Personalised Medicine and Disease Modification in Osteoarthritis: On our journey but still a way to go!

By Alan Reynolds and Nicholas Larkins

Introduction and Background

Osteoarthritis (OA) is the most prevalent joint disease, estimated to affect 250 million people worldwide with prevalence increasing due to aging populations and growing levels of obesity.1 The estimated global incident cases from the 2017 Global Burden of Diseases (GBD) study were almost 15 million with an age standardised incidence of 181.19 per 100,000, which was highest in the USA at 316.87 per 100,000.2

OA causes a substantial burden of disability, ranking third behind Alcohol Use and Unipolar Major Depression for Years Lost to Disability (YLDs) in the USA in 1996 totalling 434,000 YLDs, 5.9% of the total.3 Based on the 2010 GBD study, the combined incident case total of hip and knee OA was ranked as the 11th highest contributor to global disability and 38th highest in Disability Adjusted Life Years.4 Mobility disability due to OA is greater than any other disease and OA of the knee contributes 83% of the total burden attributed to OA.1 The quality of life (QoL) of patients with OA is substantially reduced, not only with respect to physical function but across other domains, including mental health.5-7

The socioeconomic impact is significant. In the USA, annual total costs for 2013 were estimated at $139.8 – $161.8 billion (direct $115.3 billion and indirect $11.6 – $13.0 billion).8 The social cost of OA could be as high as 2.5%9 of Gross Domestic Product although a recent systematic review reported values between 0.25 and 0.5%.10 A study from Sweden found that patients with knee OA have double the risk of sick leave with 2% of all days off work resulting from the disease and an increased risk of receiving a disability pension.11 In a survey of five European countries involving 3750 subjects, the mean SF-12 score* was 40.53 (perfect health =100] and 21.5% reported depression. Among the one third who were employed, 7% reported absenteeism and 24% presenteeism.12

*SF-12 is a self-reported short form outcome measure assessing the impact of health on an individual’s everyday life. It is often used as a quality-of-life measure.

The economic and QoL impact of OA led the Osteoarthritis Research Society International (OARSI) to submit a white paper to the US Food and Drug Administration (FDA) in 2016, “Osteoarthritis: A Serious Disease”,13 which resulted in the publication of draft guidance in 2018 that  stated the “FDA recognizes that OA can be a serious disease with an unmet medical need for therapies that modify the underlying pathophysiology of the disease and potentially change its natural course to prevent long-term disability.”14

In this article we will discuss the challenges in demonstrating disease modification in OA, illustrate this with examples of recently published clinical trials, and then describe the current status of APPA, a novel, patented potential Disease Modifying Osteoarthritic Drug (DMOAD). APPA is an oral combination of two synthetically produced isomers, originally of plant origin: 4-hydroxy-3-methoxyacetophenone (apocynin) and 2-hydroxy-4-methoxyacetophenone (paeonol).

Pathophysiology of OA

OA is a disease of diarthrodial joints and had historically been considered to result from ‘wear and tear,’ leading to cartilage loss and reduction in the joint space resulting in pain and damage to the bone. Research over the last decades has established that OA involves the whole joint with not only loss of cartilage, but also changes in the subchondral bone, synovium, tendons, ligaments and muscles. It is now recognised that the disease process is more complex than originally assumed and, to a lesser or greater degree, involves chronic inflammation,15 mainly involving the innate immune system,16 triggered for example, by aging or obesity.17,18  Ageing of itself has important consequences with regard to OA, including low grade12 inflammation (inflamm-aging), mitochondrial dysfunction with oxidative stress, and cell senescence.19,20 It has now been established, however, that OA is a heterogenous disease with several different phenotypes and possible triggers that lead to a common final pathway resulting in jointdestruction.21-26 The roles of bone, cartilage, and synovium in OA with cross talk between them involves many different pathways and provides numerous potential treatment targets.27-29

Current Treatment of OA

Despite the importance of OA on patient QoL, the human cost of pain and disability caused, and economic costs, treatment options are limited. Unlike rheumatoid arthritis (RA), no interventions have been approved that alter the course of the disease – a pharmacopeia of DMOAD as the holy grail of OA treatment.

The primary reason subjects visit physicians with OA is because of pain. Recommendations for treatment pathways are available in a number of publications,30-33 most of which start with education, self-management and lifestyle interventions such as weight loss and increased exercise. Studies of pharmacological interventions most often involve the knee rather than other sites of OA and have been of limited duration focussing on pain as the primary outcome measure. It must be noted, however, that many subjects have multiple joint involvement and that the pain is chronic.

Oral analgesics such as acetaminophen (paracetamol) and non-steroidal anti-inflammatory drugs (NSAIDs) are widely recommended but use of NSAIDs is limited due to toxicity34,35 and should be used at the lowest dose and the shortest possible time.33 Recent evidence however has questioned the efficacy of acetaminophen in OA.36-38 Topical NSAIDs are recommended as effective but without the safety concerns associated with the oral forms. In the event that pain relief is insufficient then opioids are an option with the associated concerns over long term use.

A number of injectable treatments are included (for example, corticosteroids) although there is evidence that repeated injections of corticosteroids may be associated with additional joint damage.39 These restricted treatment options and their limited effectiveness result in inadequate pain relief with associated impact on function.40 Intractable pain, loss of function and joint damage are criteria for surgical joint replacement which is frequently highly successful.34

Notwithstanding these interventions, there remains a high unmet need for effective analgesics for patients with osteoarthritis and treatments that slow, halt, or even reverse the slowly progressing joint damage.

Challenges for DMOAD clinical trials

The hope that drugs in development would achieve DMOAD status was initially discussed over 10 years ago,41,42however we seem still a long way  from achieving that goal. Many challenges exist in the design and conduct of OA clinical trials43 particularly those attempting to identify DMOAD status.

Following submission of the OARSI white paper to the FDA in 2016,13 the Agency published draft industry guidance on structural endpoints for OA.14 The document laid out the considerations for approval of a drug as a DMOAD mainly for scenarios where an intervention has beneficial effects on a surrogate end point such as a soluble biomarker or structure that in the longer term is associated with clinical benefits such as reduction in pain, improvement in function, or delay or avoidance of joint replacement.

FDA concluded, however, “At this time, the ability of treatment effects on common measures of structural progression to reliably predict treatment effects on direct measures of how patients function and feel, has not been established.” Many challenges remain in design and implementation of such trials, not least being the different endotypes and phenotypes that have been identified, validation of the imaging endpoints, qualification of soluble biomarkers, and high pain placebo response rates.43 Several publications have discussed possible trial options, but much work remains to be done to optimise designs.44,45

Surrogate Endpoints and Biomarkers

Most of the current potential DMOADs do not have direct effects on clinical outcomes (pain, function, joint replacement). As outlined above, in order to receive marketing approval, studies will need to establish the interventions have beneficial effects on surrogate endpoints (e.g., imaging) or biomarkers which have been shown to predict clinical outcomes in the longer term.

The imaging standard in OA clinical trials has been radiographically measured minimum joint space width (mJSW), which equates to tibiofemoral cartilage loss. This has a number of drawbacks, including the contribution of other structures to changes in JSW as well as issues with alignment, positioning and sensitivity to change in a disease that progresses slowly in the majority of patients. In a large 1683 patient study treated with sodium ranelate, improvements were seen in both Joint Space Width (JSW) and Western Ontario and McMaster Universities Osteoarthritis (WOMAC) scores,46 but concordance was not evaluated at the individual patient level. In 2015 OARSI published recommendations for use of radiography and Magnetic Resonance Imaging (MRI) in clinical trials to set standards and improve quality.47 Recent research has highlighted a range of different MRI approaches that are claimed to have greater sensitivity which are able to assess changes in overall joint structure48,49 and correlate with risk of joint replacement,50 although more data are needed to gain acceptance by regulatory authorities as a suitable surrogate endpoint.

In OA, biomarkers have a number of potential roles which include stratification of patient subgroups, measurement of disease activity and response to treatment. This would also involve identification of patients who will likely show progression of disease assessed by imaging, which provides potential surrogate endpoints and has the potential to identify the most appropriate treatment for an individual (i.e., personalised medicine, which Is still an aspirational goal).

The landscape of biomarkers has been reviewed recently51-53 and a number of important observations can be made. Firstly, none of the biomarkers to date have been approved for use by a regulator. However, serum cartilage-oligomeric matrix protein (sCOMP), urine Carboxy-terminal telopeptide fragments of type II collagen (uCTX-II), and MMP-generated fragment of C-reactive protein (CRPM) levels show promise as predictors of disease progression.54-56

Such promise is dampened by a study utilizing the Osteoarthritis Initiative Biomarkers cohort that reported that systemic biomarkers of bone turnover had only had weak associations with bone features.57 A proposed model from a recent analysis of two phase 3 clinical trials with salmon calcitonin combines baseline age, sex, BMI, u-CTX-II and X-ray KL-grade to predict total joint replacement (TJR) during a two-year period with an AUC of 0.75 (95% CI: 0.72-0.77).58 These approaches could improve patient selection for clinical trials of potential DMOADs.

Current DMOAD candidates

The increasing understanding of the complex pathways involved in OA has resulted in novel potential therapeutic targets being proposed with development of interventions designed to modify disease progression.59-63 These may be broadly classified into agents that:

  • target cartilage turnover;
  • target subchondral bone metabolism; or
  • inhibit the inflammatory pathways that drive cartilage and bone 59,61

Table 1 summarises drugs that have recently completed or are currently in Phase 2 clinical trials as potential DMOADs. We will discuss four examples of agents that have reported results from these early clinical trials that illustrate the challenges faced with OA clinical trials and in demonstrating disease modification.

Lorecivivint

Lorecivivint (SM04690), a small molecule, is a Wnt pathway modulator acting via CDC-like kinase 2 (CLK2) and dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) leading to reduced STAT3 and NF-κB signalling, resulting in reduced in matrix metalloproteinase activity and reduced inflammatory cytokine production.64,65

A small Phase 1 study in subjects with knee OA showed potential analgesic and DMOAD effects.66 In the Phase 2A study however, there was no significant difference in improvement in WOMAC pain score when compared with placebo at week 13, and no effects on mJSW at 52 weeks were seen.67 Results from the biomarkers ß-CTX, PINP and COMP were inconclusive. Post hoc analyses however, demonstrated significant improvements in pain scores in all dose groups in subjects with unilateral OA and those with unilateral involvement and absence of widespread pain. Unilateral knee OA subjects treated with the 0.07 mg dose had significantly increased mJSW compared to placebo. Importantly there was concordance between WOMAC pain and mJSW measurements.

A Phase 2B 24-week study enrolled patients with unilateral knee OA. The primary endpoint, change in pain Numerical Rating Scale (NRS), was achieved by the 0.07 and 0.23 mg dose groups but no dose group demonstrated benefit on mJSW.68 A post hoc analysis showed that subjects with a baseline mJSW of 2-4mm had increased sensitivity to radiographic change.69

The variable results seen between the studies clearly illustrate the challenges faced in studies evaluating potential DMOADs. Post hoc analyses have been used, however, to inform the design of the Phase 3 studies, which started enrolling subjects in June 2019.70 The completed Phase 2 studies discussed above did not include biomarkers in the information posted on ClinTrials.gov. However, a recently completed (but not reported) Phase 2 study (NCT03727022) with a primary  endpoint of change in bone mineral density does include the serum bone turnover biomarkers and cartilage metabolism.71

Sprifermin

Sprifermin, a recombinant form of truncated human fibroblast growth factor 18, is administered by intra-articular injection that induces expansion of hyaline cartilage producing chondrocytes72 and remodelling of cartilage in human OA explants.73

Two Phase 1 studies have been completed: the first study being a single ascending dose study (SADS) followed by the second study, a multiple ascending dose (MAD) safety study that showed no evidence of any safety issues.74 A Phase 1B Proof of Concept study compared 3 doses of sprifermin with placebo in a SAD/MAD design. The MAD portion evaluated 3 injections one week apart with a second round of 3 injections 12 weeks after the first injection. The study did not achieve the primary endpoint, quantitative magnetic resonance imaging (qMRI) change in central medial femorotibial compartment cartilage thickness. Evidence of benefit was reported though on prespecified secondary qMRI endpoints. Subjects receiving sprifermin reported less improvement in WOMAC pain than those given placebo, which was significant for the highest dose group, 100µg.75

The Phase 2 study compared two doses of sprifermin, administered every 6 or 12 months, with placebo in patients with symptomatic OA. Injections were given as weekly injections over a 3-week period. The primary end point was change in qMRI total femorotibial joint cartilage thickness after 2 years compared with placebo which was achieved by both 100μg doses but not the 30μg doses. Dose dependant increases in cartilage thickness and volume were also reported. None of the 4 sprifermin doses achieved significant differences from placebo for WOMAC total scores or subscale scores although all groups showed improvements from baseline.76 At the Year 3 follow- up, 18 months after the last injection however, the WOMAC pain scores versus placebo were significant.77

Synovial fluid biomarkers of cartilage metabolism, PRO-C2 (type II collagen formation), huARGS (aggrecan degradation), and FBN-C (fibronectin degradation), were evaluated in a subset of subjects from the Phase 2 study and showed a phasic pattern. There was an overall increase in ARGS over time in the sprifermin group but after each cycle of injections levels decreased, whereas ARGS simply decreased over time in the placebo group. There was an initial decrease in collagen formation measured by PRO-C2 after sprifermin injections, followed by increases, so that at the end of follow-up, levels had increased from baseline, whereas no change was seen over time in the placebo group. Similar results were seen with fibronectin.78 These temporal changes seen with the biomarkers suggest that with respect to sprifermin, these biomarkers may not be ideal for assessing response to treatment or acting as surrogates for clinical response.

MV-711

Cathepsin K is a protease found in osteoclasts and is involved in bone resorption through cleavage of type I collagen. It is also expressed in synovium and chondrocytes and causes degradation of type II collagen in cartilage.79 MV-711 is an oral, highly selective inhibitor of cathepsin K and has been shown to reduce bone and cartilage degradation dose-dependently in monkeys and humans80 and to reduce bone and cartilage loss in animal models of OA.81

The Phase 2A study compared 200 mg/day and 100 mg/day with placebo.82 The primary endpoint was change from baseline in NRS pain score for the target knee over the 26-week study period. In both MIV-711 treatment and placebo groups, pain scores reduced from baseline but there were no differences between the groups.

The key secondary imaging outcomes were assessed by MRI. Both doses of MIV-711 significantly reduced medial femoral bone area progression and medial femoral cartilage thinning versus placebo. Levels of biomarkers of bone resorption (CTX-I) and cartilage degradation (CTX-II) were reduced by both doses compared with placebo. Although MIV-711 clearly had effects on bone and cartilage seen by imaging and biomarker analysis over 26 weeks there was no symptomatic benefit. On the other hand, data from the Osteoarthritis Initiative found that changes in bone structure over 2 years do not translate into pain worsening until 4 years,83 so the study may have been too short to detect symptomatic benefit.

APPA

APPA is being developed by AKL Research and Development Ltd for treatment of  osteoarthritis in association with NBCD A/S (previously Nordic Bioscience Clinical Development).  Recruitment of subjects into the Phase 2 clinical trial (NCT04657926) began in September 2020.

In vitro studies with activated human neutrophils have found that APPA does not interfere with neutrophil host defence against infections but does inhibit neutrophil degranulation and production of neutrophil extracellular traps.84 APPA also down-regulated TNFα-stimulated NF-κB gene expression, but up-regulated expression of Nrf2, transcription factors involved in control of inflammation and response to redox stress, both of which (Nrf2 and NF- κB) interact with each other.85 In addition, both have been identified as potential targets in OA.86,87

Research utilising ex vivo tissue explants conducted by Nordic Biosciences A/S** found that APPA reduces inflammation-derived tissue turnover in human cartilage explants and inhibits RANKL-mediated osteoclastogenesis and bone resorption by human osteoclasts. Studies** using human chondrocytes undertaken by Instituto de Investigación Biomedica da Coruña reported that APPA significantly reduced the gene expression induced by IL-1β of IL-8, TNF-α, MMP-13 and MMP-3. In addition, in experiments with human cartilage explants stimulated with IL-1β, APPA significantly increased levels of proteoglycans in the intermedial layer. The results from these sets of experiments suggest that APPA affects a number of processes in joint tissues that contribute to damage in patients with OA. This conclusion is supported by results from the rat meniscal tear model of OA.  APPA treatment reduced modification of the OARSI Total Joint Score88 by 21% compared to animals that received vehicle alone.89

**Submitted to the 2021 OARSI meeting. 

In a cross-over study in dogs with naturally occurring OA that compared APPA with underwater treadmill therapy and massage, APPA reduced pain assessed by force plate measurements and normalised gait symmetry.90 A second 28-day dog study compared APPA with meloxicam plus famotidine and placebo. Both active treatment groups had significant improvements from baseline for orthopaedic score. The APPA group had significantly better scores for lameness at the walk and lameness at trot compared with the placebo group.91

The effects of APPA in a case series of human subjects with OA was recently reported in abstract form. Twenty-three subjects with a diagnosis of OA of whom 7 were scheduled for surgery have been treated with APPA. Treatment was deemed effective in 19 and, in the 16 subjects where information was available, duration of treatment ranged from 9 to 120 (median 24) months and was well tolerated.92

A Phase 2 study comparing APPA to placebo in 150 patients with knee OA has recently started. In view of the data from the animal studies and the human case series, the primary endpoint is reduction in pain over 28 days. Evidence to support potential DMOAD effects, as demonstrated in the rat meniscal tear model of OA, will be determined with biomarkers. Information from the ex vivo experiments has been evaluated to select the most appropriate biomarkers that reflect the pathways impacted by APPA, cartilage degradation by aggrecanase and MMPs, and bone turnover.

This approach differs from the examples discussed above as there is evidence that the effect on symptoms occurs early in treatment and is not dependent on structural effects. In this respect APPA is more like biological treatments for RA than other potential DMOADs currently in clinical trials.

Discussion and Conclusions

Addressing the substantial unmet needs in the treatment of OA, both for reduction of pain and for reducing the progression of disease starts with an understanding of the nature of OA as a heterogenous disease with a number of phenotypes with  different pathways to joint damage. Personalised medicine has long been an aspirational goal of OA treatment93-94 and the scientific and clinical community and pharmaceutical companies have devoted much time, effort and money to achieve that target, but many obstacles remain.

OA is usually a slowly progressing disease so demonstrating reduction in the rate of damage is challenging. These difficulties have been reviewed recently43 and proposals for study designs in the light of the FDA draft guidance have been published.44,45 A premise of current investigational DMOADs is that effects on structural changes will lead to improvements in clinical endpoints  such as pain and function. This assumption has been challenged recently95 as analysis of data from the Osteoarthritis Initiative led to the conclusion that cartilage loss resulted in limited worsening of pain.

The difficulties in assessing and demonstrating disease modification has been illustrated by the 3 examples with published results as discussed above. We look forward to resolving these difficulties  eventually through additional research studies enriched with OA subjects whose disease is likely to progress and to explore the correlations between biomarkers, surrogate endpoints and clinical outcomes.

References

  1. Hunter DJ, Bierma-Zeinstra Osteoarthritis. Lancet (London, England). 2019;393(10182):1745-59.
  2. Jin Z, Wang D, Zhang H, Liang J, Feng X, Zhao J, et Incidence trend of five common musculoskeletal disorders from 1990 to 2017 at the global, regional and national level: results from the global burden of disease study 2017. Annals of the rheumatic diseases. 2020;79(8):1014-22.
  3. McKenna MT, Michaud CM, Murray CJ, Marks Assessing the burden of disease in the United States using disability-adjusted life years. Am J Prev Med. 2005;28(5):415-23.
  4. Cross M, Smith E, Hoy D, Nolte S, Ackerman I, Fransen M, et The global burden of hip and knee osteoarthritis: estimates from the Global Burden of Disease 2010 study. Annals of the rheumatic diseases. 2014;73(7):1323-30.
  5. Vitaloni M, Botto-van Bemden A, Sciortino Contreras RM, Scotton D, Bibas M, Quintero M, et Global management of patients with knee osteoarthritis begins with quality of life assessment: a systematic review. BMC Musculoskelet Disord. 2019;20(1):493.
  6. Xie F, Kovic B, Jin X, He X, Wang M, Silvestre Economic and Humanistic Burden of Osteoarthritis: A Systematic Review of Large Sample Studies. Pharmacoeconomics. 2016;34(11):1087-100.
  7. Zhao X, Shah D, Gandhi K, Wei W, Dwibedi N, Webster L, et Clinical, humanistic, and economic burden of osteoarthritis among noninstitutionalized adults in the United States. Osteoarthritis Cartilage. 2019;27(11):1618-26.
  8. Ma VY, Chan L, Carruthers KJ. Incidence, prevalence, costs, and impact on disability of common conditions requiring rehabilitation in the United States: stroke, spinal cord injury, traumatic brain injury, multiple sclerosis, osteoarthritis, rheumatoid arthritis, limb loss, and back pain. Arch Phys Med Rehabil. 2014;95(5):986-95 e1.March LM, Bachmeier CJ. Economics of osteoarthritis: a global perspective. Baillieres Clin Rheumatol. 1997;11(4):817-34.
  9. March LM, Bachmeier CJ. Economics of osteoarthritis: a global perspective. Baillieres Clin Rheumatol. 1997;11(4):817-34.
  10. Puig-Junoy J, Ruiz Zamora Socio-economic costs of osteoarthritis: a systematic review of cost-of-illness studies. Semin Arthritis Rheum. 2015;44(5):531-41.
  11. Hubertsson J, Petersson IF, Thorstensson CA, Englund Risk of sick leave and disability pension in working-age women and men with knee osteoarthritis. Annals of the rheumatic diseases. 2013;72(3):401-5.
  12. Kingsbury SR, Gross HJ, Isherwood G, Conaghan Osteoarthritis in Europe: impact on health status, work productivity and use of pharmacotherapies in five European countries. Rheumatology (Oxford, England). 2014;53(5):937-47.
  13. Osteoarthritis: A Serious Disease, Submitted to the U.S. Food and Drug Administration December 1, 2016. https://wwwoarsiorg/sites/default/files/library/2018/pdf/oarsi_white_paper_oa_serious_disease121416_1 pdf 2016;Accessed 12 June 2019.
  14. Osteoarthritis: Structural Endpoints for the Development of Drugs, Devices, and Biological Products for Treatment. Guidance for Industry. DRAFT GUIDANCE. 2018.
  15. Berenbaum Osteoarthritis as an inflammatory disease (osteoarthritis is not osteoarthrosis!). Osteoarthritis Cartilage. 2013;21(1):16-21.
  16. Robinson WH, Lepus CM, Wang Q, Raghu H, Mao R, Lindstrom TM, et Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nature reviews Rheumatology. 2016;12(10):580-92.
  17. Abramoff B, Caldera Osteoarthritis: Pathology, Diagnosis, and Treatment Options. Med Clin North Am. 2020;104(2):293-311.
  18. Mora JC, Przkora R, Cruz-Almeida Knee osteoarthritis: pathophysiology and current treatment modalities. Journal of pain research. 2018;11:2189-96.
  19. Loeser RF, Collins JA, Diekman Ageing and the pathogenesis of osteoarthritis. Nature reviews Rheumatology. 2016;12(7):412-20.
  20. Toh WS, Brittberg M, Farr J, Foldager CB, Gomoll AH, Hui JH, et Cellular senescence in aging and osteoarthritis. Acta Orthop. 2016;87(sup363):6-14.
  21. Berenbaum F, Walker Osteoarthritis and inflammation: a serious disease with overlapping phenotypic patterns. Postgraduate Medicine. 2020;132(4):377-84.
  22. Dell’Isola A, Steultjens Classification of patients with knee osteoarthritis in clinical phenotypes: Data from the osteoarthritis initiative. PloS one. 2018;13(1):e0191045.
  23. Deveza LA, Melo L, Yamato TP, Mills K, Ravi V, Hunter Knee osteoarthritis phenotypes and their relevance for outcomes: a systematic review. Osteoarthritis Cartilage. 2017;25(12):1926-41.
  24. Mobasheri A, van Spil WE, Budd E, Uzieliene I, Bernotiene E, Bay-Jensen AC, et al. Molecular taxonomy of osteoarthritis for patient stratification, disease management and drug development: biochemical markers associated with emerging clinical phenotypes and molecular endotypes. Current opinion in rheumatology. 2019;31(1):80-9.
  25. Nelson The Value of Phenotypes in Knee Osteoarthritis Research. Open Orthop J. 2018;12:105-14.
  26. van der Esch M, Knoop J, van der Leeden M, Roorda LD, Lems WF, Knol DL, et al. Clinical phenotypes in patients with knee osteoarthritis: a study in the Amsterdam osteoarthritis cohort. Osteoarthritis Cartilage. 2015;23(4):544-9.
  27. Goldring MB, Berenbaum Emerging targets in osteoarthritis therapy. Curr Opin Pharmacol. 2015;22:51-63.
  28. Goldring SR, Goldring Changes in the osteochondral unit during osteoarthritis: structure, function and cartilage-bone crosstalk. Nature reviews Rheumatology. 2016;12(11):632-44.
  29. Mathiessen A, Conaghan Synovitis in osteoarthritis: current understanding with therapeutic implications. Arthritis Res Ther. 2017;19(1):18.
  30. Bannuru RR, Osani MC, Vaysbrot EE, Arden NK, Bennell K, Bierma-Zeinstra SMA, et OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthritis Cartilage. 2019;27(11):1578-89.
  31. Geenen R, Overman CL, Christensen R, Asenlof P, Capela S, Huisinga KL, et EULAR recommendations for the health professional’s approach to pain management in inflammatory arthritis and osteoarthritis. Annals of the rheumatic diseases. 2018;77(6):797-807.
  32. Kolasinski SL, Neogi T, Hochberg MC, Oatis C, Guyatt G, Block J, et 2019 American College of Rheumatology/Arthritis Foundation Guideline for the Management of Osteoarthritis of the Hand, Hip, and Knee. Arthritis & rheumatology (Hoboken, NJ). 2020;72(2):220- 33.
  33. National Institute for Health and Clinical Osteoarthritis: care and management. CG177. 2014;nice.org.uk/guidance/cg177.
  34. Charlesworth J, Fitzpatrick J, Perera NKP, Orchard Osteoarthritis- a systematic review of long-term safety implications for osteoarthritis of the knee. BMC Musculoskelet Disord. 2019;20(1):151.
  35. Curtis E, Fuggle N, Shaw S, Spooner L, Ntani G, Parsons C, et Safety of Cyclooxygenase-2 Inhibitors in Osteoarthritis: Outcomes of a Systematic Review and Meta-Analysis. Drugs Aging. 2019;36(Suppl 1):25-44.
  36. Bannuru RR, Schmid CH, Kent DM, Vaysbrot EE, Wong JB, McAlindon Comparative effectiveness of pharmacologic interventions for knee osteoarthritis: a systematic review and network meta-analysis. Ann Intern Med. 2015;162(1):46-54.
  37. Ennis ZN, Dideriksen D, Vaegter HB, Handberg G, Pottegård Acetaminophen for Chronic Pain: A Systematic Review on Efficacy. Basic Clin Pharmacol Toxicol. 2016;118(3):184-9.
  38. Machado GC, Maher CG, Ferreira PH, Pinheiro MB, Lin CW, Day RO, et Efficacy and safety of paracetamol for spinal pain and osteoarthritis: systematic review and meta-analysis of randomised placebo controlled trials. Bmj. 2015;350:h1225.
  39. Ayub S, Kaur J, Hui M, Hall M, Doherty M, Zhang Efficacy and Safety of Multiple Intra-articular Corticosteroid Injections for Osteoarthritis – a Systematic Review and Meta-analysis of Randomised Controlled Trials and Observational Studies. Arthritis and Rheumatology. 2020;72 (Suppl 10) Abstract 1651.
  40. Conaghan PG, Peloso PM, Everett SV, Rajagopalan S, Black CM, Mavros P, et Inadequate pain relief and large functional loss among patients with knee osteoarthritis: evidence from a prospective multinational longitudinal study of osteoarthritis real- world therapies. Rheumatology (Oxford, England). 2015;54(2):270- 7.
  41. Hunter Pharmacologic therapy for osteoarthritis–the era of disease modification. Nature reviews Rheumatology. 2011;7(1):13- 22.
  42. Qvist P, Bay-Jensen AC, Christiansen C, Dam EB, Pastoureau P, Karsdal The disease modifying osteoarthritis drug (DMOAD): Is it in the horizon? Pharmacol Res. 2008;58(1):1-7.
  43. Felson DT, Neogi Emerging Treatment Models in Rheumatology: Challenges for Osteoarthritis Trials. Arthritis & rheumatology (Hoboken, NJ). 2018;70(8):1175-81.
  44. Katz JN, Neogi T, Callahan JF, Block JA, Conaghan PG, Simon LS, et Disease modification in osteoarthritis; pathways to drug approval. Osteoarthritis and Cartilage Open. 2020;2:100059.
  45. Kraus VB, Simon LS, Katz JN, Neogi T, Hunter D, Guermazi A, et Proposed study designs for approval based on a surrogate endpoint and a post-marketing confirmatory study under FDA’s accelerated approval regulations for disease modifying osteoarthritis drugs. Osteoarthritis Cartilage. 2019;27(4):571-9.
  46. Reginster J-Y, Badurski J, Bellamy N, Bensen W, Chapurlat R, Chevalier X, et Efficacy and safety of strontium ranelate in the treatment of knee osteoarthritis: results of a double-blind, randomised placebo-controlled trial. Annals of the rheumatic diseases. 2013;72(2):179-86.
  47. Hunter DJ, Altman RD, Cicuttini F, Crema MD, Duryea J, Eckstein F, et OARSI Clinical Trials Recommendations: Knee imaging in clinical trials in osteoarthritis. Osteoarthritis Cartilage. 2015;23(5):698-715.
  48. Hayashi D, Roemer FW, Guermazi Magnetic resonance imaging assessment of knee osteoarthritis: current and developing new concepts and techniques. Clinical and experimental rheumatology. 2019;37 Suppl 120(5):88-95.
  49. Oo WM, Linklater JM, Hunter Imaging in knee osteoarthritis. Current opinion in rheumatology. 2017;29(1):86-95.
  50. Hafezi-Nejad N, Guermazi A, Demehri S, Roemer New imaging modalities to predict and evaluate osteoarthritis progression. Best practice & research Clinical rheumatology. 2017;31(5):688-704.
  51. Bay-Jensen AC, Thudium CS, Gualillo O, Mobasheri Biochemical marker discovery, testing and evaluation for facilitating OA drug discovery and development. Drug Discov Today. 2018;23(2):349-58.
  52. Bay-Jensen AC, Thudium CS, Mobasheri Development and use of biochemical markers in osteoarthritis: current update. Current opinion in rheumatology. 2018;30(1):121-8.
  53. Kraus VB, Karsdal Osteoarthritis: Current Molecular Biomarkers and the Way Forward. Calcif Tissue Int. 2020; https://doi. org/10.1007/s00223-020-00701-7.
  54. Hao HQ, Zhang JF, He QQ, Wang Cartilage oligomeric matrix protein, C-terminal cross-linking telopeptide of type II collagen, and matrix metalloproteinase-3 as biomarkers for knee and hip osteoarthritis (OA) diagnosis: a systematic review and meta-analysis. Osteoarthritis Cartilage. 2019;27(5):726-36.
  55. Kluzek S, Bay-Jensen AC, Judge A, Karsdal MA, Shorthose M, Spector T, et Serum cartilage oligomeric matrix protein and development of radiographic and painful knee osteoarthritis. A community-based cohort of middle-aged women. Biomarkers. 2015;20(8):557-64.
  56. Saberi Hosnijeh F, Siebuhr AS, Uitterlinden AG, Oei EH, Hofman A, Karsdal MA, et Association between biomarkers of tissue inflammation and progression of osteoarthritis: evidence from the Rotterdam study cohort. Arthritis Res Ther. 2016;18:81.
  57. Deveza LA, Kraus VB, Collins JE, Guermazi A, Roemer FW, Bowes M, et Association Between Biochemical Markers of Bone Turnover and Bone Changes on Imaging: Data From the Osteoarthritis Initiative. Arthritis Care Res (Hoboken). 2017;69(8):1179-91.
  58. Bihlet AR, Bjerre-Bastos JJ, Andersen JR, Byrjalsen I, Karsdal MA, Bay-Jensen Clinical and biochemical factors associated with risk of total joint replacement and radiographic progression in osteoarthritis: Data from two phase III clinical trials. Semin Arthritis Rheum. 2020.
  59. Alcaraz MJ, Guillen MI, Ferrandiz Emerging therapeutic agents in osteoarthritis. Biochem Pharmacol. 2019.
  60. Ghouri A, Conaghan Prospects for Therapies in Osteoarthritis. Calcif Tissue Int. 2020.
  61. Oo WM, Yu SP, Daniel MS, Hunter Disease-modifying drugs in osteoarthritis: current understanding and future therapeutics. Expert opinion on emerging drugs. 2018;23(4):331-47.
  62. Sun MM, Beier F, Pest Recent developments in emerging therapeutic targets of osteoarthritis. Current opinion in rheumatology. 2017;29(1):96-102.
  63. Van Spil WE, Kubassova O, Boesen M, Bay-Jensen AC, Mobasheri Osteoarthritis phenotypes and novel therapeutic targets. Biochem Pharmacol. 2019.
  64. Deshmukh V, Hu H, Barroga C, Bossard C, Kc S, Dellamary L, et 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.
  65. Deshmukh V, O’Green AL, Bossard C, Seo T, Lamangan L, Ibanez M, 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 and Cartilage. 2019;27(9):1347-60.
  66. Yazici Y, McAlindon TE, Fleischmann R, Gibofsky A, Lane NE, Kivitz AJ, et A novel Wnt pathway inhibitor, SM04690, for the treatment of moderate to severe osteoarthritis of the knee: results  of a 24-week, randomized, controlled, phase 1 study. Osteoarthritis Cartilage. 2017;25(10):1598-606.
  67. Yazici Y, McAlindon TE, Gibofsky A, Lane NE, Clauw D, Jones M, et Lorecivivint, a Novel Intraarticular CDC-Like Kinase 2 and Dual-Specificity Tyrosine Phosphorylation-Regulated Kinase 1A Inhibitor and Wnt Pathway Modulator for the Treatment of Knee Osteoarthritis: A Phase II Randomized Trial. Arthritis & rheumatology (Hoboken, NJ). 2020;72:1694 – 706.
  68. Yazici Y, Mcalindon T, Gibofsky A, Lane N, Lattermann C, Skrepnik N, et Efficacy and safety from a phase 2B trial of SM04690, a novel intra-articular Wnt pathway inhibitor for the treatment of osteoarthritis of the knee. Annals of the rheumatic diseases. 2019;78(Suppl 2):519 Abstract THU0458.
  69. Kennedy S, Swearingen C, Tambiah J, Conaghan Optimizing subject selection in knee osteoarthritis clinical trials by radiographic joint space width: Post-hoc clinical response analysis from a phase 2B trial of Wnt pathway inhibitor, SM04690. Annals of the rheumatic diseases. 2019;78(Suppl 2):144 Abstract OP0137
  70. Samumed Doses First Subject in Phase 3 STRIDES-X-ray Trial of Lorecivivint for the Treatment of Knee Osteoarthritis 2019 [cited Accessed 2 December 2020,]; Available from: https://www.samumed.com/medium/image/samumed-doses-first-subject-in-phase-3-strides-x-ray-trial-of-lorecivivint-for-the-treatment-of-knee-osteoarthritis_591/view.aspx
  71. gov. NCT03727022 A Study Evaluating the Safety, Tolerability and Efficacy of Two Injections of SM04690 for the Treatment of Moderately to Severely Symptomatic Knee Osteoarthritis. [cited 2 December 2020]; Available from: https://clinicaltrials.gov/ct2/show/NCT03727022?term=samumed
  72. Gigout A, Guehring H, Froemel D, Meurer A, Ladel C, Reker D, et Sprifermin (rhFGF18) enables proliferation of chondrocytes producing a hyaline cartilage matrix. Osteoarthritis Cartilage. 2017;25(11):1858-67.
  73. Reker D, Siebuhr AS, Thudium CS, Gantzel T, Ladel C, Michaelis M, et Sprifermin (rhFGF18) versus vehicle induces a biphasic process of extracellular matrix remodeling in human knee OA articular cartilage ex vivo. Scientific reports. 2020;10(1):6011.
  74. Dahlberg LE, Aydemir A, Muurahainen N, Gühring H, Fredberg Edebo H, Krarup-Jensen N, et al. A first-in-human, double- blind, randomised, placebo-controlled, dose ascending study of intra-articular rhFGF18 (sprifermin) in patients with advanced knee osteoarthritis. Clinical and experimental 2016;34(3):445-50.
  75. Lohmander LS, Hellot S, Dreher D, Krantz EF, Kruger DS, Guermazi A, et al. Intraarticular sprifermin (recombinant human fibroblast growth factor 18) in knee osteoarthritis: a randomized, double- blind, placebo-controlled Arthritis & rheumatology (Hoboken, NJ). 2014;66(7):1820-31.
  76. Hochberg MC, Guermazi A, Guehring H, Aydemir A, Wax S, Fleuranceau-Morel P, et Effect of Intra-Articular Sprifermin vs Placebo on Femorotibial Joint Cartilage Thickness in Patients With Osteoarthritis: The FORWARD Randomized Clinical Trial. JAMA. 2019;322(14):1360-70.
  77. Gühring H, Kraines J, Moreau F, Daelken B, Ladel C, Wirth W, et Cartilage thickness modification with sprifermin in knee osteoarthritis patients translates into symptomatic improvement over placebo in patients at risk of further structural and symptomatic progression: Post-hoc analysis of the phase ii FORWARD trial. Annals of the Rheuamtic Diseases. 2019;78 Suppl 2:70-1 Abstract OPO010.
  78. Bay-Jensen AC, Manginelli A, Moreau F, He Y, Luo Y, Andersen JR, et Assessment of cartilage degradation and protective markers in synovial fluid from osteoarthritis patients before and after cycles of intra-articular injections with sprifermin. Annals of the rheumatic diseases. 2020;79(Suppl 1):117-8 Abstract OP0189
  79. Salminen-Mankonen HJ, Morko J, Vuorio Role of cathepsin K in normal joints and in the development of arthritis. Current drug targets. 2007;8(2):315-23.
  80. Lindstrom E, Rizoska B, Henderson I, Terelius Y, Jerling M, Edenius C, et Nonclinical and clinical pharmacological characterization of the potent and selective cathepsin K inhibitor MIV-711. Journal of translational medicine. 2018;16(1):125.
  81. Lindstrom E, Rizoska B, Tunblad K, Edenius C, Bendele AM, Maul D, et The selective cathepsin K inhibitor MIV-711 attenuates joint pathology in experimental animal models of osteoarthritis. Journal of translational medicine. 2018;16(1):56.
  82. Conaghan PG, Bowes MA, Kingsbury SR, Brett A, Guillard G, Rizoska B, et Disease-Modifying Effects of a Novel Cathepsin K Inhibitor in Osteoarthritis: A Randomized Controlled Trial. Ann Intern Med. 2020;172(2):86-95.
  83. Hunter D, Nevitt M, Lynch J, Kraus VB, Katz JN, Collins JE, et Longitudinal validation of periarticular bone area and 3D shape as biomarkers for knee OA progression? Data from the FNIH OA Biomarkers Consortium. Annals of the rheumatic diseases. 2016;75(9):1607-14.
  84. Cross AL, Hawkes J, Wright HL, Moots RJ, Edwards APPA (apocynin and paeonol) modulates pathological aspects of human neutrophil function, without supressing antimicrobial ability, and inhibits TNF expression and signalling. Inflammopharmacology. 2020;28(5):1223-35.
  85. Wardyn JD, Ponsford AH, Sanderson Dissecting molecular cross-talk between Nrf2 and NF-kappaB response pathways. Biochem Soc Trans. 2015;43(4):621-6.
  86. Alcaraz MJ, Ferrándiz Relevance of Nrf2 and heme oxygenase-1 in articular diseases. Free Radic Biol Med. 2020;157:83-93.
  87. Roman-Blas JA, Jimenez NF-kappaB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis. Osteoarthritis Cartilage. 2006;14(9):839-48.
  88. Gerwin N, Bendele AM, Glasson S, Carlson The OARSI histopathology initiative – recommendations for histological assessments of osteoarthritis in the rat. Osteoarthritis Cartilage. 2010;18 Suppl 3:S24-34.
  89. Glasson S, Bendele A, Larkins APPA provides disease modification in preclinical osteoarthritis. Osteoarthritis and Cartilage. 2012;20:S72 -S3 Abstract 132.
  90. Glasson S, Larkins APPA provides symptom relief in clinical canine osteoarthritis. Osteoarthritis and Cartilage. 2012;20 (Suppl 1):S287 Abstract 572.
  91. Larkins N, King Effectiveness of apocynin-paeonol (APPA) for the management of osteoarthritis in dogs: comparisons with placebo and meloxicam in client-owned dogs. Matters. 2017;10.19185/ matters.201608000001.
  92. Larkins Efficacy and safety of the combination of apocynin and paeonol (APPA): an uncontrolled patient case series. Annals of the rheumatic diseases. 2020;79 Suppl 1:1738 Abstract AB0872.
  93. Karsdal MA, Christiansen C, Ladel C, Henriksen K, Kraus VB, Bay-Jensen AC. Osteoarthritis–a case for personalized health care? Osteoarthritis Cartilage. 2014;22(1):7-16.
  94. Tonge DP, Pearson MJ, Jones The hallmarks of osteoarthritis and the potential to develop personalised disease-modifying pharmacological therapeutics. Osteoarthritis Cartilage.

Alan Reynolds

Alan is Chief Scientific Officer and Director of AKL Research and Development Ltd. He graduated from Brunel University with an honours degree in Applied Biology before working in the antimicrobial discovery section at Glaxo, undertaking microbiology research at The Royal Free Hospital in London and acting as a Teaching Fellow at the University of London School of Pharmacy. He then rejoined the pharmaceutical industry with Ayerst (later merged with Wyeth and Lederle) in the Medical Affairs Department. Over the years he worked in a number of therapeutic areas including female health, gastroenterology and diabetes. As a member of the global strategy teams he was involved in the development of the block buster drugs Effexor and Enbrel. He was project lead for the successful submissions to the National Institute for Health and Clinical Excellence (NICE) for Enbrel and was appointed Clinical Science Director for Europe in 2007. He received several company awards including two President’s Awards for Excellence. Following the takeover of Wyeth by Pfizer in 2009 he chose to leave the corporation and worked as a consultant for a number of companies. He joined AKL in 2018. He is a member of the Treatment Subcommittee of the UK Charity Versus Arthritis and the author of 1 book chapter, 29 papers, 5 letters and 47 abstracts.

Nicholas Larkins

Nicholas is the Chief Research Officer and a Director of AKL Research and Development Ltd. A graduate in veterinary science from Sydney University and a Member of the Royal College of Veterinary Surgeons, he has been an invited Lecturer to Conferences in Australia, United Kingdom, Dubai, Spain, Italy, Belgium, The Netherlands, USA and Switzerland. Since 1998 he has been involved in ongoing post-graduate study, research with published papers in pharmacognosy, nutritional medicine and reactive oxygen species (biology and pathology). He has extensive research interests in pharmacognosy specifically focused on immunomodulatory phyto-pharmaceuticals and the roles they play in inflammation and the resolution of inflammation. He was a veterinary advisor and clinical consultant to the U.K., Spanish, New Zealand and Swedish Equestrian Teams for the Seoul Olympics, 1988: to U.K., New Zealand, United Kingdom and Australian Equestrian Teams for the Barcelona Olympics 1992 and to New Zealand Equestrian Team for the Sydney Olympics 2000. Ongoing research projects and clinical trials involve collaborations with the University of Aberdeen (UK), The Animal Health Trust (Newmarket- UK), The University of Utrecht (Holland), The University of Vienna (Austria), University of East Anglia (UK), Knight Scientific Laboratories (UK), Warwickshire Agricultural College (UK), Royal Agricultural University (UK), Bolder Biopath (USA), The University of Surrey (UK), The University of Liverpool. (UK), INIBIC-Instituto de Investigación Biomedica da Coruña (Spain) and Nordic Bioscience Clinical Development (NBCD) (Denmark).

NEXT