This site is intended for healthcare professionals
Osteoarthritis
Managing Osteoarthritis-associated Pain

Emerging therapies

Read time: 20 mins
Last updated:4th Feb 2022
Published:10th Nov 2020
  • therapies targeting nociceptive and inflammatory pathways that promote OA pain
  • disease-modifying osteoarthritis drugs (DMOADs) that may also reduce pain
  • the challenges in conducting and interpreting clinical trials on OA treatment

Emerging therapies for osteoarthritis pain

Traditional pharmacological strategies for managing pain in osteoarthritis (OA) include paracetamol, non-steroidal anti-inflammatory drugs (NSAIDs), intra-articular treatments and, for severe pain, opioid analgesics. Updates to guidelines, while strongly recommending NSAIDS, have downgraded (or withdrawn) recommendations for paracetamol use, and recommend opioids only as a last resort as there is insufficient evidence of efficacy and the likelihood of adverse reactions1–3. These treatments are often inadequate, particularly in older patients that are less able to deal with adverse reactions or who possess comorbidities that mean some treatments are contraindicated. Adverse reactions include paracetamol-induced hepatotoxicity, gastrointestinal toxicities from NSAIDs, and the risk of falls, delirium and dependency from opioids4.

Long-lasting, effective and tolerable medical treatments for OA pain are required if core first-line therapies such as exercise and lifestyle changes prove insufficient. Current data suggest that joint damage, inflammation, and peripheral and central sensitisation to pain all contribute to OA pain, which may partly explain the relative incongruence between structural damage and perceived pain in patients with OA5.

Therapies targeting inflammation

There has been a shift in interest towards therapies that target nociceptive and inflammation pathways, both of which are implicated in OA pain; increasing evidence points to cross-talk between these pathways6–9. Therapies that target inflammation, such as intra-articular glucocorticoids, are already used in the clinic to treat OA pain. However, there has been limited success in targeting individual proinflammatory mediators, such as tissue necrosis factor (TNF) or members of the interleukin family9, with biologics5,9,10. Some evidence suggests that long-term treatment (>12 months) with anti-TNF can protect against joint damage but that TNF is less involved in pain mechanisms5.

Other inflammatory cytokines are being investigated as potential treatment targets in OA. Therapies that facilitate intra-articular delivery of genes encoding anti-inflammatory cytokines may overcome the problem of delivering large biologic molecules into the joint. Currently, multiple experimental therapies that target the cross-talk between the inflammation and nociceptive pathways are under investigation, including the granulocyte–macrophage colony-stimulating factor (GM-CSF)–CC-chemokine ligand 17 (CCL17) chemokine axis5,9.

Methotrexate is an accepted treatment for rheumatoid arthritis. A pragmatic phase III trial showed methotrexate significantly reduced knee OA pain and improved the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores for stiffness and function, but the analgesic effect was clinically borderline11. An ongoing randomised controlled trial (RCT) is examining the effect of methotrexate on pain and synovitis in patients with moderate-to-severe knee OA12, but more evidence is required for a possible role of methotrexate in relieving OA pain.

Therapies targeting the nociceptive pathway

Investigative therapies associated with the nociceptive (pain) pathway aim to dampen nociceptive signalling in various ways13

Nerve growth factor as a target

Nerve growth factor (NGF) is released during injury or inflammation and regulates neuronal survival and pain signalling by binding to and activating tropomyosin kinase A (TrkA) (Figure 1). NGF also binds to the p75NTR receptor that triggers a cellular signalling cascade known to be associated with mechanical hyperalgesia13. Both NGF and p75NTR appear to be overexpressed in the serum and synovial fluid of patients experiencing pain from conditions such as OA14. Investigative therapeutic strategies aimed at disrupting NGF-induced pain, therefore, include antibodies and small molecule inhibitors against NGF, TrkA and p75NTR 13(Figure 1).

OA_T4_Fig_1_redraw.png

Figure 1. Pathways of investigative drugs targeting pain in osteoarthritis (Adapted). NGF, nerve growth factor; TrkA, tropomyosin kinase A; TRPV1, transient receptor potential vanilloid 1 (ion channel).

Anti-NGF antibodies

Currently, only one anti-NGF monoclonal antibody, fasinumab, is still being explored in Phase III clinical trials. Trials with two other candidates, fulranumab and tanezumab15, have been discontinued because of safety concerns, including osteonecrosis and rapidly progressive OA5.

Fasinumab is a fully human IgG4 monoclonal antibody. In randomised, double-blind Phase III clinical trials, OA patients treated with fasinumab at different doses from 1 to 9 mg experienced less knee and hip pain and displayed better physical function than the placebo group5,16. In a Phase II-III trial, 9 mg of subcutaneous fasinumab every 4 or 8 weeks improved pain and function in patients with chronic low back pain at 16 weeks17. However, the incidence of arthropathies also increased in both these trials, suggesting that joint-related adverse reactions are a class effect of NGF inhibitors. Data from a long-term safety study of fasinumab (NCT02683239) are expected in 202218.

Anti-TrkA and p75NTR treatments

With the aim of altering NGF-mediated pain, multiple specific TrkA and pan-Trk inhibitors have been assessed in Phase I and II clinical trials for OA pain. Most studies have been small and results have been mixed19–21. Indeed, many clinical trials have been abandoned or halted in patients with OA, and more studies are needed to confirm the utility of TrkA inhibition in clinical practice.

Another possible strategy to dampen NGF-mediated pain is to target its binding partner, p75NTR. A Phase I clinical trial (NCT03227796) of the p75NTR-Fc fusion protein (LEVI-04) in patients with OA knee pain was completed in March 202122. To date, study results have not been published.

Ion channel modulators

Ion channels have been implicated in OA pain, one of which is the transient receptor potential vanilloid 1 (TRPV1) channel that is localised in sensory neurons13. TRPV1 modulators desensitise the end terminal of these primary afferent pain fibres (Figure 1). The best studied TRPV1 agonist is CNTX-4975, a synthetic capsaicin administered by intra-articular injection. There are currently two ongoing Phase III clinical trials for CNTX-4975 in patients with chronic moderate-to-severe OA knee pain (NCT0342904923 and NCT0366199624), after the efficacy of a single intra-articular dose of 0.5 mg was previously demonstrated5,25. VICTORY-2, another Phase III trial of intra-articular CNTX-4975-05 for moderate-to-severe OA knee pain (NCT03660943), was completed in June 2021, with results pending5,26. An oral TRPV1 antagonist, NEO6860, is also being investigated for OA knee pain27.

Disease-modifying osteoarthritis drugs (DMOADs)

A DMOAD is a drug that modifies the underlying OA pathophysiology, thereby inhibiting structural damage to prevent or reduce long-term disability and offer potential symptomatic5. The symptom–structure discordance in OA complicates the application of these potential therapeutics for pain relief, at least in the short term; long-term trials are needed to determine if structural effects translate into clinical benefits for patients or if they reduce the need for joint replacement. Many candidates are currently being investigated. Those showing promise include drugs promoting cartilage repair, TissueGene-C, Cathepsin K and sprifermin5.

Drugs promoting cartilage repair

One promising drug under investigation is sprifermin, a recombinant human fibroblast growth factor 18 (FGF18). FGF18 is an important anabolic factor in chondrogenesis, chondrocyte proliferation and cartilage repair in healthy and damaged cartilage. In a phase II trial, intra-articular injections of sprifermin every 6 or 12 months had positive effects on cartilage thickness. In post hoc analyses, the structural effect did improve WOMAC pain scores28. Whether the structural benefits persist over time and translate into clinical benefits, along with long-term safety, remains unclear. Lorecivivint, which inhibits the Wnt signalling pathway in bones and joints, also shows promise5.

TissueGene-C

Intra-articular injection of this drug delivers transforming growth factor-β (TGF-β) to the joint. Dysregulation of TGF-β signalling in chondrocytes occurs in both ageing and OA cartilage. A 52-week, placebo-controlled phase III trial (n = 163) reported that TissueGene-C improved OA symptoms and showed trends towards structural improvements on MRI5. After suspension for technical issues, one phase III trial (NCT03203330) has resumed with intra-articular injection of TissueGene-C.

Cathepsin K

Cathepsin K is involved in bone resorption and is expressed in OA synovium and chondrocytes. A cathepsin K inhibitor, MIV-711, reduced cartilage damage, prevented bone resorption, and decreased biomarkers of bone and cartilage remodelling in animal models of OA and in a phase II trial. It has a good safety profile, but there was no change in pain score, the endpoint of the trial, so clinical efficacy remains unclear29.

Challenges in interpreting findings of clinical trials on therapies for osteoarthritis pain

Several characteristics of pain in general, and osteoarthritis (OA) pain in particular, increase the challenges of interpreting the results of clinical trials exploring potential new therapies5,10:

  • Pain is a subjective experience and so is challenging to measure. The numerous methods of measuring pain that are used in clinical trials (visual analogue scales, numeric rating scales, functional assessments, patient-reported outcome measures) make comparing the results of trials difficult. A further complication is the varying degrees of psychometric robustness of the measures30.
  • The biopsychosocial model of pain recognises that physical, psychological, behavioural and social factors can modulate an individual’s experience/perception of pain31. The interrelationships of these factors may confound the outcomes of clinical trials if the measurement of pain used fails to account for key domains.
  • OA is heterogenous in both clinical and pathophysiological terms – it affects multiple joints through different processes and varies substantially in its clinical and radiographic presentation. Thus, OA in different joints, and in different patients, may respond differently to the same treatment, confounding trial results. 'Pain phenotyping' of patients would help overcome this, but is very challenging5.
  • OA can progress at different rates. For example, knee OA often progresses slowly. Therefore, if clinical trials of disease-modifying drugs have follow-ups of <12 months, they may miss meaningful changes in structure and possible translation into clinical benefits5.

References

  1. Bannuru R, Osani M, Vaysbrot E, Arden N, Bennell K, Bierma-Zeinstra SMA, et al. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthritis and cartilage. 2019;27(11):1578–1589.
  2. Bruyere O, Honvo G, Veronese N, Arden NK, Branco J, Curtis EM, et al. An updated algorithm recommendation for the management of knee osteoarthritis from the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO). Seminars in Arthritis and Rheumatism. 2019;49(3):337–350.
  3. Kolasinski SL, Neogi T, Hochberg MC, Oatis C, Guyatt G, Block J, et al. 2019 American College of Rheumatology/Arthritis Foundation guideline for the management of osteoarthritis of the hand, hip, and knee. Arthritis and Rheumatology. 2020;72(2):220–233.
  4. O’Neil CK, Hanlon JT, Marcum ZA. Adverse effects of analgesics commonly used by older adults with osteoarthritis: Focus on non-opioid and opioid analgesics. American Journal of Geriatric Pharmacotherapy. 2012;10(6):331–342.
  5. Latourte A, Kloppenburg M, Richette P. Emerging pharmaceutical therapies for osteoarthritis. Nature reviews Rheumatology. 2020;16(12):673–688.
  6. Cook AD, Christensen AD, Tewari D, McMahon SB, Hamilton JA. Immune Cytokines and Their Receptors in Inflammatory Pain. Trends in immunology. 2018;39(3):240–255.
  7. Raoof R, Willemen HLDM, Eijkelkamp N. Divergent roles of immune cells and their mediators in pain. Rheumatology (Oxford, England). 2018;57(3):429–440.
  8. Malfait A-M, Schnitzer TJ. Towards a mechanism-based approach to pain management in osteoarthritis. Nature Reviews Rheumatology 2013 9:11. 2013;9(11):654–664.

  9. Conaghan PG, Cook AD, Hamilton JA, Tak PP. Therapeutic options for targeting inflammatory osteoarthritis pain. Nature Reviews Rheumatology. 2019;15(6):355–363.
  10. Cai X, Yuan S, Zeng Y, Wang C, Yu N, Ding C. New Trends in Pharmacological Treatments for Osteoarthritis. Frontiers in Pharmacology. 2021;12:645842.
  11. Kingsbury SR, Tharmanathan P, Keding A, Corbacho B, Watt FE, Scott DL, et al. Significant pain reduction with oral methotrexate in knee osteoarthritis; results from the promote randomised controlled phase iii trial of treatment effectiveness. Osteoarthritis and Cartilage. 2019;27:S84–S85.
  12. Zhu Z, Yu Q, Leng X, Han W, Li Z, Huang C, et al. Can low-dose methotrexate reduce effusion-synovitis and symptoms in patients with mid- to late-stage knee osteoarthritis? Study protocol for a randomised, double-blind, and placebo-controlled trial. Trials 2020 21:1. 2020;21(1):1–10.
  13. Alcaraz M, Guillén M, Ferrándiz M. Emerging therapeutic agents in osteoarthritis. Biochemical pharmacology. 2019;165:4–16.
  14. Montagnoli C, Tiribuzi R, Crispoltoni l, Pistilli A, Stabile A, Manfreda F, et al. β-NGF and β-NGF receptor upregulation in blood and synovial fluid in osteoarthritis. Biological chemistry. 2017;398(9):1045–1054.
  15. PharmaLive.com. Eli Lilly and Pfizer End Tanezumab Development. https://www.pharmalive.com/eli-lilly-and-pfizer-end-tanezumab-development/. Accessed 6 December 2021.
  16. Dakin P, DiMartino SJ, Gao H, Maloney J, Kivitz AJ, Schnitzer TJ, et al. The efficacy, tolerability, and joint safety of fasinumab in osteoarthritis pain: A phase IIb/III double-blind, placebo-controlled, randomized clinical trial. Arthritis and Rheumatology. 2019;71(11):1824–1834.
  17. Onuora S. Fasinumab effective for chronic low back pain. Nature Reviews Rheumatology 2020 17:1. 2020;17(1):2–2.
  18. Long-Term Safety and Efficacy Study of Fasinumab in Patients With Pain Due to Osteoarthritis (OA) of the Knee or Hip (NCT02683239). https://clinicaltrials.gov/ct2/show/NCT02683239. Accessed 6 December 2021.
  19. Krupka E, Jiang GL, Jan C. Efficacy and safety of intra-articular injection of tropomyosin receptor kinase A inhibitor in painful knee osteoarthritis: a randomized, double-blind and placebo-controlled study. Osteoarthritis and cartilage. 2019;27(11):1599–1607.
  20. Watt FE, Blauwet MB, Fakhoury A, Jacobs H, Smulders R, Lane NE. Tropomyosin-related kinase A (TrkA) inhibition for the treatment of painful knee osteoarthritis: results from a randomized controlled phase 2a trial. Osteoarthritis and cartilage. 2019;27(11):1590–1598.
  21. Ishiguro N, Oyama S, Higashi R, Yanagida K. Efficacy, safety, and tolerability of ONO-4474, an orally available pan-tropomyosin receptor kinase inhibitor, in Japanese patients with moderate to severe osteoarthritis of the knee: A randomized, placebo-controlled, double-blind, parallel-group comparative study. Journal of Clinical Pharmacology. 2020;60(1):28–36.
  22. Safety, Tolerability & Pharmacokinetics of LEVI-04 in Healthy Volunteers and Patients With Osteoarthritis Knee Pain (NCT03227796). https://clinicaltrials.gov/ct2/show/record/NCT03227796. Accessed 7 December 2021.
  23. A clinical study to test efficacy and safety of CNTX-4975-05 in patients with osteoarthritis knee pain. Available at: https://clinicaltrials.gov/ct2/show/NCT03429049. Accessed 1 April 2020.
  24. NCT03661996. A study to compare five different treatment regimens of CNTX-4975 for participants with chronic, moderate-to-severe osteoarthritis pain. https://clinicaltrials.gov/ct2/show/NCT03661996?term=NCT03661996&draw=2&rank=1. Accessed 7 December 2021.
  25. Stevens R, Ervin J, Nezzer J, Nieves Y, Guedes K, Burges R, et al. Randomized, Double-Blind, Placebo-Controlled Trial of Intraarticular Trans-Capsaicin for Pain Associated With Osteoarthritis of the Knee. Arthritis & rheumatology (Hoboken, NJ). 2019;71(9):1524–1533.
  26. A Clinical Study to Test Efficacy and Safety of Repeat Doses of CNTX-4975-05 in Patients With Osteoarthritis Knee Pain - Tabular View - ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/record/NCT03660943?term=NCT03660943&draw=2&rank=1. Accessed 7 December 2021.
  27. Arsenault P, Chiche D, Brown W, Miller J, Treister R, Leff R, et al. NEO6860, modality-selective TRPV1 antagonist: a randomized, controlled, proof-of-concept trial in patients with osteoarthritis knee pain. Pain reports. 2018;3(6). doi:10.1097/PR9.0000000000000696.
  28. Latourte A, Kloppenburg M, Richette P. Emerging pharmaceutical therapies for osteoarthritis. Nature Reviews Rheumatology. 2020;16(12):673–688.
  29. Conaghan PG, Bowes MA, Kingsbury SR, Brett A, Guillard G, Rizoska B, et al. Disease-Modifying Effects of a Novel Cathepsin K Inhibitor in Osteoarthritis: A Randomized Controlled Trial. Annals of internal medicine. 2020;172(2):86–95.
  30. Lundgren-Nilsson Å, Dencker A, Palstam A, Person G, Horton MC, Escorpizo R, et al. Patient-reported outcome measures in osteoarthritis: a systematic search and review of their use and psychometric properties. RMD Open. 2018;4(2):e000715.
  31. Turk D, Gatchel R. Biopsychosocial perspective on chronic pain. In: Psychological approaches to pain management: A practitioner’s handbook (3rd ed.). 2018 https://www.guilford.com/books/Psychological-Approaches-to-Pain-Management/Turk-Gatchel/9781462528530/contents. Accessed 23 November 2021.

 

Welcome: