Study TPX-100-5: intra-articular TPX-100 significantly delays pathological bone shape change and stabilizes cartilage in moderate to severe bilateral knee OA

Dawn McGuire, Michael Bowes, Alan Brett, Neil A Segal, Meghan Miller, David Rosen, Yoshinari Kumagai, Dawn McGuire, Michael Bowes, Alan Brett, Neil A Segal, Meghan Miller, David Rosen, Yoshinari Kumagai

Abstract

Background: TPX-100, a promotor of osteoblast and chondroblast differentiation, is a potential osteoarthritis (OA) therapy. This retrospective study compared MRI 3D femoral bone shape changes (B-scores) after intra-articular TPX-100 or placebo and analyzed the relationship between cartilage thickness and bone shape change over 12 months.

Methods: One hundred and four participants with bilateral moderate to severe knee cartilage defects were randomized to receive TPX-100 (200 mg) or placebo. Each subject's contralateral placebo-treated knee served as a paired internal control. After MRI quality control, 78/93 subjects (84%; 156 knees) were analyzed for quantitative femoral B-score and cartilage thickness. All analyses were performed centrally, blind to treatment assignment and clinical data.

Results: TPX-100-treated knees (n = 78) demonstrated a statistically significant decrease in pathologic bone shape change compared with placebo-treated knees at 6 and 12 months: 0.0298 (95% C.I. - 0.037, 0.097) vs 0.1246 (95% C.I. 0.067, 0.182) (P = 0.02), and 0.0856 (95% C.I. 0.013, 0.158) vs. 0.1969 (95% C.I. 0.123, 0.271) (P = 0.01), respectively. The correlation between bone shape change and medial and total tibiofemoral cartilage thickness changes at 12 months was statistically significant in TPX-100-treated knees (P < 0.01).

Conclusions: This is the first report of a potential therapy demonstrating a significant effect on bone shape measured by B-score in knee OA. These data, in combination with previously reported statistically significant and clinically meaningful improvements in WOMAC physical function versus placebo, support TPX-100 as a candidate for disease modification in knee OA.

Trial registration: NIH ClinicalTrials.gov, NCT01925261 . Registered 15 August 2013.

Keywords: B-score; Bone shape; DMOAD; Machine learning; Osteoarthritis; TPX-100.

Conflict of interest statement

AB and MB are employees of Imorphics, Manchester, UK. DM, MM, DR, and YK are employees of OrthoTrophix, Inc., Oakland, CA. NS reports no competing interests.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
CONSORT diagram for the TPX-100-1 and TPX-100-5 studies
Fig. 2
Fig. 2
Change in shape for the anterior femur (top row) and posterior femur (bottom row), for various B-scores. Red indicates where there is an increase in size (locally calculated, based on anatomically corresponded triangles from the shape model), blue indicates decrease in size (locally); scale shows percentage in area size change of each triangle. Change tends to be greatest around the edge of the cartilage plate (osteophyte region), but it also occurs in central subchondral regions where the bone flattens out. Adapted from Bowes et al. [11]
Fig. 3
Fig. 3
Femoral bone shape changes in TPX-100-treated knees and placebo-exposed knees in bilateral OA subjects and their comparison with historical bone shape change from the Osteoarthritis Initiative (OAI). All error bars represent standard errors of the mean
Fig. 4
Fig. 4
12-month femoral bone shape stabilization is associated with medial femoral condyle cartilage thickening/stabilization in the index knee. Subjects are stratified by B-score change quartiles. All error bars are standard errors of the mean

References

    1. Hopwood B, Tsykin A, Findlay DM, Fazzalari NL. Microarray gene expression profiling of osteoarthritic bone suggests altered bone remodelling, WNT and transforming growth factor-β/bone morphogenic protein signalling. Arthritis Res Ther. 2007;9(5):R100. doi: 10.1186/ar2301.
    1. Hayashibara T, Hiraga T, Yi B, Motoyoshi N, Kumagai Y, Nishimura R, et al. A synthetic peptide fragment of human MEPE stimulates new bone formation in vitro and in vivo. J Bone Miner Res. 2004;19(3):455–462. doi: 10.1359/JBMR.0301263.
    1. Nagel DE, Khosla S, Sanyal A, Rosen DM, Kumagai Y, Riggs L. A fragment of hypophospatemic factor, MEPE, requires inducible cyclooxygenase-2 to exert potent anabolic effects on normal human marrow osteoblast precursors. J Cell Biochem. 2004;93(6):1107–1114. doi: 10.1002/jcb.20249.
    1. Middleton-Hardie CA, Aberman H, Simon T, Alliston T, Mortazavi A, Rosen DM. AC-100 Promotes Cartilage Defect Repair In Vivo and Chondrocyte Differentiation and Function In Vitro [abstract]. Proceedings of the 56th Annual Meeting of the Orthopedic Research Society, poster # 0974. Presented March 6, 2010. New Orleans, LA
    1. McGuire D, Lane N, Segal N, Metyas S, Barthel H, Miller M, et al. TPX-100 leads to marked, sustained improvements in subjects with knee osteoarthritis: pre-clinical rationale and results of a controlled clinical trial [abstract] Osteoarthr Cartil. 2018;26:S243. doi: 10.1016/j.joca.2018.02.50219.
    1. McGuire D, Segal N, Metyas S, Barthel H, Miller M, Rosen D, et al. Intra-articular TPX-100 in knee osteoarthritis: robust functional response at 6 and 12 months is associated with increased tibiofemoral cartilage thickness [abstract L16]. Arthritis Rheumatol. 2018;70(suppl 10):3405-3406.
    1. Neogi T, Bowes MA, Niu J, De Souza KM, Vincent GR, Goggins J, et. al. Magnetic Resonance Imaging-Based Three-Dimensional Bone Shape of the Knee Predicts Onset of Knee Osteoarthritis: Data From the Osteoarthritis Initiative. Arthritis Rheum 2013;65(8):2048-2058. doi: 10.1002/art.37987.
    1. Barr AJ, Dube B, Hensor E, Kingsbury S, Peat G, Bowes M, et. al. The relationship between three-dimensional knee MRI bone shape and total knee replacement—a case control study: data from the Osteoarthritis Initiative. Rheumatology 2016; 55:1585–1593. doi: 10.1093/rheumatology/kew191.
    1. Bowes MA, Vincent GR, Wolstenholme CB, Conaghan PG. A novel method for bone area measurement provides new insights into osteoarthritis and its progression. Ann Rheum Dis. 2013;74:519–525. doi: 10.1136/annrheumdis-2013-204052.
    1. Hunter D, Nevitt M, Lynch J, Kraus VB, Katz JN, Collins JE, et. al. Longitudinal validation of periarticular bone area and 3D shape as biomarkers for knee OA progression? Data from the FNIH OA Biomarkers Consortium. Ann Rheum Dis 2016; 75:1607–1614. doi: 10.1136/annrheumdis-2015-207602.
    1. Bowes MA, Kacena K, Alabas OA, Brett A, Dube B, Bodick N, . Machine-learning, MRI bone shape and important clinical outcomes in osteoarthritis: data from the Osteoarthritis Initiative. Ann Rheum Dis. 2020;80(4):502-508. doi: 10.1136/annrheumdis-2020-217160.
    1. Eckstein F, Ateshian G, Burgkart R, Burnstein D, Cicuttini F, Dardzinski B, et al. Proposal for a nomenclature for Magnetic Resonance Imaging based measures of articular cartilage in osteoarthritis. Osteoarthr Cartil. 2006;14(10):974–983. doi: 10.1016/j.joca.2006.03.005.
    1. Wirth W, Eckstein F. A technique for regional analysis of femorotibial cartilage thickness based on quantitative magnetic resonance imaging. IEEE Trans Med Imaging. 2008;27:737–744. doi: 10.1109/TMI.2007.907323.
    1. Williams TG, Holmes A, Waterton J, Maciewicz R, Hutchinson CE, Moots R, et al. Anatomically corresponded regional analysis of cartilage in asymptomatic and osteoarthritic knees by statistical shape modelling of the bone. IEEE Trans Med Imaging. 2010;29(8):1541–1559. doi: 10.1109/TMI.2010.2047653.
    1. Hunter DJ, Nevitt M, Lynch J, Kraus V, Katz JN, Collins J, et. al. Preliminary assessment of predictive validity of periarticular bone area and shape markers in knee OA. Osteoarthr Cartil 2013; 21: S175. doi: 10.1016/j.joca.2013.02.376.
    1. Bowes MA, Lohmander LS, Wolstenholme CBH, Vincent GR, Conaghan PG, Frobell RB. Marked and rapid change of bone shape in acutely ACL injured knees – an exploratory analysis of the Kanon trial. Osteoarthr Cartil. 2019;27(4):638–645. doi: 10.1016/j.joca.2018.12.021.
    1. Conaghan PG, Bowes MA, Kingsbury S, Brett A, Guillard G, Rizoska B, et. al. Disease-Modifying Effects of a Novel Cathepsin K Inhibitor in Osteoarthritis. Ann Intern Med 2020;172(2):86-95. doi: 10.7326/M19-0675.
    1. Liu H, Li W, Gao C, Kumagai Y, Blacher RW, DenBesten PK. Dentonin, a Fragment of MEPE, Enhanced Dental Pulp Stem Cell Proliferation. J Dent Res. 2004;83(6):496–499. doi: 10.1177/154405910408300612.

Source: PubMed

Подписаться