Abstract
As the aging population grows and lifestyle factors become more prevalent, the incidence of knee osteoarthritis (KOA) is expected to continue to increase in the coming decades. This presents a substantial public health challenge with an impact on the quality of life of the affected individuals. The absence of targeted therapeutic interventions tailored specifically for KOA underscores the recognition of this condition as a significant medical concern characterized by an urgent unmet need for effective treatments. Despite advances in understanding its pathophysiology and progression, there remains a gap in the availability of therapies capable of adequately addressing the diverse clinical manifestations and underlying mechanisms of KOA. Fortunately, numerous novel targeted therapies, including biological, nanotechnology, gene, and cell therapies, are currently undergoing clinical trials for KOA treatment. Advancements in drug nanocarriers and delivery systems have demonstrated the potential to enhance the efficacy of therapeutic agents for KOA. In this review, we summarize all the advancements in targeted therapy for KOA, including small-molecule inhibitors, monoclonal antibodies, nanocarrier-based therapy, gene therapy and cytotherapy. By analyzing the latest breakthroughs in pharmaceutical therapies and relevant clinical data, this review serves as a valuable resource for clinicians and researchers involved in the ongoing quest for effective KOA treatments and provides hopes for improved management and outcomes for patients with this debilitating condition.
Keywords: clinical trials, inhibitors, knee osteoarthritis, nanocarriers, targeted therapy
1. Introduction
Osteoarthritis (OA) is recognized as the predominant degenerative arthropathy, characterized by intricate dysregulation of the entire synovial joint, encompassing structural aberrations in hyaline articular cartilage, depletion of integral subchondral bone, tissue hypertrophy, heightened vascularity in the synovium, and instability in tendons and ligaments, affecting a global population exceeding 500 million individuals.[1,2] Knee osteoarthritis (KOA), the most common type of OA, constitutes approximately 85% of the total prevalence of OA cases worldwide, thereby presenting a substantial public health challenge anticipated to persist in the forthcoming decades.[3,4] KOA exerts its influence not only on the cartilage but also on the entirety of the joint structure[5] (Fig. 1A). The central characteristic of KOA is the degradation of articular cartilage, in which an imbalanced biomechanical microenvironment and diverse biological factors disrupt cartilage homeostasis, leading to degradation of the extracellular matrix (ECM) enriched in collagen and proteoglycans, as well as articular surface fibrosis, cell death, vascular invasion, and so on.[6] This progressive destruction stimulates chondrocytes to augment anabolism through compensatory hypertrophy, resulting in the concurrent generation of matrix degradation products and pro-inflammatory mediators, thereby expediting the development of KOA. In addition, the involvement of the tissues in and around the joint is frequently followed by the KOA progression.[7] These multifactorial changes collectively worsen the clinical symptoms of KOA and complicate its management.
Figure 1.
Phenotypes of KOA (A) and inhibitors for KOA therapy binding with the effector molecules in cartilage cells (B) to regulate the expression of proteins and genes (C). KOA = knee osteoarthritis.
Over an extended period, significant efforts and resources have been dedicated to the development of therapies aimed at enhancing care, quality of life, and pain relief in individuals with KOA. Non-pharmaceutical approaches, which include patient education, exercise, and weight management, constitute fundamental elements in the management of KOA.[8,9] Pharmaceutical strategies have primarily focused on alleviating symptoms and attempting to impede or halt the underlying biological processes that contribute to tissue damage. Despite numerous studies, a conclusive treatment for KOA remains elusive, and there are currently no medications to halt the progression of KOA. Current guidelines derive recommendations for diverse OA treatment modalities from medical literature. Nonsteroidal anti-inflammatory drugs (NSAIDs), analgesics, and intra-articular corticosteroid injections show limited efficacy in the extended duration, whereas the use of opioids may lead to adverse outcomes.[10–12] In recent years, innovative treatments have emerged owing to enhanced insights into KOA and the rapid development in nanotechnology and drug delivery system. Moreover, scientists and clinicians have invested substantial efforts in elucidating pivotal molecules, signaling pathways, biological processes and the suitable time of the intervention, which holds promise as a potential therapeutic targets to attenuate or limit damage to synovial joints.[2,13]
In this review, we perform a comprehensive search of English-language articles using databases such as PubMed, Google Scholar, and the NIH, containing the following terms: “knee osteoarthritis,” “targeted therapy,” “‘clinical trials,’” “inhibitors,” and “nanocarriers” and explore all the advancements in targeted therapy for KOA, as well as the relevant clinical data, elucidating the efficacy and safety profiles of these novel therapeutic approaches in clinical settings. Additionally, we engage in a comprehensive discussion regarding the main challenges that need to be addressed for the continued advancement and refinement of these targeted therapeutic strategies, aiming to provide a deep understanding of the current landscape of KOA therapy and offer valuable insights for future research and clinical practice in the field.
2. Management of KOA
2.1. Pharmaceutical approaches
Clinically pharmaceutical approaches to KOA have primarily concentrated on alleviating symptoms or modifying the disease by topical, oral, or injectable administration (Table 1). First-line medications, such as NSAIDs, analgesics, and glucocorticoids, have been shown to be effective for symptom control.[14,15] However, there are notable cardiovascular, gastrointestinal, and renal risks associated with the oral administration of NSAIDs. Concerns have also arisen regarding the effectiveness and safety of opioid analgesics in oral therapy. Consistent daily use of opioid analgesics can lead to a degree of tolerance and physical dependence.[16] It is essential to acknowledge that acetaminophen is no longer the first-line recommended analgesic in guidelines, primarily because of its effectiveness and the presence of various unwanted side effects.[17,18] An additional category of commonly employed medications falls under the designation of cartilage protectors, encompassing products containing substances like glucosamine,[19,20] chondroitin sulfate[20] and collagen hydrolysates.[21] Despite of the fact that many studies have suggested anti-inflammatory and analgesic effects of these cartilage protectors in KOA, leading to relief from clinical symptoms and a deceleration in disease progression, the observed effects are only marginally better than those of a placebo. Limited favorable evidence and relevance support these findings.[22,23] Therefore, some clinical guidelines have issued either negative or weak recommendations for all glucosamine and chondroitin products, including those of pharmaceutical grade.[17] Several innovative targeted therapies for KOA have been developed to enhance drug delivery specificity and efficacy. We will discuss these in detail in the following “Targeted therapy strategies” section.
Table 1.
Management and mechanisms of KOA.
| Management of KOA | Mechanisms of KOA | |||
|---|---|---|---|---|
| Pharmaceutical approaches | Non-pharmaceutical approaches | Surgical management | Pathogenesis | Molecular mechanisms of KOA |
| NSAIDS | Health education | OAK | Excessive proliferation of chondrocytes | MMPs, ADAMTS↑ |
| Analgesics | Regular exercise including aerobic, strengthening, and resistance exercises | UKA | Abundance of ECM | IL-1β, IL-6, TNF-α↑ |
| Glucocorticoids | Weight loss | TKA | Synovial inflammation | NF-κB, MAPK, PI3K/Akt signaling pathway |
| Targeted therapy strategies | Physical therapy including therapeutic ultrasound, electrical stimulation, phototherapy, hydrotherapy, magnet therapy, acupuncture and moxibustion | – | Breakdown of cartilage tissue | – |
ADAMTS = a disintegrin and metalloproteinase with thrombospondin motifs, ECM = extracellular matrix, IL-1β = interleukin-1, IL-6 = interleukin-6, KOA = knee osteoarthritis, MMPs = matrix metalloproteinases, NSAIDs = nonsteroidal anti-inflammatory drugs, OAK = osteotomy around the knee, TKA = total knee arthroplasty, TNF-α = tumor necrosis factor-alpha, UKA = uknee arthroplasty.
2.2. Non-pharmaceutical approaches
Virtually all guidelines endorse the incorporation of health education, regular exercise and weight loss throughout the distinct pathological stages of KOA (Table 1), which should precede the initiation of first-line treatment.[17] The recommendations from the guidelines support the inclusion of aerobic, strengthening, and resistance exercises.[24] Nevertheless, the impact of exercise intensity on the outcomes of KOA rehabilitation, particularly in the acute stage, remains unclear. Improper exercise prescription may exacerbate KOA.[25] Weight loss contributes to a decrease in the pressure on the knee joint, leading to improvements in physical function and biomechanics when combined with exercise. Additionally, physical therapy has shown a significant therapeutic effect on KOA through modalities such as therapeutic ultrasound, electrical stimulation, phototherapy, hydrotherapy, and magnet therapy, which can effectively alleviate symptoms.[26] This renders it suitable for emergency management during the acute phase of KOA.[27] Acupuncture and moxibustion as physical therapies in traditional Chinese medicine also play specific roles in pain relief and functional restoration in the treatment of KOA.[28]
2.3. Surgical management
Osteotomy around the knee as a conservative treatment offers value in the surgical management of KOA in younger patients[29] (Table 1). This approach facilitates significant improvements in pain reduction and functional outcomes without the need for irreversible arthroplasty.[30] The preservation of the joint’s anatomical structure through osteotomy offers advantages such as maintaining proprioception and facilitating swift recovery of joint functional efficacy. This, in turn, significantly slows the progression of KOA. Knee arthroplasty is a nonconservative surgical management for severe KOA and is regarded as a secure, widely accepted, and cost-effective therapeutic intervention, offering enduring benefits.[30] Unquestionably, unicompartmental knee arthroplasty has evolved as the established surgical strategy for individuals with total cartilage loss limited to a single femorotibial compartment.[31,32] Upon satisfying the appropriate criteria and adhering to surgical principles, full knee functionality can be restored using unicompartmental knee arthroplasty. In cases of severe arthritis, consideration for total knee arthroplasty is warranted, contingent upon the failure of nonoperative interventions.[33]
3. Molecular mechanisms of KOA
KOA is primarily characterized by the excessive proliferation of chondrocytes, which are specialized cells within cartilage tissue, coupled with a gradual decline in the abundance of ECM proteins, leading to the breakdown of cartilage tissue, contributing to joint degeneration and dysfunction (Table 1). Among the aberrant molecular mechanisms of KOA, there is a notable upregulation in the synthesis of degrading proteases, including matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) (Fig. 1C). Specifically, these enzymes target type II collagen, a key structural component of cartilage that leads to cartilage degradation.[34] Interleukin-1 (IL-1β), which is primarily secreted by chondrocytes, osteoblasts, synovial cells, and leukocytes, is involved in the pathogenesis of KOA. IL-1β triggers downstream signaling cascades involving NF-κB and MAPK pathways by binding to its receptor on synoviocytes (Fig. 2C), and then induces the transcription of genes encoding inflammatory mediators, including MMPs and cyclooxygenase-2 (COX-2), which are responsible for cartilage degradation.[35,36] Similarly, Interleukin-6 (IL-6) collectively accelerates KOA progression by stimulating MMP production, promoting cartilage permeability, and enhancing osteoclast formation.[37] Additionally, upon binding to its receptors, tumor necrosis factor-alpha (TNF-α) can activates the NF-κB and PI3K/Akt signaling pathways to initiate a cascade of events that culminate in the production of pro-inflammatory mediators.[38] These findings highlight the possible molecular mechanisms mediating various aspects of KOA pathophysiology and underscore its potential as a therapeutic target for disease management.
Figure 2.
Schematic of nanocarrier-based therapy. (A) the various nanocarriers loading active compounds, (B) intra-articular administration for targeted therapy of KOA, (C) the therapeutic effects are obtained by the inhibition of NF-κB and MAPK signaling pathway. KOA = knee osteoarthritis.
4. Targeted therapy strategies
Several innovative approaches have been devised to enhance drug delivery specificity and efficacy in joint tissues for KOA according to aberrant molecular mechanisms. Active and passive targeting techniques have been tailored to target specific cells and tissues within the joint, such as cartilage, synovium/synoviocytes, chondrocytes, and the joint microenvironment, including the synovial fluid[39] (Fig. 1B). These targeted therapeutic strategies aim to improve drug retention and concentration at the joint by leveraging the unique characteristics and physiological properties of joint tissues, such as cell surface markers or tissue-specific receptors, thereby enhancing therapeutic outcomes while minimizing off-target effects. Herein, we summarize the recent advancements in targeted therapy for KOA, with a focus on a diverse array of therapeutic approaches, including small-molecule inhibitors, monoclonal antibodies, nanocarrier-based therapy, gene therapy and cytotherapy.
4.1. Small-molecule inhibitors
Small-molecule inhibitors, characterized by their molecular weight of <1000 Da, constitute a fundamental class of organic compounds widely utilized in biomedical research and targeted drug development.[40] Small-molecule inhibitors offer a versatile approach for manipulating cellular processes and influencing disease pathways.[41] Their relatively small size allows them to penetrate cellular membranes efficiently, enabling them to interact with intracellular targets and exert precise regulatory effects on biological pathways.[42] Notably, the inhibitors exhibited promising inhibitory effects on KOA progression, suggesting their utility in mitigating the disease’s pathological processes and offering potential avenues for therapeutic intervention in KOA.[43,44] Among them, MMPs inhibitors, ADAMTS inhibitors, Cathepsin K inhibitor and Wingless-type MMTV integration site family (Wnt) signaling inhibitors have been well evaluated, offering potential therapeutic benefits for KOA patients.
4.1.1. MMPs inhibitors
MMPs are a family of enzymes implicated in KOA owing to their involvement in cartilage degradation.[45,46] The specific mechanism involves MMPs breaking down the extracellular matrix components of cartilage, contributing to its deterioration in KOA. Consequently, the development of synthetic small-molecule inhibitors targeting MMPs holds promise as a potential therapeutic strategy for KOA (Fig. 1C). By inhibiting MMP activity, these molecules aim to mitigate cartilage degradation and thereby alleviate KOA symptoms, thereby offering a potential route for disease management and treatment. As recently reported, the allosteric MMP-13 inhibitor, AQU-019 is a promising candidate that has been optimized to enhance its potency, metabolic stability, and oral bioavailability.[47] However, available preclinical data remains insufficient to support their commercialization.
4.1.2. ADAMTS inhibitors
ADAMTS is also known to play crucial roles in the degradation of cartilage matrix components, contributing to the progression of KOA.[48] Consequently, the development of small-molecule inhibitors targeting ADAMTS has emerged as a logical therapeutic approach for mitigating cartilage degradation and attenuating KOA progression (Fig. 1C).[49] Inhibition of ADAMTS presents a potentially safer alternative to targeting MMP in the context of KOA.[50,51] GLPG1972/S201086, identified as a small-molecule inhibitor targeting ADAMTS5, exhibits promising chondroprotective properties in preclinical models of OA.[52,53] In phase I clinical trials involving healthy individuals (NCT03311009, https://clinicaltrials.gov/), GLPG1972/S201086 demonstrated a favorable safety profile, indicating its tolerability.[54,55] In a global phase II clinical trials, significant reductions in cartilage thickness of patients for 52 weeks were observed, indicating that GLPG1972/S201086 can be considered a notable structural advantage of a disease-modifying drug candidate for KOA (NCT03595618). However, the study did not achieve its primary and secondary endpoints, and no dose-response relationship was observed.[56] The treatment was generally well-tolerated, but the lack of efficacy led to the discontinuation of OA development. While GLPG1972/S201086 did not demonstrate efficacy as a monotherapy for OA, its potential synergies with other therapeutic agents have not been fully explored. Combining it with other treatments could theoretically enhance therapeutic outcomes, however, further research is necessary to evaluate such combinations to explore their potential for clinical application.
4.1.3. Cathepsin K inhibitor
Cathepsin K, a cysteine protease, exhibits predominant expression within osteoclasts, playing a pivotal role in bone resorption by facilitating the degradation of bone matrix proteins (Fig. 1C).[57,58] In preclinical studies, MIV-711 a cathepsin K inhibitor, was shown to reduce cartilage damage and preventing bone resorption in animal models.[59] Moreover, it has been shown to decrease biomarkers associated with bone and cartilage remodeling, suggesting its potential as a protease inhibitor for the management of KOA progression. In a phase II clinical trial involving individuals with symptomatic KOA, MIV-711 treatment demonstrated notable outcomes (NCT02705625). MIV-711 was generally well-tolerated over both the initial 26-week period and subsequent 6-month extension. Serious adverse events were infrequent and none were deemed treatment-related. Specifically, patients receiving MIV-711 exhibited reduced bone remodeling, as evidenced by magnetic resonance imaging assessments indicating decreased bone area, compared to those receiving a placebo. Moreover, over the 26-week study period, individuals treated with MIV-711 experienced less cartilage loss than those in the placebo group.[60] A primary limitation of these studies was their relatively short duration, which may have been insufficient to observe symptom benefits following structural modification. Additionally, the lack of statistically significant pain reduction despite structural improvements suggests that longer treatment periods or combination therapies may be necessary to achieve symptomatic relief. Given the ability of MIV-711 to modify the joint structure, combining it with agents that provide symptomatic relief could offer a comprehensive treatment approach for OA. Further research is warranted to explore such combination therapies and determine whether extended treatment durations with MIV-711 alone can yield significant symptom improvement in larger-scale clinical trials.
4.1.4. Wnt signaling inhibitors
Wnt signaling pathways, primarily mediated by β-catenin and involving a myriad of proteins, play multifaceted roles in various physiological processes not only within joints but also in other tissues throughout the body.[61] The widespread involvement of Wnt signaling complicates the selective targeting of this pathway for the treatment of KOA. By modulating Wnt-related proteins, interventions may mitigate the aberrant signaling cascades implicated in KOA development and progression.[62,63] Lorecivivint (LOR), an inhibitor of the Wnt signaling pathway, was discovered using high-throughput screening methods. Preclinical studies have shown that the inhibitory effects of LOR on the Wnt signaling pathway contribute to its anti-inflammatory and chondroprotective properties. Interestingly, these effects appear to be independent of β-catenin and instead involve the inhibition of 2 intranuclear kinases: cdc2-like kinase (CLK2) and tyrosine phosphorylation-regulated kinase (DYRK1A).[64] Moreover, phase I-III clinical trials for LOR have been completed (NCT02536833, NCT03122860, NCT04385303, NCT03928184, NCT05603754) and the results showed that the administration of LOR via intra-articular injection had a favorable safety profile, without indications of systemic drug exposure. Additionally, LOR led to improvements in KOA symptoms, including pain reduction and enhanced joint function, compared with placebo. Moreover, imaging analyses have suggested potential disease-modifying effects, such as reduced cartilage degradation.[65–68] While the results are promising, the study duration was limited to 52 weeks, which may not capture long-term efficacy and safety outcomes. Further studies are necessary to confirm the sustained benefits and monitor any potential long-term adverse effects.
4.1.5. Others
In addition to MMPs, ADAMTS, Cathepsin K inhibitors, Wnt signaling inhibitors, and various other small-molecule inhibitors, including IL-6, TNF and NF-κB signaling pathways, have demonstrated potential in inhibiting the onset and progression of KOA. Recently, a small-molecule compound, SC75741, was reported to exhibit protective effects against articular joint destruction in preclinical studies by inhibiting key inflammatory mediators, including NF-κB, TNF-α, and IL-6. By targeting the NF-κB signaling pathway, SC75741 exhibited therapeutic potential as a small-molecule inhibitor capable of attenuating miR-21/NF-κB-driven KOA progression.[69,70] Further investigation of its mechanism of action and safety profile is warranted to validate its clinical utility in KOA management. Indeed, by employing advanced screening methodologies, such as combinatorial chemistry and computational modeling techniques, researchers have identified promising small-molecule inhibitors that exhibit inhibitory activity against key enzymes or signaling pathways implicated in KOA progression. These findings highlight the potential of high-throughput screening as a valuable approach for discovering novel therapeutics for KOA and advancing our understanding of the molecular mechanisms underlying the disease.[71–73]
4.2. Monoclonal antibodies
Monoclonal antibodies (mAbs) have also been developed as versatile therapeutic agents for modulating cellular processes and impacting KOA pathology. They selective target specific proteins or interfer with crucial biochemical reactions, including ADAMTS,[51,74,75] Interleukins (ILs),[76,77] TNF,[78] nerve growth factor (NGF),[79] and CC-chemokine ligand 17 (CCL17).[80,81] MAbs offer a precise and potent means of modulating disease pathways. Through their ability to bind with high affinity to specific antigens, mAbs can block key signaling pathways involved in KOA pathogenesis, thereby attenuating inflammation, cartilage degradation, and joint damage. Their specificity and targeted action make them promising candidates for the development of novel KOA treatments with potentially fewer off-target effects than small-molecule inhibitors. Many clinical trials have been performed for the evaluation of mAbs, including NCT03583346 for neutralizing antibodies against ADAMTS, NCT03304379 and NCT02528188 for Fasinumab and Tanezumab (neutralizing antibodies against NGF), and NCT03485365 for anti-CCL17 antibodies. However, no mAbs have been successfully approved for use as alternative clinical therapies for KOA. Tanezumab, the most promising neutralizing antibody, was announced that a Phase III study showed the 10 mg dose of tanezumab significantly improved chronic low back pain at 16 weeks compared to placebo (NCT02528253). Some Phase III trials have reported that tanezumab effectively reduced hip pain (NCT00863304) and pain associated with KOA (NCT02528188). However, its use raised concerns regarding joint-related adverse events, leading to the denial of its application by the Food and Drug Administration (FDA) during phase III studies.[82] Similarly, Fasinumab, another monoclonal antibody targeting NGF, was developed to manage pain associated with OA. A clinical trial identified by NCT03304379 evaluated its efficacy and safety in patients with moderate-to-severe KOA. However, Fasinumab faces challenges and uncertainties regarding its safety profile and clinical outcomes, posing dilemmas for further development and regulatory approval. Therefore, the primary limitation of Tanezumab and Fasinumab lies in their safety profile, particularly the risk of joint-related adverse events, which has led to the suspension of certain clinical trials and regulatory hesitancy. Additionally, the variability in efficacy between different dosages, suggests that the optimal dosing requires further investigation. In addition, as with any monoclonal antibody therapy, there is a possibility of immunogenicity, which could affect the long-term efficacy and safety.
4.3. Nanocarrier-based therapy
4.3.1. Nanoparticles
Nanoparticles (NPs) are frequently employed in studies focusing on targeted therapy of KOA by enhancing drug penetration across the cartilage matrix regulating drug pharmacokinetics, improving efficacy, and reducing the toxicity of therapeutic agents (Fig. 2A and B).[83–85] Through the manipulation of physicochemical properties or surface modifications with specific moieties, NPs can be engineered with functional groups to selectively target components or cells in KOA therapy. Natural polymers, such as chitosan,[86,87] silk fibroin,[88,89] albumin,[90,91] and chemically synthesized nanomaterials, such as poly (lactic-co-glycolic) acid (PLGA),[92,93] polylactic acid (PLA),[94] polyurethane,[95] and different polymer combinations,[96] are the most frequently used to prepare NPs for drug delivery in KOA therapy. In addition to chemically polymeric nanoparticles, organometallic or inorganic materials have also been utilized to form nanoparticles for targeted drug delivery in KOA therapy. Hollow mesoporous silica nanoparticles capped with chitosan to construct pH-responsive nanoparticle are recognized as promising entities in the field of KOA therapy.[97]
Recently, there has been a notable increase in the development of multifunctional NPs for KOA therapy to amplify the targeting profile and reduce side effects, including microenvironment sensitive NPs,[98,99] ligand-modified NPs,[100] antibody-modified NPs,[101] magnetic NPs,[102] and bionic NPs.[103] Among these, the most prevalent and effective strategy is cartilage targeting, which involves the modification of NPs with specific functional groups or ligands that possess an inherent affinity for cartilage tissue. By tailoring the surface properties of NPs through the incorporation of cartilage-targeting moieties, such as peptides or antibodies, researchers aim to enhance the specificity and selectivity of drug delivery to affected joint tissues. As reported in the literature, drug delivery systems based on NPs hold potential for KOA therapy, offering benefits such as specific drug distribution, prolonged drug release, and enhanced drug retention. To date, however, there have been no reports on the use of NPs as drug carriers in clinical trials for KOA. The complex preparation procedures associated with NPs and potential toxicity concerns may hinder their clinical application for KOA treatment.
4.3.2. Liposomes
Liposomes possess a unique bilayer membrane, where the hydrophilic head groups face the aqueous environment, whereas the lipophilic tails are sequestered within the interior (Fig. 2A).[104] This amphiphilic nature enables liposomes to efficiently encapsulate both hydrophilic and lipophilic molecules within their aqueous core and lipid bilayer, respectively.[105] Beyond their structural elegance, liposomes have earned distinction as premier nano-drug carriers, securing approval from the stringent regulatory standards set forth by the FDA.[106] Recent studies have focused on incorporating active, small-molecule compounds such as rapamycin and lornoxicam, which typically have low bioavailability, into liposomal formulations for the treatment of KOA. These findings have demonstrated enhanced therapeutic efficacy and improved bioavailability compared with conventional formulations.[107,108] Similarly, surface modifications of liposomes with functional targeting groups is a prevalent approach in drug delivery strategies. By capitalizing on the abundant presence of type II collagen in cartilage, researchers have explored the modification of liposomes with type II collagen antibodies to achieve cartilage-specific targeting for drug delivery.[109] Studies have reported promising outcomes, suggesting that these tailored liposomes can effectively be accumulated into cartilage tissues, thereby enhancing the precision and efficacy of drug delivery to target sites within the joint. Other advanced strategies have been applied to liposomes.[110,111] This targeted approach holds significant potential for optimizing therapeutic outcomes while minimizing off-target effects, thereby advancing the field of precision medicine for KOA treatment.
In clinical trials, liposomal bupivacaine has attracted much attentions and positive results have been obtained. In this review we summarized the targeted therapy strategies being investigated in KOA clinical trials (Table 2). A double-blinded, randomized controlled trial (NCT04910165) was perform recently, which contrasted the use of liposomal bupivacaine with ropivacaine as preoperative interventions before TKAs. Liposomal bupivacaine in peripheral regional nerve blocks is associated with reductions in pain intensity, shorter hospital stays, decreased use of opioids among inpatients, enhanced WOMAC scores, and good safety.[112] Additionally, a phase III, randomized, double-blind, placebo- and active-controlled study was conducted to evaluate the efficacy and safety of liposomal formulation of dexamethasone (TLC599) in patients with KOA (NCT04123561). TLC599 was designed to provide prolonged pain relief and improved joint function in patients with KOA through sustained delivery of dexamethasone, with the aim of minimizing systemic exposure and associated risks. Adverse events, vital signs, and laboratory parameters were monitored to assess the safety profile of TLC599. The results showed that TLC599 provided prolonged pain relief and improved joint function in KOA patients through the sustained delivery of dexamethasone. Compared with low-dose dexamethasone (4mg) administration and placebo, the incidence rate of adverse events did not increase after the administration of TLC599 with a dose of 12mg dexamethasone. As pioneers in the realm of nanomedicine for KOA, liposomes continue to fuel innovation and inspire the development of novel drug delivery systems aimed at address the complex challenges of modern pharmacotherapy.
Table 2.
Targeted therapy strategies being investigated in KOA clinical trials.
| Targeted therapy strategies | Type of drug/carrier | Drug name | Administration routes | Safety and | Effectiveness | Clinicaltrials.gov identifier | Current phase of development | References of clinical data |
|---|---|---|---|---|---|---|---|---|
| Small-molecule inhibitors | ADAMTS5 inhibitor | GLPG1972/S201086 | Oral | Well-tolerated | Inefficacy | NCT03311009, NCT03595618 | Phase II | [54–56] |
| Cathepsin K inhibitor | MIV-711 | Oral | Well-tolerated | Significant reduction of the progression of bone area and cartilage thinning | NCT02705625 | Phase II | [60] | |
| Wnt signaling inhibitors | Lorecivivint | Intra-articular | Well-tolerated | Pain reduction and enhanced joint function | NCT02536833, NCT03122860, NCT04385303, NCT03928184, NCT05603754 | Phase III | [65–68] | |
| Monoclonal antibodies | Neutralizing antibody against ADAMTS5 | M6495 | Subcutaneous | Well-tolerated | Effective inhibition of aggrecan degradation | NCT03583346 | Phase II | / |
| Neutralizing antibody against NGF | Tanezumab, Fasinumab | Subcutaneous | The potential for joint-related adverse events | Significant pain relief | NCT03304379, NCT02528188 | Phase III | [82] | |
| Anti-CCL17 antibody | GSK3858279 | Intravenous, subcutaneous | Favorable safety and tolerability | Significant improvements in knee pain | NCT03485365 | Phase I | / | |
| Nanocarrier-based therapy | Liposomes | Bupivacaine | Intra-articular | Good safety | The prolonged analgesia | NCT04910165 | Phase III | [112] |
| Liposomes | Dexamethasone | Intra-articular | Minimization of systemic exposure and associated risks | The prolonged pain relief and improved joint function | NCT04123561 | Phase III | / | |
| Gene therapy | Chondrocytes | Tissuegene-C | Intra-articular | Well-tolerated | Increase of cartilage thickness and slower rates of subchondral bone surface area growth | NCT02072070 | Phase III | [113] |
| Nonviral gene therapy | XT-150 | Intra-articular | Well-tolerated | Significant improvements in pain and function | NCT04124042 | Phase II | / | |
| Adeno-associated viral vectors | Sc-raav2.5IL-1Ra | Intra-articular | – | – | NCT02790723 | Phase I | / | |
| Adeno-associated viral vectors | GNSC-001 | Intra-articular | – | – | NCT05835895 | Phase I | / | |
| Adeno-associated viral vectors | FX201 | Intra-articular | – | – | NCT04119687 | Phase I | / | |
| Cytotherapy | Mesenchymal stem cells | AT-mscs | Intra-articular | Joint-related adverse events | Limited pain relief and functional improvement | NCT01183728, NCT03869229, NCT05081921, NCT05783154 | Phase II | [114–117] |
| MSC-exos | Exooa-1 | Intra-articular | – | – | NCT05060107 | Phase I | / |
ADAMTS = a disintegrin and metalloproteinase with thrombospondin motifs, KOA = knee osteoarthritis, MSC-Exos = mesenchymal stem cell-derived exosomes.
“–” represents no results posted; “/” represents no reference available.
4.3.3. Other
Micelles with diameters typically ranging between 5 and 100 nm arise through the spontaneous arrangement of amphiphilic polymers within water-based solutions,[118,119] making them suitable carriers for a wide range of therapeutic agents in nanomedicine applications (Fig. 2A and B).[120] The acidic microenvironment and elevated levels of matrix metalloproteinase-13 (MMP-13) serve as characteristic biomarkers associated with KOA, providing valuable targets for the development of micelle-based drug delivery systems.[121] Among these systems, MMP-13 responsive and pH sensitive polymer (MR-PPL) micelles, poly (β-amino ester) micelles, have emerged as promising platforms for KOA treatment.[121–123] These micelles can respond to the acidic conditions prevalent in KOA-affected joints, facilitating targeted drug release with heightened specificity and efficacy. Using these OA markers, researchers aim to enhance the therapeutic outcomes of micellar drug delivery, offering a promising direction for precision medicine in KOA management.
Dendrimers comprising 3 distinct components – core, branches, and shell – exhibit a well-defined architecture that lends itself to precise functionalization and manipulation (Fig. 2A and B).[124,125] The outer shell of dendrimers serves as a versatile platform for the attachment of cargo molecules or targeting ligands, allowing tailored modifications to enhance their therapeutic or diagnostic capabilities.[126,127] Through conjugation strategies, dendrimers can be engineered to selectively deliver payloads to specific cellular targets or tissues, offering immense potential for drug delivery applications.[128] A novel dendrimer approach involving kartogenin (KGN), polyamidoamine (PAMAM), and polyethylene glycol (PEG) has been reported and 2 distinct conjugates, PEG-PAMAM-KGN and KGN-PEG-PAMAM, were synthesized. Through this strategy, they enhanced the release profile of KGN and amplified the chondrogenic effects within KOA joints.[129] The conjugation of insulin-like growth factor 1 with PAMAM and PEG not only facilitated controlled release kinetics but also potentiated the therapeutic efficacy of the drug, suggesting a promising avenue for the development of targeted and sustained drug delivery systems for KOA treatment.[130]
Despite the growing interest in nanomedicine for KOA treatment, the clinical application of micelles or dendrimers remains relatively limited. This restraint may stem from inherent drawbacks associated with micellar or dendrimer systems, such as their inefficacy in encapsulating hydrophilic drugs, the uncontrolled burst release effect, and concerns regarding potential toxicity. These limitations underscore the need for continued research and innovation to overcome challenges associated with nanocarrier-based therapies for KOA. Addressing these concerns could unlock the full potential of micelles and dendrimers in delivering therapeutics for KOA management, paving the way for more effective and safer treatment options.
4.4. Gene therapy
In the context of KOA treatment, gene therapy does not primarily aim to restore aberrant KOA-related genes to their normal states. Instead, it serves as a delivery platform designed to inhibit the expression of genes implicated in KOA progression or to induce the overexpression of therapeutic factors to mitigate KOA pathology.[131] This approach involves targeted modulation of gene expression within affected joint tissues, with the ultimate goal of alleviating symptoms and halting disease progression. By manipulating gene expression profiles, the gene therapy holds promise for providing long-term therapeutic benefits and potentially altering the underlying mechanisms driving KOA pathogenesis.[132]
The clinical effectiveness of TissueGene-C (TG-C) as a cell and gene therapy designed for KOA treatment was evaluated in a multicenter, double-blind, phase III clinical trial (NCT02072070).[113,133] Patients receiving TG-C treatment displayed increased cartilage thickness, slower rates of subchondral bone surface area growth in various knee regions and improved serum index levels. These findings suggest potential of TG-C as a disease-modifying drug for KOA treatment. TG-C is generally well-tolerated in clinical studies. The most commonly reported adverse events were mild to moderate in severity, including localized joint pain and swelling. No serious adverse events related to the treatment have been reported. Other clinical trials of gene therapy for KOA are available at https://clinicaltrials.gov/ (NCT04124042, NCT02790723, NCT05835895, and NCT04119687). XT-150 is an investigational nonviral gene therapy designed to deliver IL-10v, a modified variant of the anti-inflammatory cytokine interleukin-10, aimed at treating moderate-to-severe KOA in NCT04124042. Although the primary endpoint, – achieving at least 30% improvement in WOMAC pain score by day 180, – showed no statistically significant difference between the XT-150 and placebo groups, post hoc analyses in stage B indicated that participants receiving 2 doses of 0.45 mg XT-150 experienced significant improvements in pain and function by day 360 compared to those receiving a single dose. The primary endpoint of the study was not met, suggesting the need for further research to optimize dosing regimens and confirm efficacy. Additionally, the improvements observed in post hoc analyses require validation in larger, randomized trials. A recent clinical study on adeno-associated virus gene therapy, including Sc-rAAV2.5IL-1Ra, GNSC-001 and FX201 for KOA by intra-articular injection was also conducted (https://clinicaltrials.gov/, NCT05835895). As these are ongoing clinical trials, comprehensive data on efficacy and safety are not yet available.
Therefore, the advancement of gene therapy in KOA is rapidly progressing and fueled by continuous research and technological innovations. Additionally, advancements in gene delivery systems, such as viral vectors and nanoparticles, have facilitated the effective and targeted delivery of therapeutic genes to KOA-affected tissues. Therefore, the development of gene therapy holds promise for revolutionizing KOA treatment in the future. However, the research and development of gene therapies involve substantial investments, often exceeding 100s of millions of dollars and producing viral vectors or other gene delivery systems requires highly specialized facilities and stringent quality control, contributing to high production costs, resulting in many approved gene therapies having extremely high prices, limiting widespread adoption.[134,135] In addition, current gene therapy production relies on specialized bioprocessing techniques that are difficult to scale, limiting availability, and the variability in gene expression, vector integration, and dosing makes standardization possible.[136,137]
4.5. Cytotherapy
4.5.1. Stem cell therapy
Mesenchymal stem cells (MSCs) have garnered considerable attention in KOA research owing to their unique biological properties.[127] These cells possess the ability to self-renew and differentiate into various cells, including chondrocytes, which are crucial for cartilage regeneration.[138,139] Additionally, MSCs exhibit immunomodulatory effects that can help mitigate the inflammatory processes associated with KOA pathogenesis. Their multilineage differentiation potential, particularly their capacity to differentiate into chondrocytes, makes them attractive candidates for cell-based therapies aimed at repairing damaged cartilage in KOA joints. Clinical trials investigating the efficacy of autologous MSC therapy in KOA have primarily yielded limited evidence regarding pain relief and functional improvement in patients (NCT01183728, NCT03869229, NCT05081921, NCT05783154). Furthermore, these trials have not provided convincing evidence of significant changes in cartilage thickness as assessed by magnetic resonance imaging.[114–116,140] Despite the promising regenerative potential of MSCs, the outcomes of these clinical studies suggest that further research is necessary to optimize treatment protocols and better understand the mechanisms underlying MSC-based therapies for KOA.[117]
Exosomes are a subtype of extracellular vesicles secreted by cells that facilitate intercellular communication.[141] These nanosized extracellular vesicles carry diverse cargoes of bioactive molecules, including proteins, lipids, and nucleic acids, which can modulate cellular processes involved in tissue repair and regeneration. Mesenchymal stem cell-derived exosomes (MSC-Exos) have garnered considerable attention in biomedical research owing to their potential therapeutic applications in treating KOA.[142] Studies have demonstrated the ability of MSC-Exos to promote chondrogenesis and autophagy, inhibit inflammation, and enhance extracellular matrix synthesis in preclinical models of cartilage damage.[143–145] MSC-Exos can also be developed as delivery vehicles loading active compounds for targeted therapy under KOA conditions.[146] Recently, a phase I study was performed to assess the safety profile of MSC-Exos administered via intra-articular injection in the knee joints of patients diagnosed with mild to moderate symptomatic osteoarthritis (NCT05060107). Further research is warranted to elucidate the mechanisms of action and optimize the therapeutic efficacy of MSC-Exos in clinical settings.
4.5.2. Platelet therapy
Emerging autologous cellular therapies have shown potential as adjunctive components in various regenerative medicine treatment protocols.[147] Platelet therapy based on platelet-rich plasma (PRP) is characterized as a component of autologous blood with an elevated platelet concentration compared to baseline levels.[148] PRP therapies have been used for various indications for over 3 decades, garnering considerable attention for their potential in regenerative medicine.[149] Platelet growth factors facilitate 3 phases of the wound healing and repair cascade: inflammation, proliferation, and remodeling. However, recommendations derived from in vitro and animal research often yield divergent clinical outcomes because the challenge of translating nonclinical study findings and clinical treatment protocols.[150] While intra-articular injections of PRP may be advisable for patients with KOA, it is important to note that this approach lacks FDA approval.[151]
While cell therapy holds great promise for treating various diseases, potential risks include immune rejection, tumorigenicity (e.g., uncontrolled proliferation of stem cells), and complications from immune-modulating therapies. Challenges related to cost, scalability, and standardization hinder its widespread adoption.[152,153] Addressing these issues through technological advancements, automation in manufacturing, and improved regulatory frameworks is essential for making cell therapies more accessible and feasible on a global scale.
5. Conclusions and future prospects
Preclinical studies and genetic analyses in KOA research have identified numerous novel therapeutic targets through comprehensive investigations into disease mechanisms and genetic factors, offering potential avenues for the development of innovative targeted therapy strategies. Some targeted drugs have progressed to phase II and III clinical trials, where they have shown promising results in terms of efficacy and safety profiles. Nanocarrier-based therapy offers precise control over drug delivery, allowing for enhanced therapeutic efficacy, sustained drug release and reduced systemic side effects. Depending on the therapeutic goals, these systems can target specific tissues within the joint, such as cartilage, synovium, or cells residing within these tissues. By precisely delivering therapeutic agents to the desired sites of action, nano-scale drug delivery systems hold great promise for improving KOA treatment outcomes. Their ability to selectively target affected tissues while minimizing off-target effects represents a significant advancement in the field of KOA therapy. The majority of targeted therapy strategies used clinically consist of small-molecule inhibitors and mAbs, which are continuously under investigation for their potential applications in KOA treatment. Research in this area is ongoing with a focus on developing disease-modifying osteoarthritis drug (DMOADs). Over the next decade, substantial advancements have been made in OA therapeutics, driven by ongoing research and development efforts focused on small-molecule inhibitors and mAbs. Currently, gene therapy and cytotherapy for KOA are gaining significant attentions with advancements made in addressing the challenge of sustaining transgene expression in joints and supplying cartilage regeneration therapy over the long term. However, there is a lack of consensus regarding the optimal selection of transgenes and promoters for effective treatment. There is a growing interest in utilizing multiple therapeutic gene products as they may offer enhanced efficacy. Additionally, exploring combination therapeutic approaches that integrate gene therapy with other medical or surgical interventions warrants further investigation. Despite significant progress in the field of MSC-based regenerative medicine, several challenges persist, indicating that there is still a long road ahead to effectively and economically repair articular cartilage defects and osteochondral interface. To achieve efficient osteogenesis and chondrogenesis, innovative strategies and technologies are required to enhance the potential of cytotherapies for KOA. In conclusion, all advancements in targeted therapy underscore the importance of translational research in elucidating new therapeutic options for KOA and provide hope for improved management and outcomes for patients with this debilitating condition.
Author contributions
Conceptualization: Qi Tang.
Formal analysis: Juan-Hong Liang.
Investigation: Feng-Lan Huang, Zhen-Yan Ye.
Methodology: Feng-Lan Huang.
Visualization: Zhen-Yan Ye.
Writing –original draft: Li Chen.
Writing – review & editing: Zhi-Kai Zhao, Juan-Hong Liang.
Abbreviations:
- ADAMTS
- a disintegrin and metalloproteinase with thrombospondin motifs
- CCL17
- CC-chemokine ligand 17
- CLKs
- Cdc2-like kinases
- COX-2
- cyclooxygenase-2
- DMOADs
- the disease-modifying osteoarthritis drug
- DYRKs
- tyrosine phosphorylation-regulated kinases
- ECM
- extracellular matrix
- FDA
- Food and Drug Administration
- HMSNs
- hollow mesoporous silica nanoparticles
- IL-1β
- interleukin-1
- IL-6
- interleukin-6
- ILs
- interleukins
- KGN
- kartogenin
- KOA
- knee osteoarthritis
- KPP
- Kgn-peg-pamam
- LOR
- lorecivivint
- mAbs
- monoclonal antibodies
- MMP-13
- matrix metalloproteinase-13
- MMPs
- matrix metalloproteinases
- MR-PPL
- MMP-13 responsive and ph sensitive polymer
- MSC-Exos
- mesenchymal stem cell-derived exosomes
- MSCs
- mesenchymal stem cells
- NGF
- nerve growth factor
- NPs
- nanoparticles
- NSAIDs
- nonsteroidal anti-inflammatory drugs
- OA
- osteoarthritis
- OAK
- osteotomy around the knee
- PAMAM
- polyamidoamine
- PEG
- polyethylene glycol
- PGFs
- platelet growth factors
- PLA
- polylactic acid
- PLGA
- poly (lactic-co-glycolic) acid
- PPK
- peg-pamam-kgn
- PRP
- platelet-rich plasma
- TG-C
- tissuegene-C
- TKA
- total knee arthroplasty
- TNF-α
- tumor necrosis factor-alpha
- UKA
- unicompartmental knee arthroplasty
- WNT
- wingless-type MMTV integration site family
This work was funded by the Fundamental Research Funds for the Central Universities of Sichuan University (Grant Number: KY202208-1-119).
The authors have no conflicts of interest to disclose.
All data generated or analyzed during this study are included in this published article [and its supplementary information files].
How to cite this article: Chen L, Huang F-L, Tang Q, Zhao Z-K, Ye Z-Y, Liang J-H. Targeted therapy for knee osteoarthritis: From basic to clinics. Medicine 2025;104:33(e43686).
LC and FLH contributed to this article equally.
References
- [1].Boer CG, Hatzikotoulas K, Southam L, et al. Deciphering osteoarthritis genetics across 826,690 individuals from 9 populations. Cell. 2021;184:4784–818. e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Yao Q, Wu X, Tao C, et al. Osteoarthritis: pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther. 2023;8:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Lv Y, Sui L, Lv H, Zheng J, Feng H, Jing F. Burden of knee osteoarthritis in China and globally from 1992 to 2021, and projections to 2030: a systematic analysis from the global burden of disease study 2021. Front Public Health. 2025;13:1543180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Kang Y, Liu C, Ji Y, et al. The burden of knee osteoarthritis worldwide, regionally, and nationally from 1990 to 2019, along with an analysis of cross-national inequalities. Arch Orthop Trauma Surg. 2024;144:2731–43. [DOI] [PubMed] [Google Scholar]
- [5].Park JH, Lee H, J-s C, Kim I, Lee J, Jang SH. Effects of knee osteoarthritis severity on inter-joint coordination and gait variability as measured by hip-knee cyclograms. Sci Rep. 2021;11:1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Chen W, Lin T, He Q, et al. Study on the potential active components and molecular mechanism of Xiao Huoluo Pills in the treatment of cartilage degeneration of knee osteoarthritis based on bioinformatics analysis and molecular docking technology. J Orthop Surg Res. 2021;16:1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Sanchez-Lopez E, Coras R, Torres A, Lane NE, Guma M. Synovial inflammation in osteoarthritis progression. Nat Rev Rheumatol. 2022;18:258–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Zhao R-l, Ma P-h, Liu B-y, et al. Short-term and long-term effectiveness of acupuncture and Tuina on knee osteoarthritis: study protocol for a randomized controlled trial. Front Neurol. 2023;14:1301217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Zhou X, Liu G, Han B, Li H, Zhang L, Liu X. Different prevention and treatment strategies for knee osteoarthritis (KOA) with various lower limb exoskeletons – a comprehensive review. Robotica. 2021;39:1345–67. [Google Scholar]
- [10].Nowaczyk A, Szwedowski D, Dallo I, Nowaczyk J. Overview of first-line and second-line pharmacotherapies for osteoarthritis with special focus on intra-articular treatment. Int J Mol Sci. 2022;23:1566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].da Costa BR, Pereira TV, Saadat P, et al. Effectiveness and safety of non-steroidal anti-inflammatory drugs and opioid treatment for knee and hip osteoarthritis: network meta-analysis. BMJ. 2021;375:n2321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Kurteva S, Abrahamowicz M, Gomes T, Tamblyn R. Association of opioid consumption profiles after hospitalization with risk of adverse health care events. JAMA Network Open. 2021;4:e218782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Latourte A, Kloppenburg M, Richette P. Emerging pharmaceutical therapies for osteoarthritis. Nat Rev Rheumatol. 2020;16:673–88. [DOI] [PubMed] [Google Scholar]
- [14].Kolasinski SL, Neogi T, Hochberg MC, et al. 2019 American College of Rheumatology/Arthritis Foundation guideline for the management of osteoarthritis of the hand, hip, and knee. Arthritis Rheumatol. 2020;72:220–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Gibbs AJ, Gray B, Wallis JA, et al. Recommendations for the management of hip and knee osteoarthritis: a systematic review of clinical practice guidelines. Osteoarthritis Cartilage. 2023;31:1280–92. [DOI] [PubMed] [Google Scholar]
- [16].Losina E, Song S, Bensen GP, Katz JN. Opioid use among medicare beneficiaries with knee osteoarthritis: prevalence and correlates of chronic use. Arthritis Care Res (Hoboken). 2023;75:876–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Arden NK, Perry TA, Bannuru RR, et al. Non-surgical management of knee osteoarthritis: comparison of ESCEO and OARSI 2019 guidelines. Nat Rev Rheumatol. 2021;17:59–66. [DOI] [PubMed] [Google Scholar]
- [18].Richard MJ, Driban JB, McAlindon TE. Pharmaceutical treatment of osteoarthritis. Osteoarthritis Cartilage. 2023;31:458–66. [DOI] [PubMed] [Google Scholar]
- [19].Fernández-Martín S, González-Cantalapiedra A, Muñoz F, García-González M, Permuy M, López-Peña M. Glucosamine and chondroitin sulfate: is there any scientific evidence for their effectiveness as disease-modifying drugs in knee osteoarthritis preclinical studies?—a systematic review from 2000 to 2021. Animals (Basel). 2021;11:1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Meng Z, Liu J, Zhou N. Efficacy and safety of the combination of glucosamine and chondroitin for knee osteoarthritis: a systematic review and meta-analysis. Arch Orthop Trauma Surg. 2023;143:409–21. [DOI] [PubMed] [Google Scholar]
- [21].Larder CE, Iskandar MM, Kubow S. Collagen hydrolysates: a source of bioactive peptides derived from food sources for the treatment of osteoarthritis. Medicines (Basel). 2023;10:50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Babur MN, Siddiqi FA, Tassadaq N, Tareen A, Osama M. Effects of glucosamine and chondroitin sulfate supplementation in addition to resistance exercise training and manual therapy in patients with knee osteoarthritis: a randomized controlled trial. J Pak Med Assoc. 2022;72:1272–77. [DOI] [PubMed] [Google Scholar]
- [23].Sharma L. Osteoarthritis of the knee. N Engl J Med. 2021;384:51–9. [DOI] [PubMed] [Google Scholar]
- [24].Hall M, Allison K, Knox G, et al. Addition of aerobic physical activity to resistance exercise for hip osteoarthritis (PHOENIX): a randomised comparative effectiveness trial. Lancet Rheumatol. 2025;7:e343–54. [DOI] [PubMed] [Google Scholar]
- [25].Long H, Cao R, Yin H, Yu F, Guo A. Associations between obesity, diabetes mellitus, and cardiovascular disease with progression states of knee osteoarthritis (KOA). Aging Clin Exp Res. 2023;35:333–40. [DOI] [PubMed] [Google Scholar]
- [26].Wei G, Lu K, Umar M, et al. Risk of metabolic abnormalities in osteoarthritis: a new perspective to understand its pathological mechanisms. Bone Res. 2023;11:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Dantas LO, de Fátima Salvini T, McAlindon TE. Knee osteoarthritis: key treatments and implications for physical therapy. Braz J Phys Ther. 2021;25:135–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Matos LC, Machado JP, Monteiro FJ, Greten HJ. editors. Understanding traditional Chinese medicine therapeutics: an overview of the basics and clinical applications. Healthcare. MDPI; 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Peng H, Ou A, Huang X, et al. Osteotomy around the knee: the surgical treatment of osteoarthritis. Orthop Surg. 2021;13:1465–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Sappey-Marinier E, Batailler C, Lustig S. Osteoarthritis, osteotomies, and knee arthroplasty. Textbook of Musculoskeletal Disorders. Springer; 2023:481–88. [Google Scholar]
- [31].Crawford DA, Berend KR, Thienpont E. Unicompartmental knee arthroplasty: US and global perspectives. Orthop Clin North Am. 2020;51:147–59. [DOI] [PubMed] [Google Scholar]
- [32].Wignadasan W, Thompson J, Ibrahim M, Kayani B, Magan A, Haddad F. Day-case unicompartmental knee arthroplasty: a literature review and development of a novel hospital pathway. Ann R Coll Surg Engl. 2022;104:165–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Tripathy SK, Varghese P, Srinivasan A, et al. Joint awareness after unicompartmental knee arthroplasty and total knee arthroplasty: a systematic review and meta‐analysis of cohort studies. Knee Surg Sports Traumatol Arthrosc. 2021;29:3478–87. [DOI] [PubMed] [Google Scholar]
- [34].Liu S, Deng Z, Chen K, et al. Cartilage tissue engineering: from proinflammatory and anti‑inflammatory cytokines to osteoarthritis treatments. Mol Med Rep. 2022;25:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Lin X, Wang W, McDavid A, Xu H, Boyce BF, Xing L. The E3 ubiquitin ligase Itch limits the progression of post-traumatic osteoarthritis in mice by inhibiting macrophage polarization. Osteoarthritis Cartilage. 2021;29:1225–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Mailhot B, Christin M, Tessandier N, et al. Neuronal interleukin-1 receptors mediate pain in chronic inflammatory diseases. J Exp Med. 2020;217:e20191430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Liao Y, Ren Y, Luo X, et al. Interleukin-6 signaling mediates cartilage degradation and pain in post-traumatic osteoarthritis. bioRxiv 2021;09.08:459303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Xiao SQ, Cheng M, Wang L, et al. The role of apoptosis in the pathogenesis of osteoarthritis. Int Orthop. 2023;47:1895–919. [DOI] [PubMed] [Google Scholar]
- [39].Ma L, Zheng X, Lin R, et al. Knee osteoarthritis therapy: recent advances in intra-articular drug delivery systems. Drug Des Devel Ther. 2023;16:1311–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Roskoski R, Jr. Properties of FDA-approved small molecule protein kinase inhibitors: a 2021 update. Pharmacol Res. 2021;165:105463. [DOI] [PubMed] [Google Scholar]
- [41].Ayala-Aguilera CC, Valero T, Lorente-Macias A, Baillache DJ, Croke S, Unciti-Broceta A. Small molecule kinase inhibitor drugs (1995–2021): medical indication, pharmacology, and synthesis. J Med Chem. 2021;65:1047–131. [DOI] [PubMed] [Google Scholar]
- [42].Bonekamp NA, Peter B, Hillen HS, et al. Small-molecule inhibitors of human mitochondrial DNA transcription. Nature. 2020;588:712–6. [DOI] [PubMed] [Google Scholar]
- [43].Park J, Lee SY. A review of osteoarthritis signaling intervention using small-molecule inhibitors. Medicine (Baltimore). 2022;101:e29501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Song C, Hu Z, Xu D, et al. STING signaling in inflammaging: a new target against musculoskeletal diseases. Front Immunol. 2023;14:1227364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Du X, Zy L, Xx T, et al. Research progress on the pathogenesis of knee osteoarthritis. Orthop Surg. 2023;15:2213–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Zhao QH, Lin LP, Guo YX, et al. Matrix metalloproteinase‑13, NF‑κB p65 and interleukin‑1β are associated with the severity of knee osteoarthritis. Exp Ther Med. 2020;19:3620–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Bendele AM, Neelagiri M, Neelagiri V, Sucholeiki I. Development of a selective matrix metalloproteinase 13 (MMP-13) inhibitor for the treatment of osteoarthritis. Eur J Med Chem. 2021;224:113666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Li S-H, Wu Q-F. MicroRNAs target on cartilage extracellular matrix degradation of knee osteoarthritis. Eur Rev Med Pharmacol Sci. 2021;25:1185–97. [DOI] [PubMed] [Google Scholar]
- [49].Song X-Y, Xie W-P, Zhang P, Zhao M, Bi R-X. Cangxitongbi capsule protects articular cartilage of the knee in rats by regulating ADAMTS-5. Ann Transl Med. 2020;8:1511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Latourte A, Richette P. Inhibition of ADAMTS-5: the right target for osteoarthritis? Osteoarthritis Cartilage. 2022;30:175–7. [DOI] [PubMed] [Google Scholar]
- [51].Bihlet AR, Balchen T, Goteti K, et al. Safety, tolerability, and pharmacodynamics of the ADAMTS-5 nanobody M6495: two phase 1, single-center, double-blind, randomized, placebo-controlled studies in healthy subjects and patients with osteoarthritis. ACR Open Rheumatol. 2024;6:205–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Clement-Lacroix P, Meurisse S, Lepescheux L, et al. ADAMTS-5 inhibition with the potent and highly selective inhibitor GLPG1972 results in strong disease-modifying OA drug effects in the rat meniscectomy model. Osteoarthritis Cartilage. 2018;26:S26. [Google Scholar]
- [53].Clement-Lacroix P, Little CB, Smith MM, et al. Pharmacological characterization of GLPG1972/S201086, a potent and selective small-molecule inhibitor of ADAMTS5. Osteoarthritis Cartilage. 2022;30:291–301. [DOI] [PubMed] [Google Scholar]
- [54].van der Aar E, Deckx H, Dupont S, et al. Safety, pharmacokinetics, and pharmacodynamics of the ADAMTS-5 inhibitor GLPG1972/S201086 in healthy volunteers and participants with osteoarthritis of the knee or hip. Clin Pharmacol Drug Dev. 2022;11:112–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Schnitzer T, Pueyo M, Deckx H, et al. Efficacy and safety of s201086/GLPG1972, an ADAMTS-5 inhibitor, in patients with knee osteoarthritis: roccella, a 52-week, randomized, double-blind, dose-ranging phase 2 study. Osteoarthritis Cartilage. 2021;29:S264. [Google Scholar]
- [56].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;31:985–94. [DOI] [PubMed] [Google Scholar]
- [57].Mijanović O, Jakovleva A, Branković A, et al. Cathepsin K in pathological conditions and new therapeutic and diagnostic perspectives. Int J Mol Sci. 2022;23:13762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Zou N, Liu R, Li C. Cathepsin K+ non-osteoclast cells in the skeletal system: function, models, identity, and therapeutic implications. Front Cell Dev Biol. 2022;10:1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].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;16:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].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;172:86–95. [DOI] [PubMed] [Google Scholar]
- [61].Feng J, Zhang Q, Pu F, et al. Signalling interaction between beta-catenin and other signalling molecules during osteoarthritis development. Cell Prolif. 2024;57:e13600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Cherifi C, Monteagudo S, Lories RJ. Promising targets for therapy of osteoarthritis: a review on the Wnt and TGF-β signalling pathways. Ther Adv Musculoskelet Dis. 2021;13:1759720X211006959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Lories RJ, Monteagudo S. Is Wnt signaling an attractive target for the treatment of osteoarthritis? Rheumatol Ther. 2020;7:259–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Song M, Pang L, Zhang M, Qu Y, Laster KV, Dong Z. Cdc2-like kinases: structure, biological function, and therapeutic targets for diseases. Signal transduction and targeted therapy. Signal Transduct Target Ther. 2023;8:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Yazici Y, McAlindon T, Fleischmann R, et al. 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;25:1598–606. [DOI] [PubMed] [Google Scholar]
- [66].Yazici Y, McAlindon TE, Gibofsky A, et al. Results from a 52-week randomized, double-blind, placebo-controlled, phase 2 study of a novel, intra-articular wnt pathway inhibitor (SM04690) for the treatment of knee osteoarthritis. Osteoarthritis Cartilage. 2018;26:S293–4. [Google Scholar]
- [67].Yazici Y, McAlindon T, Gibofsky A, et al. 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. Osteoarthritis Cartilage. 2019;27:S503. [Google Scholar]
- [68].Yazici Y, Swearingen C, Ghandehari H, Simsek I, Kennedy S, Tambiah J, et al. Pos1365 Structural Severity in Knee Osteoarthritis Impacts Treatment Response: A Post Hoc Pooled Analysis of Lorecivivint Clinical Trials. BMJ Publishing Group Ltd; 2023. [Google Scholar]
- [69].Weng P-W, Yadav VK, Pikatan NW, et al. Novel NFκB inhibitor SC75741 mitigates chondrocyte degradation and prevents activated fibroblast transformation by modulating miR-21/GDF-5/SOX5 signaling. Int J Mol Sci. 2021;22:11082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Xu Z, Shen Z, Wu B, Gong S, Chen B. Small molecule natural compound targets the NF‐κB signaling and ameliorates the development of osteoarthritis. J Cell Physiol. 2021;236:7298–307. [DOI] [PubMed] [Google Scholar]
- [71].Al Jundi S, Martinez JR, Cresta J, et al. Identifying small molecules for protecting chondrocyte function and matrix integrity after controlled compressive injury. Osteoarthr Cartil Open. 2022;4:100289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Martinez J, Cresta J, DeSantis G, et al. Identification of potential therapeutics for post-traumatic osteoarthritis that effect apoptosis, matric degradation and inflammatory biomarkers using a in vitro injury platform. Osteoarthritis Cartilage. 2020;28:S487. [Google Scholar]
- [73].Su C-H, Lin C-Y, Tsai C-H, et al. Betulin suppresses TNF-α and IL-1β production in osteoarthritis synovial fibroblasts by inhibiting the MEK/ERK/NF-κB pathway. J Funct Foods. 2021;86:104729. [Google Scholar]
- [74].Werkmann D, Buyse M-A, Dejager L, et al. In vitro characterization of the ADAMTS-5 specific nanobody® M6495. Osteoarthritis Cartilage. 2018;26:S178. [Google Scholar]
- [75].Pereira JNS, Ottevaere I, Serruys B, Guehring H, Ladel C, Lindemann S. Translational pharmacokinetic and pharmacodynamic modelling of the anti-ADAMTS-5 NANOBODY((R)) (M6495) using the neo-epitope ARGS as a biomarker. J Pharmacokinet Pharmacodyn. 2024;52:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].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;173:509–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Yu L, Luo R, Qin G, Zhang Q, Liang W. Efficacy and safety of anti-interleukin-1 therapeutics in the treatment of knee osteoarthritis: a systematic review and meta-analysis of randomized controlled trials. J Orthop Surg Res. 2023;18:100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Croft M, Salek-Ardakani S, Ware CF. Targeting the TNF and TNFR superfamilies in autoimmune disease and cancer. Nat Rev Drug Discovery. 2024;23:939–61. [DOI] [PubMed] [Google Scholar]
- [79].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;71:1824–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Shin H, Prasad V, Lupancu T, et al. The GM-CSF/CCL17 pathway in obesity-associated osteoarthritic pain and disease in mice. Osteoarthritis Cartilage. 2023;31:1327–41. [DOI] [PubMed] [Google Scholar]
- [81].Luo H, Li L, Han S, Liu T. The role of monocyte/macrophage chemokines in pathogenesis of osteoarthritis: a review. Int J Immunogenet. 2024;51:130–42. [DOI] [PubMed] [Google Scholar]
- [82].Hochberg MC, Carrino J, Schnitzer T, Guermazi A, Walsh D, White A, et al., editors. Subcutaneous Tanezumab Versus NSAID for the Treatment of Osteoarthritis: Joint Safety Events in a Randomized, Double-blind, Active-Controlled, 80-Week, Phase-3 Study. Arthritis & Rheumatology; 2019. [Google Scholar]
- [83].Jin G-Z. Current nanoparticle-based technologies for osteoarthritis therapy. Nanomaterials (Basel). 2020;10:2368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Pontes AP, Welting TJ, Rip J, Creemers LB. Polymeric nanoparticles for drug delivery in osteoarthritis. Pharmaceutics. 2022;14:2639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Younas A, Gu H, Zhao Y, Zhang N. Novel approaches of the nanotechnology-based drug delivery systems for knee joint injuries: a review. Int J Pharm. 2021;608:121051. [DOI] [PubMed] [Google Scholar]
- [86].Ramírez-Noguera P, Marín I Z, Chavarin BM G, Valderrama ME, López-Barrera LD, Díaz-Torres R. Study of the early effects of chitosan nanoparticles with glutathione in rats with osteoarthrosis. Pharmaceutics. 2023;15:2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Yang L, Zhao X, Zhang J, et al. Synthesis of charged chitosan nanoparticles as functional biolubricant. Colloids Surf B. 2021;206:111973. [DOI] [PubMed] [Google Scholar]
- [88].Su X, Wei L, Xu Z, et al. Evaluation and application of silk fibroin based biomaterials to promote cartilage regeneration in osteoarthritis therapy. Biomedicines. 2023;11:2244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Wang Z, Yin X, Zhuang C, et al. Injectable regenerated silk fibroin micro/nanosphere with enhanced permeability and stability for osteoarthritis therapy. Small. 2024;20:e2405049. [DOI] [PubMed] [Google Scholar]
- [90].Gupta N, Rao SK, Jaison D, Patil S, Gupta N, Arunachalam KD. Kaempferol loaded albumin nanoparticles and dexamethasone encapsulation into electrospun polycaprolactone fibrous mat–concurrent release for cartilage regeneration. J Drug Delivery Sci Technol. 2021;64:102666. [Google Scholar]
- [91].Khandelia R, Hodgkinson T, Crean D, et al. Reproducible synthesis of biocompatible albumin nanoparticles designed for intra-articular administration of celecoxib to treat osteoarthritis. ACS Appl Mater Interfaces. 2024;16:14633–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Shin HJ, Park H, Shin N, et al. p66shc siRNA nanoparticles ameliorate chondrocytic mitochondrial dysfunction in osteoarthritis. Int J Nanomedicine. 2020;Volume 15:2379–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Wei L, Pan Q, Teng J, Zhang H, Qin N. Intra-articular administration of PLGA resveratrol sustained-release nanoparticles attenuates the development of rat osteoarthritis. Mater Today Bio. 2024;24:100884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Abbas H, El-Deeb NM, Zewail M. PLA-coated Imwitor((R)) 900 K-based herbal colloidal carriers as novel candidates for the intra-articular treatment of arthritis. Pharm Dev Technol. 2021;26:682–92. [DOI] [PubMed] [Google Scholar]
- [95].Zhang H, Xiong H, Ahmed W, et al. Reactive oxygen species-responsive and scavenging polyurethane nanoparticles for treatment of osteoarthritis in vivo. Chem Eng J. 2021;409:128147. [Google Scholar]
- [96].Salama AH, Abdelkhalek AA, Elkasabgy NA. Etoricoxib-loaded bio-adhesive hybridized polylactic acid-based nanoparticles as an intra-articular injection for the treatment of osteoarthritis. Int J Pharm. 2020;578:119081. [DOI] [PubMed] [Google Scholar]
- [97].Jin T, Wu D, Liu X-M, et al. Intra-articular delivery of celastrol by hollow mesoporous silica nanoparticles for pH-sensitive anti-inflammatory therapy against knee osteoarthritis. J Nanobiotechnol. 2020;18:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Hu B, Gao F, Li C, et al. Rhein laden pH-responsive polymeric nanoparticles for treatment of osteoarthritis. AMB Express. 2020;10:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Xiong W, Lan Q, Liang X, et al. Cartilage-targeting poly (ethylene glycol)(PEG)-formononetin (FMN) nanodrug for the treatment of osteoarthritis. J Nanobiotechnol. 2021;19:197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Ren K, Ke X, Chen Z, et al. Zwitterionic polymer modified xanthan gum with collagen II-binding capability for lubrication improvement and ROS scavenging. Carbohydr Polym. 2021;274:118672. [DOI] [PubMed] [Google Scholar]
- [101].Wang X, Cai Y, Wu C, et al. Conversion of senescent cartilage into a pro-chondrogenic microenvironment with antibody-functionalized copper sulfate nanoparticles for efficient osteoarthritis therapy. J Nanobiotechnol. 2023;21:258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Partain BD, Unni M, Rinaldi C, Allen KD. The clearance and biodistribution of magnetic composite nanoparticles in healthy and osteoarthritic rat knees. J Control Release. 2020;321:259–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Zhou K, Yang C, Shi K, et al. Activated macrophage membrane-coated nanoparticles relieve osteoarthritis-induced synovitis and joint damage. Biomaterials. 2023;295:122036. [DOI] [PubMed] [Google Scholar]
- [104].Nakhaei P, Margiana R, Bokov DO, et al. Liposomes: structure, biomedical applications, and stability parameters with emphasis on cholesterol. Front Bioeng Biotechnol. 2021;9:705886. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- [105].Pande S. Liposomes for drug delivery: review of vesicular composition, factors affecting drug release and drug loading in liposomes. Artif Cells Nanomed Biotechnol. 2023;51:428–40. [DOI] [PubMed] [Google Scholar]
- [106].Liu P, Chen G, Zhang J. A review of liposomes as a drug delivery system: current status of approved products, regulatory environments, and future perspectives. Molecules. 2022;27:1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].He K, Huang X, Shan R, et al. Intra-articular injection of lornoxicam and MicroRNA-140 co-loaded cationic liposomes enhanced the therapeutic treatment of experimental osteoarthritis. Aaps Pharmscitech. 2021;23:9. [DOI] [PubMed] [Google Scholar]
- [108].Chen C-H, Kuo SM, Tien Y-C, Shen P-C, Kuo Y-W, Huang HH. Steady augmentation of anti-osteoarthritic actions of rapamycin by liposome-encapsulation in collaboration with low-intensity pulsed ultrasound. Int J Nanomedicine. 2020;15:3771–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Mitsou E, Klein J. Liposome-based interventions in knee osteoarthritis. Small. 2025;21:e2410060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Deng C, Chen Y, Zhao X, et al. Apoptotic neutrophil membrane-camouflaged liposomes for dually targeting synovial macrophages and fibroblasts to attenuate osteoarthritis. ACS Appl Mater Interfaces. 2023;15:39064–80. [DOI] [PubMed] [Google Scholar]
- [111].He Y, Sun M, Wang J, et al. Chondroitin sulfate microspheres anchored with drug-loaded liposomes play a dual antioxidant role in the treatment of osteoarthritis. Acta Biomater. 2022;151:512–27. [DOI] [PubMed] [Google Scholar]
- [112].Malige A, Pellegrino AN, Kunkle K, Konopitski AK, Brogle PJ, Nwachuku CO. Liposomal bupivacaine in adductor canal blocks before total knee arthroplasty leads to improved postoperative outcomes: a randomized controlled trial. J Arthroplasty. 2022;37:1549–56. [DOI] [PubMed] [Google Scholar]
- [113].Kim M-K, Ha C-W, 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;29:48–59. [DOI] [PubMed] [Google Scholar]
- [114].Freitag J, Bates D, Wickham J, et al. Adipose-derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: a randomized controlled trial. Regen Med. 2019;14:213–30. [DOI] [PubMed] [Google Scholar]
- [115].Kim SH, Ha C-W, Park Y-B, Nam E, Lee J-E, Lee H-J. Intra-articular injection of mesenchymal stem cells for clinical outcomes and cartilage repair in osteoarthritis of the knee: a meta-analysis of randomized controlled trials. Arch Orthop Trauma Surg. 2019;139:971–80. [DOI] [PubMed] [Google Scholar]
- [116].Sadri B, Hassanzadeh M, Bagherifard A, et al. Cartilage regeneration and inflammation modulation in knee osteoarthritis following injection of allogeneic adipose-derived mesenchymal stromal cells: a phase II, triple-blinded, placebo controlled, randomized trial. Stem Cell Res Ther. 2023;14:162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Wang G, Xing D, Liu W, et al. Preclinical studies and clinical trials on mesenchymal stem cell therapy for knee osteoarthritis: a systematic review on models and cell doses. Int J Rheum Dis. 2022;25:532–62. [DOI] [PubMed] [Google Scholar]
- [118].Zhou T, Xiong H, Wang S, et al. An injectable hydrogel dotted with dexamethasone acetate-encapsulated reactive oxygen species-scavenging micelles for combinatorial therapy of osteoarthritis. Mater Today Nano. 2022;17:100164. [Google Scholar]
- [119].Maboudi AH, Lotfipour MH, Rasouli M, et al. Micelle-based nanoparticles with stimuli-responsive properties for drug delivery. Nanotechnol Rev. 2024;13:20230218. [Google Scholar]
- [120].Zhang J, Zhang J, Li H, Zhang H, Meng H. Research progress on biodegradable polymer-based drug delivery systems for the treatment of knee osteoarthritis. Front Bioeng Biotechnol. 2025;13:1561708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Lan Q, Lu R, Chen H, et al. MMP-13 enzyme and pH responsive theranostic nanoplatform for osteoarthritis. J Nanobiotechnol. 2020;18:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Kang C, Jung E, Hyeon H, Seon S, Lee D. Acid-activatable polymeric curcumin nanoparticles as therapeutic agents for osteoarthritis. Nanomed Nanotechnol Biol Med. 2020;23:102104. [DOI] [PubMed] [Google Scholar]
- [123].Prokopovich P. Different approaches in poly-beta-amino-esters based drug localisation in cartilage. Osteoarthritis Cartilage. 2024;32:S291–2. [Google Scholar]
- [124].Pérez-Ferreiro M, Abelairas AM, Criado A, Gómez IJ, Mosquera J. Dendrimers: exploring their wide structural variety and applications. Polymers. 2023;15:4369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Sarode RJ, Mahajan HS. Dendrimers for drug delivery: an overview of its classes, synthesis, and applications. J Drug Delivery Sci Technol. 2024;98:105896. [Google Scholar]
- [126].Chis AA, Dobrea C, Morgovan C, et al. Applications and limitations of dendrimers in biomedicine. Molecules. 2020;25:3982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Shang Z, Wanyan P, Zhang B, Wang M, Wang X. A systematic review, umbrella review, and quality assessment on clinical translation of stem cell therapy for knee osteoarthritis: are we there yet? Stem Cell Res Therapy. 2023;14:91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Dias AP, da Silva Santos S, da Silva JV, et al. Dendrimers in the context of nanomedicine. Int J Pharm. 2020;573:118814. [DOI] [PubMed] [Google Scholar]
- [129].Zeng W-N, Zhang Y, Wang D, et al. Intra-articular injection of kartogenin-enhanced bone marrow–derived mesenchymal stem cells in the treatment of knee osteoarthritis in a rat model. Am J Sports Med. 2021;49:2795–809. [DOI] [PubMed] [Google Scholar]
- [130].Geiger BC, Wang S, Padera RF, Jr, Grodzinsky AJ, Hammond PT. Cartilage-penetrating nanocarriers improve delivery and efficacy of growth factor treatment of osteoarthritis. Sci Transl Med. 2018;10:eaat8800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Hosseinkhani H, Domb AJ, Sharifzadeh G, Nahum V. Gene therapy for regenerative medicine. Pharmaceutics. 2023;15:856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Collon K, Gallo MC, Lieberman JR. Musculoskeletal tissue engineering: regional gene therapy for bone repair. Biomaterials. 2021;275:120901. [DOI] [PubMed] [Google Scholar]
- [133].Conaghan P, Wang M, Kim S. Post hoc analyses of 52 week pain responder indices to explore clinically meaningful outcomes after intra-articular tissuegene-C (Tg-C): data from a knee osteoarthritis randomized trialpost hoc analyses of 52 week pain responder indices to explore clinically meaningful outcomes after intra-articular tissuegene-C (Tg-C): data from a knee osteoarthritis randomized trial. Osteoarthritis Cartilage. 2023;31:S177–8. [Google Scholar]
- [134].Scotti C, Aiuti A, Naldini L. Challenges and solutions to the sustainability of gene and cell therapies. Nat Rev Genet. 2025;26:437–8. [DOI] [PubMed] [Google Scholar]
- [135].Phares S, Trusheim M, Emond SK, Pearson SD. Managing the challenges of paying for gene therapy: strategies for market action and policy reform in the United States. J Comp Eff Res. 2024;13:e240118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Gao J, Gunasekar S, Xia ZJ, et al. Gene therapy for CNS disorders: modalities, delivery and translational challenges. Nat Rev Neurosci. 2024;25:553–72. [DOI] [PubMed] [Google Scholar]
- [137].Lee NK, Chang JW. Manufacturing cell and gene therapies: challenges in clinical translation. Ann Lab Med. 2024;44:314–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Kangari P, Talaei-Khozani T, Razeghian-Jahromi I, Razmkhah M. Mesenchymal stem cells: amazing remedies for bone and cartilage defects. Stem Cell Res Ther. 2020;11:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Jiang S, Tian G, Li X, et al. Research progress on stem cell therapies for articular cartilage regeneration. Stem Cells Int. 2021;2021:1–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Emadedin M, Labibzadeh N, Liastani MG, et al. Intra-articular implantation of autologous bone marrow–derived mesenchymal stromal cells to treat knee osteoarthritis: a randomized, triple-blind, placebo-controlled phase 1/2 clinical trial. Cytotherapy. 2018;20:1238–46. [DOI] [PubMed] [Google Scholar]
- [141].Ni Z, Zhou S, Li S, et al. Exosomes: roles and therapeutic potential in osteoarthritis. Bone Res. 2020;8:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Jeyaraman M, Muthu S, Shehabaz S, et al. Current understanding of MSC-derived exosomes in the management of knee osteoarthritis. Exp Cell Res. 2022;418:113274. [DOI] [PubMed] [Google Scholar]
- [143].Wu J, Kuang L, Chen C, et al. miR-100-5p-abundant exosomes derived from infrapatellar fat pad MSCs protect articular cartilage and ameliorate gait abnormalities via inhibition of mTOR in osteoarthritis. Biomaterials. 2019;206:87–100. [DOI] [PubMed] [Google Scholar]
- [144].Jin Y, Xu M, Zhu H, et al. Therapeutic effects of bone marrow mesenchymal stem cells‐derived exosomes on osteoarthritis. J Cell Mol Med. 2021;25:9281–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Taghiyar L, Jahangir S, Ravari MK, Shamekhi MA, Eslaminejad MB. Cartilage repair by mesenchymal stem cell-derived exosomes: preclinical and clinical trial update and perspectives. Cell Biol Transl Med. 2021;1326:73–93. [DOI] [PubMed] [Google Scholar]
- [146].Duan L, Xu X, Xu L, et al. Exosome-mediated drug delivery for cell-free therapy of osteoarthritis. Curr Med Chem. 2021;28:6458–83. [DOI] [PubMed] [Google Scholar]
- [147].Giannotti L, Di Chiara Stanca B, Spedicato F, et al. Progress in regenerative medicine: exploring autologous platelet concentrates and their clinical applications. Genes. 2023;14:1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Acebes-Huerta A, Arias-Fernández T, Bernardo A, et al. Platelet-derived bio-products: classification update, applications, concerns and new perspectives. Transfus Apher Sci. 2020;59:102716. [DOI] [PubMed] [Google Scholar]
- [149].Everts P, Onishi K, Jayaram P, Lana JF, Mautner K. Platelet-rich plasma: new performance understandings and therapeutic considerations in 2020. Int J Mol Sci. 2020;21:7794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Gato-Calvo L, Magalhaes J, Ruiz-Romero C, Blanco FJ, Burguera EF. Platelet-rich plasma in osteoarthritis treatment: review of current evidence. Ther Adv Chronic Dis. 2019;10:2040622319825567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Sebbagh P, Cannone A, Gremion G, et al. Current status of PRP manufacturing requirements & European regulatory frameworks: practical tools for the appropriate implementation of PRP therapies in musculoskeletal regenerative medicine. Bioengineering (Basel). 2023;10:292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Bashor CJ, Hilton IB, Bandukwala H, Smith DM, Veiseh O. Engineering the next generation of cell-based therapeutics. Nat Rev Drug Discovery. 2022;21:655–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Claridge B, Lozano J, Poh QH, Greening DW. Development of extracellular vesicle therapeutics: challenges, considerations, and opportunities. Front Cell Dev Biol. 2021;9:734720. [DOI] [PMC free article] [PubMed] [Google Scholar]


