 200 pm). (d) Quantification of the number of Sox9 positive

84 cells in (c) (n=36 images, Mean ± 95% CI, ***p<0.0001, t-test). (e) hMSCs treated with

85 SM04690 (30nM) or DMSO in 3-D sphere culture for 21 days and stained with Safranin

86 O for mature chondrocytes (scale bars, 200 pm). (f) hMSCs treated with SM04690

87 (30nM) or DMSO for 21 days and stained for various markers for mature chondrocytes

88 (scale bars, 200 pm). (g) Total sulphated GAG in hMSCs treated with SM04690 (30nM)

89 or DMSO for 21 days measured by DMMB assay. Total GAG levels relative to the

90 weight of the cell aggregates shown (n=3, Mean ± 95% CI, *p=0.041, t-test).

99 SM04690 induced chondrocyte differentiation from hMSCs

100 (a) Dose response quantification of the number of Rhodamine B stained chondrogenic

101 colonies from hMSCs treated with SM04690 or DMSO or TGFp3 for 5 days (n=4, Mean

102 ± 95% CI, *p=0.013, ***p<0.0001, one-way ANOVA). (b) Expression of Sox9 in hMSCs

103 following treatment with various doses of SM04690 or DMSO for 3 days as measured

104 by Western blot. Tubulin binding protein (TBP) serves as a loading control.

110 111 112

116 Figure 3

117 SM04690 induced chondrocyte differentiation from hMSCs

118 hMSCs, ATDC5 cells and primary mouse calvarial cells treated with DMSO or SM04690

119 (30nM) or TGFp3 for 21 days (a) Gene expression of mature chondrocyte markers in

120 treated hMSCs measured by qRT-PCR. Fold change relative to DMSO (n=3, Mean ±

121 95% CI, SOX9 **p=0.002, COMP ***p<0.0001, ACAN **p=0.009, COL2A1 ***p=0.0001,

122 TGFp1 ***p=0.0009, TIMP1 *p=0.046, CD44 **p= 0.0031, COL10A1 ***p<0.0001, t-

123 test). (b) Expression of osteocyte and tendon/ligament markers in treated hMSCs

124 measured by qRT-PCR. Fold change relative to DMSO (n=3, Mean ± 95% CI, COL1A1,

125 BGLAP, ALPL, BMP4: ***p<0.0001, RUNX2 **p=0.0009, t-test). (c) Safranin O and

126 Alcian Blue staining of treated ATDC5 cells (scale bars, 200 pm). (d) Alcian Blue

127 staining of treated primary mouse calvaria cells (scale bar, 50 pm). (e) Quantification of

128 chondrocytes in (d) (n=3, Mean ± 95% CI, *p=0.044, t-test).

137 SM04690 induced chondrocyte differentiation from hMSCs

138 hMSCs treated with SM04690 (30nM or 10nM) or CX-4945 (10 pM) or KY02111 (10

139 pM) or DMSO for 21 days and gene expression measured by qRT-PCR (a) Gene

140 expression of mature chondrocyte markers in treated hMSCs. Fold change relative to

141 DMSO (n=3, Mean ± 95% CI, *p<0.05, **p<0.01, ***p<0.0001, one-way ANOVA). (b)

142 Expression of osteocyte and tendon/ligament markers in treated hMSCs. Fold change

143 relative to DMSO (n=3, Mean ± 95% CI, *p<0.05, p<0.01, ***p<0.001, one-way

144 ANOVA).

156 Supplementary Figure 6

157 SM04690 induced chondrocyte differentiation from hMSCs

158 (a) Dose response quantification of the number of Rhodamine B stained chondrogenic

159 colonies from hMSCs treated with Wnt pathway inhibitors (n=3, Mean ± 95% CI). (b)

160 EC5o values for compounds in (a). (c) Dose response quantification of Alcian blue

161 stained chondrocytes from hMSCs treated with DMSO or SM04690 or TGFp3 or Wnt

162 pathway inhibitors for 21 days (n=6, Mean ± 95% CI). (d) EC50 values for compounds in

163 (c). (e) Representative images of chondrocytes from (c). (f) Dose response

164 quantification of Safranin O stained chondrocytes from hMSCs treated with DMSO or

165 SM04690 or TGFp3 or Wnt pathway inhibitors for 21 days (n=6, Mean ± 95% CI). (g)

166 EC50 values for compounds in (f). (h) Representative images of chondrocytes from (f).

175 Figure 4

176 SM04690 protected chondrocytes from catabolic breakdown in vitro

177 Chondrocytes treated with cytokines- TNFa (20ng/ml) + Oncostatin M (10ng/ml) or IL1p

178 (10ng/ml) and SM04690 (30nM) or DMSO control for 72hrs (a, b) Gene expression of

179 proteases (MMP1, MMP3, MMP13, IHH) in chondrocytes measured by qRT-PCR. Fold

180 change relative to unstimulated control (n=3, Mean ± 95% CI, TNFa + Oncostatin M:

181 MMP1 *p=0.04, MMP3 *p=0.014, MMP13 **p=0.0046, IHH *p=0.022, IL1p: MMP1

182 *p=0.019, MMP3 *p=0.017, MMP13 *p=0.015, IHH *p=0.026, t-test). (c) Levels of

183 secreted GAG expressed as a ratio of intracellular GAG, measured by the DMMB

184 assay. (n=6, Mean ± 95% CI, TNFa+OM ***p<0.0001, IL1p *p=0.032, t-test). (d) Levels

185 of secreted Nitric Oxide (NO) measured using the Griess reagent assay. (n=6, Mean ±

186 95% CI, TNFa+OM ***p<0.0001, IL1p **p=0.005, t-test).

195 Supplementary Figure 7

196 SM04690 protected hMSCs and chondrocytes from catabolic breakdown in vitro

197 (a, b) Gene expression of proteases (MMP1, MMP3, MMP13, IHH) in hMSCs treated

198 with either (a) TNFa (20ng/ml) + Oncostatin M (10ng/ml) or (b) IL1p (10ng/ml) and

199 SM04690 (30nM) or DMSO for 72hrs measured by qRT-PCR. Fold change relative to

200 unstimulated control (n=3, Mean ± 95% CI, TNFa + Oncostatin M: MMP1 ***p=0.0004,

201 MMP3 p=0.148, MMP13 **p=0.01, IHH **p=0.0075, IL1p: MMP1 p=0.077, MMP3

202 p=0.141, MMP13 p=0.158, IHH *p=0.014, t-test). (c, d) Gene expression of proteases

203 (MMP1, MMP3, MMP13) in human chondrocytes treated with either (c) TNFa (20ng/ml)

204 + Oncostatin M (10ng/ml) or (d) IL1p (10ng/ml) and SM04690 (30nM) or CX-4945 (10

205 pM) or KY02111 (10 pM) or DMSO for 72hrs as measured by qRT-PCR. Fold change

206 relative to unstimulated control (n=3, Mean ± 95% CI, *p<0.05, **p<0.01, ***p<0.001,

207 one-way ANOVA).

210 211 212

215 Figure 5

216 SM04690 promoted cartilage repair and protection in the rat ACLT+pMMx OA model.

217 ACLT+pMMx rats treated with either vehicle or SM04690 (0.3|jg) (a) Representative

218 images of medial tibial plateau of the knee joint stained with Safranin O-Fast Green

219 from naive or vehicle-treated or SM04690 (0.3jg) treated rats 12 weeks from treatment.

220 (b) The medial joint score based on the OARSI scoring system (n= 12 rats, Mean ± 95%

221 CI, **p=0.008, Mann-Whitney U test). (c) Circulating PIIANP levels following treatment

222 (n= 12 rats, Mean ± 95% CI, week 3 *p=0.018, week 5 p=0.13, t-test). (d) Gene

223 expression of chondrocyte markers in the cartilage of rats 4 weeks from treatment

224 measured by qRT-PCR (n= 7 for vehicle, n= 8 for treatment, Mean ± 95% CI, Col2a1

225 *p=0.024, COMP ***p=0.0007, aggrecan ***p=0.0014, t-test). (e) Representative images

226 of the superficial zone of articular cartilage from the ACLT+pMMx model stained for

227 Doublecortin (Dcx). (f) Quantification of Dcx expressing chondrocytes in (e) (n= 9

228 rats/group for vehicle, n=12 rats/group for treatment, Mean ± 95% CI, **p=0.001, t-test).

229 (g) Gene expression of proteases in the cartilage of rats 4 weeks from treatment

230 measured by qRT-PCR (n= 7 for vehicle, n= 8 for treatment, Mean ± 95% CI, MMP1

231 ***p=0.0006, MMP3 ***p=0.0006, MMP13 **p=0.0046, ADAMTS5 *p=0.016, t-test). (h)

232 Circulating COMP levels following treatment (n= 12 rats, Mean ± 95% CI, week 2

233 p=0.19, week 5 **p=0.0064, t-test).

238 Supplementary Figure 8

239 SM04690 promoted cartilage repair and protection in the ACLT OA model in rats.

240 (a) Pharmacokinetics of SM04690 in rat cartilage, bone and plasma following a single IA

241 injection of SM04690 (0.3|jg). (b) The medial joint score in the ACLT + pMMx model,

242 based on the OARSI scoring system (n= 12 rats, Mean ± 95% CI, ***p=0.0005, Mann-

243 Whitney U test) (c) Cartilage thickness measurement and (d) Safranin O staining

244 intensity of knee sections from the ACLT+pMMx model (n=12 rats, Mean ± 95% CI,

245 **p=0.0042, ***p<0.0001 respectively, t-test).

257 Supplementary Figure 9

258 SM04690 inhibited Wnt signaling in the ACLT+pMMx OA model in rats.

259 ACLT+pMMx rats treated with either vehicle or single IA injection of SM04690 (0.3jg)

260 and cartilage analyzed 4 weeks from treatment for gene expression by qRT-PCR. (a)

261 Col10a1 expression. (b) Expression of Wnt pathway genes, represented on a color

262 scale as shown. Expression of Wnt pathway genes (c) Axin2 and (d) p-catenin (n= 7 for

263 vehicle, n= 8 for treatment, Mean ± 95% CI, Col10a p=0.48, Axin 2 *p=0.017, p-catenin

264 *p=0.04, t-test).

276 Supplementary Figure 10

277 SM04690 inhibited Wnt signaling in the ACLT+pMMx OA model in rats.

278 ACLT+pMMx rats treated with either vehicle or SM04690 (0.3pg) and analyzed by IHC

279 12 weeks from treatment. (a) Representative images of the superficial zone of articular

280 cartilage stained for p-catenin. (b, c) Quantification of staining intensity of p-catenin in

281 (b) the total cell and in (c) the nucleus of the cells, from images in (a) (n= 6 rats/group,

282 Mean ± 95% CI, **p=0.0091 and *p=0.044 respectively, t-test).

Relative Expression CT

Relative Expression

i i I i i

F Relative Expression

3 I 8 §

Relative Expression S è 8 8 S

Relative E xpress ion

. s I I I I

Relative Expression