SYSADOAs: do their origin and quality make a difference in efficacy and safety?

24 Febrero, 2021
11 Marzo, 2021
24 Marzo, 2021
Citar como:
Citar como Vitaloni M, Möller I, Verges J. SYSADOAs: su origen y calidad hacen la diferencia en eficacia y seguridad? [Internet]. Global Rheumatology. Vol 2 / Ene - Jun [2021]. Available from: https://doi.org/10.46856/grp.10.e072

Lecturas 1976Lecturas

Licencia

Este es un artículo de acceso abierto, distribuido bajo los términos de Creative Commons Attribution (CC .BY. NC-4). Esta permitido copiar y redistribuir el material en cualquier medio o formato. Remezclar, transformar y construir a partir del material . Usted debe dar crédito de manera adecuada, brindar un enlace a la licencia, e indicar si se han realizado cambios. Puede hacerlo en cualquier forma razonable, pero no de forma tal que sugiera que usted o su uso tienen el apoyo de la licenciante. Usted no puede hacer uso del material con propósitos comerciales.

SYSADOAs: do their origin and quality make a difference in efficacy and safety?

Resumen

Resumen /Abstract

Resumen en Español

Los medicamentos sintomáticos de acción lenta para la artrosis (SYSADOA) comprenden un grupo heterogéneo de medicamentos con efecto a largo plazo sobre los síntomas de la artrosis. Los más utilizados (condroitina sulfato, glucosamina y diacereína) han sido recomendados por el EULAR y por el ESCEO. Sin embargo, la prescripción de SYSADOA es controvertida debido a la gran cantidad de productos disponibles y a que las variaciones en las técnicas de extracción y purificación pueden dar lugar a diferencias en el contenido, la composición, la pureza, los efectos biológicos, la seguridad. Las diferencias en la regulación de productos de grado farmacéutico y nutracéutico provocan variaciones en calidad y contenido. Aunque las críticas de que sus efectos modificadores de los síntomas son leves o insignificantes, está demostrado que los SYSADOA reducen dolor y rigidez  aumentando la capacidad funcional mientras ejercen un efecto condroprotector en pacientes con artrosis de rodilla y mano. Guías internacionales han propuesto que la condroitina y la glucosamina sean el tratamiento de elección en la artrosis, especialmente en pacientes con comorbilidades que toman múltiples medicamentos. La seguridad y la eficacia deben evaluarse teniendo en cuenta estas diferencias, y los proveedores de atención médica deben hacer todo lo posible para que los pacientes sean conscientes de las diferencias existentes entre los productos. Esta revisión analiza el uso de SYSADOA en el tratamiento de la artrosis, con énfasis en la seguridad y eficacia y cómo estas se ven afectadas por la calidad y el origen de la materia prima.

Resumen en Inglés

Symptomatic slow-acting drugs for osteoarthritis (SYSADOAs) comprise a heterogeneous group of drugs with a long-term effect on the symptoms of osteoarthritis. The most widely used agents—chondroitin sulfate, glucosamine, and diacerein—have been recommended by the European League Against Rheumatism (EULAR) and by the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis, and Musculoskeletal Diseases (ESCEO). However, prescription of SYSADOAs is challenging owing to the large number of agents available and the fact that variations in extraction and purification techniques can lead to differences in content, composition, purity, biological effects, and safety. Moreover, differences in the regulation of pharmaceutical-grade products and nutraceutical-grade products lead to variations in quality and content between the two classes of products. Despite criticisms that their symptom-modifying effects are mild or insignificant, SYSADOAs have been shown to reduce pain and stiffness and increase functional capacity while exerting a chondroprotective effect in patients with knee and hand osteoarthritis. Recent international guidelines have proposed that chondroitin and glucosamine be the treatment of choice in osteoarthritis, especially in patients with comorbidities taking multiple medications, owing to their safety profile. Safety and efficacy should therefore be evaluated when prescribing SYSADOAs taking into account these differences, and health care providers should make every effort to make patients aware of the existing differences between products. This review discusses the use of SYSADOAs in the treatment of osteoarthritis, with emphasis on safety and effectiveness and how these are affected by the quality and origin of the agents.

Resumen en Portugués

Os medicamentos sintomáticos de ação lenta para a osteoartrite (SYSADOAs) constituem um grupo heterogêneo de medicamentos com efeito de longo prazo nos sintomas da osteoartrite. Os agentes mais amplamente usados - sulfato de condroitina, glucosamina e diacereína - foram recomendados pela EULAR e pela ESCEO. Entretanto, a prescrição de SYSADOAs é desafiadora devido ao grande número de agentes disponíveis e ao fato de que variações nas técnicas de extração e purificação podem levar a diferenças no conteúdo, composição, pureza, efeitos biológicos e segurança. As diferenças na regulamentação de produtos de grau farmacêutico e produtos de grau nutracêutico levam a variações na qualidade e no conteúdo entre as duas classes de produtos. Apesar das críticas de que seus efeitos modificadores dos sintomas são leves ou insignificantes, os SYSADOAs demonstraram reduzir a dor e a rigidez e aumentar a capacidade funcional enquanto exercem um efeito condroprotetor em pacientes com osteoartrite de joelho e mão. Diretrizes internacionais recentes propuseram que a condroitina e a glucosamina sejam o tratamento de escolha na osteoartrite, especialmente em pacientes com comorbidades em uso de vários medicamentos, devido ao seu perfil de segurança. A segurança e eficácia devem, portanto, ser avaliadas ao prescrever SYSADOAs levando em consideração essas diferenças, e os profissionais de saúde devem fazer todos os esforços para alertar os pacientes sobre as diferenças existentes entre os produtos. Esta revisão discute o uso de SYSADOAs no tratamento da osteoartrite, com ênfase na segurança e eficácia e como estes são afetados pela qualidade e origem dos agentes

Introducción

Osteoarthritis, the most common form of arthritis, is a frequent, progressive, degenerative joint disease that leads to functional limitation and diminished quality of life [1]. It is characterized by loss of cartilage and synovial fluid, bone degradation, and inflammation and is a leading cause of chronic pain [2]. The complex and heterogeneous nature of the pathophysiology of osteoarthritis involves a dynamic interaction between biological, biomechanical, and genetic components [2, 3]. Overuse of joints and altered joint mechanics can result in destruction of chondrocytes and disruption of the extracellular matrix, and, eventually, detrimental changes in cartilage function that lead to osteoarthritis [3]. Activation of chondrocytes in response to chemical and mechanical stimuli results in the production of pro-inflammatory cytokines that play an important role in the pathogenesis of osteoarthritis [2].

Today, osteoarthritis has been reported to affect about 300 million people worldwide, and its prevalence is increasing owing to the aging of the population and obesity [4, 5]. As a disabling condition, osteoarthritis impacts daily living. It is associated with different grades of disability, ranging from mild, intermittent pain with only minimal difficulty performing daily activities to severe chronic pain, progressive structural damage, and loss of function, all of which are often associated with a decline in mental health, as well as an increase in mortality when a person is no longer able to walk or live independently. Osteoarthritis generates considerable disability, and the cost of the disease could reach 0.25%-0.50% of gross domestic product. The real burden of the disease may have been underestimated, and it could prove very difficult to calculate the indirect costs of the disease in terms of social support, lost productivity, and wage losses [6].

The different phenotypes and degrees of severity and the chronic nature of the disease mean that various therapeutic options may be applied and combined for the management of osteoarthritis during the disease [7]. First-line treatment is usually nonpharmacologic, such as weight loss, physical therapy, and exercise programs. These approaches can improve osteoarthritis symptoms and slow disease progression and have no adverse effects when properly applied. In parallel, pharmacologic treatment includes opioids, nonsteroidal anti-inflammatory drugs (NSAIDs), and antidepressants [8, 9], which aim to reduce the pain associated with osteoarthritis [10, 11]. However, the adverse effects associated with most of these drugs prevent their long-term use. Besides, because of their potential toxicity, interest in alternative treatments is increasing, and approximately 70% of patients with osteoarthritis are now turning to natural products to alleviate their condition [12].

Natural products to treat osteoarthritis include the symptomatic slow-acting drugs for osteoarthritis (SYSADOAs), which are available as nutritional supplements and oral pharmaceutical-grade products. Two of the more common agents, chondroitin sulfate and glucosamine, are frequently used in the treatment of osteoarthritis. Both agents are biologically active molecules that are substrates for proteoglycan, which is an essential component of the cartilage matrix. According to the criteria of the European Medicines Agency (EMA), chondroitin can be considered a biologically active substance [13, 14]. Also, these products have been combined in pharmaceutical preparations and food supplements and are recommended by the European League Against Rheumatism (EULAR) and by the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) [7, 15]. The use and effectiveness of SYSADOAs are controversial issues, although, a study on the perceived effectiveness and safety of these agents revealed that patients report an improvement in mobility, quality of life, and mood and generally considered these drugs to be effective [16].

The purpose of the present review is to examine the safety and effectiveness of SYSADOAs in the treatment of osteoarthritis. Given that the efficacy of SYSADOAs is controversial, the association between quality and origin of the drugs and efficacy is reviewed.

We performed a review of the literature using the key words oral SYSADOAs, chondroitin sulfate, glucosamine, diacerein, molecular weight, quality, purity, origin, extraction, and quality control. We applied no filters with respect to language or year of publication

Discusión y Conclusión

FINDINGS

SYSADOAs

SYSADOAs are slow-acting agents, meaning that their benefits appear some weeks after initiation of treatment and persist after treatment is withdrawn [17].

Two of the more common agents, chondroitin sulphate and glucosamine, exercise a chondroprotective effect and are considered disease-modifying osteoarthritis drugs (DMOADs). Administered alone or in combination, they have a corrective effect on degeneration of connective tissue by supporting new cellular growth in bone and cartilage, as well as by inhibiting cytokines, metalloproteinase activity, and degradative enzymes [18, 19]. As a consequence, they can delay the progression of knee osteoarthritis [18]. In particular, for chondroitin sulfate, the efficacy in decreasing the pain of osteoarthritis and slowing cartilage destruction is clinically meaningful [2], and its use decreases the need for drugs with more harmful adverse effects, such as NSAIDs.

Chondroitin sulfate

Chondroitin sulfate is a glycosaminoglycan that is found in animal cartilaginous tissue and has been recommended for symptomatic pain relief and for improving joint function, with evidence that it delays the progression of osteoarthritis [20]. It is a high-molecular-weight (50-100 kDa), long-chain polymer that can be extracted from various sources (bovine, porcine, or marine cartilage), with bovine being the most effective type [21, 22]. After extraction, its molecular weight is 10-40 kDa. Chondroitin 4-6 sulfate is available as a pharmaceutical-grade product and as a nutritional supplement. The large number of different agents available can hamper prescription of the most appropriate product. Consequently, extrapolation of efficacy data from pharmaceutical-grade chondroitin sulfate to food supplements may be inaccurate depending on the sources and different degrees of purity. The ESCEO specifically recommends pharmaceutical-grade chondroitin sulfate and glucosamine products, since evidence for these agents is robust [7].

In vitro, chondroitin sulfate has immunomodulatory and anti-inflammatory effects: it reduces NF-kB nuclear translocation and decreases production of pro-inflammatory cytokines such as IL-1b, IFN-g, and TNF-a [23]. Moreover, in vitro results indicate that chondroitin sulfate has a chondroprotective effect in osteoarthritis [24]. In their review on evidence for the benefits of pharmaceutical-grade chondroitin sulfate, Hochberg et al [19] reported that this agent exerted a beneficial effect in vitro on the cell types involved in osteoarthritis. Chondroitin sulfate was also shown to increase type II collagen and proteoglycan synthesis in human articular chondrocytes, reduce production of pro-inflammatory factors and proteases, slow the cellular death process, and improve the anabolic/catabolic balance of the extracellular matrix.

 

Exogenous chondroitin sulfate is absorbed rapidly after oral administration and has been reported to reach peak plasma concentrations after 2.4 hours [25]. As for safety and efficacy, a Cochrane Collaboration systematic review showed that while chondroitin sulfate had small-to-moderate benefits compared with placebo, these were clinically meaningful [26]. Clinical trials confirm that it exerts structure-modifying effects. A meta-analysis published in 2010, reported a small but significant effect of chondroitin sulfate on the reduction in the rate of decline in joint space width of 0.13 mm (95%CI, 0.06-0.19; p=0.0002), which corresponded to an effect size of 0.23 (95%CI, 0.11-0.35; p=0.0001) [27]. Consequently, chondroitin sulfate is classified as a SYSADOA and a DMOAD. The incidence and severity of adverse effects related to chondroitin sulfate at 1200 mg/d are low, and findings in this respect are similar to those reported for placebo [26, 28].

Michel et al [18] reported that long-term treatment could delay radiographic progression in knee osteoarthritis. Furthermore, in patients with symptomatic osteoarthritis of the hand, chondroitin sulfate was demonstrated to improve hand pain and function and have a good safety profile [29]. Its beneficial effects include anti-inflammatory action, increased type II collagen and proteoglycans, reduced bone absorption, and a better anabolic/catabolic balance in chondrocytes [21].  

In their literature review, Honvo et al [30] report that chondroitin had a beneficial effect on pain, symptoms, function, and radiological progression and an excellent safety profile. The CONCEPT trial compared chondroitin sulfate with celecoxib and found that 800 mg/d was superior to placebo and similar to celecoxib for reducing pain and improving function over 6 months in symptomatic knee osteoarthritis. The authors recommend that chondroitin sulfate be considered first-line treatment in the medical management of this condition. In addition, bovine-derived chondroitin sulfate has been shown to suppress osteoclast activity and, therefore, bone resorption at concentrations as low as 1 µg/mL [31]. In fact, in one placebo-controlled trial, chondroitin sulfate was shown to lead to a slight but not significant decrease in pain intensity during activity between weeks 24 and 32 and to be slightly more effective than placebo with respect to quality of life [32]. However, the chondroitin sulfate preparation studied was of avian origin.

Glucosamine

Glucosamine is a monosaccharide that is naturally produced in the human body and can be extracted from crustacean shells. It acts as a substrate for the biosynthesis of glycosaminoglycan chains and the production of aggrecan, which gives cartilage hydrophilicity, thus making it beneficial in osteoarthritis [33]. Glucosamine hydrochloride is a simple molecule that is obtained by extraction and is used as a nutraceutical. Glucosamine sulfate, on the other hand, is obtained semi synthetically. It is found only as a pharmaceutical-grade product (prescription crystalline glucosamine sulfate) [7] and has a stronger inhibitory effect than glucosamine hydrochloride on the cellular processes involved in the physiopathology of osteoarthritis [34].

Glucosamine sulfate inhibits the IL-1 intracellular signaling cascade and gene expression. In vitro, it has been shown to reduce levels of prostaglandin E2 production and interfere with NF-kB DNA binding in chondrocytes and synovial cells [35, 36]. It has also been suggested that enzymatic breakdown of the extracellular matrix might be reduced with glucosamine. The chondroprotective properties of glucosamine in vivo may be based on the inhibition of catabolic activity and cartilage degradation, as opposed to an ability to rebuild cartilage [37].

The effect of glucosamine sulfate on pain in patients with knee osteoarthritis has been shown to be greater than the effect of paracetamol and similar to that of NSAIDs 7 [7]. In addition, glucosamine sulfate has been shown to have disease-modifying effects [38], to reduce the need for concomitant medication, and to delay the need for total joint replacement surgery [39, 40]. The ESCEO strongly recommends prescription crystalline glucosamine sulfate as Step 1 long-term background therapy in knee osteoarthritis. Herrero-Beaumont et al [41] reported a significant improvement in the Lequesne algofunctional index, as well as in the Osteoarthritis Research Society International (OARSI) responder indices with glucosamine sulfate compared with placebo. The symptomatic benefit of glucosamine has been controversial [34], although a Cochrane review suggested that conflicting trial results could be due to the different formulations of glucosamine studied [42].

Combined chondroitin sulfate and glucosamine

Dietary supplements combining chondroitin sulphate and glucosamine are increasingly popular [43, 44]. However, the paucity of formulations containing both products in their prescription grade makes it difficult to recommend them [7].

In the GAIT trial, Clegg et al [45] reported that combination therapy was effective in a subgroup of patients with moderate-to-severe knee pain. The authors observed that the rate of response was significantly higher with combined therapy than with placebo (79.2% vs 54.3%, p=0.002). The results of another randomized controlled trial [46] indicate that the combination of both agents proved to be as effective as celecoxib in patients with painful knee osteoarthritis. Combination therapy has also been shown to have significant effects on pain relief and function compared with placebo [47].

In a study performed in Australia, Fransen et al [48] analyzed whether glucosamine sulfate, chondroitin sulfate, or the combination of both nutraceutical grade supplements limited or reduced structural disease progression (cartilage loss) or provided pain relief to people with chronic knee pain due to osteoarthritis. While the combination resulted in a statistically significant reduction in joint space narrowing at 2 years, no significant symptomatic benefits were demonstrated compared with placebo. Nevertheless, the authors believe that future research should examine the combination over longer periods.

Diacerein

Diacerein, also known as diacetylrhein, is an anthraquinone derivative, whose active metabolite is rhein. It displays anti-inflammatory, anticatabolic, and pro-anabolic properties in cartilage and synovium [49]. Diacerein acts by inhibiting the IL-1b signaling pathway and related downstream metalloproteases [50]. Data from animal models have shown that IL-1b plays a key role in cartilage degradation, subchondral bone remodeling, chondrocyte apoptosis, and joint inflammation [51]. In animal models, diacerein has been very effective in the prevention of cartilage destruction and may have disease-modifying properties in individuals with hip and knee osteoarthritis [52]. It also has a protective effect against subchondral bone remodeling [53]. In a rabbit model of surgically induced knee osteoarthritis, diacerein elicited an anti-inflammatory effect on the synovial membrane and modified the orientation of the subchondral trabecular lattice [54].

Evidence from clinical trials indicates that diacerein could provide effective symptomatic relief in patients with osteoarthritis. Pelletier et al [55] found diacerein to be superior to placebo at 100 mg/day with respect to pain on movement. Similarly, data from the ECHODIAH trial [52] showed that diacerein was superior to placebo in terms of its structure-modifying effect and radiographic progression in patients with hip osteoarthritis. The OARSI recommendations report that diacerein was more efficacious for pain reduction than paracetamol in patients with osteoarthritis [56]. As for NSAIDs, while their onset of action is more rapid than that of diacerein, their efficacy with respect to joint function and pain was comparable to that of diacerein after 1 month of treatment [49].

The incidence of adverse events with diacerein was similar to that of piroxicam; however, the percentage of patients with adverse events in the piroxicam group was higher for dyspepsia (32.9% vs 22.1%) and edema (9.4% vs 4.7%) [57]. Diacerein has been associated with a higher risk of diarrhea, especially in long-term therapy, although this is generally considered to be mild to moderate. It has also been shown to induce rash, pruritus, eczema, and a mild/moderate increase in liver enzymes. Few severe events have been recorded [49].

All in all, compared with NSAIDs, diacerein seems to have similar efficacy and an acceptable safety profile in patients with knee osteoarthritis. The ESCEO considers diacerein to be a first-line pharmacological background treatment in this condition [49]. It could prove particularly interesting in patients with known upper gastrointestinal problems or heart disease, for whom NSAIDs are contraindicated [58].

Quality

The biological activity of nutritional supplements and pharmaceutical-grade products can vary considerably owing to current regulatory restrictions [2, 59].

Quality and consistency issues have arisen with chondroitin sulfate as a food supplement, and these have important implications for efficacy and safety. Wide variations have been reported between the labeled amount of chondroitin sulfate and the amount actually present in the product [60, 61], possibly as a result of factors such as source material, manufacturing processes, and contaminants. In their quantitative and qualitative evaluation of chondroitin sulfate in dietary supplements based on a very high pure European Pharmacopeia chondroitin sulfate reference standard, Volpi and Maccari [60] found that the content of chondroitin sulfate in finished products evaluated using 2 validated methods (agarose gel electrophoresis and SAX-HPLC) was almost 100% and that it met the label claim, although the molecular weight and the disaccharide content varied by about 30%. Adebowale et al [43] found that only 5 of 32 tested supplements contained ±10% of the labeled amount, and that 17 of 32 contained less than 40% of the label claim.

Data from well-designed studies on pharmaceutical-grade chondroitin sulfate should not be extrapolated to food supplements, and vice versa [2]. When the drug is supplied as a food supplement or nutraceutical, its quality may not match that of the pharmaceutical-grade product due to the absence of controls. In vitro studies analyzing the composition of chondroitin sulfate using various techniques revealed variations in molecular weight depending on the origin of the product [62, 63] and in disaccharide content [64, 65]. Variations in origin, production, and purification processes can lead to differences in biological effects [63]. Cantley et al [31] showed that the effects of bovine-derived chondroitin sulfate were more consistent than those of fish- and pig-derived products.

The purification process involves a certain degree of degradation, which reduces the molecular weight of the product. While the objective of the purification protocol is to minimize contaminants (eg, other glycosaminoglycans, proteins, small organic molecules, viruses, prions, and solvents) [21], the extraction and purification conditions can have a critical effect on molecular mass, which is associated with pharmacological activity [60]. Therefore, accurate and practical analytical methods are necessary to ensure quality control of these products.

Relationship between quality and efficacy/safety

Preparations of glucosamine and chondroitin sulfate could vary considerably, with the result that use of incorrect formulations could result in suboptimal outcomes, in turn leading to poor adherence and dissatisfaction with treatment. [66] According to the ESCEO recommendations, clinical benefit, adherence, and satisfaction can only be ensured through a judicious choice of formulation. The patented prescription-grade crystalline glucosamine sulfate is formulated in a stabilized delivery system, which maximizes bioavailability in humans and has been shown to reverse the pro-inflammatory and degenerative effects on cartilage. In their review of the literature, Vlad et a [67]l assessed studies on glucosamine to investigate potential sources of heterogeneity and found that the results reported for the efficacy of glucosamine hydrochloride in knee osteoarthritis are markedly heterogeneous, thus making it difficult to draw definitive conclusions, probably due, in part, to differences in the formulations. The importance of the formulation was highlighted in an analysis of trials based on the patented formulation. The authors examined randomized clinical trials evaluating the effectiveness and safety of glucosamine in osteoarthritis based on various indexes (WOMAC, Lequesne index), which showed that a commercial preparation of glucosamine was superior to placebo with respect to pain and function [42].

DISCUSSION

SYSADOAs encompass a heterogeneous group of drugs with a long-term effect on the symptoms of osteoarthritis. Their use in clinical practice is a controversial issue, often depending on the specific type of patient and associated comorbidities, on the severity of knee and hand osteoarthritis, and on the characteristics of the product used. The case of nutraceuticals (food supplements) is even more controversial, given loose research requirements and regulation in production standards. The variations in the extraction and purification techniques can lead to differences in content, composition, purity, biological effects, and safety [21]. Consequently, many preparations are of poor quality, contain amounts that differ from those shown on the label, and do not provide information on the structural characteristics or mass [68]. However, in the absence of data from clinical trials, we cannot state that nutraceuticals are effective or not.

One of the main criticisms of SYSADOAs is that their symptom-modifying effects are mild or insignificant. Guidelines do see a clear role for SYSADOAs in the management of knee osteoarthritis, and partially in hand osteoarthritis. In their updated stepwise treatment algorithm, the ESCEO considers long-term administration of SYSADOAs, specifically pharmaceutical-grade glucosamine sulfate and/or chondroitin sulfate, as suitable therapy for symptomatic disease, stating that the evidence for this approach is “unequivocal” [7]. However, long-term treatment with chondroitin sulfate has been reported to exert a disease-modifying effect by delaying radiographic progression in patients with knee osteoarthritis [18, 68], and patients perceive these drugs to be effective [16].

In their 2019 update on the role of pharmaceutical-grade chondroitin sulfate for symptomatic management of knee osteoarthritis, Honvo et al [30] report that chondroitin sulfate consistently demonstrated a beneficial effect on pain and function with an excellent safety profile and cost-effectiveness. When evaluating safety and efficacy, it is important to ensure that data are interpreted taking into account differences between pharmaceutical-grade products and nutraceutical-grade products and that the source and purity of the preparation under investigation are clearly stated [2]. Given the different physicochemical characteristics and the lack of clinical studies, the risk-benefit of nutraceutical-grade preparations cannot be extrapolated from pharmaceutical-grade products [14]. However, despite differences observed in vitro, a pharmaceutical-grade SYSADOA has never been shown to be better than a nutraceutical, simply because no comparative studies have been performed in humans.

In order to shed some light on the controversy surrounding SYSADOAs, further studies are required to reveal subgroups of patients who are more likely to benefit from this approach, as well as other functions of chondroitin sulfate. Furthermore, results from studies investigating the structure-modifying properties of these agents could lead to the design of better, more effective products [68]. Much of the controversy surrounding SYSADOAs arises as a result of differences in quality, purity, and efficacy. It would be interesting to perform clinical trials that specifically compare pharmaceutical-grade products with nutraceuticals based on an assessment of these factors. No randomized double-blind studies have shown that the combination of pharmaceutical-grade chondroitin sulfate and glucosamine can be substituted by other formulations found as nutraceuticals.

Indeed, differences in quality between nutraceuticals can be observed not only between products, but also between batches of the same product [69], and the lack of clear regulatory definitions on content makes it very difficult to guarantee quality. The quality and purity of pharmaceutical-grade formulations of SYSADOAs are generally high, whereas those of nutraceuticals are often controversial or demonstrably poor. Consequently, closer regulation of manufacturers is necessary to ensure that only high-quality nutraceuticals are manufactured. In addition, purity should be guaranteed with the use of specific and accurate analytical procedures [61].

Conflicto de intereses

Ingrid Möller is a member of the editorial board of the journal.

Financiación

The authors did not receive any funding for the volunteer research work
which produced the submitted manuscript.

Referencias

  1. Hellio Le Graverand-Gastineau MP. OA clinical trials: current targets and trials for OA. Choosing molecular targets: what have we learned and where we are headed? Osteoarthritis Cartilage. 2009;17(11):1393-401. doi: 10.1016/j.joca.2009.04.009. Epub 2009 Apr 22.

 

  1. Chevalier X, Conrozier T. Access to highly purified chondroitin sulfate for appropriate treatment of osteoarthritis: a review. Med Access. 2017;1(1):e134-e144. Doi: 10.5301/maapoc.000002.

 

  1. Glyn-Jones S, Palmer AJ, Agricola R, Price AG, Vincent TL, Weinans H, et al. Osteoarthritis. Lancet. 2015;386(9991):376-87.

 

  1. Lawrence RC, Felson DT, Helmick CG, Arnold LM, Choi H, Deyo RA, et al. Estimates of the prevalence of arthritis and other rheumatic diseases in the United States. Part II. Arthritis Rheum. 2008; 58(1):26-35. doi: 10.1002/art.23176

 

  1. OARSI. Osteoarthritis: A Serious Disease, Submitted to the U.S. Food and Drug Administration, December 1, 2016. Accessed at:

https://www.oarsi.org/sites/default/files/docs/2016/oarsi_white_paper_oa_serious_disease_121416_1.pdf

 

  1. Cross M, Smith E, Hoy D, Nolte S, Ackerman I, Fransen M, et al. The global burden of hip and knee osteoarthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis. 2014;73(7):1323-30. doi: 10.1136/annrheumdis-2013-204763. Epub 2014 Feb 19.

 

  1. Bruyère O, Honvo G, Veronese N, Arden NK, Branco J, Curtis EM, et al. An updated algorithm recommendation for the management of knee osteoarthritis from the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO). Semin Arthritis Rheum. 2019;49(3):337-350. doi: 0.1016/j.semarthrit.2019.04.008. Epub 2019 Apr 30.

 

  1. Kolasinski SL, Neogi T, Hochberg MC, Oatis C, Guyatt G, Block J, et al. 2019 American College of Rheumatology/Arthritis Foundation Guideline for the Management of Osteoarthritis of the Hand, Hip, and Knee. Arthritis Rheumatol. 2020;72(2):220-33. doi: 10.1002/art.41142. Epub 2020 Jan 6.

 

  1. McAlindon TE, Bannuru RR, Sullivan MC, Arden NK, Berenbaum F, Bierma-Zeinstra SM, et al. OARSI guidelines for the non-surgical management of knee osteoarthritis. Osteoarthritis Cartilage. 2014;22(3):363-88.

 

  1. Machado GC, Maher CG, Ferreira PH, Pinheiro MB, Lin CW, Day Ro, et al. Efficacy and safety of paracetamol for spinal pain and osteoarthritis: systematic review and meta-analysis of randomized placebo controlled trials. BMJ. 2015;350:h1225. doi: 10.1136/bmj.h1225.

 

  1. da Costa BR, Reichenbach S, Keller N, Nartey L, Wandel S, Jüni P, et al. Effectiveness of non-steroidal anti-inflammatory drugs for the treatment of pain in knee and hip osteoarthritis: a network meta-analysis. Lancet. 2017;390(10090):e21-e33. doi: 10.1016/S0140-6736(17)31744-0

 

  1. Basedow M, Runciman WB, March L, Esterman A. Australians with osteoarthritis; the use of and beliefs about complementary and alternative medicines. Complement Ther Clin Pract. 2014;20(4):237-42. doi: 10.1016/j.ctcp.2014.08.002. Epub 2014 Aug 20.

 

  1. EMA (2014) Guideline on similar biological medicinal products containing biotechnology-derived proteins as active substance: non-clinical and clinical issues. EMEA/CHMP/BMWP/42832/05 Rev.1. http://www.ema.europa.eu/docs/en_GB/document_library/ Scientific_guideline/2015/01/WC500180219.pdf

 

  1. Bruyère O, Cooper C, Al-Daghri NM, Dennison EM Rizzoli R, Reginster JY. Inappropriate claims from non-equivalent medications in osteoarthritis: a position paper endorsed by the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO). Aging Clin Exp Res. 2018;30(2):111-7. doi: 10.1007/s40520-017-0861-1. Epub 2017 Nov 24.

 

  1. Zhang W, Doherty M, Arden N, Bannwarth B, Bijlsma J, Gunther KP, et al. EULAR evidence based recommendations for the management of hip osteoarthritis:

Report of a task force of the EULAR Standing Commitee for the International

Clinical Studies including Therapeutics (ESCISIT). Ann Rheum Dis. 2005;64:669-81.

 

  1. Adán C, Molinero A, Castellano L, Peral V, Araño B, de-Juan B, Fernández A, García-Alcalde P, Matute E. Perceived effectiveness and safety of symptomatic slow action drugs for osteoarthritis (sysadoa) for treating osteoarthrosis from community pharmacy. Farmacéuticos Comunitarios. 2016 Dec 30; 8(4):5-17 DOI: 10.5672/FC.2173-9218.(2016/Vol8).004.02 

 

  1. https://www.saludcastillayleon.es/portalmedicamento/es/terapeutica/ojo-markov/sysadoa-controversias

 

  1. Michel BA, Stucki G, Frey D, De Vathaire F, Vignon E, Bruehlmann P, et al. Chondroitins 4 and 6 sulfate in osteoarthritis of the knee: A randomized, controlled trial. Arthritis Rheum. 2005;52:779-86.

 

  1. Hochberg M, Chevalier X, Henrotin Y, Hunter DJ, Uebelhart D. Symptom and structure modification in osteoarthritis with pharmaceutical-grade chondroitin sulfate: what’s the evidence? Curr Med Res Opin. 2013;29(3):259-267. doi: 10.1185/03007995.2012.753430. Epub 2013 Jan 31.

 

  1. Rillo O, Riera H, Acosta C, Liendo V, Bolaños J, Monterola L, et al. PANLAR Consensus Recommendations for the Management of Osteoarthritis of Hand, Hip, and Knee. J Clin Rheumatol. 2016;22(7):345-354. doi: 10.1097/RHU.0000000000000449.

 

  1. Martel-Pelletier J, Farran A, Montell E, Vergés J, Pelletier JP. Discrepancies in composition and biological effects of different formulations of chondroitin sulfate. Molecules. 2015;20:4277-89.

 

  1. Henrotin Y, Mathy M, Sanchez C, Lambert C. Chondroitin sulfate in the treatment of osteoarthritis: From in vitro studies to clinical recommendations. Ther Adv Musculoskelet Dis. 2010;2(6):335-48.

 

  1. du Souich P, García AG, Vergés J, Montell E. Immunomodulatory and anti-inflammatory effects of chondroitin sulphate. J Cell Mol Med. 2009;13(8A):1451-63. doi: 10.1111/j.1582-4934.2009.00826.x. Epub 2009 Jun 11.

 

  1. Volpi N. Anti-inflammatory activity of chondroitin sulphate: new functions from an old natural macromolecule. Inflammopharmacology. 2011;19(6):299-306. doi: 10.1007/s10787-011-0098-0. Epub 2011 Nov 1.

 

 

  1. Volpi N. Oral bioavailability of chondroitin sulphate (Condrosulf) and its constituents in healthy male volunteers. Osteoarthritis Cartilage. 2002;10(10):768-77.

 

  1. Singh JA, Noorbaloochi S, MacDonald R, Maxwell LJ. Chondroitin for osteoarthritis. Cochrane Database Syst Rev. 2015;1:CD005614. doi: 10.1002/14651858.CD005614.pub2

 

  1. Hochberg MC. Structure-modifying effects of chondroitin sulfate in knee osteoarthritis: an updated meta-analysis of randomized placebo-controlled trials of 2-year duration. Osteoarthritis Cartilage. 2010;18 Suppl 1:S28-31.  doi: 10.1016/j.joca.2010.02.016. Epub 2010 Apr 27.

 

  1. Rainsford KD. Importance of pharmaceutical composition and evidence from clinical trials and pharmacological studies in determining effectiveness of chondroitin sulphate and other glycosaminoglycans: a critique. J Pharm Pharmacol. 2009;61(10):1263-1270.

 

  1. Gabay C, Medinger-Sadowski C, Gascon D, Kolo F, Finckh A. Symptomatic effects of choindroitin 4 and chondroitin 6 sulfate on hand osteoarthritis : A randomized, double-blind, placebo-controlled clinical trial at a single center. Arthritis Rheum. 2011;63(11) :3383-91.

 

  1. Honvo G, Bruyère O, Reginster J-Y. Update on the role of pharmaceutical-grade chondroitin sulfate in the symptomatic management of knee arthritis. Aging Clin Exp Res. 2019;31(8):1163-

 

  1. Cantley MD, Rainsford DK, Haynes DR. Comparison of the ability of chondroitin sulfate derived from bovine, fish, and pigs to suppress human osteoclast activity in vitro. Inflammopharmacology. 2013;21:407-12. doi: 10.1007/s10787-013-0171-y. Epub 2013 May 4

 

  1. Mazières B, Hucher M, Zaïm M, Garnero P. Effect of chondroitin sulphate in symptomatic knee osteoarthritis: a multicenter, randomized, double-blind, placebo-controlled study. Ann Rheum Dis. 2007;66(5):639-45.

 

  1. Reginster J-Y, Bruyere O, Neuprez A. Current role of glucosamine in the treatment of osteoarthritis. Rheumatology. 2007;46:731-5. doi:10.1093/rheumatology/kem026

 

  1. Altman RD, Abramson S, Bruyere O, Clegg D, Herrero-Beaumont G, Maheu E, Moskowitz R, Pavelka K, Reginster JY. Commentary: osteoarthritis of the knee and glucosamine. Osteoarthritis Cartilage. 2006;14(10):963-6.

 

  1. Largo R, Alvarez-Soria MA, Diez Ortego J, Calvo E, Sanchez-Pernuate O, Egido J, et al. Glucosamine inhibits IL-1 beta-induced NFkappaB activation in human osteoarthritis chondrocytes. Osteoarthritis Cartilage 2003;11:290-8.

 

  1. Alvarez-Soria MA, Largo R, Calvo E, Egido J, Herrero-Beaumont G. Differential anticatabolic profile of glucosamine sulfate versus other anti-osteoarthritic drugs on human osteoarthritic chondrocytes and synovial fibroblasts in culture. Osteoarthritis Cartilage. 2005;13:S153.

 

  1. Uitterlinden EJ, Jahr H, Koevoet JL, Jenniskens YM, Bierma-Zeinstra, DeGroot J, et al. Glucosamine decreases expression of anabolic and catabolic genes in human osteoarthritic cartilage explants. Osteoarthritis Cartilage. 2006;14(3):250-7.

 

  1. Bruyère O, Altman RD, Reginster JY. Efficacy and safety of glucosamine sulfate in the management of osteoarthritis: Evidence from real-life setting trials and surveys. Semin Arthritis Rheum. 2016;45(4 Suppl):S12-7. doi: 10.1016/j.semarthrit.2015.11.011. Epub 2015 Dec 2.

 

  1. Bruyere O, Pavelka K, Rovati LC, Gatterová J, Giacovelli G, Deroisy R, et al. Total joint replacement after glucosamine sulphate treatment in knee osteoarthritis: results of a mean 8-year observation of patients from two previous 3-year, randomized, placebo-controlled trials. Osteoarthritis Cartilage. 2008;16(2):254-60.

 

  1. Rovati LC, Girolami R, D’Amato M, Giacovelli G. Effects of glucosamine sulfate on the use of rescue non-steroidal anti-inflammatory drugs in knee osteoarthritis: Results from the Pharmaco-Epidemiology of GonArthroSis (PEGASus) study. Semin Arthritis Rheum. 2016;45(4 Suppl):S34-41. doi: 10.1016/j.semarthrit.2015.10.009. Epub  2015 Oct 29.

 

  1. Herrero-Beaumont G, Ivorra JA, Del Carmen Trabado M, Blanco FJ, Benito P, Martín-Mola E, et al. Glucosamine sulfate in the treatment of knee osteoarthritis symptoms: a randomized, double-blind, placebo-controlled study using acetaminophen as a side comparator. Arthritis Rheum. 2007;56(2):555-67.

 

  1. Towheed TE, Maxwell L, Anastassiades TP, Shea B, Houpt J, Robinson V, et al. Glucosamine therapy for treating osteoarthritis. Cochrane Database Syst Rev. 2005;(2):CD002946.

 

  1. Adebowale AO, Cox DS, Liang Z, Eddington ND. Analysis of glucosamine and chondroitin sulfate content in marketed products and the Caco-2 permeability of chondroitin sulfate raw materials. JANA. 2000;3(1):37-44.

 

  1. Santos GR, Piquet AA, Glauser BF, Tovar AM, Pereira MS, Vilanova E, et al. Systematic

analysis of pharmaceutical preparations of chondroitin sulfate combined

with glucosamine. Pharmaceuticals (Basel) 2017;10(2).

 

  1. Clegg DO, Reda DJ, Harris CL, Klein MA, O’Dell JR, Hooper MM, et al. Glucosamine, chondroitin sulfate, and the two in combination for painful knee osteoarthritis. N Engl J Med. 2006;354(8):795-808.

 

  1. Hochberg MC, Martel-Pelletier J, Monfort J, Möller I, Castillo JR, Arden N, et al. Combined chondroitin sulfate and glucosamine for painful knee osteoarthritis: a multicenter, randomized, double-blind, non-inferiority trial versus celecoxib. Ann Rheum Dis. 2016;75(1):37-44. doi: 10.1136/annrheumdis-2014-206792. Epub 2015 Jan 14.

 

  1. Zeng C, Wei J, Li H, Wang YL, Xie DX, Yang T, et al. 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. doi: 10.1038/srep16827.

 

  1. Fransen M, Agalotis M, Nairn L, Votrubec M, Bridgett L, Su S, 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(5):851-8. doi: 10.1136/annrheumdis-2013-203954. Epub 2014 Jan 6.

 

  1. Pavelka K, Bruyère O, Cooper C, Kanis JA, Leeb BF, Maheu E, et al. Diacerein: benefits, risks and place in the management of osteoarthritis. An opinion-based report from the ESCEO. Drugs Aging. 2016;33(2):75-85. doi: 10.1007/s40266-016-0347-4

 

  1. Martel-Pelletier J, Pelletier JP. Effects of diacerein at the molecular level in the osteoarthritis disease process. Ther Adv Musculoskelet Dis. 2010;2(2):95-104. doi: 10.1177/1759720X09359104.
  2. Pavelka K, Trc T, Karpas K, Vitek P, Sedlácková M, Vlasáková V, et al. The efficacy and safety of diacerein in the treatment of painful osteoarthritis of the knee: a randomized, multicenter, double-blind, placebo-controlled study with primary end points at two months after the end of a three-month treatment period. Arthritis Rheum. 2007;56(12):4055-64.

 

  1. Dougados M, Nguyen M, Berdah L, Maziéres B, Vignon E, Lequesne M, et al. Evaluation of the structure-modifying effects of diacerein in hip osteoarthritis: ECHODIAH, a three-year, placebo-controlled trial. Evaluation of the Chondromodulating Effect of Diacerein in OA of the hip. Arthritis Rheum. 2001;44(11):2539-2547.

 

  1. Pelletier JP, Lajeunesse D, Reboul P, Mineau F, Fernandes JC, Sabouret P, et al. Diacerein reduces the excess synthesis of bone remodeling factors by human osteoblast cells from osteoarthritic subchondral bone. J Rheumatol. 2001;28(4):814-24.

 

  1. Permuy M, Guede D, López-Peña M, Muñoz F, Caeiro J-R, González-Cantalapiedra A. Comparison of various SYSADOA for the osteoarthritis treatment: an experimental study in rabbits. BMC Musculoskeletal Disord. 2015;16:120.

 

  1. Pelletier JP, Yaron M, Haraoui B, Cohen P, Nahir MA, Choquette D, et al. Efficacy and safety of diacerein in osteoarthritis of the knee: a double-blind, placebo-controlled trial. The Diacerein Study Group. Arthritis Rheum. 2000;43(10):2339-48.

 

  1. Zhang W, Nuki G, Moskowitz RW, Abramson S, Altman RD, Arden NK, et al. OARSI recommendations for the management of hip and knee osteoarthritis: part III: changes in evidence following systematic cumulative update of research published through January 2009. Osteoarthritis Cartilage. 2010;18(4):476-99. doi: 10.1016/j.joca.2010.01.013. Epub 2010 Feb 11.

 

  1. Louthrenoo W, Nilganuwong S, Aksaranugraha S, Asavatanabodee P, Saengnipanthkul S, Thai Study Group. The efficacy and safety of diacerein in the treatment of painful knee osteoarthritis: a randomized, double-blind, NSAID-controlled study. Osteoarthritis Cartilage. 2007;15(6):4055-64.

 

  1. Panova E, Jones G. Benefit–risk assessment of diacerein in the treatment of osteoarthritis. Drug Saf. 2015;38(3):245-52. doi: 10.1007/s40264-015-0266-z.

 

  1. Restaino OF, Finamore R, Stellavato A, Diana P, Bedini E, Trifuoggi M, et al. European chondroitin sulfate and glucosamine food supplements: A systematic quality and quantity assessment compared to pharmaceuticals. Carbohydr Polym. 2019;222:114984. doi: 10.1016/j.carbpol.2019.114984. Epub 2019 Jun 20.

 

  1. Volpi N, Maccari F. Quantitative and qualitative evaluation of chondroitin sulfate in dietary supplements. Food Anal Methods. 2008;1:195-204. DOI 10.1007/s12161-008-9020-9

 

  1. Volpi N, Maccari F. Two analytical approaches to the evaluation of chondroitin sulfate in European food supplements. Sep Sci. 2009;1(1):23-8.

 

  1. Volpi N. Analytical aspects of pharmaceutical grade chondroitin sulfates. J Pharm Sci. 2007;96:3168-80.

 

  1. Tat SK, Pelletier JP, Mineau F, Duval N, Martel-Pelletier J. Variable effects of 3 different chondroitin sulfate compounds on human osteoarthritic cartilage/chondrocytes: Relevance of purity and production process. J Rheumatol. 2010;37:656-64. doi: 10.3899/jrheum.090696. Epub 2010 Jan 28.

 

  1. Sakai S, Otake E, Toida T, Goda Y. Identification of the origin of chondroitin sulfate in “health foods”. Chem Pharm Bull (Tokyo). 2007;55(2):299-303.

 

  1. Malavaki CG, Asimakopoulou AP, Lamari FN, Theocharis AD, Tzanakakis GN, Karamanos NK. Capillary electrophoresis for the quality control of chondroitin sulfates in raw materials and formulations. Anal Biochem. 2008;374(1):213-20.

 

  1. Bruyère O. Pharmaceutical-grade chondroitin sulfate in the management of knee osteoarthritis. Expert Opin Pharmacother. 2018;19(4):409-12. doi: 10.1080/14656566.2018.1442438.

 

  1. Vlad SC, LaValley M, McAlindon TE, Felson DT. Glucosamine for pain in osteoarthritis: why do trial results differ? Arthritis Rheum. 2007;56(7):2267-77.
  2. Lamari FN. The potential of chondroitin sulfate as a therapeutic agent. Connect Tissue Res. 2008;49(3):289-92. doi: 10.1080/03008200802148314.

 

  1. Lockwood GB. The quality of commercially available nutraceutical supplements and food sources. J Pharm Pharmacol 2011;63(1):3-10. doi: 10.1111/j.2042-7158.2010.01159.x. Epub 2010 Oct 11.
enviar Envía un artículo