ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Investigational approaches to the management of osteoarthritis

Investigational approaches to the management of osteoarthritis
Literature review current through: Jan 2024.
This topic last updated: Dec 19, 2023.

INTRODUCTION — The management of osteoarthritis (OA) consists of nonpharmacologic, pharmacologic, and surgical approaches. Conventional pharmacologic therapy for OA has mainly targeted symptomatic relief, with agents such as nonsteroidal anti-inflammatory drugs (NSAIDs), acetaminophen, opioid analgesia, and intra-articular injections.

As we gain a better understanding of the pathogenetic mechanisms in OA, there has been a growing interest in the development of drugs whose mechanism of action are directed towards different pain pathways, as well as the inhibition of catabolic processes or stimulation of anabolic processes in the OA joint. These drugs are referred to as disease-modifying OA drugs (DMOADs) or structure-modifying OA drugs (SMOADs).

To date, no pharmacologic agents have been approved by regulatory authorities for disease modification in OA. Existing trial strategies have failed to detect clinical efficacy of potential DMOADs based on the lack of responsive outcome measures to record treatment-related improvements in pain, function, or joint structure, or just due to lack of efficacy of these agents. While current evidence-based guidelines either advise against or weakly recommend these treatments, public interest remains high [1,2]. Clinician awareness of these treatments and the limited demonstrated efficacy in clinical trials is therefore a key factor in supporting patients with informed decision-making.

A review of investigational approaches to the pharmacotherapy of OA will be discussed here. Established therapies for OA and surgical interventions are discussed elsewhere. (See "Overview of the management of osteoarthritis" and "Management of knee osteoarthritis" and "Management of moderate to severe knee osteoarthritis" and "Management of hand osteoarthritis" and "Management of hip osteoarthritis" and "Overview of surgical therapy of knee and hip osteoarthritis".)

TARGETS FOR TREATMENT — The major goals of OA management are pain control, functional improvement, education about the disease, self-management, and prevention or slowing of structural changes to the joints. However, developing agents to address these goals is challenging given the complexity of pain pathways and the etiology of structural changes.

As is discussed elsewhere, the etiology of OA is multifactorial and affects the composition and structure of all tissues of the joints, including articular cartilage, bone, ligaments, synovium, and synovial fluid. In addition, there is a poor correlation between symptoms and structural changes, as is often demonstrated on plain radiographic imaging, reinforcing the concept that the pain experienced in OA is likely due to a combination of nociceptive (including inflammatory) and/or neuropathic pain. It is also likely that there are multiple local structural sources of pain, including subchondral bone marrow lesions, bone, soft tissue around the joint, and bursae. (See "Pathogenesis of osteoarthritis" and "Epidemiology and risk factors for osteoarthritis".)

In the drug development process, the application of preclinical models of OA to human models has been filled with challenges. Due to difficulties in demonstrating clinical efficacy, many potentially promising disease-modifying OA drug (DMOAD) trials have been terminated. Some of the major categories of investigational agents for the treatment of OA are presented below.

Pain modification — Investigational agents targeting pain in OA have focused on inhibiting nerve growth factor or direct disruption of nerves in affected joints.

Inhibition of nerve growth factor — There is evidence supporting the role of nerve growth factor in the development of sensory neurons such as nociceptors, which are involved in sensing tissue damage. Expression of nerve growth factor is increased in injured or inflamed tissue and is associated with increased pain. However, safety concerns related to a potential increased risk of rapidly progressive OA have limited the development of agents targeting this factor.

Tanezumab, a humanized monoclonal antibody that blocks nerve growth factor (anti-NGF), was shown to reduce knee pain while walking compared with placebo in a randomized trial including 450 patients with moderate to severe knee OA [3]. However, trials of tanezumab were put on partial clinical hold by the US Food and Drug Administration (FDA) in 2010 due to secondary osteonecrosis [4]. A subsequent study found that despite initial reports, tanezumab was not associated with an increase in osteonecrosis but rather rapidly progressive OA [5]. There were additional concerns regarding peripheral nervous system effects [6].

In a trial including 696 patients with moderate to severe hip or knee OA, tanezumab-treated patients (2.5 mg and 5 mg groups) showed greater improvements in scores assessing pain and physical function compared with patients in the placebo group after 16 weeks [7]. However, rapidly progressive OA occurred only in tanezumab-treated patients, and the incidence of total joint replacements was also higher in the tanezumab-treated groups. Furthermore, the improvements in pain and function were relatively modest. Due to safety concerns regarding the risk of rapidly progressive OA, tanezumab is no longer under development [8].

Another anti-NGF agent, fasinumab, appeared to reduce pain and improve function in a trial including 421 patients with moderate to severe knee or hip OA [9]. Adverse effects were higher in the treatment group, particularly with higher rates of nervous system and musculoskeletal symptoms. A dose-dependent increase in arthropathies (7 percent of fasinumab-treated patients and 1 percent of placebo-treated patients) was also observed during the study. Trials of fulranumab, which showed similar efficacy and adverse events to other anti-NGF agents, were reportedly discontinued during phase III development due to the company's portfolio strategy rather than concerns about the drug's safety [10].

Nerve block injections and radiofrequency ablation — Nerve block injections and radiofrequency ablation (RFA) targeting the genicular or saphenous nerves are minimally invasive, direct approaches to address knee OA pain [11-14]. These procedures target one or more of the nerves supplying the knee joint, thereby disrupting pain signals. In one randomized trial of 38 patients with knee OA, RFA of the genicular nerve was associated with improved knee pain at 4 and 12 weeks when compared with sham procedure [11]. Post-RFA studies demonstrate sustained improvements in knee pain at 24 months, with no serious adverse events reported [12,15]. Further sham-controlled trials with larger sample sizes and longer follow-up are warranted.

The use of these techniques in other chronic pain conditions is discussed separately. (See "Interventional therapies for chronic pain".)

Regenerative medicine therapies

Platelet-rich plasma — Platelet-rich plasma (PRP) has gained considerable attention; however, well-designed trials do not show evidence of benefit in improving OA symptoms [16-18]. PRP is derived from autologous blood, with platelets being the main constituent. The mechanism of action is not well understood, but it is thought to provide high concentrations of growth factors, including tissue growth factor and platelet-derived growth factors, which can mediate the proliferation of mesenchymal stem or stromal cells (MSCs) and increase matrix synthesis and collagen formation [19]. PRP is thought to reduce inflammation in an OA joint by enhancing the expression of NF-kappa-beta inhibitor, thus reducing NF-kappa-beta signaling and dampening its downstream inflammatory cytokine activation [19]. It can switch off the inhibition of type II collagen and aggrecan gene expression in interleukin (IL) 1beta-activated NF-kappa-beta in chondrocytes [20]. Thus far, PRP has predominantly been used to treat musculoskeletal conditions, particularly tendon-related pathologies. In general, no major adverse events have been reported, with only minor and transient risks and adverse effects of PRP seen, which include pain/bleeding, tenderness, and swelling and bruising at the site of injection [21]. However, general risks associated with intra-articular injections should be applied in this context, which includes risk of local infection. (See "Joint aspiration or injection in adults: Complications" and "Elbow tendinopathy (tennis and golf elbow)", section on 'Platelet-rich plasma and other biologic injections'.)

Increasing evidence from well-designed trials does not support the use of PRP for the management of OA [16-18]. As an example, a randomized trial including 100 patients with ankle OA found that intra-articular PRP injections, compared with placebo injections, did not demonstrate a statistically significant improvement in a composite score of pain and function during the 26-week follow-up period [16]. Similarly, another randomized trial including patients with knee OA did not demonstrate a benefit in terms of pain or volume loss [17]. Prior data suggested that PRP may provide short-term (up to 12 months) symptomatic relief for knee and hip OA [22], but the majority of those trials revealed a high likelihood of bias with low-to-moderate methodologic quality and variability in their PRP protocols and lack of longer-term follow-up [18,22,23]. Factors that may contribute to differences in results among trials include the lack of standardization of preparations, with different preparation protocols with varying concentrations of platelets, presence/absence of white cells, and mode of activation (ie, mechanically with freeze-thawing cycles, chemically, or endogenously).

Additional information regarding trials of PRP for knee OA is presented separately. (See "Management of knee osteoarthritis", section on 'Platelet-rich plasma'.)

Stem cell therapy — MSCs derived from bone marrow, adipose, synovium, and other tissues have been investigated for their potential role in regenerating chondrocytes, mediating tissue repair, and stimulating growth factors. Published trials have demonstrated that intra-articular injection is well tolerated; small sample size and heterogeneity in selection criteria, MSC tissue source, number of MSCs, and analyzed outcomes limit generalizability and conclusive determination of benefit [24-26]. In a randomized, placebo-controlled trial of adipose-derived intra-articular MSC injection in 252 patients with moderate knee OA, the treatment group showed improved Visual Analog Scale and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain scores at six months when compared with placebo and no difference in structural change by magnetic resonance imaging (MRI) evaluation [27]. Given the heterogeneity and small sample size issues with prior studies, this study is notable for its large sample size and inclusion of only patients with moderate OA. Further clinical studies are needed to determine if MSCs are an effective long-term treatment for OA and, if effective, to establish the optimal MSC-harvesting source, MSC cell dose, and number of injections.

Tissue gene therapy — Transforming growth factor-beta (TGF-beta) is part of cellular differentiation and growth synthesis of extracellular matrix and chondrogenesis, and TGF-beta signaling pathways are thought to be involved in early cartilage development and maintenance of cartilage over time. TissueGene-C is a novel gene and cell therapy consisting of human allogeneic chondrocytes and irradiated GP2-293 cells overexpressing TGF-beta1 [28]. Results from two small phase II trials [29,30] and one phase III trial [31] demonstrated statistically significant improvements in pain and function in patients. Trends of structural improvement were observed but were not statistically significant.

Structural modification — Drugs targeting structural changes in OA include agents that modify inflammatory pathways, cartilage catabolism and anabolism, and bone remodeling.

Inflammatory pathways

Tumor necrosis factor-alpha inhibitors — Tumor necrosis factor (TNF)-alpha, predominantly produced by activated synoviocytes, phagocytic mononuclear cells, and articular cartilage, is one of the major cytokines involved in the destruction of articular matrix. Together with IL-1beta, it activates inflammatory signaling pathways that end in the activation of NF-kappa-beta, a transcription factor that regulates the expression of chemokines, cytokines, adhesion molecules, and matrix-degrading enzymes. (See "Pathogenesis of osteoarthritis", section on 'Inflammatory mediators'.)

The concept of TNF-alpha inhibition, which has been effective for the treatment of rheumatoid and psoriatic arthritis, has been applied to OA with mixed results with different TNF-alpha inhibitors.

Infliximab, a chimeric monoclonal antibody directed against TNF-alpha, appears to have a potential effect on disease progression. In a 12-month open-label pilot study in erosive hand OA, patients who received monthly intra-articular infliximab injections into the affected joints of the more severely affected hand were found to have a reduction in the anatomical lesion radiologic score when compared with joints injected with saline [32]. In another cohort of rheumatoid arthritis patients, intra-articular infliximab injections reduced the radiologic progression of incident secondary hand OA after three years of follow-up [33].

The data for adalimumab, a fully human monoclonal antibody also directed against TNF-alpha, have shown mixed results in the treatment of OA. Several studies have failed to demonstrate improvements in hand OA pain with subcutaneous adalimumab injections [34,35]. However, another randomized trial including 60 patients with erosive hand OA did show a significant decrease in the number of new erosions with subcutaneous adalimumab injections after 12 months of follow-up [36].

TNF-alpha blockade may not be the appropriate agent in a low-level inflammatory condition. Despite limited effects on pain in OA patients, there may be a potential positive effect on disease progression and the development of erosions.

Interleukin 1beta inhibitors — IL-1 is capable of inducing the expression of matrix metalloproteinases (MMPs) and other catabolic genes, which stimulate extracellular matrix remodeling. IL-1beta antagonists have been shown to prevent cartilage breakdown in a multitude of animal models [37,38]. However, translation to human models failed to demonstrate a reduction in pain in patients with knee OA. As an example, in a randomized trial with 170 patients with symptomatic knee OA, intra-articular injections with anakinra, a recombinant human IL-1 receptor antagonist, was well tolerated but was not associated with improvements in OA symptoms compared with placebo [39]. Another trial with a monoclonal antibody that binds type 1 IL-1 receptor also failed to elicit a statistically significant difference in WOMAC pain score in knee OA after three months of treatment [40].

Findings from an exploratory analysis suggest that there may be a role for IL-1 inhibition in large joint OA; however, these results are limited by the fact that the parent trial was not designed to examine the efficacy of IL-1beta inhibitors for OA [41]. The analysis included 10,061 patients with elevated C-reactive protein (CRP) levels and a history of myocardial infarction who were randomly assigned to receive placebo or canakinumab, a human anti-IL-1beta monoclonal antibody. After a median follow-up of 3.7 years, the incidence rates for total hip and knee arthroplasty were lower among the patients receiving canakinumab compared with the placebo group (0.31 versus 0.54 events per 100 person-years, respectively; hazard ratio [HR] 0.58 [95% CI 0.61-0.87]). However, direct trial evidence is needed before conclusions can be made regarding a potential role for canakinumab in the management of OA. Also, of note, there were more deaths attributed to infection or sepsis in the canakinumab groups compared with the placebo group in the parent trial [42].

Initial studies show good tolerability of anti-IL-1 blockade in healthy individuals, but a trend towards decreased absolute neutrophil count was observed [43]. Further studies of an anti-IL-1alpha/beta dual variable domain-immunoglobulin are underway, targeting a specific patient population in whom there may be a benefit.

Additional information on the role of IL-1 in the pathogenesis of OA can be found separately. (See "Pathogenesis of osteoarthritis", section on 'Inflammatory mediators'.)

Inducible nitric oxide synthase inhibition — Inducible nitric oxide synthase (iNOS), an isoform of nitric oxide synthase, is highly expressed in chondrocytes. It activates MMPs, inhibits collagen and proteoglycan synthesis, and induces chondrocyte cell death [44].

Studies of cindunistat hydrochloride maleate, an orally administered irreversible inhibitor of iNOS, did not demonstrate a reduction in the rate of joint space narrowing in knee OA. A post-hoc analysis showed a beneficial effect at 48 weeks in patients with radiographically less advanced OA, but this effect was not sustained at the 96-week follow-up [45].

Bradykinin receptor B2 antagonist — The OA joint synovium generates bradykinin, a vasodilator and inflammatory peptide. Bradykinin, through the B2 receptor, via the activation of chondrocytes and transcription factor NF-kappa-beta, induces the release of proinflammatory cytokines. It also induces angiogenesis and promotes vessel permeability, growth, and remodeling.

Icatibant, an intra-articular bradykinin receptor B2 antagonist, showed some initial potential analgesic effects [46]. However, further drug trials were suspended due to a possible lack of efficacy [47]. Additional studies of the effect of fasitibant, another bradykinin receptor B2 antagonist, on arthritis pain is also underway [48].

Inhibition of microtubule assembly — Colchicine inhibits microtubule assembly and disrupts inflammasome activation. In an exploratory analysis of 5522 patients with coronary artery disease randomized to daily colchicine or placebo, the incidence rates for total hip and knee arthroplasty were lower among patients receiving colchicine when compared with the placebo group (0.90 versus 1.30 per 100 person-years, respectively; HR 0.69 [95% CI 0.51-0.95]) [49]. The effect remained consistent after patients with gout and patients receiving arthroplasty within the first six months after randomization were excluded. However, these results are limited by the fact that the parent trial was not designed to examine the effect of colchicine on OA and median follow-up was 29 months. In contrast, in another trial, patients randomized to twice daily colchicine or placebo had no difference in knee OA symptoms after 16 weeks of treatment [50]. Further investigation of colchicine therapy is needed to determine its role in the management of OA.

Cartilage catabolism and anabolism

Bone morphogenetic protein-7 — Bone morphogenetic protein (BMP)-7, previously termed osteogenic protein-1, is expressed by human chondrocytes and contributes to the repair of mature tissues [51]. BMP-7 also suppresses catabolic activities of cytokines, including IL-1, IL-6, IL-8, MMP-1, and MMP-13 [52].

An initial randomized trial including 33 patients with knee OA showed promise with the use of weekly intra-articular recombinant human BMP-7 injections [53]. At 12-week follow-up, there was a trend towards symptomatic improvement in those who received different doses of recombinant BMP-7 compared with placebo. In the treatment groups, similar trends of improvement were observed in WOMAC pain and function subscales. Further studies evaluating the use of recombinant BMP-7 in the treatment of OA are underway.

Fibroblast growth factor — Based on preclinical study evidence, fibroblast growth factor (FGF)-18 (sprifermin) is thought to be a potential therapeutic agent for OA. In animal models of injury-induced OA, FGF-18 has been shown to have an anabolic effect on cartilage, stimulating proteoglycan synthesis and cartilage matrix formation [54,55]. However, the available evidence for its use in patients with knee OA is limited and still of uncertain clinical importance.

Two phase I trials of intra-articular recombinant human FGF-18 for knee OA demonstrated a statistically significant dose-dependent improvement in tibiofemoral cartilage volume and a reduction in joint space narrowing after 12 months [56,57]. In a larger phase II trial including 549 patients with symptomatic knee OA, intra-articular administration of 100 micrograms of sprifermin every 6 or 12 months was associated with an increase in total femorotibial cartilage thickness after 2 years when compared with placebo [58]. However, there were no improvements in total WOMAC pain scores among those receiving sprifermin. Although the difference in cartilage thickness was statistically significant, the clinical relevance of these findings remains uncertain. Subgroup analysis of patients with low joint space width and moderate-to-high pain showed improved pain outcomes alongside improved cartilage thickness, suggesting that future trials should target this subgroup [59].

Further clinical studies are warranted to gain a better understanding of the effects of sprifermin at optimal doses for possible disease modification in OA.

Matrix metalloproteinase inhibitors — MMP inhibition was thought to be a potential target for OA as MMP genes are expressed in all aspects of the articular joint, and one of its main roles is the enzymatic collagen breakdown in articular cartilage. Of all the MMPs identified to date, collagenases MMP-1, MMP-8, MMP-13, and MMP-14 are associated with cartilage collagen destruction [60,61]. (See "Pathogenesis of osteoarthritis", section on 'Proteases'.)

Thus far, MMP inhibition for treating OA has been relatively unsuccessful due to intolerable musculoskeletal side effects, including musculoskeletal pain, Dupuytren's contracture, and frozen shoulder [62,63].

ADAMTS — Aggrecanase activity increase is a known trigger for cartilage degradation. A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5 are two key aggrecanases. Collagen and aggrecan are the primary structural components of the cartilage extracellular matrix, and the progression of OA correlates with their degradation [64,65]. Thus, collagenases and aggrecanases are prospective targets for the development of DMOAD; however, trials evaluating the efficacy of ADAMTS-5 inhibition have not shown benefit in symptomatic knee OA [66].

Mitogen-activated protein kinase inhibitor — Alternate signal transduction pathways, which regulate MMP, are being explored, based on the side effect profile of direct MMP inhibition. Mitogen-activated protein kinases (MAPK) are central regulators of cell signaling pathways that control cell proliferation, survival, and matrix synthesis [67] and regulate chondrocyte production of MMPs [68].

Several MAPK inhibitors have been investigated in clinical trials, with only a few progressing to phase II trials due to associated liver and central nervous system adverse effects [69].

Cathepsin-K inhibitor — Cathepsin-K (CatK) is expressed in osteoclasts and is a key enzyme involved in bone resorption. It is also expressed in chondrocytes in cartilage, where it can cleave type II collagen and aggrecan, which are main components of the cartilage matrix [70]. Inhibition of CatK has been shown to reduce biomarkers of bone resorption and cartilage degradation in animal models [71,72]. Clinical trials with CatK inhibitors have shown variable outcomes. Balicatib did not reduce cartilage volume loss, and morphea-like skin reactions have been reported [73]. In another trial including 244 patients with symptomatic and radiographic knee OA, pain scores at 26 weeks among patients taking MIV-711 (a potent CatK inhibitor) were similar to those in the placebo group [74]. However, a reduction in bone and cartilage progression was observed in the MIV-711 group. The safety profile was reassuring and was without an increased risk of morphea-like skin reactions.

Wnt signaling pathway inhibitors — The Wnt pathway has a prominent role in OA joints, particularly with cartilage catabolism and regeneration through modulation of the differentiation of osteoblasts and chondrocytes and production of catabolic proteases [75,76]. SM04690 (lorecivivint) is a novel small-molecule Wnt pathway inhibitor proposed to have multiple effects on the articular cartilage, including slowing down cartilage degradation and reducing inflammation [77]. A proof-of-concept trial consisting of 455 patients with unilateral symptomatic knee OA did not meet the primary endpoint of change from baseline in WOMAC pain score in the target knee [78]. However, in an analysis of secondary endpoints, symptomatic and medial joint space width improvements were observed in patients in the lorecivivint group at 52 weeks. Findings from a phase IIb randomized trial that included 695 patients with moderate to severe knee OA who were randomized to receive either lorecivivint or placebo were more promising [79]. Overall, statistically significant improvements were observed in patient-reported outcomes that included WOMAC pain, function, and patient global assessments compared with placebo [80].

Bone remodeling

Calcitonin — The main role of calcitonin is to regulate calcium homeostasis. It is produced primarily by the parafollicular C cells in the thyroid gland. It binds to calcitonin receptors on osteoclasts and promotes the inhibition of bone resorption, and thus increases the activity of osteoblasts. In preclinical studies, calcitonin has been shown to induce inhibition of MMP activity and cartilage degradation, both in vivo and ex vivo [81]. It also may have an effect on the inhibition of toll-like receptors (TLRs), which release cytokines and inflammatory mediators [82].

Pilot studies of salmon calcitonin comparing two doses of salmon calcitonin formulated with a 5-CNAC carrier against placebo demonstrated no significant differences in pain scores [83]. However, a considerable improvement was seen in the function score among patients receiving oral salmon calcitonin.

However, the results from two phase III trials of oral salmon calcitonin failed to provide any reproducible clinical benefits in patients with symptomatic knee OA [84].

Bisphosphonates — Commonly used as the treatment for osteoporosis, the therapeutic basis of bisphosphonates is based on their direct inhibitory effect on the function of osteoclasts (see "Bisphosphonate therapy for the treatment of osteoporosis"). They have the potential to retard subchondral bone remodeling. Patients with progressive OA have been found to have higher urinary levels of C-terminal telopeptide of type II collagen (CTX-II), a marker of cartilage degradation [85]; thus, the application of bisphosphonates seems ideal in such cases. Bisphosphonates can also exert a slight immunomodulating effect via the inhibition of proinflammatory cytokines, with etidronate in vitro, showing some inhibitory effect on MMPs when they binds to human cartilage [86].

The use of bisphosphonates in OA has yielded mixed results. The British study of risedronate in structure and symptoms of knee OA (BRISK) trial was a randomized control trial of patients with mild to moderate OA of the medial compartment of the knee [87]. Patients were randomized to once daily risedronate (5 or 15 mg) or placebo. Those who received 15 mg of risedronate showed improvement in the WOMAC index, significant improvement in the patient global assessment, and decreased use of walking aids.

In another large study, the Knee OA Structural Arthritis (KOSTAR) study, 2483 patients with medial compartment knee OA from North America and the European Union were given risedronate or placebo and followed for two years [85]. The risedronate-treated groups did not show significant improvements in signs and symptoms of OA compared with placebo, and there was no significant reduction in radiographic progression.

In a hip OA study with alendronate versus placebo, significant improvements in WOMAC pain score were seen with alendronate, but there were no significant differences in structural OA progression [88]. Another trial including 223 patients with knee OA found that yearly zoledronic infusions compared with placebo did not result in statistically significant differences in cartilage volume loss over the two-year follow-up period [89].

Zoledronic acid, a once-yearly bisphosphonate infusion, may be of potential promise in OA. In the initial trial of a single infusion of zoledronic acid compared with placebo, there was a significant reduction in bone marrow lesion size at six months with an ongoing trend after 12 months [90]. A significant reduction in Visual Analogue Scale pain score was also seen at six months, which was not sustained by 12 months. Despite promising results from early studies, a larger, well-powered trial found no significant benefits in pain, bone marrow lesions, or cartilage loss with zoledronic acid [89].

Strontium — Strontium ranelate, a treatment for osteoporosis in postmenopausal women, is classified as a dual-action bone agent given its ability to both stimulate cartilage matrix formation and inhibit subchondral bone resorption [91], thereby improving the overall architecture of the bone. It has been shown to reduce the progression of radiographic features of spinal OA and back pain in females with concomitant osteoporosis and spinal OA [92]. (See "Overview of the management of low bone mass and osteoporosis in postmenopausal women", section on 'Other'.)

A three-year phase III randomized trial of patients randomly allocated to strontium ranelate 1 g/day, 2 g/day, or placebo found that treatment with strontium ranelate was associated with smaller degradations in joint space width, improved WOMAC total score, and pain subscores in the group receiving the higher dose of strontium [93]. MRI assessing cartilage volume loss and bone marrow lesions was further done in a subset of patients. Strontium ranelate dosed at 2 g/day was associated with a significant reduction in both cartilage volume loss in the plateau and bone marrow lesion progression in the medial compartment at 36 months.

Adverse effects of strontium ranelate use, including myocardial infarction, will affect its consideration for future treatment of OA [94]. (See "Overview of the management of low bone mass and osteoporosis in postmenopausal women", section on 'Other'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Osteoarthritis".)

SUMMARY

Investigational agents are not yet recommended – None of the investigational agents discussed in this topic are recommended for the treatment of osteoarthritis (OA). The major goals of treatment are pain control and prevention or slowing of structural changes to the joints. (See 'Targets for treatment' above.)

Investigational agents for pain control – Investigational agents targeting pain have focused on inhibiting nerve growth factor or disrupting the nerves supplying the affected joint through injections or radiofrequency ablation. (See 'Inhibition of nerve growth factor' above and 'Nerve block injections and radiofrequency ablation' above.)

Investigational regenerative medicine therapies – Platelet-rich plasma, stem cell therapy, and tissue gene therapy aim to stimulate local growth factors and tissue repair. (See 'Regenerative medicine therapies' above.)

Investigational agents for slowing structural changes – Drugs targeting structural changes in OA include agents that modify inflammatory pathways, cartilage catabolism and anabolism, and bone remodeling. (See 'Inflammatory pathways' above and 'Cartilage catabolism and anabolism' above and 'Bone remodeling' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Kenneth Kalunian, MD, who contributed to an earlier version of this topic review.

  1. Duong V, Hunter DJ. Osteoarthritis research is failing to reach consumers. Nat Rev Rheumatol 2023; 19:464.
  2. Jang CW, Bang M, Park JH, Cho HE. Impact of changes in clinical practice guidelines for intra-articular injection treatments for knee osteoarthritis on public interest and social media. Osteoarthritis Cartilage 2023; 31:793.
  3. Lane NE, Schnitzer TJ, Birbara CA, et al. Tanezumab for the treatment of pain from osteoarthritis of the knee. N Engl J Med 2010; 363:1521.
  4. Wood JN. Nerve growth factor and pain. N Engl J Med 2010; 363:1572.
  5. Hochberg MC, Tive LA, Abramson SB, et al. When Is Osteonecrosis Not Osteonecrosis?: Adjudication of Reported Serious Adverse Joint Events in the Tanezumab Clinical Development Program. Arthritis Rheumatol 2016; 68:382.
  6. Schnitzer TJ, Ekman EF, Spierings EL, et al. Efficacy and safety of tanezumab monotherapy or combined with non-steroidal anti-inflammatory drugs in the treatment of knee or hip osteoarthritis pain. Ann Rheum Dis 2015; 74:1202.
  7. Schnitzer TJ, Easton R, Pang S, et al. Effect of Tanezumab on Joint Pain, Physical Function, and Patient Global Assessment of Osteoarthritis Among Patients With Osteoarthritis of the Hip or Knee: A Randomized Clinical Trial. JAMA 2019; 322:37.
  8. McKenzie H. Eli Lilly and Pfizer put osteoarthritis pain drug out of its misery. PharmaLive, October 27, 2021. https://www.pharmalive.com/eli-lilly-and-pfizer-end-tanezumab-development/.
  9. Dakin P, DiMartino SJ, Gao H, 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 Rheumatol 2019; 71:1824.
  10. Sanga P, Katz N, Polverejan E, et al. Long-Term Safety and Efficacy of Fulranumab in Patients With Moderate-to-Severe Osteoarthritis Pain: A Phase II Randomized, Double-Blind, Placebo-Controlled Extension Study. Arthritis Rheumatol 2017; 69:763.
  11. Choi WJ, Hwang SJ, Song JG, et al. Radiofrequency treatment relieves chronic knee osteoarthritis pain: a double-blind randomized controlled trial. Pain 2011; 152:481.
  12. Caragea M, Woodworth T, Curtis T, et al. Genicular nerve radiofrequency ablation for the treatment of chronic knee joint pain: a real-world cohort study with evaluation of prognostic factors. Pain Med 2023; 24:1332.
  13. Shanahan EM, Robinson L, Lyne S, et al. Genicular Nerve Block for Pain Management in Patients With Knee Osteoarthritis: A Randomized Placebo-Controlled Trial. Arthritis Rheumatol 2023; 75:201.
  14. Liu J, Wang T, Zhu ZH. Efficacy and safety of radiofrequency treatment for improving knee pain and function in knee osteoarthritis: a meta-analysis of randomized controlled trials. J Orthop Surg Res 2022; 17:21.
  15. Hunter C, Davis T, Loudermilk E, et al. Cooled Radiofrequency Ablation Treatment of the Genicular Nerves in the Treatment of Osteoarthritic Knee Pain: 18- and 24-Month Results. Pain Pract 2020; 20:238.
  16. Paget LDA, Reurink G, de Vos RJ, et al. Effect of Platelet-Rich Plasma Injections vs Placebo on Ankle Symptoms and Function in Patients With Ankle Osteoarthritis: A Randomized Clinical Trial. JAMA 2021; 326:1595.
  17. Bennell KL, Paterson KL, Metcalf BR, et al. Effect of Intra-articular Platelet-Rich Plasma vs Placebo Injection on Pain and Medial Tibial Cartilage Volume in Patients With Knee Osteoarthritis: The RESTORE Randomized Clinical Trial. JAMA 2021; 326:2021.
  18. Costa LAV, Lenza M, Irrgang JJ, et al. How Does Platelet-Rich Plasma Compare Clinically to Other Therapies in the Treatment of Knee Osteoarthritis? A Systematic Review and Meta-analysis. Am J Sports Med 2023; 51:1074.
  19. Andia I, Maffulli N. Platelet-rich plasma for managing pain and inflammation in osteoarthritis. Nat Rev Rheumatol 2013; 9:721.
  20. van Buul GM, Koevoet WL, Kops N, et al. Platelet-rich plasma releasate inhibits inflammatory processes in osteoarthritic chondrocytes. Am J Sports Med 2011; 39:2362.
  21. Kanchanatawan W, Arirachakaran A, Chaijenkij K, et al. Short-term outcomes of platelet-rich plasma injection for treatment of osteoarthritis of the knee. Knee Surg Sports Traumatol Arthrosc 2016; 24:1665.
  22. Bennell KL, Hunter DJ, Paterson KL. Platelet-Rich Plasma for the Management of Hip and Knee Osteoarthritis. Curr Rheumatol Rep 2017; 19:24.
  23. Shen L, Yuan T, Chen S, et al. The temporal effect of platelet-rich plasma on pain and physical function in the treatment of knee osteoarthritis: systematic review and meta-analysis of randomized controlled trials. J Orthop Surg Res 2017; 12:16.
  24. Wong KL, Lee KB, Tai BC, et al. Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years' follow-up. Arthroscopy 2013; 29:2020.
  25. Vangsness CT Jr, Farr J 2nd, Boyd J, et al. Adult human mesenchymal stem cells delivered via intra-articular injection to the knee following partial medial meniscectomy: a randomized, double-blind, controlled study. J Bone Joint Surg Am 2014; 96:90.
  26. Lamo-Espinosa JM, Mora G, Blanco JF, et al. Intra-articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: multicenter randomized controlled clinical trial (phase I/II). J Transl Med 2016; 14:246.
  27. Kim KI, Lee MC, Lee JH, et al. Clinical Efficacy and Safety of the Intra-articular Injection of Autologous Adipose-Derived Mesenchymal Stem Cells for Knee Osteoarthritis: A Phase III, Randomized, Double-Blind, Placebo-Controlled Trial. Am J Sports Med 2023; 51:2243.
  28. Noh MJ, Copeland RO, Yi Y, et al. Pre-clinical studies of retrovirally transduced human chondrocytes expressing transforming growth factor-beta-1 (TG-C). Cytotherapy 2010; 12:384.
  29. Lee MC, Ha CW, Elmallah RK, et al. A placebo-controlled randomised trial to assess the effect of TGF-ß1-expressing chondrocytes in patients with arthritis of the knee. Bone Joint J 2015; 97-B:924.
  30. Ha CW, Cho JJ, Elmallah RK, et al. A Multicenter, Single-Blind, Phase IIa Clinical Trial to Evaluate the Efficacy and Safety of a Cell-Mediated Gene Therapy in Degenerative Knee Arthritis Patients. Hum Gene Ther Clin Dev 2015; 26:125.
  31. Kim MK, Ha CW, In Y, et al. A Multicenter, Double-Blind, Phase III Clinical Trial to Evaluate the Efficacy and Safety of a Cell and Gene Therapy in Knee Osteoarthritis Patients. Hum Gene Ther Clin Dev 2018; 29:48.
  32. Fioravanti A, Fabbroni M, Cerase A, Galeazzi M. Treatment of erosive osteoarthritis of the hands by intra-articular infliximab injections: a pilot study. Rheumatol Int 2009; 29:961.
  33. Güler-Yüksel M, Allaart CF, Watt I, et al. Treatment with TNF-α inhibitor infliximab might reduce hand osteoarthritis in patients with rheumatoid arthritis. Osteoarthritis Cartilage 2010; 18:1256.
  34. Magnano MD, Chakravarty EF, Broudy C, et al. A pilot study of tumor necrosis factor inhibition in erosive/inflammatory osteoarthritis of the hands. J Rheumatol 2007; 34:1323.
  35. Chevalier X, Ravaud P, Maheu E, et al. Adalimumab in patients with hand osteoarthritis refractory to analgesics and NSAIDs: a randomised, multicentre, double-blind, placebo-controlled trial. Ann Rheum Dis 2015; 74:1697.
  36. Verbruggen G, Wittoek R, Vander Cruyssen B, Elewaut D. Tumour necrosis factor blockade for the treatment of erosive osteoarthritis of the interphalangeal finger joints: a double blind, randomised trial on structure modification. Ann Rheum Dis 2012; 71:891.
  37. Pelletier JP, Caron JP, Evans C, et al. In vivo suppression of early experimental osteoarthritis by interleukin-1 receptor antagonist using gene therapy. Arthritis Rheum 1997; 40:1012.
  38. Santangelo KS, Nuovo GJ, Bertone AL. In vivo reduction or blockade of interleukin-1β in primary osteoarthritis influences expression of mediators implicated in pathogenesis. Osteoarthritis Cartilage 2012; 20:1610.
  39. Chevalier X, Goupille P, Beaulieu AD, et al. Intraarticular injection of anakinra in osteoarthritis of the knee: a multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum 2009; 61:344.
  40. Cohen SB, Proudman S, Kivitz AJ, et al. A randomized, double-blind study of AMG 108 (a fully human monoclonal antibody to IL-1R1) in patients with osteoarthritis of the knee. Arthritis Res Ther 2011; 13:R125.
  41. Schieker M, Conaghan PG, Mindeholm L, et al. Effects of Interleukin-1β Inhibition on Incident Hip and Knee Replacement : Exploratory Analyses From a Randomized, Double-Blind, Placebo-Controlled Trial. Ann Intern Med 2020; 173:509.
  42. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med 2017; 377:1119.
  43. Doorenspleet ME, Klarenbeek PL, de Hair MJ, et al. Rheumatoid arthritis synovial tissue harbours dominant B-cell and plasma-cell clones associated with autoreactivity. Ann Rheum Dis 2014; 73:756.
  44. Schnitzer TJ. New pharmacologic approaches in the management of osteoarthritis. Arthritis Care Res (Hoboken) 2010; 62:1174.
  45. Hellio le Graverand MP, Clemmer RS, Redifer P, et al. A 2-year randomised, double-blind, placebo-controlled, multicentre study of oral selective iNOS inhibitor, cindunistat (SD-6010), in patients with symptomatic osteoarthritis of the knee. Ann Rheum Dis 2013; 72:187.
  46. Song IH, Althoff CE, Hermann KG, et al. Contrast-enhanced ultrasound in monitoring the efficacy of a bradykinin receptor 2 antagonist in painful knee osteoarthritis compared with MRI. Ann Rheum Dis 2009; 68:75.
  47. Meini S, Maggi CA. Knee osteoarthritis: a role for bradykinin? Inflamm Res 2008; 57:351.
  48. Gomis A, Meini S, Miralles A, et al. Blockade of nociceptive sensory afferent activity of the rat knee joint by the bradykinin B2 receptor antagonist fasitibant. Osteoarthritis Cartilage 2013; 21:1346.
  49. Heijman MWJ, Fiolet ATL, Mosterd A, et al. Association of Low-Dose Colchicine With Incidence of Knee and Hip Replacements : Exploratory Analyses From a Randomized, Controlled, Double-Blind Trial. Ann Intern Med 2023; 176:737.
  50. Leung YY, Haaland B, Huebner JL, et al. Colchicine lack of effectiveness in symptom and inflammation modification in knee osteoarthritis (COLKOA): a randomized controlled trial. Osteoarthritis Cartilage 2018; 26:631.
  51. Griffith DL, Keck PC, Sampath TK, et al. Three-dimensional structure of recombinant human osteogenic protein 1: structural paradigm for the transforming growth factor beta superfamily. Proc Natl Acad Sci U S A 1996; 93:878.
  52. Elshaier AM, Hakimiyan AA, Rappoport L, et al. Effect of interleukin-1beta on osteogenic protein 1-induced signaling in adult human articular chondrocytes. Arthritis Rheum 2009; 60:143.
  53. Hunter DJ, Pike MC, Jonas BL, et al. Phase 1 safety and tolerability study of BMP-7 in symptomatic knee osteoarthritis. BMC Musculoskelet Disord 2010; 11:232.
  54. Moore EE, Bendele AM, Thompson DL, et al. Fibroblast growth factor-18 stimulates chondrogenesis and cartilage repair in a rat model of injury-induced osteoarthritis. Osteoarthritis Cartilage 2005; 13:623.
  55. Ellman MB, An HS, Muddasani P, Im HJ. Biological impact of the fibroblast growth factor family on articular cartilage and intervertebral disc homeostasis. Gene 2008; 420:82.
  56. Lohmander LS, Hellot S, Dreher D, et al. Intraarticular sprifermin (recombinant human fibroblast growth factor 18) in knee osteoarthritis: a randomized, double-blind, placebo-controlled trial. Arthritis Rheumatol 2014; 66:1820.
  57. Dahlberg LE, Aydemir A, Muurahainen 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. Clin Exp Rheumatol 2016; 34:445.
  58. Hochberg MC, Guermazi A, Guehring H, et al. Effect of Intra-Articular Sprifermin vs Placebo on Femorotibial Joint Cartilage Thickness in Patients With Osteoarthritis: The FORWARD Randomized Clinical Trial. JAMA 2019; 322:1360.
  59. Guehring H, Moreau F, Daelken B, et al. The effects of sprifermin on symptoms and structure in a subgroup at risk of progression in the FORWARD knee osteoarthritis trial. Semin Arthritis Rheum 2021; 51:450.
  60. Davidson RK, Waters JG, Kevorkian L, et al. Expression profiling of metalloproteinases and their inhibitors in synovium and cartilage. Arthritis Res Ther 2006; 8:R124.
  61. Ohuchi E, Imai K, Fujii Y, et al. Membrane type 1 matrix metalloproteinase digests interstitial collagens and other extracellular matrix macromolecules. J Biol Chem 1997; 272:2446.
  62. Krzeski P, Buckland-Wright C, Bálint G, et al. Development of musculoskeletal toxicity without clear benefit after administration of PG-116800, a matrix metalloproteinase inhibitor, to patients with knee osteoarthritis: a randomized, 12-month, double-blind, placebo-controlled study. Arthritis Res Ther 2007; 9:R109.
  63. Tu G, Xu W, Huang H, Li S. Progress in the development of matrix metalloproteinase inhibitors. Curr Med Chem 2008; 15:1388.
  64. Verma P, Dalal K. ADAMTS-4 and ADAMTS-5: key enzymes in osteoarthritis. J Cell Biochem 2011; 112:3507.
  65. Yang CY, Chanalaris A, Troeberg L. ADAMTS and ADAM metalloproteinases in osteoarthritis - looking beyond the 'usual suspects'. Osteoarthritis Cartilage 2017; 25:1000.
  66. Schnitzer T, Pueyo M, Deckx H, et al. Evaluation of S201086/GLPG1972, an ADAMTS-5 inhibitor, for the treatment of knee osteoarthritis in ROCCELLA: a phase 2 randomized clinical trial. Osteoarthritis Cartilage 2023; 31:985.
  67. Loeser RF, Erickson EA, Long DL. Mitogen-activated protein kinases as therapeutic targets in osteoarthritis. Curr Opin Rheumatol 2008; 20:581.
  68. Starkman BG, Cravero JD, Delcarlo M, Loeser RF. IGF-I stimulation of proteoglycan synthesis by chondrocytes requires activation of the PI 3-kinase pathway but not ERK MAPK. Biochem J 2005; 389:723.
  69. Zhang J, Shen B, Lin A. Novel strategies for inhibition of the p38 MAPK pathway. Trends Pharmacol Sci 2007; 28:286.
  70. Salminen-Mankonen HJ, Morko J, Vuorio E. Role of cathepsin K in normal joints and in the development of arthritis. Curr Drug Targets 2007; 8:315.
  71. Lindström E, Rizoska B, Tunblad K, et al. The selective cathepsin K inhibitor MIV-711 attenuates joint pathology in experimental animal models of osteoarthritis. J Transl Med 2018; 16:56.
  72. Hayami T, Zhuo Y, Wesolowski GA, et al. Inhibition of cathepsin K reduces cartilage degeneration in the anterior cruciate ligament transection rabbit and murine models of osteoarthritis. Bone 2012; 50:1250.
  73. Rünger TM, Adami S, Benhamou CL, et al. Morphea-like skin reactions in patients treated with the cathepsin K inhibitor balicatib. J Am Acad Dermatol 2012; 66:e89.
  74. Conaghan PG, Bowes MA, Kingsbury SR, et al. Disease-Modifying Effects of a Novel Cathepsin K Inhibitor in Osteoarthritis: A Randomized Controlled Trial. Ann Intern Med 2020; 172:86.
  75. Luyten FP, Tylzanowski P, Lories RJ. Wnt signaling and osteoarthritis. Bone 2009; 44:522.
  76. Kahn M. Can we safely target the WNT pathway? Nat Rev Drug Discov 2014; 13:513.
  77. Wang Y, Fan X, Xing L, Tian F. Wnt signaling: a promising target for osteoarthritis therapy. Cell Commun Signal 2019; 17:97.
  78. Yazici Y, McAlindon TE, Gibofsky A, et al. 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 Rheumatol 2020; 72:1694.
  79. Yazici Y, McAlindon TE, Gibofsky A, et al. A Phase 2b randomized trial of lorecivivint, a novel intra-articular CLK2/DYRK1A inhibitor and Wnt pathway modulator for knee osteoarthritis. Osteoarthritis Cartilage 2021; 29:654.
  80. Sabha M, Siaton BC, Hochberg MC. Lorecivivint, an intra-articular potential disease-modifying osteoarthritis drug. Expert Opin Investig Drugs 2020; 29:1339.
  81. Karsdal MA, Sondergaard BC, Arnold M, Christiansen C. Calcitonin affects both bone and cartilage: a dual action treatment for osteoarthritis? Ann N Y Acad Sci 2007; 1117:181.
  82. Li J, Xie ZG, Xie Y, Dong QR. Calcitonin treatment is associated with less severe osteoarthritis and reduced toll-like receptor levels in a rat model. J Orthop Sci 2014; 19:1019.
  83. Manicourt DH, Azria M, Mindeholm L, et al. Oral salmon calcitonin reduces Lequesne's algofunctional index scores and decreases urinary and serum levels of biomarkers of joint metabolism in knee osteoarthritis. Arthritis Rheum 2006; 54:3205.
  84. Karsdal MA, Byrjalsen I, Alexandersen P, et al. Treatment of symptomatic knee osteoarthritis with oral salmon calcitonin: results from two phase 3 trials. Osteoarthritis Cartilage 2015; 23:532.
  85. Bingham CO 3rd, Buckland-Wright JC, Garnero P, et al. Risedronate decreases biochemical markers of cartilage degradation but does not decrease symptoms or slow radiographic progression in patients with medial compartment osteoarthritis of the knee: results of the two-year multinational knee osteoarthritis structural arthritis study. Arthritis Rheum 2006; 54:3494.
  86. Saag KG. Bisphosphonates for osteoarthritis prevention: "Holy Grail" or not? Ann Rheum Dis 2008; 67:1358.
  87. Spector TD, Conaghan PG, Buckland-Wright JC, et al. Effect of risedronate on joint structure and symptoms of knee osteoarthritis: results of the BRISK randomized, controlled trial [ISRCTN01928173]. Arthritis Res Ther 2005; 7:R625.
  88. Nishii T, Tamura S, Shiomi T, et al. Alendronate treatment for hip osteoarthritis: prospective randomized 2-year trial. Clin Rheumatol 2013; 32:1759.
  89. Cai G, Aitken D, Laslett LL, et al. Effect of Intravenous Zoledronic Acid on Tibiofemoral Cartilage Volume Among Patients With Knee Osteoarthritis With Bone Marrow Lesions: A Randomized Clinical Trial. JAMA 2020; 323:1456.
  90. Laslett LL, Doré DA, Quinn SJ, et al. Zoledronic acid reduces knee pain and bone marrow lesions over 1 year: a randomised controlled trial. Ann Rheum Dis 2012; 71:1322.
  91. Reginster JY, Badurski J, Bellamy N, et al. Efficacy and safety of strontium ranelate in the treatment of knee osteoarthritis: results of a double-blind, randomised placebo-controlled trial. Ann Rheum Dis 2013; 72:179.
  92. Bruyere O, Delferriere D, Roux C, et al. Effects of strontium ranelate on spinal osteoarthritis progression. Ann Rheum Dis 2008; 67:335.
  93. Pelletier JP, Roubille C, Raynauld JP, et al. Disease-modifying effect of strontium ranelate in a subset of patients from the Phase III knee osteoarthritis study SEKOIA using quantitative MRI: reduction in bone marrow lesions protects against cartilage loss. Ann Rheum Dis 2015; 74:422.
  94. Protelos and Osseor. European Medicines Agency. Available at: https://www.ema.europa.eu/en/medicines/human/referrals/protelos-osseor (Accessed on June 11, 2023).
Topic 5493 Version 44.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟