Next Article in Journal
Profiles of Elderly Patients with Obesity Hypoventilation Syndrome in Martinique: A Single-Center Study
Previous Article in Journal
Inhaled Corticosteroids and the Risk of Nontuberculous Mycobacterial Pulmonary Disease in Chronic Obstructive Pulmonary Disease: Findings from a Nationwide Population-Based Study
Previous Article in Special Issue
Current Surgical Techniques in the Treatment of Adult Developmental Dysplasia of the Hip
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmatherapeutic Treatment of Osteoarthrosis—Does the Pill against Already Exist? A Narrative Review

1
Department of Orthopedic Surgery and Traumatology, Hospital Barmherzige Brüder Munich, Romanstr. 93, 80639 München, Germany
2
Department of Traumatology and Orthopedic Surgery, Hospital Ludwigsburg, Posilipostr. 4, 71640 Ludwigsburg, Germany
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2023, 13(7), 1087; https://doi.org/10.3390/jpm13071087
Submission received: 12 June 2023 / Revised: 23 June 2023 / Accepted: 26 June 2023 / Published: 30 June 2023
(This article belongs to the Special Issue Cutting-Edge in Arthroplasty: Before, While and after Surgery)

Abstract

:
The aim of this narrative review is to summarize the current pharmacotherapeutic treatment options for osteoarthritis (OA). Is therapy still mainly symptomatic or does the pill against arthrosis already exist? Causal and non-causal, as well as future therapeutic approaches, are discussed. Various surgical and non-surgical treatment options are available that can help manage symptoms, slow down progression, and improve quality of life. To date, however, therapy is still mainly symptomatic, often using painkilling and anti-inflammatory drugs until the final stage, which is usually joint replacement. These “symptomatic pills against” have side effects and do not alter the progression of OA, which is caused by an imbalance between degenerative and regenerative processes. Next to resolving mechanical issues, the goal must be to gain a better understanding of the cellular and molecular basis of OA. Recently, there has been a lot of interest in cartilage-regenerative medicine and in the current style of treating rheumatoid arthritis, where drug therapy (“the pill against”) has been established to slow down or even stop the progression of rheumatoid arthritis and has banned the vast majority of former almost regular severe joint destructions. However, the “causal pill against” OA does not exist so far. First, the early detection of osteoarthritis by means of biomarkers and imaging should therefore gain more focus. Second, future therapeutic approaches have to identify innovative therapeutic approaches influencing inflammatory and metabolic processes. Several pharmacologic, genetic, and even epigenetic attempts are promising, but none have clinically improved causal therapy so far, unfortunately.

Graphical Abstract

1. Introduction

Osteoarthritis (OA) is a disease with a degenerative and inflammatory component that affects a large proportion of the ageing population [1]. The joints of the lower extremities are particularly affected, especially the hip and knee. Cartilage loss is the most visible change, but all joint structures are affected, some occurring quite a bit earlier than cartilage, such as the synovia or subchondral bone. OA involves a variety of factors, such as mechanical loading, ageing, inflammation, and metabolic changes, and the activation of different signaling pathways and enzymatic processes, ultimately leading to a progressive loss of joint function. Consequently, osteoarthritis is a heterogeneous disease with a common end route but many different starting points. Accordingly, there are diverse treatment modalities. They can either be conservative such as thermal, pharmacological, orthotic or physiotherapeutic, or surgical. The latter can be reconstructive, such as in cartilage stimulation or cartilage transplantation, load allocating, such as in osteotomies, or finally replacing, such as in terms of artificial joint replacement. However, especially in the early stages, the therapeutic approach often remains purely symptomatic. Currently, there is no causal drug for the treatment of OA. The most common treatment form is the use of painkilling and anti-inflammatory drugs until the final stage of treatment, which is usually joint replacement. However, the actual cause of OA is often not being treated. Another recent narrative review summarized the guidelines and symptomatic pharmacotherapeutic treatments [2].
The current review’s aim is to describe future perspectives. In recent years, the pathogenesis of osteoarthritis has become increasingly well understood, and new therapeutic approaches are emerging. There are clear similarities to rheumatoid arthritis, which led to a large number of rapid and severe joint destructions several decades ago. Today, those cases are rare due to new medical approaches, mainly the so-called biologicals [3]. The goal is to implement causal therapy and to abandon singular symptomatic therapy in OA as well. By influencing the underlying signaling pathways of OA or by using stem cells, attempts have been made to prevent the destruction of cartilage or to reduce the pain-triggering inflammatory reaction. The development of OA pharmacotherapies is primarily focused on the protection or even the regeneration of cartilage tissue, led by the assumption that the protection of cartilage structure also influences the clinical symptoms. Contradictorily, an MRI-based study has shown that a loss of cartilage thickness is associated with only a small amount of worsening knee pain, an association mediated in part by worsening synovitis [4]. Possible therapeutic approaches are, therefore, in addition to chondroprotection, the causal treatment of synovitis.
The aim of this narrative review is to summarize the current pharmacotherapeutic treatment options for OA therapy beyond the solely symptomatic attempts. Causal and non-causal, as well as future, therapeutic approaches are discussed below. This review attempts to think outside the box by also mentioning the approaches in the field of gene therapy and epigenetics. OA therapies that are not drug therapies are not addressed in this review. Lifestyle modifications, such as a change in eating habits and dietary weight management, as well as activity, physiotherapy, and mechanical issues also play a large part in successful therapy. A claim to the completeness of all possible therapy alternatives for OA can therefore not be made. The question addressed here is whether OA therapy will remain mainly symptomatic or if the pill against arthrosis already exists?

2. Symptomatic OA Therapy

2.1. Pain Killers

The most common therapy for OA is still the use of painkillers. In principle, there are different classes of substances available. These drugs are a purely symptomatic therapy that can alleviate but cannot influence the progression of OA. As this topic has been well described in several guidelines and in a recent narrative review [2], just a short summary of the most used painkillers will be presented in the following.
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most commonly used and prescribed pain medications in the treatment of OA and are routinely recommended in clinical practice guidelines [5]. Pain management is an essential part of OA treatment. Inflammation occurs in all joint tissues and is thus an essential target of drug therapy. Inflammation leads to the release of various neuromodulatory mediators such as cytokines and prostaglandins which are synthesized by the enzyme cyclooxygenase (COX). The release of proinflammatory mediators leads to the classic symptoms of synovitis, joint swelling and hyperthermia. Furthermore, inflammation can induce joint tissue damage [6,7,8].
NSAIDs can counteract these processes by reducing the corresponding inflammatory cascades by inhibiting the COX enzyme. Additionally, NSAIDs can reduce the sensation of pain relief by desensitizing nociceptors and are thus effective drugs in the symptomatic therapy of OA [9]. Smith et al. and Stewart et al. report in their meta-analysis that oral NSAIDs are similarly effective as opioids in relieving pain in patients with OA [10,11]. However, the use of NSAIDS is limited due to their known but rare risk of gastrointestinal, cardiovascular, and renal adverse events [12].
Studies have concluded that the oral use of NSAIDs is mainly recommended for short-term or intermittent therapy, rather than prolonged treatment [13]. Osani et al. reported in their meta-analysis that the therapeutic peak of NSAID-induced benefits in patients with knee OA was reached after 2 weeks and decreased over time, while cardiovascular and gastrointestinal side effects were already significantly increased after 4 weeks of treatment.
The usage of selective COX-2 inhibitors has increased in the past decades, due to their benefit of combining both anti-inflammatory and analgesic properties, while providing better gastrointestinal tolerability and a reduction in gastroduodenal ulcers in comparison with non-selective COX inhibitors [14].
Coxibs, however, have been associated with an increase in cardiovascular events due to a reduction in prostaglandin synthesis [15]. On the contrary, in a meta-analysis conducted by Cooper et al., recent data has shown that celecoxib does not significantly increase cardiovascular risk when compared with conventional NSAIDs and placebos, regardless of the dose and the duration of treatment [16]. However, recommendations on the analgesic use of various NSAIDs for patients with underlying health conditions remain conflicting. When prescribing any type of NSAIDs, all risk factors (age, gastrointestinal, cardiovascular, and renal events) should be taken into account. Due to adverse effects, NSAIDs are not intended for long term treatment.
A simple and effective way to reduce the risk of gastrointestinal side effects is the topical application of NSAIDs. Amemiya et al. compared the effects of esflurbiprofen patches versus flurbiprofen tablets in patients with gonarthrosis in 2021. Maximum esflurbiprofen concentrations were observed in the synovium, synovial fluids, and plasma after esflurbiprofen plaster (SFPP) application for 12 h. The numeric rating scale (NRS) results indicated a long-lasting effect of SFPP. Through transdermal application, a continuously high drug effect level was achieved. Overall, no dose peaks need to be accepted to achieve the same effect as with repeated daily oral administration [17].
Depending on the risk profile and the patient’s pain perception, it must be decided on a case-by-case basis whether oral or topical application is preferable.
Opioids are an alternative for more severe pain states when usual pain medication or other pain treatments are not sufficient or may not be used. In a direct comparison with NSAIDs, tramadol was inferior in terms of analgesia [18]. When prescribing opioids, the side effect profile, such as central nervous effects with fatigue, dizziness, and impaired balance, should be taken into account. This is where opioids differ adversely from NSAIDs. Opioids such as tramadol should not be prescribed as a first-line therapy. If other drugs are not used due to their side effect profile or if a non-drug therapy (e.g., surgery) is not currently available, they should be used. Opioids are not used long-term or routinely for osteoarthritis. However, they may be indicated for short-term therapy [2].

2.2. Symptomatic OA Therapy with Potentially Causal Impact

2.2.1. Hyaluronic Acid (Intra-Articular)

Hyaluronic acid (HA) is reduced in both concentration and molecular size in patients with osteoarthritis; hence, the intra-articular injection of HA aims to substitute the physiological synovial HA. The individual hyaluronic acid (HA) products differ in terms of production, molecular weight, degree of cross-linking, viscosity, and frequency of application per series. Despite a large number of scientific studies, the effectiveness of this therapy is still disputed in the literature. While some meta-analyses show tangible benefits, they often include studies with a high risk of bias. When meta-analyses are restricted to studies with a low risk of bias, the effect of IAHA is similar to that of saline injections [2,19]. There is a growing amount of literature demonstrating that product differences, particularly HA molecular weight, may have a significant effect on treatment outcomes, with a higher molecular weight showing better results [19]. Due to the invasive mode of application, the indication for intra-articular HA injection should, however, only be made when the prescription of NSAIDs is not possible due to side effects or contraindications or when these are not sufficiently effective. The possible side effects such as joint infection or irritation of the knee joint must be discussed with the patient in advance; severe adverse events are rare.

2.2.2. Corticosteroids (Intra-Articular)

The intra-articular injection of a glucocorticoid for the relief of the acute inflammatory symptoms of activated arthrosis can be a useful measure. The aim of the treatment is to reduce pain and restore mobility. Randomised and placebo-controlled studies have shown that the intra-articular injection of a glucocorticoid into an osteoarthritic knee joint can significantly reduce symptoms over a period of at least 1 week. Occasionally, however, a prolonged effect lasting 16–24 weeks is observed after the intra-articular application of glucocorticoids [20]. They should be used in the short term at the lowest possible but effective dosage for painful arthrosis that does not respond to other therapeutic measures. This may be the case, for example, in inflamed arthrosis with acute pain exacerbation.
There is a lack of data on the long-term effects of cortisone injections on articular cartilage and on a possible association with adverse joint effects. However, in some in vivo studies, corticosteroids were found to be cytotoxic to articular cartilage [21]. In a 2-year randomised trial, the cortisone-infiltration group showed higher cartilage loss than the saline-injection group. Taken together, cortisone injections are still a common treatment option, but they are not without side effects. Frequent use must therefore be considered critically [22]. Injections at too short intervals increase the risk of infection. The patient should be informed about the possible complications of infection and/or tissue atrophy as well as about possible treatment alternatives.

2.2.3. Platelet-Rich Plasma (Intra-Articular)

Aside from the available oral drugs against OA, the usage of autologous growth factors, e.g., intra-articular injections of platelet-rich plasma (PRP), can be used for OA treatment, especially for knee OA. The autologous fluid, which is obtained by centrifuging whole blood, is a highly concentrated cocktail of inflammatory mediators and growth factors capable of reducing inflammatory distress and stimulating cell proliferation and cartilaginous matrix production [16]. Between 2011 and 2021, 867 studies on the topic of PRP were published, with an upward trend over the years [23].
Multiple studies have confirmed effective pain relief and the improvement of physical function after PRP injections as well as an acceptable safety profile [24,25].
PRP injections also showed stronger effects compared with conventional injections with corticosteroids and hyaluronic acid [24]. Furthermore, regular injections of intra-articular corticosteroid can lead to the loss of cartilage structure and thus more rapid disease progression.
Consequently, PRP injections may not only contribute to pain relief through anti-inflammatory effects but can also provide lasting pain relief and functional restoration through targeted structural reconstruction when used over an extended period of time.
It was shown that PRP injections significantly improved physical function and WOMAC scores at 3, and up to 12, months [24].
Patients undergoing treatment with PRP injections experience both pain relief and improved joint function. However, it remains unclear whether the short-term effect of PRP injection is due to the temporary changes in the joint environment or whether PRP injections actually lead to structural changes, thus preventing the progression of OA. Another unresolved question surrounds which components of PRP cause this effect. In particular, the proportion of leukocytes (leukocyte-rich or leukocyte-poor) is still the subject of research. The first positive results have been achieved with leukocyte-poor plasma [26]. In summary, the use of PRP is showing very encouraging preliminary results; however, its use is not yet recommended as first-line-therapy in the guidelines.

2.2.4. Chondroitin and Glucosamine

Chondroitin and glucosamine have chondroprotective, analgesic, and anti-inflammatory effects. They are symptomatic, slow-acting drugs used against osteoarthritis. Glucosamine is a component of glycosaminoglycans and can be found in high amounts in articular cartilage and synovial fluid. Chondroitin is found in the extracellular matrix of articular cartilage and plays a role in maintaining osmotic pressure. Thus, it could improve elasticity and the resistance of cartilage [27]. Both chondroitin and glucosamine seem to develop their effects—partly in different forms—through the use of many different pathways. However, only a few selected ones will be mentioned below. Glucosamine was shown to decrease the levels of proinflammatory interleukin-1 (IL-1), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and C-reactive protein (CRP) in studies with rats. In contrast, the anti-inflammatory interleukins IL-2 and IL-10 were increased [28,29,30]. Glucosamine also appears to have immunomodulatory effects that affect the activity of phospholipase A2, matrix metalloproteinases, or aggrecans [31]. Moreover, chondroitin and glucosamine block the pathways involved in inflammation in osteoarthritis, such as the mitogen-activated protein kinase (MAPK) pathway [32]. In addition, both chondroitin and glucosamine have antioxidant effects [33].
In a meta-analysis by Zhu et al. from 2018, it was shown that chondroitin—via oral administration—significantly alleviates pain and leads to an improvement in physical function compared with a placebo. Glucosamine, on the other hand, may improve stiffness [34]. A combination of glucosamine and chondroitin appears to provide better pain relief than acetaminophen in hip and knee OA, but celecoxib showed the best results in this study [35]. It was found that both chondroitin alone and glucosamine alone could significantly reduce the decrease in joint space. Moreover, intraarticular injections of hyaluronic acid in combination with glucosamine hydrochloride led to a significantly higher reduction in IL-6, IL-1β, and TGF-β compared with hyaluronic acid alone in patients with temporomandibular OA [36].
Kwoh et al. and Fransen et al. failed to demonstrate any changes in joint structure with chondroitin or glucosamine administration in patients with chronic knee pain in OA [37,38]. Another meta-analysis summarized seventeen studies, of which only seven studies demonstrated a statistically significant reduction in pain and four studies demonstrated a reduction in joint space narrowing [39].
Several smaller dosages of glucosamine throughout the day appear to be more effective than one large dose per day [40].
Taken together, chondroitin and glucosamine seem to have an effect on the milder forms of OA, reducing joint inflammation and pain. The administration is safe and shows only a small number of adverse effects, such as headache or nausea. Overall, however, there are conflicting results regarding their clinical efficacy. Thus, in patients with contraindications to NSAIDs or with an increased risk of gastrointestinal or cardiovascular risks, the use of oral glucosamine and chondroitin may be considered as a treatment trial before more invasive therapies are undertaken.

2.2.5. Collagen

Collagen is a protein of the extracellular matrix that occurs mainly as collagen type II in the articular cartilage. The enteral absorption of undenatured type II collagen is very low, but di- or tripeptides containing the amino acids proline or hydroxyproline can be absorbed and show an effect [41]. Hydrolyzed collagen could contribute to cartilage regeneration by increasing the synthesis of macromolecules in the extracellular matrix [42]. In addition, collagen is able to modulate both humoral and cellular components of the immune system. It contributes to the body’s ability to distinguish between harmless molecules and potentially harmful pathogens [43]. This leads, for example, to the transformation of naive T cells into T regulatory cells that produce anti-inflammatory substances such as TGF-β and IL-10 [44]. Proline and hydroxyproline can induce hyaluronic acid synthesis [45] and the chondrocytes to synthesize glycosaminoglycans [46].
Pain in patients with hip and knee OA can be alleviated using an oral supplementation of collagen. WOMAC scores, VAS scores, and quality of life improve significantly compared with a placebo [47]. Trc et al. compared a supplementation of hydrolyzed collagen and glucosamine sulphate for 90 days, respectively. The supplementation of hydrolyzed collagen led to a statistically significant improvement in WOMAC and VAS scores compared with glucosamine sulphate [48].
Joint conditions seem to improve following the administration of collagen. It may induce cartilage repair to maintain structure and function. The clinical use of collagen is safe and has minimal adverse effects, mainly gastrointestinal. However, further studies are needed to show the benefits in the treatment of patients with OA and to determine the optimal dosage and duration.

3. Causal OA Therapy

3.1. Monoclonal Antibodies

Over the last few decades, monoclonal antibodies have emerged as a revolutionary tool in the field of medicine with many promising clinical applications. One of the biggest advantages of monoclonal antibodies is their high specificity as they are designed to target molecules in the body such as cytokines, growth factors, and receptors. In the context of osteoarthritis, antibodies revolutionized the treatment of rheumatoid arthritis. Currently, drug therapy (“the pill against”) has been established to slow down or even stop the progression of the autoimmune disease and has caused the end of the vast majority of formerly regular severe joint destructions. Although the effects and goals of treatment are partly comparable to OA, they are not established as standard therapy. The question arises, will this change? Up to now, there have been some approaches that use monoclonal antibodies in pain therapy of OA, some of which will be presented in the following.

3.1.1. TNF and IL-1 Inhibitors

In one randomized controlled trial, patients with erosive hand OA were treated with the anti-TNF antibody adalimumab (subcutaneous administration once a week) or a placebo for 12 weeks each. Pain intensity was measured with the VAS score. There was no significant difference [49]. Another anti-TNF antibody (etanercept) failed to demonstrate any benefit in a treatment duration of 24 weeks compared with a placebo in hand OA [50]. Canakinumab is an IL-1 inhibitor that showed a reduced rate of joint arthroplasties in patients with atherosclerotic disease [51]. However, further studies failed to show any benefits with respect to pain alleviation in patients with OA when IL-1 was blocked [52]. Overall, TNF and IL-1 inhibitors seem to be rather unsuitable for patients with OA.

3.1.2. Anti-NGF

Joint tissues have been innervated using nociceptors, except for cartilage. Nerve growth factor is an important neurotrophin in inflamed synovium. It is upregulated in patients with OA and leads to an increase in pain. There are three different monoclonal antibodies used in therapy: Tanezumab, Ulranumab, and Fasinumab. They lead to impressive pain relief in patients with knee and hip OA but accelerate the progression of OA [53]. The administration of fewer doses showed a reduced but still substantial effect on pain and function. Nevertheless, 3% of the patients suffered from progressive OA [54]. There are a few other adverse effects such as peripheral neuropathies, headaches, upper respiratory tract infections, oedema, or joint pain [55]. NGF seems to be a relevant factor for cartilage integrity or the repair of cartilage, so that a complete blockade is not an effective treatment in patients with OA. Anti-NGF treatment is promising, but studies are needed to find the optimal dosage to alleviate pain and reduce the adverse effects.

3.2. Stem Cell Treatments

Mesenchymal stem cells (MSCs), as a specific type of adult stem cell, possess great potential in regenerative therapy due to their capacity for self-renewal and differentiation [56]. Great attention has been paid to cell-based therapy that may influence cartilage repair such as mesenchymal stem cell therapy. Most studies have been conducted in the context of knee joint osteoarthritis. MSCs are primarily used as intra-articular injection therapy. MSCs modulate immune or inflammatory effects and tissue regeneration in knee osteoarthritis [57,58]. The exact mechanism of MSC therapy remains unclear. It is known that cartilage repair and protection against OA-induced cartilage degeneration is promoted by MSC-derived extracellular vesicles.
Injected MSCs are expected to repair damaged issues due to the trilineage potential and immunomodulatory properties of MSCs. MSCs can be harvested from different sites. The best known or most accessible sites are bone marrow or fat tissue. Other sources include muscle tissue, synovial membranes, or placenta. In addition, the cells can be obtained either from autogenic or allogenic sources. The advantage of allogenic stem cells is that they can be harvested from healthy donors and expanded in vitro to obtain a clinically relevant amount for injection. The disadvantage of allogeneic cell collection is a possible reaction of the recipient’s immune system after injection [59].
Studies in humans have reported variable structural outcomes after MSC injection from hyaline-like cartilage to fibrous tissue. A meta-analysis including 582 knee-OA-patients in 11 trials was performed to assess the efficacy and safety of MSC treatment for knee OA patients using VAS, IKDC, WOMAC, Lequesne, Lysholm, and Tegner scores. MSC-treatment groups from the identified trials were compared with their respective control groups. It shows that VAS decreases and IKDC increases significantly after 24 months follow up. MSC therapy also showed significant decreases in WOMAC and Lequesne scores after the 12-month follow up. The evaluation of Lysholm (24-month) and Tegner (12- and 24-month) scores also demonstrated favorable results for MSC treatment. The effects of MSC therapy on short-term primary endpoints still need to be evaluated in a larger number of patients [60].
Another important question is the dosage at which the stem cells should be injected. A larger amount of injected MSCs may be expected to induce better effects. Interestingly, in studies with allogeneic stem cells, it was found that no improvement was observed in relation to “high dose” as opposed to “low dose” stem cell transplantation. The clinical symptoms and MRI imaging of the cartilage were the main factors assessed. There were also differences in the dose effect of stem cells depending on their origin. These results suggest that appropriate MSC doses applied in intra-articular injections to OA patients need to be determined for each origin of MSCs [61,62]. Furthermore, MSC injection combined with other agents such as hyaluronic acid [63] or PRP [64] has better therapeutic effects than MSC injection alone. This implies the possible value of drug cocktail therapy when using MSC injection in knee OA patients.
Overall, MSC transplantation treatment was shown to be safe and has great potential as an efficacious clinical therapy, especially for patients with knee OA. Further clinical and in vitro studies are needed to better clarify the underlying molecular and biochemical mechanisms. Particularly, it is yet to be determined whether MSCs should be injected as a single agent or in combination with another drug or as a complementary therapy to surgical treatment.

4. Future Directions

4.1. Gene Therapy

Gene therapy consists of using a vector to bring genes directly into cells and tissues to treat a specific disease. Viral vectors include RNA viruses and DNA viruses. Two different gene therapy strategies are currently in preclinical and clinical development for OA [65]. The first approach consists of ex vivo modifying and amplifying cells, followed by their intra-articular injection. The aim is to over-express TGF-ß-1 in irradiated allogenic chondrocytes [66]. The second approach is an in vivo gene therapy through the local or systemic injection of viral vectors containing the transgene of interest. In general, OA gene therapy aims to reduce inflammation through overexpressing transgenes such as IL-1Ra or a soluble TNF receptor [67]. In the future, gene therapy could become a strategy to regulate the intra-articular expression of therapeutic targets in OA.

4.2. Epigenetics

Epigenetics is a field of research that analyzes the changes in gene expression or cell phenotype occurring without the modification of the DNA sequence. Several epigenetic regulators appear to be involved in the pathogenesis of OA. The epigenetic profiling of articular chondrocytes has revealed the existence of an activating sequence that is present in billions of people with a risk locus (GDF5-UQCC1) that is involved in OA progression. These epigenetic modifications can also suppress the expression of protective genes in OA [61]. Abnormal changes in DNA methylation occur in the promoter regions of related genes and signaling pathways in OA chondrocytes. Epigenetic regulation typically involves DNA methylation, histone modification, and noncoding RNA-mediated regulation. Epigenetic mechanisms can control several signaling pathways simultaneously. For this reason, epigenetic modifications have been considered a potential therapeutic target to manage OA [68].

5. Conclusions

OA is caused by an imbalance between degenerative and regenerative processes. To date, therapy is still mainly symptomatic. As well as improving mechanical issues, the future therapeutic goal must be to gain an even better understanding of the cellular and molecular causes of OA. Non-surgical therapy comprises basic measures such as weight reduction, exercise therapy (water and land), and health education. Specific measures comprise biomechanical interventions, physiotherapy, physical measures, and drug therapy. Several pharmacologic, genetic, and even epigenetic attempts are promising, but unfortunately, so far none have proven causal therapy to work or cure OA. The early detection of osteoarthritis by means of biomarkers and imaging must also gain focus to allow for early and targeted treatment.
With regards to drug therapy, the individual risk profile as well as the level of suffering or pain intensity must be taken into account before treatment is started. For this reason, it is not possible to give general advice on drug therapy for OA. A non-causal but proven treatment option for OA is the use of painkillers (mainly NSAIDs), which are beyond the focus of this review. Their duration of therapy is limited due to side effects, especially in patients with corresponding underlying diseases. They are solely symptomatic, therefore alleviate but do not alter the progression of OA. The use of PRP injections seems to clearly overcome hyaluronic acid which has recently shown conflicting results. PRP can potentially stimulate cell proliferation and cartilaginous matrix production and provide lasting pain relief and functional restoration through targeted structural reconstruction when used over an extended period of time. Therapies with chondroprotective substances such as chondroitin, glucosamine, collagen, or monoclonal antibodies lead to a reduction in pain. However, a significant therapeutic effect in singular application has not been detected so far. The use of stem cells in arthrosis therapy, however, is a promising therapy. Its possibility for cell regeneration or conversion into functional cells holds great potential, especially in the context of the therapy of degenerative diseases such as OA. Favored cell sources, dosage, and therapy duration remain unclear.
Due to the multifactorial genesis of OA, most therapeutic approaches are still symptomatic and the “causal pill against” OA does not yet exist. Future therapeutic approaches have to identify innovative therapeutic targets aimed at influencing the inflammatory and metabolic processes underlying the pathogenesis and progression of OA.

Author Contributions

Conceptualization, J.B. and F.W.; methodology, J.B. and F.W.; software, F.W.; validation, J.B. and F.W.; formal analysis, J.B. and F.W.; investigation, J.B. and F.W.; resources, J.B. and F.W.; data curation, F.W.; writing—original draft preparation, F.W., P.B., A.-M.B., and C.B.; writing—review and editing, J.B. and F.W.; visualization, J.R.; supervision, J.B. and F.W.; project administration, J.B. and F.W.; funding acquisition, J.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Das, S.K.; Farooqi, A. Osteoarthritis. Best Pract. Res. Clin. Rheumatol. 2008, 22, 657–675. [Google Scholar] [CrossRef]
  2. Richard, M.J.; Driban, J.B.; McAlindon, T.E. Pharmaceutical treatment of osteoarthritis. Osteoarthr. Cartil. 2023, 31, 458–466. [Google Scholar] [CrossRef] [PubMed]
  3. Smolen, J.S.; Landewe, R.B.M.; Bijlsma, J.W.J.; Burmester, G.R.; Dougados, M.; Kerschbaumer, A.; McInnes, I.B.; Sepriano, A.; van Vollenhoven, R.F.; de Wit, M.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann. Rheum. Dis. 2020, 79, 685–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Bacon, K.; LaValley, M.P.; Jafarzadeh, S.R.; Felson, D. Does cartilage loss cause pain in osteoarthritis and if so, how much? Ann. Rheum. Dis. 2020, 79, 1105–1110. [Google Scholar] [CrossRef]
  5. Arden, N.K.; Perry, T.A.; Bannuru, R.R.; Bruyere, O.; Cooper, C.; Haugen, I.K.; Hochberg, M.C.; McAlindon, T.E.; Mobasheri, A.; Reginster, J.Y. Non-surgical management of knee osteoarthritis: Comparison of ESCEO and OARSI 2019 guidelines. Nat. Rev. Rheumatol. 2021, 17, 59–66. [Google Scholar] [CrossRef]
  6. Dawes, J.M.; Kiesewetter, H.; Perkins, J.R.; Bennett, D.L.; McMahon, S.B. Chemokine expression in peripheral tissues from the monosodium iodoacetate model of chronic joint pain. Mol. Pain 2013, 9, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Driscoll, C.; Chanalaris, A.; Knights, C.; Ismail, H.; Sacitharan, P.K.; Gentry, C.; Bevan, S.; Vincent, T.L. Nociceptive Sensitizers Are Regulated in Damaged Joint Tissues, Including Articular Cartilage, When Osteoarthritic Mice Display Pain Behavior. Arthritis Rheumatol. 2016, 68, 857–867. [Google Scholar] [CrossRef]
  8. Pinho-Ribeiro, F.A.; Verri, W.A., Jr.; Chiu, I.M. Nociceptor Sensory Neuron-Immune Interactions in Pain and Inflammation. Trends Immunol. 2017, 38, 5–19. [Google Scholar] [CrossRef] [Green Version]
  9. Osani, M.C.; Vaysbrot, E.E.; Zhou, M.; McAlindon, T.E.; Bannuru, R.R. Duration of Symptom Relief and Early Trajectory of Adverse Events for Oral Nonsteroidal Antiinflammatory Drugs in Knee Osteoarthritis: A Systematic Review and Meta-Analysis. Arthritis Care Res. 2020, 72, 641–651. [Google Scholar] [CrossRef]
  10. Smith, S.R.; Deshpande, B.R.; Collins, J.E.; Katz, J.N.; Losina, E. Comparative pain reduction of oral non-steroidal anti-inflammatory drugs and opioids for knee osteoarthritis: Systematic analytic review. Osteoarthr. Cartil. 2016, 24, 962–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Stewart, M.; Cibere, J.; Sayre, E.C.; Kopec, J.A. Efficacy of commonly prescribed analgesics in the management of osteoarthritis: A systematic review and meta-analysis. Rheumatol. Int. 2018, 38, 1985–1997. [Google Scholar] [CrossRef] [PubMed]
  12. O′Neil, C.K.; Hanlon, J.T.; Marcum, Z.A. Adverse effects of analgesics commonly used by older adults with osteoarthritis: Focus on non-opioid and opioid analgesics. Am. J. Geriatr. Pharmacother. 2012, 10, 331–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bannuru, R.R.; Osani, M.C.; Vaysbrot, E.E.; Arden, N.K.; Bennell, K.; Bierma-Zeinstra, S.M.A.; Kraus, V.B.; Lohmander, L.S.; Abbott, J.H.; Bhandari, M.; et al. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthr. Cartil. 2019, 27, 1578–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Coxib and Traditional NSAID Trialists’ (CNT) Collaboration; Bhala, N.; Emberson, J.; Merhi, A.; Abramson, S.; Arber, N.; Baron, J.A.; Bombardier, C.; Cannon, C.; Farkouh, M.E.; et al. Vascular and upper gastrointestinal effects of non-steroidal anti-inflammatory drugs: Meta-analyses of individual participant data from randomised trials. Lancet 2013, 382, 769–779. [Google Scholar] [CrossRef] [Green Version]
  15. Gunter, B.R.; Butler, K.A.; Wallace, R.L.; Smith, S.M.; Harirforoosh, S. Non-steroidal anti-inflammatory drug-induced cardiovascular adverse events: A meta-analysis. J. Clin. Pharm. Ther. 2017, 42, 27–38. [Google Scholar] [CrossRef] [Green Version]
  16. Cooper, D.L.; Harirforoosh, S. Effect of formulation variables on preparation of celecoxib loaded polylactide-co-glycolide nanoparticles. PLoS ONE 2014, 9, e113558. [Google Scholar] [CrossRef]
  17. Amemiya, M.; Nakagawa, Y.; Yoshimura, H.; Takahashi, T.; Inomata, K.; Nagase, T.; Ju, Y.J.; Shimaya, M.; Tsukada, S.; Hirasawa, N.; et al. Comparison of tissue pharmacokinetics of esflurbiprofen plaster with flurbiprofen tablets in patients with knee osteoarthritis: A multicenter randomized controlled trial. Biopharm. Drug Dispos. 2021, 42, 418–426. [Google Scholar] [CrossRef] [PubMed]
  18. Welsch, P.; Sommer, C.; Schiltenwolf, M.; Hauser, W. Opioids in chronic noncancer pain-are opioids superior to nonopioid analgesics? A systematic review and meta-analysis of efficacy, tolerability and safety in randomized head-to-head comparisons of opioids versus nonopioid analgesics of at least four week’s duration. Schmerz 2015, 29, 85–95. [Google Scholar] [CrossRef]
  19. Phillips, M.; Vannabouathong, C.; Devji, T.; Patel, R.; Gomes, Z.; Patel, A.; Dixon, M.; Bhandari, M. Differentiating factors of intra-articular injectables have a meaningful impact on knee osteoarthritis outcomes: A network meta-analysis. Knee Surg. Sports Traumatol. Arthrosc. 2020, 28, 3031–3039. [Google Scholar] [CrossRef] [Green Version]
  20. Hepper, C.T.; Halvorson, J.J.; Duncan, S.T.; Gregory, A.J.; Dunn, W.R.; Spindler, K.P. The efficacy and duration of intra-articular corticosteroid injection for knee osteoarthritis: A systematic review of level I studies. J. Am. Acad. Orthop. Surg. 2009, 17, 638–646. [Google Scholar] [CrossRef] [Green Version]
  21. Kijowski, R. Risks and Benefits of Intra-articular Corticosteroid Injection for Treatment of Osteoarthritis: What Radiologists and Patients Need to Know. Radiology 2019, 293, 664–665. [Google Scholar] [CrossRef] [PubMed]
  22. Sabha, M.; Hochberg, M.C. Non-surgical management of hip and knee osteoarthritis; comparison of ACR/AF and OARSI 2019 and VA/DoD 2020 guidelines. Osteoarthr. Cartil. Open 2022, 4, 100232. [Google Scholar] [CrossRef] [PubMed]
  23. Cui, Y.; Lin, L.; Wang, Z.; Wang, K.; Xiao, L.; Lin, W.; Zhang, Y. Research trends of platelet-rich plasma therapy on knee osteoarthritis from 2011 to 2021: A review. Medicine 2023, 102, e32434. [Google Scholar] [CrossRef] [PubMed]
  24. Shen, L.; Yuan, T.; Chen, S.; Xie, X.; Zhang, C. 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. [Google Scholar] [CrossRef] [Green Version]
  25. Di Martino, A.; Di Matteo, B.; Papio, T.; Tentoni, F.; Selleri, F.; Cenacchi, A.; Kon, E.; Filardo, G. Platelet-Rich Plasma Versus Hyaluronic Acid Injections for the Treatment of Knee Osteoarthritis: Results at 5 Years of a Double-Blind, Randomized Controlled Trial. Am. J. Sports Med. 2019, 47, 347–354. [Google Scholar] [CrossRef]
  26. Belk, J.W.; Kraeutler, M.J.; Houck, D.A.; Goodrich, J.A.; Dragoo, J.L.; McCarty, E.C. Platelet-Rich Plasma Versus Hyaluronic Acid for Knee Osteoarthritis: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Am. J. Sports Med. 2021, 49, 249–260. [Google Scholar] [CrossRef]
  27. Jomphe, C.; Gabriac, M.; Hale, T.M.; Heroux, L.; Trudeau, L.E.; Deblois, D.; Montell, E.; Verges, J.; du Souich, P. Chondroitin sulfate inhibits the nuclear translocation of nuclear factor-kappaB in interleukin-1beta-stimulated chondrocytes. Basic Clin. Pharmacol. Toxicol. 2008, 102, 59–65. [Google Scholar] [CrossRef]
  28. Aghazadeh-Habashi, A.; Kohan, M.H.; Asghar, W.; Jamali, F. Glucosamine dose/concentration-effect correlation in the rat with adjuvant arthritis. J. Pharm. Sci. 2014, 103, 760–767. [Google Scholar] [CrossRef]
  29. Li, Y.; Chen, L.; Liu, Y.; Zhang, Y.; Liang, Y.; Mei, Y. Anti-inflammatory effects in a mouse osteoarthritis model of a mixture of glucosamine and chitooligosaccharides produced by bi-enzyme single-step hydrolysis. Sci. Rep. 2018, 8, 5624. [Google Scholar] [CrossRef] [Green Version]
  30. Waly, N.E.; Refaiy, A.; Aborehab, N.M. IL-10 and TGF-beta: Roles in chondroprotective effects of Glucosamine in experimental Osteoarthritis? Pathophysiology 2017, 24, 45–49. [Google Scholar] [CrossRef]
  31. Imagawa, K.; de Andres, M.C.; Hashimoto, K.; Pitt, D.; Itoi, E.; Goldring, M.B.; Roach, H.I.; Oreffo, R.O. The epigenetic effect of glucosamine and a nuclear factor-kappa B (NF-kB) inhibitor on primary human chondrocytes—Implications for osteoarthritis. Biochem. Biophys. Res. Commun. 2011, 405, 362–367. [Google Scholar] [CrossRef] [Green Version]
  32. Wen, Z.H.; Tang, C.C.; Chang, Y.C.; Huang, S.Y.; Hsieh, S.P.; Lee, C.H.; Huang, G.S.; Ng, H.F.; Neoh, C.A.; Hsieh, C.S.; et al. Glucosamine sulfate reduces experimental osteoarthritis and nociception in rats: Association with changes of mitogen-activated protein kinase in chondrocytes. Osteoarthr. Cartil. 2010, 18, 1192–1202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kuptniratsaikul, V.; Dajpratham, P.; Taechaarpornkul, W.; Buntragulpoontawee, M.; Lukkanapichonchut, P.; Chootip, C.; Saengsuwan, J.; Tantayakom, K.; Laongpech, S. Efficacy and safety of Curcuma domestica extracts compared with ibuprofen in patients with knee osteoarthritis: A multicenter study. Clin. Interv. Aging 2014, 9, 451–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhu, X.; Sang, L.; Wu, D.; Rong, J.; Jiang, L. Effectiveness and safety of glucosamine and chondroitin for the treatment of osteoarthritis: A meta-analysis of randomized controlled trials. J. Orthop. Surg. Res. 2018, 13, 170. [Google Scholar] [CrossRef] [Green Version]
  35. Zhu, X.; Wu, D.; Sang, L.; Wang, Y.; Shen, Y.; Zhuang, X.; Chu, M.; Jiang, L. Comparative effectiveness of glucosamine, chondroitin, acetaminophen or celecoxib for the treatment of knee and/or hip osteoarthritis: A network meta-analysis. Clin. Exp. Rheumatol. 2018, 36, 595–602. [Google Scholar] [PubMed]
  36. Zeng, C.; Wei, J.; Li, H.; Wang, Y.L.; Xie, D.X.; Yang, T.; Gao, S.G.; Li, Y.S.; Luo, W.; Lei, G.H. Effectiveness and safety of Glucosamine, chondroitin, the two in combination, or celecoxib in the treatment of osteoarthritis of the knee. Sci. Rep. 2015, 5, 16827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Kwoh, C.K.; Roemer, F.W.; Hannon, M.J.; Moore, C.E.; Jakicic, J.M.; Guermazi, A.; Green, S.M.; Evans, R.W.; Boudreau, R. Effect of oral glucosamine on joint structure in individuals with chronic knee pain: A randomized, placebo-controlled clinical trial. Arthritis Rheumatol. 2014, 66, 930–939. [Google Scholar] [CrossRef] [PubMed]
  38. Fransen, M.; Agaliotis, M.; Nairn, L.; Votrubec, M.; Bridgett, L.; Su, S.; Jan, S.; March, L.; Edmonds, J.; Norton, R.; et al. Glucosamine and chondroitin for knee osteoarthritis: A double-blind randomised placebo-controlled clinical trial evaluating single and combination regimens. Ann. Rheum. Dis. 2015, 74, 851–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Knapik, J.J.; Pope, R.; Hoedebecke, S.S.; Schram, B.; Orr, R.; Lieberman, H.R. Effects of Oral Glucosamine Sulfate on Osteoarthritis-Related Pain and Joint-Space Changes: Systematic Review and Meta-Analysis. J. Spec. Oper Med. 2018, 18, 139–147. [Google Scholar] [CrossRef]
  40. Chiu, H.W.; Li, L.H.; Hsieh, C.Y.; Rao, Y.K.; Chen, F.H.; Chen, A.; Ka, S.M.; Hua, K.F. Glucosamine inhibits IL-1beta expression by preserving mitochondrial integrity and disrupting assembly of the NLRP3 inflammasome. Sci. Rep. 2019, 9, 5603. [Google Scholar] [CrossRef] [Green Version]
  41. Ohara, H.; Matsumoto, H.; Ito, K.; Iwai, K.; Sato, K. Comparison of quantity and structures of hydroxyproline-containing peptides in human blood after oral ingestion of gelatin hydrolysates from different sources. J. Agric. Food Chem. 2007, 55, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  42. Henrotin, Y.; Sanchez, C.; Balligand, M. Pharmaceutical and nutraceutical management of canine osteoarthritis: Present and future perspectives. Vet. J. 2005, 170, 113–123. [Google Scholar] [CrossRef] [PubMed]
  43. Bagchi, D.; Misner, B.; Bagchi, M.; Kothari, S.C.; Downs, B.W.; Fafard, R.D.; Preuss, H.G. Effects of orally administered undenatured type II collagen against arthritic inflammatory diseases: A mechanistic exploration. Int. J. Clin. Pharmacol. Res. 2002, 22, 101–110. [Google Scholar] [PubMed]
  44. Tong, T.; Zhao, W.; Wu, Y.Q.; Chang, Y.; Wang, Q.T.; Zhang, L.L.; Wei, W. Chicken type II collagen induced immune balance of main subtype of helper T cells in mesenteric lymph node lymphocytes in rats with collagen-induced arthritis. Inflamm. Res. 2010, 59, 369–377. [Google Scholar] [CrossRef]
  45. Ohara, H.; Iida, H.; Ito, K.; Takeuchi, Y.; Nomura, Y. Effects of Pro-Hyp, a collagen hydrolysate-derived peptide, on hyaluronic acid synthesis using in vitro cultured synovium cells and oral ingestion of collagen hydrolysates in a guinea pig model of osteoarthritis. Biosci. Biotechnol. Biochem. 2010, 74, 2096–2099. [Google Scholar] [CrossRef] [Green Version]
  46. Gordon, M.K.; Hahn, R.A. Collagens. Cell Tissue Res. 2010, 339, 247–257. [Google Scholar] [CrossRef]
  47. Garcia-Coronado, J.M.; Martinez-Olvera, L.; Elizondo-Omana, R.E.; Acosta-Olivo, C.A.; Vilchez-Cavazos, F.; Simental-Mendia, L.E.; Simental-Mendia, M. Effect of collagen supplementation on osteoarthritis symptoms: A meta-analysis of randomized placebo-controlled trials. Int. Orthop. 2019, 43, 531–538. [Google Scholar] [CrossRef]
  48. Trc, T.; Bohmova, J. Efficacy and tolerance of enzymatic hydrolysed collagen (EHC) vs. glucosamine sulphate (GS) in the treatment of knee osteoarthritis (KOA). Int. Orthop. 2011, 35, 341–348. [Google Scholar] [CrossRef] [Green Version]
  49. Aitken, D.; Laslett, L.L.; Pan, F.; Haugen, I.K.; Otahal, P.; Bellamy, N.; Bird, P.; Jones, G. A randomised double-blind placebo-controlled crossover trial of HUMira (adalimumab) for erosive hand OsteoaRthritis—The HUMOR trial. Osteoarthr. Cartil. 2018, 26, 880–887. [Google Scholar] [CrossRef] [Green Version]
  50. Kloppenburg, M.; Ramonda, R.; Bobacz, K.; Kwok, W.Y.; Elewaut, D.; Huizinga, T.W.J.; Kroon, F.P.B.; Punzi, L.; Smolen, J.S.; Vander Cruyssen, B.; et al. Etanercept in patients with inflammatory hand osteoarthritis (EHOA): A multicentre, randomised, double-blind, placebo-controlled trial. Ann. Rheum. Dis. 2018, 77, 1757–1764. [Google Scholar] [CrossRef]
  51. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, S.X.; Abramson, S.B.; Attur, M.; Karsdal, M.A.; Preston, R.A.; Lozada, C.J.; Kosloski, M.P.; Hong, F.; Jiang, P.; Saltarelli, M.J.; et al. Safety, tolerability, and pharmacodynamics of an anti-interleukin-1alpha/beta dual variable domain immunoglobulin in patients with osteoarthritis of the knee: A randomized phase 1 study. Osteoarthr. Cartil. 2017, 25, 1952–1961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Schnitzer, T.J.; Easton, R.; Pang, S.; Levinson, D.J.; Pixton, G.; Viktrup, L.; Davignon, I.; Brown, M.T.; West, C.R.; Verburg, K.M. 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, 37–48. [Google Scholar] [CrossRef] [PubMed]
  54. Berenbaum, F.; Blanco, F.J.; Guermazi, A.; Miki, K.; Yamabe, T.; Viktrup, L.; Junor, R.; Carey, W.; Brown, M.T.; West, C.R.; et al. Subcutaneous tanezumab for osteoarthritis of the hip or knee: Efficacy and safety results from a 24-week randomised phase III study with a 24-week follow-up period. Ann. Rheum. Dis. 2020, 79, 800–810. [Google Scholar] [CrossRef] [PubMed]
  55. Lane, N.E.; Schnitzer, T.J.; Birbara, C.A.; Mokhtarani, M.; Shelton, D.L.; Smith, M.D.; Brown, M.T. Tanezumab for the treatment of pain from osteoarthritis of the knee. N. Engl. J. Med. 2010, 363, 1521–1531. [Google Scholar] [CrossRef]
  56. Liu, D.; Kou, X.; Chen, C.; Liu, S.; Liu, Y.; Yu, W.; Yu, T.; Yang, R.; Wang, R.; Zhou, Y.; et al. Circulating apoptotic bodies maintain mesenchymal stem cell homeostasis and ameliorate osteopenia via transferring multiple cellular factors. Cell Res. 2018, 28, 918–933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Marks, P.W.; Witten, C.M.; Califf, R.M. Clarifying Stem-Cell Therapy’s Benefits and Risks. N. Engl. J. Med. 2017, 376, 1007–1009. [Google Scholar] [CrossRef] [Green Version]
  58. Jones, I.A.; Togashi, R.; Wilson, M.L.; Heckmann, N.; Vangsness, C.T., Jr. Intra-articular treatment options for knee osteoarthritis. Nat. Rev. Rheumatol. 2019, 15, 77–90. [Google Scholar] [CrossRef]
  59. Ankrum, J.A.; Ong, J.F.; Karp, J.M. Mesenchymal stem cells: Immune evasive, not immune privileged. Nat. Biotechnol. 2014, 32, 252–260. [Google Scholar] [CrossRef] [Green Version]
  60. Yubo, M.; Yanyan, L.; Li, L.; Tao, S.; Bo, L.; Lin, C. Clinical efficacy and safety of mesenchymal stem cell transplantation for osteoarthritis treatment: A meta-analysis. PLoS ONE 2017, 12, e0175449. [Google Scholar] [CrossRef] [PubMed]
  61. Gupta, P.K.; Chullikana, A.; Rengasamy, M.; Shetty, N.; Pandey, V.; Agarwal, V.; Wagh, S.Y.; Vellotare, P.K.; Damodaran, D.; Viswanathan, P.; et al. Efficacy and safety of adult human bone marrow-derived, cultured, pooled, allogeneic mesenchymal stromal cells (Stempeucel(R)): Preclinical and clinical trial in osteoarthritis of the knee joint. Arthritis Res. Ther. 2016, 18, 301. [Google Scholar] [CrossRef] [Green Version]
  62. Lu, L.; Dai, C.; Du, H.; Li, S.; Ye, P.; Zhang, L.; Wang, X.; Song, Y.; Togashi, R.; Vangsness, C.T.; et al. Intra-articular injections of allogeneic human adipose-derived mesenchymal progenitor cells in patients with symptomatic bilateral knee osteoarthritis: A Phase I pilot study. Regen. Med. 2020, 15, 1625–1636. [Google Scholar] [CrossRef] [PubMed]
  63. Lamo-Espinosa, J.M.; Mora, G.; Blanco, J.F.; Granero-Molto, F.; Nunez-Cordoba, J.M.; Sanchez-Echenique, C.; Bondia, J.M.; Aquerreta, J.D.; Andreu, E.J.; Ornilla, E.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bastos, R.; Mathias, M.; Andrade, R.; Bastos, R.; Balduino, A.; Schott, V.; Rodeo, S.; Espregueira-Mendes, J. Intra-articular injections of expanded mesenchymal stem cells with and without addition of platelet-rich plasma are safe and effective for knee osteoarthritis. Knee Surg. Sports Traumatol. Arthrosc. 2018, 26, 3342–3350. [Google Scholar] [CrossRef] [PubMed]
  65. Delplace, V.; Boutet, M.A.; Le Visage, C.; Maugars, Y.; Guicheux, J.; Vinatier, C. Osteoarthritis: From upcoming treatments to treatments yet to come. Jt. Bone Spine 2021, 88, 105206. [Google Scholar] [CrossRef]
  66. Cherian, J.J.; Parvizi, J.; Bramlet, D.; Lee, K.H.; Romness, D.W.; Mont, M.A. Preliminary results of a phase II randomized study to determine the efficacy and safety of genetically engineered allogeneic human chondrocytes expressing TGF-beta1 in patients with grade 3 chronic degenerative joint disease of the knee. Osteoarthr. Cartil. 2015, 23, 2109–2118. [Google Scholar] [CrossRef] [Green Version]
  67. Nixon, A.J.; Grol, M.W.; Lang, H.M.; Ruan, M.Z.C.; Stone, A.; Begum, L.; Chen, Y.; Dawson, B.; Gannon, F.; Plutizki, S.; et al. Disease-Modifying Osteoarthritis Treatment With Interleukin-1 Receptor Antagonist Gene Therapy in Small and Large Animal Models. Arthritis Rheumatol. 2018, 70, 1757–1768. [Google Scholar] [CrossRef] [Green Version]
  68. Tong, L.; Yu, H.; Huang, X.; Shen, J.; Xiao, G.; Chen, L.; Wang, H.; Xing, L.; Chen, D. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res. 2022, 10, 60. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wilken, F.; Buschner, P.; Benignus, C.; Behr, A.-M.; Rieger, J.; Beckmann, J. Pharmatherapeutic Treatment of Osteoarthrosis—Does the Pill against Already Exist? A Narrative Review. J. Pers. Med. 2023, 13, 1087. https://doi.org/10.3390/jpm13071087

AMA Style

Wilken F, Buschner P, Benignus C, Behr A-M, Rieger J, Beckmann J. Pharmatherapeutic Treatment of Osteoarthrosis—Does the Pill against Already Exist? A Narrative Review. Journal of Personalized Medicine. 2023; 13(7):1087. https://doi.org/10.3390/jpm13071087

Chicago/Turabian Style

Wilken, Frauke, Peter Buschner, Christian Benignus, Anna-Maria Behr, Johannes Rieger, and Johannes Beckmann. 2023. "Pharmatherapeutic Treatment of Osteoarthrosis—Does the Pill against Already Exist? A Narrative Review" Journal of Personalized Medicine 13, no. 7: 1087. https://doi.org/10.3390/jpm13071087

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop