header advert
Bone & Joint Research Logo

Receive monthly Table of Contents alerts from Bone & Joint Research

Comprehensive article alerts can be set up and managed through your account settings

View my account settings

Visit Bone & Joint Research at:

Loading...

Loading...

Open Access

Editorial

Disease-modifying agents in osteoarthritis: where are we now and what does the future hold?



Download PDF

Abstract

Cite this article: Bone Joint Res 2023;12(10):654–656.

Osteoarthritis (OA) remains one of the most common chronic health conditions and a leading cause of pain and disability among adults.1 The Global Burden of Disease report estimates that over 500 million individuals are affected by OA worldwide,2 with global prevalence having increased by 113% since 1990.3 By 2030, OA is predicted to be the single greatest cause of disability globally, with one-third of the global population estimated to be affected.4

Despite the immense burden that OA already brings to the global population, the general treatment paradigm has remained largely unchanged. This comprises symptomatic treatment of the disease, combining non-pharmacological management (patient education, weight loss, activity modification, and physiotherapy) and pharmacological management (predominantly confined to analgesic and anti-inflammatory agents). Surgical intervention with joint arthroplasty may be required for end-stage disease.5

Although a vast array of research into disease-modifying osteoarthritic drugs (DMOADs) has been carried out with a range of potential pharmacological targets identified, none have yet gained regulatory approval. These agents are defined by their ability to alter the underlying pathophysiological mechanisms of OA in order to inhibit structural disease progression, while providing symptomatic relief.6 The US Food and Drug Administration (FDA) and the European Medicines Agency draft guidance states that in order for a DMOAD to be approved, there must be clear demonstration of slowing of knee or hip joint space width on radiographs with relevant symptomatic benefit.6,7 A range of these potential DMOADs remain at varying stages of investigation and development, and have traditionally centred around potentiating or inhibiting pathways related to three predominating disease-driving OA ‘endotypes’: cartilage-driven, inflammation-driven, or bone-driven.8 Given the range and growing complexity of potential agents and targets for disease modification in OA, the authors recommend that agents are categorized and considered from the main endotypes in which they act. Such agents include bone morphogenetic proteins (BMPs),9,10 Wingless-like signalling inhibitors,11-13 proteinase inhibitors,14-17 growth factors,18-20 and anabolic bone remodelling agents.21-23 More recently, there has been increasing interest in novel agents and themes, including specific plant extracts,24 the action of non-coding RNAs (ncRNAs),25,26 the influence of the gut microbiome,27,28 and gene-environment interactions,29-31 which could be harnessed in the OA disease process.

Emerging approaches and future therapies: although encouraging results have been observed from preclinical studies of several potential DMOADS including those mentioned above, currently large-scale clinical trials of such agents have proven unconvincing in achieving meaningful improvements in OA-related symptoms, despite several showing substantial structural improvements. Consequently, attention has recently turned to gene-environment-disease interactions of OA and the molecules that may modify such interactions.

Recent studies of the effects of ncRNAs in the OA disease process have revealed these molecules to be important mediators of joint inflammation,32 chondrocyte apoptosis,33 and extracellular matrix (ECM) degeneration. Until recently, these ncRNA peptide chains were thought to be ‘translation noise’, of no functional consequence, however more recently these molecules have been shown to have important effects on gene expression and molecular pathway modulation. A recent study by Zhou et al25 showed that one such long non-coding RNA, H19, significantly reduced OA-related inflammation in a mouse model, mediated through potentiation of IL-38 and IL-36R. Separately, Luobu et al26 showed the potential importance of the circular RNA ‘S-phase cyclin A-associated protein in the endoplasmic reticulum’ (circSCAPER) in OA-associated inflammation. Their in vitro study of human chondrocytes revealed that circSCAPER promoted interleukin-1 beta (IL-1β) associated cartilage degradation through chondrocyte proliferation arrest, apoptosis enhancement, and ECM degradation. Knockdown of circSCAPER reduced chondrocyte degradation.26 Further study of these and other ncRNAs as potential DMOADs may provide insightful data.

As our knowledge of the pathogenesis of OA expands, so does our awareness of the number of common pathways and interactions between OA, other diseases, environmental influences, and the microbiome. The association between OA and metabolic syndrome is now well established, but also emerging is the shared genetic basis between OA and conditions such as obesity and major depressive disorder.30,31,34 Furthermore, our knowledge of the interplay between the gut microbiome, the environment, and OA is rapidly evolving. The collection of trillions of microbial organisms that reside in the gastrointestinal tract has been shown to play a key role in host metabolism, the immune system, and inflammatory response.27 Recent evidence has shown the involvement of the gut microbiome in OA progression.35,36 The ability for exercise to directly modulate the microbiome and its relationship to the severity of OA is also being evaluated.29 Furthermore, the effect of probiotic supplementation in modulating disease progression of OA has been evaluated in a mouse model of OA, which showed significant reduction in cartilage degeneration in the probiotic supplemented cohort.36 The beneficial effects of probiotics in OA have also been shown in a recent large double-blind randomized controlled trial, where probiotic treatment with lactobacillus casei Shirota in patients with knee OA showed significant improvements in OA-related knee function (Western Ontario and McMaster Universities osteoarthritis index)37 and pain compared with placebo.28

A notable component of the bone-driven OA endotype is mediated through the subchondral bone plate.38 This endotype is characterized by hypomineralization of subchondral bone, osteophyte generation, development of microfractures & cysts, and subchondral plate sclerosis.39,40 Subchondral plate remodelling and attrition may precede and mediate cartilage degeneration in OA.41 The repurposing of established regulation-approved bone remodelling agents employed in the treatment of other bone diseases for use in bone-driven OA also shows potential. Recombinant parathyroid hormone (PTH) (Teriparatide), which is an established FDA-approved therapy for osteoporosis,42 has shown promise in preclinical studies. Systemic injection of higher-dose PTH reduced OA progression by preserving subchondral bone and reducing cartilage degradation,22,23 and more recently intra-articular injection showed similar OA-attenuating effects with much lower doses.21

Recently, new molecules have emerged as potential pharmacological agents within the inflammation-driven model of OA. The protein Peli1, which has an emerging role in IL-1R regulation and a host of other interactions in macrophage-associated inflammation, has been shown to have a substantial pro-inflammatory role in OA-associated cartilage degradation.43 Recent in vitro and in vivo models demonstrating knockdown of Peli1 showed reduced expression of IL-6, tumour necrosis factor alpha, and matrix metalloproteinase-3, and resulted in attenuated chondrocyte-related degradation.43 Similarly, Dihydrocaffeic acid (DHCA), the main bioactive component in the plant Gynura bicolor, has shown both anti-oxidative and anti-inflammatory properties44. More recently, DHCA demonstrated chondroprotective effects in vitro and in a destabilised medial meniscus OA mouse model, through downregulation of the inflammatory mediators nitric oxide synthase and IL-6.24

To conclude, advances in our understanding of the pathogenesis of OA have enabled the identification of an increasing array of potential pharmacological targets and novel disease-modifying agents for OA. However, despite encouraging preclinical data and some clinical evidence for benefit, none have yet reached the threshold of symptomatic improvements required for regulatory approval. Recent interest in novel molecular targets and agents, including ncRNAs, manipulation of gene expression, and modulation of the gut microbiome, could provide valuable avenues for further study in this rapidly evolving field.


Correspondence should be sent to Navnit S. Makaram. E-mail:

References

1. Hunter DJ , Schofield D , Callander E . The individual and socioeconomic impact of osteoarthritis . Nat Rev Rheumatol . 2014 ; 10 ( 7 ): 437 441 . Crossref PubMed Google Scholar

2. Quicke JG , Conaghan PG , Corp N , Peat G . Osteoarthritis year in review 2021: epidemiology & therapy . Osteoarthritis Cartilage . 2022 ; 30 ( 2 ): 196 206 . Crossref PubMed Google Scholar

3. Long H , Liu Q , Yin H , et al. Prevalence Trends of Site-Specific Osteoarthritis From 1990 to 2019: Findings From the Global Burden of Disease Study 2019 . Arthritis Rheumatol . 2022 ; 74 ( 7 ): 1172 1183 . Crossref PubMed Google Scholar

4. Thomas E , Peat G , Croft P . Defining and mapping the person with osteoarthritis for population studies and public health . Rheumatology (Oxford) . 2014 ; 53 ( 2 ): 338 345 . Crossref PubMed Google Scholar

5. Makaram NS , Roberts SB , Macpherson GJ . Simultaneous Bilateral Total Knee Arthroplasty Is Associated With Shorter Length of Stay but Increased Mortality Compared With Staged Bilateral Total Knee Arthroplasty: A Systematic Review and Meta-Analysis . J Arthroplasty . 2021 ; 36 ( 6 ): 2227 2238 . Crossref PubMed Google Scholar

6. No authors listed . Clinical Development Programs for Drugs, Devices, and Biological Products for the Treatment of Rheumatoid Arthritis (RA) . U.S. Food & Drug Administration . 2019 . https://www.fda.gov/regulatory-information/search-fda-guidance-documents/clinical-development-programs-drugs-devices-and-biological-products-treatment-rheumatoid-arthritis ( date last accessed 26 September 2023 ). Google Scholar

7. No authors listed . Guideline on clinical investigation of medicinal products used in the treatment of osteoarthritis . European Medicines Agency . 2010 . https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-clinical-investigation-medicinal-products-used-treatment-osteoarthritis_en.pdf ( date last accessed 20 September 2023 ). Google Scholar

8. Lv Z , Yang YX , Li J , et al. Molecular Classification of Knee Osteoarthritis . Front Cell Dev Biol . 2021 ; 9 : 725568 . Crossref PubMed Google Scholar

9. Whitty C , Pernstich C , Marris C , McCaskie A , Jones M , Henson F . Sustained delivery of the bone morphogenetic proteins BMP-2 and BMP-7 for cartilage repair and regeneration in osteoarthritis . Osteoarthr Cartil Open . 2022 ; 4 ( 1 ): 100240 . Crossref PubMed Google Scholar

10. Takahashi T , Muneta T , Tsuji K , Sekiya I . BMP-7 inhibits cartilage degeneration through suppression of inflammation in rat zymosan-induced arthritis . Cell Tissue Res . 2011 ; 344 ( 2 ): 321 332 . Crossref PubMed Google Scholar

11. Deshmukh V , Hu H , Barroga C , et al. A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee . Osteoarthritis Cartilage . 2018 ; 26 ( 1 ): 18 27 . Crossref PubMed Google Scholar

12. Deshmukh V , O’Green AL , Bossard C , et al. Modulation of the Wnt pathway through inhibition of CLK2 and DYRK1A by lorecivivint as a novel, potentially disease-modifying approach for knee osteoarthritis treatment . Osteoarthritis Cartilage . 2019 ; 27 ( 9 ): 1347 1360 . Crossref PubMed Google Scholar

13. Lories RJ , Monteagudo S . Review Article: Is Wnt Signaling an Attractive Target for the Treatment of Osteoarthritis? Rheumatol Ther . 2020 ; 7 ( 2 ): 259 270 . Crossref PubMed Google Scholar

14. Troeberg L , Nagase H . Proteases involved in cartilage matrix degradation in osteoarthritis . Biochim Biophys Acta . 2012 ; 1824 ( 1 ): 133 145 . Crossref PubMed Google Scholar

15. Hamamura K , Nishimura A , Iino T , Takigawa S , Sudo A , Yokota H . Chondroprotective effects of Salubrinal in a mouse model of osteoarthritis . Bone Joint Res . 2015 ; 4 ( 5 ): 84 92 . Crossref PubMed Google Scholar

16. Schon J , Chahla J , Paudel S , et al. Expression profile of matrix metalloproteinases in the labrum of femoroacetabular impingement . Bone Joint Res . 2020 ; 9 ( 4 ): 173 181 . Crossref PubMed Google Scholar

17. Wang M , Sampson ER , Jin H , et al. MMP13 is a critical target gene during the progression of osteoarthritis . Arthritis Res Ther . 2013 ; 15 ( 1 ): R5 . Crossref PubMed Google Scholar

18. Sennett ML , Meloni GR , Farran AJE , Guehring H , Mauck RL , Dodge GR . Sprifermin treatment enhances cartilage integration in an in vitro repair model . J Orthop Res . 2018 ; 36 ( 10 ): 2648 2656 . Crossref PubMed Google Scholar

19. Yan D , Chen D , Cool SM , et al. Fibroblast growth factor receptor 1 is principally responsible for fibroblast growth factor 2-induced catabolic activities in human articular chondrocytes . Arthritis Res Ther . 2011 ; 13 ( 4 ): R130 . Crossref PubMed Google Scholar

20. Xie Y , Zinkle A , Chen L , Mohammadi M . Fibroblast growth factor signalling in osteoarthritis and cartilage repair . Nat Rev Rheumatol . 2020 ; 16 ( 10 ): 547 564 . Crossref PubMed Google Scholar

21. Chen C-H , Kang L , Chang L-H , et al. Intra-articular low-dose parathyroid hormone (1-34) improves mobility and articular cartilage quality in a preclinical age-related knee osteoarthritis model . Bone Joint Res . 2021 ; 10 ( 8 ): 514 525 . Crossref PubMed Google Scholar

22. Shao L-T , Gou Y , Fang J-K , et al. Parathyroid hormone (1-34) ameliorates cartilage degeneration and subchondral bone deterioration in collagenase-induced osteoarthritis model in mice . Bone Joint Res . 2020 ; 9 ( 10 ): 675 688 . Crossref PubMed Google Scholar

23. Yan J , Tian F , Wang W-Y , et al. Parathyroid hormone (1-34) prevents cartilage degradation and preserves subchondral bone micro-architecture in guinea pigs with spontaneous osteoarthritis . Osteoarthritis Cartilage . 2014 ; 22 ( 11 ): 1869 1877 . Crossref PubMed Google Scholar

24. Lu R , Wang Y-G , Qu Y , et al. Dihydrocaffeic acid improves IL-1β-induced inflammation and cartilage degradation via inhibiting NF-κB and MAPK signalling pathways . Bone Joint Res . 2023 ; 12 ( 4 ): 259 273 . Crossref PubMed Google Scholar

25. Zhou Y , Li J , Xu F , Ji E , Wang C , Pan Z . Long noncoding RNA H19 alleviates inflammation in osteoarthritis through interactions between TP53, IL-38, and IL-36 receptor . Bone Joint Res . 2022 ; 11 ( 8 ): 594 607 . Crossref PubMed Google Scholar

26. Luobu Z , Wang L , Jiang D , Liao T , Luobu C , Qunpei L . CircSCAPER contributes to IL-1β-induced osteoarthritis in vitro via miR-140-3p/EZH2 axis . Bone Joint Res . 2022 ; 11 ( 2 ): 61 72 . Crossref PubMed Google Scholar

27. Shreiner AB , Kao JY , Young VB . The gut microbiome in health and in disease . Curr Opin Gastroenterol . 2015 ; 31 ( 1 ): 69 75 . Crossref PubMed Google Scholar

28. Lei M , Guo C , Wang D , Zhang C , Hua L . The effect of probiotic Lactobacillus casei Shirota on knee osteoarthritis: a randomised double-blind, placebo-controlled clinical trial . Benef Microbes . 2017 ; 8 ( 5 ): 697 703 . Crossref PubMed Google Scholar

29. Hao X , Zhang J , Shang X , et al. Exercise modifies the disease-relevant gut microbial shifts in post-traumatic osteoarthritis rats . Bone Joint Res . 2022 ; 11 ( 4 ): 214 225 . Crossref PubMed Google Scholar

30. Zhang F , Rao S , Baranova A . Shared genetic liability between major depressive disorder and osteoarthritis . Bone Joint Res . 2022 ; 11 ( 1 ): 12 22 . Crossref PubMed Google Scholar

31. Jiang L , Zhu X , Rong J , et al. Obesity, osteoarthritis and genetic risk . Bone Joint Res . 2018 ; 7 ( 7 ): 494 500 . Crossref PubMed Google Scholar

32. Mathiessen A , Conaghan PG . Synovitis in osteoarthritis: current understanding with therapeutic implications . Arthritis Res Ther . 2017 ; 19 ( 1 ): 18 . Crossref PubMed Google Scholar

33. Malemud CJ . Anticytokine therapy for osteoarthritis: evidence to date . Drugs Aging . 2010 ; 27 ( 2 ): 95 115 . Crossref PubMed Google Scholar

34. Wang Y , Meng F , Wu J , et al. Associations between adipokines gene polymorphisms and knee osteoarthritis: a meta-analysis . BMC Musculoskelet Disord . 2022 ; 23 ( 1 ): 166 . Crossref Google Scholar

35. Guan Z , Jia J , Zhang C , et al. Gut microbiome dysbiosis alleviates the progression of osteoarthritis in mice . Clin Sci (Lond) . 2020 ; 134 ( 23 ): 3159 3174 . Crossref PubMed Google Scholar

36. Sophocleous A , Azfer A , Huesa C , Stylianou E , Ralston SH . Probiotics Inhibit Cartilage Damage and Progression of Osteoarthritis in Mice . Calcif Tissue Int . 2023 ; 112 ( 1 ): 66 73 . Crossref PubMed Google Scholar

37. Bellamy N , Buchanan WW , Goldsmith CH , Campbell J , Stitt LW . Validation study of WOMAC: a health status instrument for measuring clinically important patient relevant outcomes to antirheumatic drug therapy in patients with osteoarthritis of the hip or knee . J Rheumatol . 1988 ; 15 ( 12 ): 1833 1840 . PubMed Google Scholar

38. Castañeda S , Roman-Blas JA , Largo R , Herrero-Beaumont G . Subchondral bone as a key target for osteoarthritis treatment . Biochem Pharmacol . 2012 ; 83 ( 3 ): 315 323 . Crossref PubMed Google Scholar

39. Goldring SR . The role of bone in osteoarthritis pathogenesis . Rheum Dis Clin North Am . 2008 ; 34 ( 3 ): 561 571 . Crossref PubMed Google Scholar

40. Oo WM , Yu SP-C , Daniel MS , Hunter DJ . Disease-modifying drugs in osteoarthritis: current understanding and future therapeutics . Expert Opin Emerg Drugs . 2018 ; 23 ( 4 ): 331 347 . Crossref PubMed Google Scholar

41. Neogi T . Clinical significance of bone changes in osteoarthritis . Ther Adv Musculoskelet Dis . 2012 ; 4 ( 4 ): 259 267 . Crossref PubMed Google Scholar

42. No authors listed . Forteo approved for osteoporosis treatment . FDA Consum . 2003 ; 37 ( 2 ): 4 . PubMed Google Scholar

43. Mo H , Wang Z , He Z , et al. Decreased Peli1 expression attenuates osteoarthritis by protecting chondrocytes and inhibiting M1-polarization of macrophages . Bone Joint Res . 2023 ; 12 ( 2 ): 121 132 . Crossref PubMed Google Scholar

44. Oliveira MM , Ratti BA , Daré RG , et al. Dihydrocaffeic Acid Prevents UVB-Induced Oxidative Stress Leading to the Inhibition of Apoptosis and MMP-1 Expression via p38 Signaling Pathway . Oxid Med Cell Longev . 2019 ; 2019 : 2419096 . Crossref PubMed Google Scholar

Author contributions

N. S. Makaram: Writing – original draft, Writing – review & editing.

A. H. R. W. Simpson: Writing – original draft, Writing – review & editing.

Funding statement

The authors received no financial or material support for the research, authorship, and/or publication of this article.

Twitter

Follow N. S. Makaram @makortho

© 2023 Author(s) et al. This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives (CC BY-NC-ND 4.0) licence, which permits the copying and redistribution of the work only, and provided the original author and source are credited. See https://creativecommons.org/licenses/by-nc-nd/4.0/