Next Article in Journal
Molecular and Pathophysiological Links between Metabolic Disorders and Inflammatory Bowel Diseases
Next Article in Special Issue
Detection and Evaluation of Serological Biomarkers to Predict Osteoarthritis in Anterior Cruciate Ligament Transection Combined Medial Meniscectomy Rat Model
Previous Article in Journal
Role of the Myokine Irisin on Bone Homeostasis: Review of the Current Evidence
Previous Article in Special Issue
Selection of Highly Proliferative and Multipotent Meniscus Progenitors through Differential Adhesion to Fibronectin: A Novel Approach in Meniscus Tissue Engineering
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug Delivery Systems for the Treatment of Knee Osteoarthritis: A Systematic Review of In Vivo Studies

by
Francesco Manlio Gambaro
1,2,*,
Aldo Ummarino
1,2,
Fernando Torres Andón
1,3,
Flavio Ronzoni
1,4,
Berardo Di Matteo
2,5 and
Elizaveta Kon
1,2
1
Department of Biomedical Sciences, Humanitas University Pieve Emanuele, 20090 Milan, Italy
2
IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
3
Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
4
Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
5
Department of Traumatology, Orthopaedics and Disaster Surgery, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(17), 9137; https://doi.org/10.3390/ijms22179137
Submission received: 1 August 2021 / Revised: 21 August 2021 / Accepted: 22 August 2021 / Published: 24 August 2021

Abstract

:
Many efforts have been made in the field of nanotechnology to improve the local and sustained release of drugs, which may be helpful to overcome the present limitations in the treatment of knee OA. Nano-/microparticles and/or hydrogels can be now engineered to improve the administration and intra-articular delivery of specific drugs, targeting molecular pathways and pathogenic mechanisms involved in OA progression and remission. In order to summarize the current state of this field, a systematic review of the literature was performed and 45 relevant studies were identified involving both animal models and humans. We found that polymeric nanoparticles loaded with anti-inflammatory drugs (i.e., dexamethasone or celecoxib) are the most frequently investigated drug delivery systems, followed by microparticles and hydrogels. In particular, the nanosystem most frequently used in preclinical research consists of PLGA-nanoparticles loaded with corticosteroids and non-steroidal anti-inflammatory drugs. Overall, improvement in histological features, reduction in joint inflammation, and improvement in clinical scores in patients were observed. The last advances in the field of nanotechnology could offer new opportunities to treat patients affected by knee OA, including those with previous meniscectomy. New smart drug delivery approaches, based on nanoparticles, microparticles, and hydrogels, may enhance the therapeutic potential of intra-articular agents by increasing the permanence of selected drugs inside the joint and better targeting specific receptors and tissues.

1. Introduction

Knee osteoarthritis (OA) affects around 250 million people worldwide, making it one of the leading causes of disability [1]. However, this estimated prevalence is likely going to grow as a consequence of the increased incidence of obesity and population ageing. Our understanding of its etiology has grown with time; the initial model of osteoarthritis as a simple cartilage degenerative disease has been overcome by a more complex multifactorial model encompassing an interplay among mechanical, metabolic, and inflammatory factors [2]. The complexity of the etiology is mirrored by the indications found in the treatment guidelines of the most important orthopedic societies as the Osteoarthritis Research Society International (OARSI) [3], the American College of Rheumatology (ACR) [4], and the American Academy of Orthopedic Surgeons (AAOS) [5]. They recommend addressing knee OA at multiple levels moving from nonpharmacological approaches such as weight control and adequate sport activity to oral or topical pharmacological treatments, intra-articular (IA) corticosteroid injections, and to novel promising cellular therapies such as stem cells [6]. Nevertheless, due to the multifactorial etiology and progressive nature of this condition, all the above mentioned approaches aim at controlling symptoms until the severity of the disease mandates surgical intervention with joint replacement. A recent study investigating the satisfaction of patients with the current OA treatment algorithm found that 78% of the patients had uncontrolled chronic knee pain [7]. Reasons behind the failure of current approaches may stem from both the lack of patients’ compliance and the frequent side effects related to the use of systemic drugs. Therefore, intra-articular injections seem to be more promising due to their preferential local effect. After intra-articular injections, the amount of substance inside the joint may rapidly decrease (<1% of the initial concentration) already 48 h after the injection, as observed in a rodent model by Allen et al., who employed elastin-like polypeptides [8]. Hence, new efforts have been made to develop new drug delivery approaches to overcome the present limitations in the treatment of knee OA and to allow a sustained release of the drug inside the joint.
To achieve this goal, several strategies have been investigated; in particular, nanoparticles, microparticles, or hydrogels have been used as drug delivery systems [9]. A schematic representation of these three main categories is presented in Figure 1. According to the International Organization for Standardization (ISO) [10], nanoparticles (NPs) are materials with an external dimension or an internal structure in the nanoscale (ranging from 1 nm to 1000 nm), therefore representing the smallest particles in nature after chemicals (atoms and small molecules) [11]. To date, many different materials have been exploited to manufacture NPs [12]; liposome-based NPs are so far those that have gained the most success in the medical world. Indeed, up until 2016, 18 NP drugs were approved by FDA, 12 of which were liposome-based, and 95% of them were indicated for cancer treatment [13]. In addition, in the last year, the use of lipid nanosystems for the in vivo delivery of mRNA in COVID-19 vaccines marked a breakthrough, which may be applied to other diseases in the near future [14,15]. However, if we consider those nanosystems as candidates for the treatment of OA, polymer-based NPs outstand liposomes in the number of publications. Moreover, in order to overcome the limitations of natural polymers (rapid degradation or limited mechanical properties), synthetic polymers have been developed such as polyethylene glycol (PEG) and poly-lactic-glycolic acid (PLGA) [16]. In addition to NPs, microparticles (MPs), with a size range between 1 and 1000 μm, have been investigated as drug delivery systems, especially those presenting a size similar to erythrocytes (7.5 μm) [17]. Drug delivery systems in the microscale display a more prolonged drug release versus other systems with different sizes [9]. Indeed, a study observed that drug bioactivity was recorded even after 2 months after the IA injection of MP in a mouse knee [18]. The beneficial properties of these MPs led to their investigation in a phase III clinical trial [19]. Finally, another type of nanostructure is represented by natural or synthetic hydrogels that are composed by a three-dimensional network of cross-linked polymer chains. These hydrogels can be either directly loaded with the desired drug or even with microspheres and/or nanospheres as drug carriers [20] (Figure 1). The aim of the present systematic review is to provide the readers with a comprehensive summary on the aforementioned “biotechnological strategies” explored so far to deliver drugs within the osteoarthritic knee.

2. Methods

The present systematic review was written according to the Preferred Reporting items for Systematic Reviews and Meta-analysis (PRISMA) guidelines [21], whose flowchart is depicted in Figure 2. A comprehensive search of PubMed, Google Scholar, Cochrane, and EMBASE was performed using various combinations of the following keywords: “Knee osteoarthritis,” “Drug delivery,” “Microparticles,” “Nanoparticles,” “Hydrogels.” The last research was performed on 14 June 2021 and two independent reviewers conducted the research independently (F.M.G. and A.U.). All 360 articles obtained by the literature search were screened according to the following inclusion criteria: (i) employment of a drug delivery system for the treatment of knee osteoarthritis; (ii) in vivo study; and (iii) knee intra-articular (IA) route of administration. Exclusion criteria were: (i) in vitro studies or review articles; (ii) other delivery methods than intra-articular application (e.g., transdermal application), (iii) not focused on osteoarthritis; (iv) not written in the English language.

3. Results

3.1. Drug Delivery Systems

We included 45 studies in total (Figure 2): 19 (42%) dealt with nanoparticles to improve the delivery of the pharmacological agent whereas 16 studies (36%) used microparticles and 10 studies (22%) employed hydrogels. The distribution of studies according to the year of publication is provided in Figure 3.

3.1.1. Nanoparticles

A summary of the main features of all the studies employing NPs is reported in Table 1. Among the 19 included studies using NPs, 68% exploited polymers [22,23,24,25,26,27,28,29,30,31,32,33,34], 16% carbon-based NPs [35,36,37], 5% liposomes [38], 5% metal-based NPs [39], and 5% employed zeolitic imidazolate framework-8 (ZIF-8) [33]. In the polymeric NPs group, 56% of the studies used the Poly Lactic-co-Glycolic Acid (PLGA), making the latter the most employed material. Among these studies, four out of nine studies did not use a pure form of PLGA-based NPs but a combination of PLGA with Polyethylene glycol (PEG). In addition to PLGA, materials such as Poly N-isopropylacrylamide (pNIPAM), Polyester amide (PEA), Poloxamer, Hyaluronic Acid, and/or Chitosan have been employed.
PLGA is a synthetic polymer composed by a combination of two monomers: polylactic acid (PLA) and polyglycolic acid (PGA). PLA has a slow degradation rate while PGA presents a fast degradation rate. Thus, taking advantage of the different properties of these two molecules and by choosing the ratio of PLA and PGA, the degradation profile of PLGA can be designed for particular purposes, e.g., a slow and continuous release of drugs inside the knee [40]. Interestingly, polycaprolactone (PCL) is a very tough polyester, allowing a very slow degradation rate (up to 2–4 years) compared to the previous polymers. This feature obviously gains particular interest in the treatment of chronic conditions such as knee OA [41]. On the other hand, polyethylene glycol (PEG), with a high cross-linking capacity compared to other polymers, can be engineered with different sizes allowing the modification of NP-hydrophobicity, permeability, elasticity, and also degradation rate [41]. PEGylation, a process of linking PEG to other polymers or drugs, has been widely used as a “mask” strategy to avoid interaction with proteins or cells, reducing the immunogenicity and increasing the half-life after intravenous administration [42].
Among natural polymers, Hyaluronic Acid (HA) and Chitosan (CS) have been broadly used for biomedical research. HA is an anionic non-sulfated glycosaminoglycan highly present in our body (i.e., connective, epithelial, and neural tissues) and is a major component of the extracellular matrix, able to affect cell proliferation and migration, thus having important implications in tissue healing and modulation of inflammation and angiogenesis. High molecular weight HA has been FDA-approved to treat osteoarthritis of the knee via intra-articular injection, while HA with different sizes has been used to engineer drug delivery strategies, such as nanoparticles or hydrogels. Instead, Chitosan is a natural polysaccharide derived from chitin with the advantage of having free amine groups available for chemical modification, thus offering the chance to enhance specific functional properties. In addition, it has a similar structure to human glycosaminoglycans, therefore providing a possible microenvironment for chondrocyte proliferation and extracellular matrix synthesis [43].
Moving to carbon-based NPs, three studies employed these particular constructs [35,37,44]; in particular one study used fullerene-based NPs, while the other two studies employed graphene oxide or carbon nanotubules. Among the least represented groups, we found liposome-based NPs, employing a combination of lipids and HA [38] and metal-based NPs employing gold [39]. Finally, one study dealt with NPs composed by metal organic frameworks (MOFs) made of Zinc and imidazolate [33]. Indeed, MOFs present specific features that make them suitable for drug delivery since they have high porosity, high mechanical stability, and also a pH-responsive decomposition [45].
Summarizing the observed in vivo effects of NPs, six studies described improved histological structure after treatment [23,25,26,35,38,39] and three studies showed an increase in the OARSI score compared to control [22,26,33]. Concerning the level of inflammatory mediators, a decrease in IL-1β [26,33], TNF-α [25,26], MMP-3, and MMP-13 [33,36] expression levels was documented in most studies. Moreover, an increase in collagen type II levels was reported in three studies [23,33,36]. The above-mentioned results were observed at an average follow up time of 5 weeks (range 1–12 weeks).
Concerning the animal model on which the effects of the drug-loaded NPs were analyzed, rats were employed in the majority of the studies (81%), followed by rabbits (10%), bovines (6%), and sheep (3%).

3.1.2. Microparticles

All 16 studies employing microparticles (MPs) to deliver pharmacological compounds used polymer-based MPs. In particular, 12 papers (80%) used PLGA [46,47,48,49,50,51,52,53,54,55,56,57], making it the most popular material to assemble MPs. Instead of employing a pure form of PLGA, two studies used PLGA combined with either PVA or PEA. The remaining three studies used poly-ε-caprolactone (PCL), Chitosan, or Heparin-based MPs.
Summarizing the in vivo effects of MPs, three studies reported an improvement in histological structure [53,58,59], while two studies on humans reported a significant reduction in average daily pain (ADP) [48,49]. Concerning the level of inflammatory mediators, only one study [60] observed a decrease in IL-1β, IL-6, IL-17, TNF-α, ADAMTS-5, and MMP3 expression levels.
The above-mentioned results were observed at an average follow up time of 6.8 weeks (range 2–12 weeks).
Concerning the species, four studies were performed on humans (27%) and the remaining on animals, in particular rats (60%), dogs (12%), and rabbits (12%). A summary of the features of all the studies employing MPs can be found in Table 2.

3.1.3. Hydrogels

Among the selected articles, only 11 studies employed hydrogels as the drug delivery system for the treatment of OA; 60% of them were published in the year 2020. The most commonly adopted material to assemble the hydrogel was polylactic acid (PLA).
Indeed, PLA is prepared starting from L-lactide monomers that are subject to ring-opening polymerization to form L-lactic acid oligomers (LAo). This polymer is then mixed with a gelatin solution and is ready to be loaded with drugs, as described by three studies [61,62,63]. Instead, one study employed a HA-based gel [64] and another one a combination of HA and dextran [65].
One study employed a poloxamer-based hydrogel, a copolymer composed by polypropylene glycol (PPG) and PEG [66]. The remaining five studies used respectively α-chondroitin sulfate-ethyl glycol (EG), PEG, agarose, Chitosan–glycerin–borax, and poloxamer [66,67,68,69,70].
Summarizing the in vivo effects of hydrogels, two studies reported a more favorable histological structure [64,71], and five studies showed an improvement in the OARSI score compared to control [61,62,68,69,70]. Concerning the level of inflammatory mediators, six studies [61,62,63,66,70,71] observed a decrease in IL-1β, IL-6, IL-17, TNF-α, ADAMTS-5, and MMP3 expression levels. The abovementioned results were observed in an average follow up time of 8.4 weeks (range 2–24 weeks).
Concerning the animal model, five studies employed rats, four rabbits, and only one study a canine model. A summary of the features of all the studies employing hydrogels is reported in Table 3.

3.2. Types of Pharmacological Agents Delivered

Among the 45 included studies, the most frequently loaded drug category was represented by corticosteroids (16 studies); indeed, eight studies employed triamcinolone acetonide, five dexamethasone, and three betamethasone. The second most employed drug category was represented by NSAIDs (nine studies); in particular, three studies employed lornoxicam, two indomethacin, one ibuprofen, one naproxen, one diclofenac, and one piroxicam. Among the other molecules presenting a measurable pharmacological effect, hyaluronic acid, COX-2 specific inhibitors (coxib), chondroitin sulfate, clodronate, and KAFAK have been studied. Finally, the following drugs were employed only by a single study: methotrexate, adenosine, curcumin, oleic acid, NO-Hemoglobin Notch-1 siRNA, methyl prednisolone, antisense oligomers, eicosatetraenoic acid, rapamycin, and quercetin. Concerning the effects observed after treatment according to the drug used, the coxib group showed the most consistent improvement; all the studies employing either etoricoxib [29] or celecoxib [31,38] reported a better histological outcome after treatment. Instead, focusing on the IA concentration change in cytokines, a decrease in IL-1β, IL-6, TNF- α, MMP-3 was observed with all the main drug categories, i.e., with glucocorticoids [70], NSAIDs [60], and coxibs [29]. All the included studies on humans [48,49,51,54], employed triamcinolone acetonide. A graphic representation of the distribution of loaded drugs based on the type of nanomaterials is provided in Figure 4.

4. Discussion

Knowledge of the most commonly utilized drug delivery systems, such as nanoparticles, microparticles, or hydrogels, is crucial to understanding the direction of new therapeutic advances in the management of knee osteoarthritis. The present systematic review collected all the studies documenting the application of nano and microstructures as drug carriers in both animals and humans affected by knee OA. The main finding is that nanoparticles are the most employed drug delivery system, followed by microparticles and hydrogels. Moreover, the most frequently used materials to create these drug delivery systems are polymers and in particular PLGA. The most commonly loaded drug types are corticosteroids, followed by NSAIDs. Among the 45 studies included, only four [48,49,51,54] were performed in humans and all of these employed microparticles made of PLGA loaded with triamcinolone acetonide. These four trials were all published in the last three years, underlining that the application of novel intra-articular drug delivery system is facing the animal-to-human model transition, thus getting closer to a more consistent application in clinical practice. Multiple evidence shows why NPs are considered as the preferred drug delivery system: Firstly, the experience with NPs in other fields such as cancer therapy or vaccines has helped researchers to design NPs able to reach a particular tissue and specific cellular or molecular targets. In addition, the uptake of nanostructures by cells is higher compared to microstructures (1 and 10 µm) and NPs are commonly phagocytized more effectively than microspheres by macrophages [72]. Indeed, as macrophages are among the most important immune cells regulating the level of inflammation inside the knee joint, a proper uptake of the drugs by these cells (facilitated by NPs) can result in a reprogramming of their phenotype from an M1-like (which is pro-inflammatory) to an M2-like, which instead contributes to reduce the inflammatory distress in OA patients [73]. On the contrary, microspheres might be too big to be uptaken by macrophages and the preferential option for these microparticles is to release their content in the joint’s space, with a reduced ability to target specific receptors on the surface of precise immune cells. At the same time, hydrogels have a robust advantage versus other drug delivery systems, thanks to their possible application as biomaterials for bioprinting [74]. Moving toward the materials suitable for NP, MP, or hydrogel fabrication, the most employed were polymers, and in particular PLGA. The main features of this polymer are its synthetic nature and the possibility of modelling the drug releasing profile; the timing for degradation of PLGA-based NPs can be tuned to obtain a slower release by interfering with its molecular weight and ratio of lactide to glycolic acid in order to achieve the desired dosage and release interval depending upon the drug type [75]. The second most employed particles were liposomes. These carriers have the advantages of being nontoxic, flexible, biocompatible, completely biodegradable, and nonimmunogenic after systemic and non systemic administrations but they carry also some disadvantages such as a short half-life and high production cost [76]. Aside from the nature of the employed structures, many studies have investigated the ability of liposomes to produce a timely sustained drug release over time, and this is one of the main goals to achieve in the field of intra-articular injections for the treatment of knee OA. In regard to this aspect, hydrogels showed the longest observed beneficial effects in terms of reduction in cytokines concertation, better OARSI score, and histological features up to an average time of 8.4 weeks after treatment, compared to 6.8 weeks with MP and 5 weeks with NPs. Some important findings and preliminary trends can be garnered from this review, revealing that as a whole the literature in this area is still limited and a significant knowledge gap in the field exists.

5. Limitations

The main limitations of the present review are represented by the heterogeneity of the included studies in terms of materials (Table 1, Table 2 and Table 3), loaded drugs (Figure 4), analyzed outcomes, time intervals of observation, and animal models, thus revealing the need for further studies to identify the best performing drug delivery system.

6. Conclusions

The awareness that the “right” intra-articular carrier may optimize and increase the therapeutic effects of drugs has boosted the research on NPs, MPs, and hydrogels. The inherent translational potential in orthopaedics appears relevant considering both the clinical and socioeconomic burden of knee OA and the expanding range of therapeutic agents currently being tested on humans.

Author Contributions

F.M.G. and A.U. performed the systematic review and data collection. B.D.M., F.T.A. and F.R. drafted the manuscript. E.K. performed the critical review and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This project received funding from the European Union’s Horizon 2020 Research and Innovation Programme under grant agreement No. 814444 (MEFISTO).

Institutional Review Board Statement

Not applicable (systematic review of the literature).

Informed Consent Statement

Not applicable.

Data Availability Statement

All the data retrieved have been included in the manuscript.

Conflicts of Interest

F.M. Gambaro, A. Ummarino, F. Torres Andon and F. Ronzoni have nothing to disclose. B. Di Matteo was a paid speaker for Arthrex Italy. E. Kon is a consultant for Cartiheal Ltd. (Israel) and Greenbone Ortho S.r.l. (Italy); she was a paid speaker for Zimmer Biomet (USA) and Fidia Spa (Italy). She has royalties from Cartiheal Ltd. (Israel).

Abbreviations

HAHyaluronic Acid
IAIntra-Articular
MPsMicroparticles
MMP-3Matrix-Metalloprotenase-3
NPsNanoparticles
OAOsteoarthritis
PEGPolyethylene Glycol
PLAPoly Lactic Acid
PLGAPoly Lactic-co-Glycolic Acid

References

  1. Bannuru, R.R.; Schmid, C.H.; Kent, D.M.; Vaysbrot, E.E.; Wong, J.B.; McAlindon, T.E. Comparative Effectiveness of Pharmacologic Interventions for Knee Osteoarthritis: A Systematic Review and Network Meta-Analysis. Ann. Intern. Med. 2015, 162, 46–54. [Google Scholar] [CrossRef]
  2. Mora, J.C.; Przkora, R.; Cruz-Almeida, Y. Knee Osteoarthritis: Pathophysiology and Current Treatment Modalities. J. Pain Res. 2018, 11, 2189–2196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. McAlindon, T.E.; Bannuru, R.R.; Sullivan, M.C.; Arden, N.K.; Berenbaum, F.; Bierma-Zeinstra, S.M.; Hawker, G.A.; Henrotin, Y.; Hunter, D.J.; Kawaguchi, H.; et al. OARSI Guidelines for the Non-Surgical Management of Knee Osteoarthritis. Osteoarthr. Cartil. 2014, 22, 363–388. [Google Scholar] [CrossRef] [Green Version]
  4. Hochberg, M.C.; Altman, R.D.; April, K.T.; Benkhalti, M.; Guyatt, G.; McGowan, J.; Towheed, T.; Welch, V.; Wells, G.; Tugwell, P.; et al. American College of Rheumatology 2012 Recommendations for the Use of Nonpharmacologic and Pharmacologic Therapies in Osteoarthritis of the Hand, Hip, and Knee. Arthritis Care Res. 2012, 64, 465–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Jevsevar, D.S. Treatment of Osteoarthritis of the Knee: Evidence-Based Guideline, 2nd Edition. J. Am. Acad. Orthop. Surg. 2013, 21, 571–576. [Google Scholar] [CrossRef] [PubMed]
  6. Ceccarelli, G.; Gentile, P.; Marcarelli, M.; Balli, M.; Ronzoni, F.L.; Benedetti, L.; Cusella De Angelis, M.G. In Vitro and In Vivo Studies of Alar-Nasal Cartilage Using Autologous Micro-Grafts: The Use of the Rigenera® Protocol in the Treatment of an Osteochondral Lesion of the Nose. Pharmaceuticals 2017, 10, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Xue, Q.; Long, H.; Lin, J.; Du, D.; Zhou, J.; Chen, J.; Li, S.; Zhang, Y.; Cheng, Y.; Ma, X.; et al. Quality of Life and Treatment Satisfaction with Pharmacological Interventions in Chinese Adults with Chronic Pain Due to Osteoarthritis. BMC Musculoskelet. Disord. 2021, 22, 178. [Google Scholar] [CrossRef]
  8. Allen, K.D.; Adams, S.B.; Setton, L.A. Evaluating Intra-Articular Drug Delivery for the Treatment of Osteoarthritis in a Rat Model. Tissue Eng. Part B Rev. 2010, 16, 81–92. [Google Scholar] [CrossRef] [Green Version]
  9. Kou, L.; Xiao, S.; Sun, R.; Bao, S.; Yao, Q.; Chen, R. Biomaterial-Engineered Intra-Articular Drug Delivery Systems for Osteoarthritis Therapy. Drug Deliv. 2019, 26, 870–885. [Google Scholar] [CrossRef] [PubMed]
  10. ISO/TS 80004-1:2015(En), Nanotechnologies—Vocabulary—Part 1: Core Terms. Available online: https://www.iso.org/obp/ui/#iso:std:iso:ts:80004:-1:ed-2:v1:en (accessed on 5 April 2021).
  11. Zhang, Y. Relationship between Size and Function of Natural Substance Particles. Nano Biomed. Eng. 2011, 3, 1–16. [Google Scholar] [CrossRef]
  12. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.d.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano Based Drug Delivery Systems: Recent Developments and Future Prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef] [Green Version]
  13. Anselmo, A.C.; Mitragotri, S. Nanoparticles in the Clinic: An Update. Bioeng. Transl. Med. 2019, 4, e10143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Thi, T.T.H.; Suys, E.J.A.; Lee, J.S.; Nguyen, D.H.; Park, K.D.; Truong, N.P. Lipid-Based Nanoparticles in the Clinic and Clinical Trials: From Cancer Nanomedicine to COVID-19 Vaccines. Vaccines 2021, 9, 359. [Google Scholar] [CrossRef]
  15. Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q. Lipid Nanoparticles-From Liposomes to MRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021. [Google Scholar] [CrossRef] [PubMed]
  16. Rahimi, M.; Charmi, G.; Matyjaszewski, K.; Banquy, X.; Pietrasik, J. Recent Developments in Natural and Synthetic Polymeric Drug Delivery Systems Used for the Treatment of Osteoarthritis. Acta Biomater. 2021, 123, 31–50. [Google Scholar] [CrossRef] [PubMed]
  17. Khan, Y.S.; Farhana, A. Histology, Cell. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2020. [Google Scholar]
  18. Maudens, P.; Seemayer, C.A.; Pfefferlé, F.; Jordan, O.; Allémann, E. Nanocrystals of a Potent P38 MAPK Inhibitor Embedded in Microparticles: Therapeutic Effects in Inflammatory and Mechanistic Murine Models of Osteoarthritis. J. Control. Release 2018, 276, 102–112. [Google Scholar] [CrossRef]
  19. Paik, J.; Duggan, S.T.; Keam, S.J. Triamcinolone Acetonide Extended-Release: A Review in Osteoarthritis Pain of the Knee. Drugs 2019, 79, 455–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. He, Z.; Wang, B.; Hu, C.; Zhao, J. An Overview of Hydrogel-Based Intra-Articular Drug Delivery for the Treatment of Osteoarthritis. Colloids Surf. B Biointerfaces 2017, 154, 33–39. [Google Scholar] [CrossRef] [PubMed]
  21. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gøtzsche, P.C.; Ioannidis, J.P.A.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA Statement for Reporting Systematic Reviews and Meta-Analyses of Studies That Evaluate Healthcare Interventions: Explanation and Elaboration. BMJ 2009, 339, b2700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Fan, W.; Li, J.; Yuan, L.; Chen, J.; Wang, Z.; Wang, Y.; Guo, C.; Mo, X.; Yan, Z. Intra-Articular Injection of Kartogenin-Conjugated Polyurethane Nanoparticles Attenuates the Progression of Osteoarthritis. Drug Deliv. 2018, 25, 1004–1012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Higaki, M.; Ishihara, T.; Izumo, N.; Takatsu, M.; Mizushima, Y. Treatment of Experimental Arthritis with Poly(D, L-Lactic/Glycolic Acid) Nanoparticles Encapsulating Betamethasone Sodium Phosphate. Ann. Rheum. Dis. 2005, 64, 1132–1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Horisawa, E.; Hirota, T.; Kawazoe, S.; Yamada, J.; Yamamoto, H.; Takeuchi, H.; Kawashima, Y. Prolonged Anti-Inflammatory Action of DL-Lactide/Glycolide Copolymer Nanospheres Containing Betamethasone Sodium Phosphate for an Intra-Articular Delivery System in Antigen-Induced Arthritic Rabbit. Pharm. Res. 2002, 19, 403–410. [Google Scholar] [CrossRef] [PubMed]
  25. Kamel, R.; Salama, A.H.; Mahmoud, A.A. Development and Optimization of Self-Assembling Nanosystem for Intra-Articular Delivery of Indomethacin. Int. J. Pharm. 2016, 515, 657–668. [Google Scholar] [CrossRef]
  26. Kang, C.; Jung, E.; Hyeon, H.; Seon, S.; Lee, D. Acid-Activatable Polymeric Curcumin Nanoparticles as Therapeutic Agents for Osteoarthritis. Nanomedicine 2020, 23, 102104. [Google Scholar] [CrossRef] [PubMed]
  27. Kang, M.-L.; Kim, J.-E.; Im, G.-I. Thermoresponsive Nanospheres with Independent Dual Drug Release Profiles for the Treatment of Osteoarthritis. Acta Biomater. 2016, 39, 65–78. [Google Scholar] [CrossRef] [PubMed]
  28. Kim, S.R.; Ho, M.J.; Kim, S.H.; Cho, H.R.; Kim, H.S.; Choi, Y.S.; Choi, Y.W.; Kang, M.J. Increased Localized Delivery of Piroxicam by Cationic Nanoparticles after Intra-Articular Injection. Drug Des. Dev. Ther. 2016, 10, 3779–3787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Liu, P.; Gu, L.; Ren, L.; Chen, J.; Li, T.; Wang, X.; Yang, J.; Chen, C.; Sun, L. Intra-Articular Injection of Etoricoxib-Loaded PLGA-PEG-PLGA Triblock Copolymeric Nanoparticles Attenuates Osteoarthritis Progression. Am. J. Transl. Res. 2019, 11, 6775–6789. [Google Scholar] [PubMed]
  30. Liu, X.; Corciulo, C.; Arabagian, S.; Ulman, A.; Cronstein, B.N. Adenosine-Functionalized Biodegradable PLA-b-PEG Nanoparticles Ameliorate Osteoarthritis in Rats. Sci. Rep. 2019, 9, 7430. [Google Scholar] [CrossRef] [PubMed]
  31. Villamagna, I.J.; Gordon, T.N.; Hurtig, M.B.; Beier, F.; Gillies, E.R. Poly(Ester Amide) Particles for Controlled Delivery of Celecoxib. J. Biomed. Mater. Res. A 2019, 107, 1235–1243. [Google Scholar] [CrossRef]
  32. Zerrillo, L.; Que, I.; Vepris, O.; Morgado, L.N.; Chan, A.; Bierau, K.; Li, Y.; Galli, F.; Bos, E.; Censi, R.; et al. PH-Responsive Poly(Lactide-Co-Glycolide) Nanoparticles Containing near-Infrared Dye for Visualization and Hyaluronic Acid for Treatment of Osteoarthritis. J. Control. Release 2019, 309, 265–276. [Google Scholar] [CrossRef]
  33. Zhou, F.; Mei, J.; Yang, S.; Han, X.; Li, H.; Yu, Z.; Qiao, H.; Tang, T. Modified ZIF-8 Nanoparticles Attenuate Osteoarthritis by Reprogramming the Metabolic Pathway of Synovial Macrophages. ACS Appl. Mater. Interfaces 2020, 12, 2009–2022. [Google Scholar] [CrossRef] [PubMed]
  34. Zhou, P.-H.; Qiu, B.; Deng, R.-H.; Li, H.-J.; Xu, X.-F.; Shang, X.-F. Chondroprotective Effects of Hyaluronic Acid-Chitosan Nanoparticles Containing Plasmid DNA Encoding Cytokine Response Modifier A in a Rat Knee Osteoarthritis Model. Cell. Physiol. Biochem. 2018, 47, 1207–1216. [Google Scholar] [CrossRef] [PubMed]
  35. Yudoh, K.; Shishido, K.; Murayama, H.; Yano, M.; Matsubayashi, K.; Takada, H.; Nakamura, H.; Masuko, K.; Kato, T.; Nishioka, K. Water-Soluble C60 Fullerene Prevents Degeneration of Articular Cartilage in Osteoarthritis via down-Regulation of Chondrocyte Catabolic Activity and Inhibition of Cartilage Degeneration during Disease Development. Arthritis Rheum. 2007, 56, 3307–3318. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, A.; Wang, P.; Zhang, J.; Ye, W.; Wei, Q. Restoration Effect and Tribological Behavior of Hyaluronic Acid Reinforced with Graphene Oxide in Osteoarthritis. J. Nanosci. Nanotechnol. 2019, 19, 91–97. [Google Scholar] [CrossRef] [PubMed]
  37. Sacchetti, C.; Liu-Bryan, R.; Magrini, A.; Rosato, N.; Bottini, N.; Bottini, M. Polyethylene-Glycol-Modified Single-Walled Carbon Nanotubes for Intra-Articular Delivery to Chondrocytes. ACS Nano 2014, 8, 12280–12291. [Google Scholar] [CrossRef] [Green Version]
  38. Dong, J.; Jiang, D.; Wang, Z.; Wu, G.; Miao, L.; Huang, L. Intra-Articular Delivery of Liposomal Celecoxib-Hyaluronate Combination for the Treatment of Osteoarthritis in Rabbit Model. Int. J. Pharm. 2013, 441, 285–290. [Google Scholar] [CrossRef]
  39. Sarkar, A.; Carvalho, E.; D’souza, A.A.; Banerjee, R. Liposome-Encapsulated Fish Oil Protein-Tagged Gold Nanoparticles for Intra-Articular Therapy in Osteoarthritis. Nanomedicine 2019, 14, 871–887. [Google Scholar] [CrossRef] [PubMed]
  40. Gentile, P.; Chiono, V.; Carmagnola, I.; Hatton, P.V. An Overview of Poly(Lactic-Co-Glycolic) Acid (PLGA)-Based Biomaterials for Bone Tissue Engineering. Int. J. Mol. Sci. 2014, 15, 3640–3659. [Google Scholar] [CrossRef]
  41. Reddy, M.S.B.; Ponnamma, D.; Choudhary, R.; Sadasivuni, K.K. A Comparative Review of Natural and Synthetic Biopolymer Composite Scaffolds. Polymers 2021, 13, 1105. [Google Scholar] [CrossRef]
  42. Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a Strategy for Improving Nanoparticle-Based Drug and Gene Delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef] [Green Version]
  43. Sultankulov, B.; Berillo, D.; Sultankulova, K.; Tokay, T.; Saparov, A. Progress in the Development of Chitosan-Based Biomaterials for Tissue Engineering and Regenerative Medicine. Biomolecules 2019, 9, 470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Pei, Y.; Cui, F.; Du, X.; Shang, G.; Xiao, W.; Yang, X.; Cui, Q. Antioxidative Nanofullerol Inhibits Macrophage Activation and Development of Osteoarthritis in Rats. Int. J. Nanomed. 2019, 14, 4145–4155. [Google Scholar] [CrossRef] [Green Version]
  45. Wang, X.; Chen, X.-Z.; Alcântara, C.C.J.; Sevim, S.; Hoop, M.; Terzopoulou, A.; de Marco, C.; Hu, C.; de Mello, A.J.; Falcaro, P.; et al. MOFBOTS: Metal-Organic-Framework-Based Biomedical Microrobots. Adv. Mater. 2019, 31, e1901592. [Google Scholar] [CrossRef] [PubMed]
  46. Abou-ElNour, M.; Ishak, R.A.H.; Tiboni, M.; Bonacucina, G.; Cespi, M.; Casettari, L.; Soliman, M.E.; Geneidi, A.S. Triamcinolone Acetonide-Loaded PLA/PEG-PDL Microparticles for Effective Intra-Articular Delivery: Synthesis, Optimization, in Vitro and in Vivo Evaluation. J. Control. Release 2019, 309, 125–144. [Google Scholar] [CrossRef]
  47. Bodick, N.; Williamson, T.; Strand, V.; Senter, B.; Kelley, S.; Boyce, R.; Lightfoot-Dunn, R. Local Effects Following Single and Repeat Intra-Articular Injections of Triamcinolone Acetonide Extended-Release: Results from Three Nonclinical Toxicity Studies in Dogs. Rheumatol. Ther. 2018, 5, 475–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Conaghan, P.G.; Cohen, S.B.; Berenbaum, F.; Lufkin, J.; Johnson, J.R.; Bodick, N. Brief Report: A Phase IIb Trial of a Novel Extended-Release Microsphere Formulation of Triamcinolone Acetonide for Intraarticular Injection in Knee Osteoarthritis. Arthritis Rheumatol. 2018, 70, 204–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Conaghan, P.G.; Hunter, D.J.; Cohen, S.B.; Kraus, V.B.; Berenbaum, F.; Lieberman, J.R.; Jones, D.G.; Spitzer, A.I.; Jevsevar, D.S.; Katz, N.P.; et al. Effects of a Single Intra-Articular Injection of a Microsphere Formulation of Triamcinolone Acetonide on Knee Osteoarthritis Pain. J. Bone Jt. Surg. Am. 2018, 100, 666–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Chen, X.; Liu, Y.; Wen, Y.; Yu, Q.; Liu, J.; Zhao, Y.; Liu, J.; Ye, G. A Photothermal-Triggered Nitric Oxide Nanogenerator Combined with SiRNA for Precise Therapy of Osteoarthritis by Suppressing Macrophage Inflammation. Nanoscale 2019, 11, 6693–6709. [Google Scholar] [CrossRef]
  51. Kraus, V.B.; Conaghan, P.G.; Aazami, H.A.; Mehra, P.; Kivitz, A.J.; Lufkin, J.; Hauben, J.; Johnson, J.R.; Bodick, N. Synovial and Systemic Pharmacokinetics (PK) of Triamcinolone Acetonide (TA) Following Intra-Articular (IA) Injection of an Extended-Release Microsphere-Based Formulation (FX006) or Standard Crystalline Suspension in Patients with Knee Osteoarthritis (OA). Osteoarthr. Cartil. 2018, 26, 34–42. [Google Scholar] [CrossRef] [Green Version]
  52. Ho, M.J.; Jeong, H.T.; Im, S.H.; Kim, H.T.; Lee, J.E.; Park, J.S.; Cho, H.R.; Kim, D.Y.; Choi, Y.W.; Lee, J.W.; et al. Design and In Vivo Pharmacokinetic Evaluation of Triamcinolone Acetonide Microcrystals-Loaded PLGA Microsphere for Increased Drug Retention in Knees after Intra-Articular Injection. Pharmaceutics 2019, 11, 419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kumar, A.; Bendele, A.M.; Blanks, R.C.; Bodick, N. Sustained Efficacy of a Single Intra-Articular Dose of FX006 in a Rat Model of Repeated Localized Knee Arthritis. Osteoarthr. Cartil. 2015, 23, 151–160. [Google Scholar] [CrossRef] [Green Version]
  54. Russell, S.J.; Sala, R.; Conaghan, P.G.; Habib, G.; Vo, Q.; Manning, R.; Kivitz, A.; Davis, Y.; Lufkin, J.; Johnson, J.R.; et al. Triamcinolone Acetonide Extended-Release in Patients with Osteoarthritis and Type 2 Diabetes: A Randomized, Phase 2 Study. Rheumatology 2018, 57, 2235–2241. [Google Scholar] [CrossRef] [Green Version]
  55. Rudnik-Jansen, I.; Schrijver, K.; Woike, N.; Tellegen, A.; Versteeg, S.; Emans, P.; Mihov, G.; Thies, J.; Eijkelkamp, N.; Tryfonidou, M.; et al. Intra-Articular Injection of Triamcinolone Acetonide Releasing Biomaterial Microspheres Inhibits Pain and Inflammation in an Acute Arthritis Model. Drug Deliv. 2019, 26, 226–236. [Google Scholar] [CrossRef] [Green Version]
  56. Zhang, Z.; Bi, X.; Li, H.; Huang, G. Enhanced Targeting Efficiency of PLGA Microspheres Loaded with Lornoxicam for Intra-Articular Administration. Drug Deliv. 2011, 18, 536–544. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, Z.; Huang, G. Intra-Articular Lornoxicam Loaded PLGA Microspheres: Enhanced Therapeutic Efficiency and Decreased Systemic Toxicity in the Treatment of Osteoarthritis. Drug Deliv. 2012, 19, 255–263. [Google Scholar] [CrossRef] [Green Version]
  58. Aydin, O.; Korkusuz, F.; Korkusuz, P.; Tezcaner, A.; Bilgic, E.; Yaprakci, V.; Keskin, D. In Vitro and in Vivo Evaluation of Doxycycline-Chondroitin Sulfate/PCLmicrospheres for Intraarticular Treatment of Osteoarthritis. J. Biomed. Mater. Res. B Appl. Biomater. 2015, 103, 1238–1248. [Google Scholar] [CrossRef] [PubMed]
  59. Tellier, L.E.; Treviño, E.A.; Brimeyer, A.L.; Reece, D.S.; Willett, N.J.; Guldberg, R.E.; Temenoff, J.S. Intra-Articular TSG-6 Delivery from Heparin-Based Microparticles Reduces Cartilage Damage in a Rat Model of Osteoarthritis. Biomater. Sci. 2018, 6, 1159–1167. [Google Scholar] [CrossRef]
  60. Park, J.W.; Yun, Y.-P.; Park, K.; Lee, J.Y.; Kim, H.-J.; Kim, S.E.; Song, H.-R. Ibuprofen-Loaded Porous Microspheres Suppressed the Progression of Monosodium Iodoacetate-Induced Osteoarthritis in a Rat Model. Colloids Surf. B Biointerfaces 2016, 147, 265–273. [Google Scholar] [CrossRef] [PubMed]
  61. Matsuzaki, T.; Matsushita, T.; Tabata, Y.; Saito, T.; Matsumoto, T.; Nagai, K.; Kuroda, R.; Kurosaka, M. Intra-Articular Administration of Gelatin Hydrogels Incorporating Rapamycin-Micelles Reduces the Development of Experimental Osteoarthritis in a Murine Model. Biomaterials 2014, 35, 9904–9911. [Google Scholar] [CrossRef] [PubMed]
  62. Tanaka, T.; Matsushita, T.; Nishida, K.; Takayama, K.; Nagai, K.; Araki, D.; Matsumoto, T.; Tabata, Y.; Kuroda, R. Attenuation of Osteoarthritis Progression in Mice Following Intra-Articular Administration of Simvastatin-Conjugated Gelatin Hydrogel. J. Tissue Eng. Regen. Med. 2019, 13, 423–432. [Google Scholar] [CrossRef]
  63. Tsubosaka, M.; Kihara, S.; Hayashi, S.; Nagata, J.; Kuwahara, T.; Fujita, M.; Kikuchi, K.; Takashima, Y.; Kamenaga, T.; Kuroda, Y.; et al. Gelatin Hydrogels with Eicosapentaenoic Acid Can Prevent Osteoarthritis Progression in Vivo in a Mouse Model. J. Orthop. Res. 2020, 38, 2157–2169. [Google Scholar] [CrossRef]
  64. Lu, H.-T.; Sheu, M.-T.; Lin, Y.-F.; Lan, J.; Chin, Y.-P.; Hsieh, M.-S.; Cheng, C.-W.; Chen, C.-H. Injectable Hyaluronic-Acid-Doxycycline Hydrogel Therapy in Experimental Rabbit Osteoarthritis. BMC Vet. Res. 2013, 9, 68. [Google Scholar] [CrossRef] [Green Version]
  65. García-Fernández, L.; Olmeda-Lozano, M.; Benito-Garzón, L.; Pérez-Caballer, A.; San Román, J.; Vázquez-Lasa, B. Injectable Hydrogel-Based Drug Delivery System for Cartilage Regeneration. Mater. Sci. Eng. C 2020, 110, 110702. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, T.; Chen, S.; Dou, H.; Liu, Q.; Shu, G.; Lin, J.; Zhang, W.; Peng, G.; Zhong, Z.; Fu, H. Novel Glucosamine-Loaded Thermosensitive Hydrogels Based on Poloxamers for Osteoarthritis Therapy by Intra-Articular Injection. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 118, 111352. [Google Scholar] [CrossRef] [PubMed]
  67. Hui, J.H.; Chan, S.-W.; Li, J.; Goh, J.C.H.; Li, L.; Ren, X.F.; Lee, E.H. Intra-Articular Delivery of Chondroitin Sulfate for the Treatment of Joint Defects in Rabbit Model. J. Mol. Hist. 2007, 38, 483–489. [Google Scholar] [CrossRef] [PubMed]
  68. Mok, S.-W.; Fu, S.-C.; Cheuk, Y.-C.; Chu, I.-M.; Chan, K.-M.; Qin, L.; Yung, S.-H.; Kevin Ho, K.-W. Intra-Articular Delivery of Quercetin Using Thermosensitive Hydrogel Attenuate Cartilage Degradation in an Osteoarthritis Rat Model. Cartilage 2020, 11, 490–499. [Google Scholar] [CrossRef]
  69. Stefani, R.M.; Lee, A.J.; Tan, A.R.; Halder, S.S.; Hu, Y.; Guo, X.E.; Stoker, A.M.; Ateshian, G.A.; Marra, K.G.; Cook, J.L.; et al. Sustained Low-Dose Dexamethasone Delivery via a PLGA Microsphere-Embedded Agarose Implant for Enhanced Osteochondral Repair. Acta Biomater. 2020, 102, 326–340. [Google Scholar] [CrossRef]
  70. Wang, Q.-S.; Xu, B.-X.; Fan, K.-J.; Li, Y.-W.; Wu, J.; Wang, T.-Y. Dexamethasone-Loaded Thermosensitive Hydrogel Suppresses Inflammation and Pain in Collagen-Induced Arthritis Rats. Drug Des. Dev. Ther. 2020, 14, 4101–4113. [Google Scholar] [CrossRef] [PubMed]
  71. Mou, D.; Yu, Q.; Zhang, J.; Zhou, J.; Li, X.; Zhuang, W.; Yang, X. Intra-Articular Injection of Chitosan-Based Supramolecular Hydrogel for Osteoarthritis Treatment. Tissue Eng. Regen. Med. 2021, 18, 113–125. [Google Scholar] [CrossRef] [PubMed]
  72. Jeevanandam, J.; Barhoum, A.; Chan, Y.S.; Dufresne, A.; Danquah, M.K. Review on Nanoparticles and Nanostructured Materials: History, Sources, Toxicity and Regulations. Beilstein J. Nanotechnol. 2018, 9, 1050–1074. [Google Scholar] [CrossRef] [Green Version]
  73. Ummarino, A.; Gambaro, F.M.; Kon, E.; Torres Andón, F. Therapeutic Manipulation of Macrophages Using Nanotechnological Approaches for the Treatment of Osteoarthritis. Nanomaterials 2020, 10, 1562. [Google Scholar] [CrossRef] [PubMed]
  74. Pescosolido, L.; Schuurman, W.; Malda, J.; Matricardi, P.; Alhaique, F.; Coviello, T.; van Weeren, P.R.; Dhert, W.J.A.; Hennink, W.E.; Vermonden, T. Hyaluronic Acid and Dextran-Based Semi-IPN Hydrogels as Biomaterials for Bioprinting. Biomacromolecules 2011, 12, 1831–1838. [Google Scholar] [CrossRef]
  75. Makadia, H.K.; Siegel, S.J. Poly Lactic-Co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier. Polymers 2011, 3, 1377–1397. [Google Scholar] [CrossRef] [PubMed]
  76. Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K. Liposome: Classification, Preparation, and Applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic representation of the sizes of the three main types of drug delivery systems: the nanoparticle (size range 1–1000 nm), the microparticle (up to 10 μm), and the hydrogel (up to 10 mm). NOTE: the images are not in real proportions.
Figure 1. Schematic representation of the sizes of the three main types of drug delivery systems: the nanoparticle (size range 1–1000 nm), the microparticle (up to 10 μm), and the hydrogel (up to 10 mm). NOTE: the images are not in real proportions.
Ijms 22 09137 g001
Figure 2. PRISMA Flowchart summarizing the papers’ selection process. Legend: OA: osteoarthritis.
Figure 2. PRISMA Flowchart summarizing the papers’ selection process. Legend: OA: osteoarthritis.
Ijms 22 09137 g002
Figure 3. Distribution of the included studies according to the year of publication.
Figure 3. Distribution of the included studies according to the year of publication.
Ijms 22 09137 g003
Figure 4. Distribution of drugs loaded on the different types of delivery systems in the included studies. Legend: HA—Hyaluronic acid, NSAID—non-steroidal anti-inflammatory drug; TA—Triamcinolone Acetonide.
Figure 4. Distribution of drugs loaded on the different types of delivery systems in the included studies. Legend: HA—Hyaluronic acid, NSAID—non-steroidal anti-inflammatory drug; TA—Triamcinolone Acetonide.
Ijms 22 09137 g004
Table 1. Synopsis of studies employing nanoparticles (NPs). Legend: ADAMTS-5—A disintegrin and metalloproteinase with thrombospondin motifs 5 2; CrmA—cytokine response modifier A; DPPC—1,2-dipalmitoyl-sn-glycero-3-phosphocholine; HA—hyaluronic acid; HDI—hexamethylene diisocyanate; IL—interleukin; MMP—matrix metalloproteinase; NGF—nerve growth factor; OARSI—Osteoarthritis Research Society International; PAE—poly(β-amino ester); PEAs—poly(ester amide)s; PEG—poly(ethylene glycol); PLA—poly(lactic acid): PLGA—poly(lactic-co-glycolic acid); pNIPAM—poly(N-isopropylacrylamide); TNF-α—tumor necrosis factor alpha; WBC—white blood cells; ZIF-8—zeolitic imidazolate framework-8.
Table 1. Synopsis of studies employing nanoparticles (NPs). Legend: ADAMTS-5—A disintegrin and metalloproteinase with thrombospondin motifs 5 2; CrmA—cytokine response modifier A; DPPC—1,2-dipalmitoyl-sn-glycero-3-phosphocholine; HA—hyaluronic acid; HDI—hexamethylene diisocyanate; IL—interleukin; MMP—matrix metalloproteinase; NGF—nerve growth factor; OARSI—Osteoarthritis Research Society International; PAE—poly(β-amino ester); PEAs—poly(ester amide)s; PEG—poly(ethylene glycol); PLA—poly(lactic acid): PLGA—poly(lactic-co-glycolic acid); pNIPAM—poly(N-isopropylacrylamide); TNF-α—tumor necrosis factor alpha; WBC—white blood cells; ZIF-8—zeolitic imidazolate framework-8.
TableArticlesDrugMaterialAnimal ModelResultsRef
Polymeric NPsFan W 2018KartogeninPEG-HDI-N-BOC SerinolRatBetter histological and OARSI score at 12 weeks compared to control[22]
Higaki M 2005BetamethasonePLGARatDecrease in inflammatory cells after 7 days[23]
Horisawa E 2002BetamethasonePLGARabbitDecreased joint swelling for 21 d[24]
Kamel R 2016IndomethacinSelf assembling PLGARat↓ diameter; favorable histology; ↓ TNF-α in serum[25]
Kang C 2020CurcuminAcid-activable PAERat↓ TNF-α and IL-1β production, favorable histology[25]
Kang M 2016Diclofenac/KartogeninPluronicRat↓ of OARSI score[26]
Kim SR 2016PiroxicamPLGA + Eudragit RLRatProlonged retention into joint compared to NPs without Eudragit RL[27]
Liu P
2019
EtoricoxibPLGA/PEGRatFavorable μCT; ↓ MMP-13 and ADAMTS-5; ↑ collagen and aggrecan[28]
Liu X
2019
AdenosinePLGA-PEGRat↓ OARSI score[30]
Villamagna IJ 2019CelecoxibPEAsSheep↓ joint effusion; ↓ WBC[31]
Zerrillo L 2019HAPLGARatNP are still in the knee after 35 days[32]
Zhou F 2015Berberine chlorideChitosanRatHigher anti-apoptosis activity and prolonged i.a. drug retention[33]
Zhou PH 2018CrmAHA and ChitosanRat↓ OARSI score; ↓ IL-1β, MMP-3, MMP-13; collagen conserved[34]
LiposomesDong J 2013celecoxibLiposome + hyaluronic acidRabbitFavorable histology[38]
Carbon-based NPsYudoh K 2007KAFAKFullereneRabbitFavorable histology[35]
Liu A
2019
Hyaluronan conjugationGraphene oxideRat↓ MMP-3 concentration in the joint[36]
Sacchetti C. 2014Antisense oligomersCarbon nanotubesRatInhibition of protein synthesis in chondrocytes and reduction in inflammation[37]
Metal-based NPsSarkar A 2019Fish oil protein, both in DPPC liposomesGoldRat↓ IL-1β, IL-12, PGE2, TNF-α[39]
Other NPsZhou F 2020S-methylisothiourea Catalase Anti-CD16/32ZIF-8 (MOF)RatFavorable histology and X-ray[33]
Table 2. Synopsis of studies employing microparticles (MPs). Legend: ADAMTS-5—A disintegrin and metalloproteinase with thrombospondin motifs 5 2; CrmA—cytokine response modifier A; DPPC—1,2-dipalmitoyl-sn-glycero-3-phosphocholine; FBR—foreign-body responses; HA—hyaluronic acid; HDI—hexamethylene diisocyanate; IL—interleukin; MMP—matrix metalloproteinase; NGF—nerve growth factor; OARSI—Osteoarthritis Research Society International; PAE—poly(β-amino ester); PEAs—poly(ester amide)s; PEG—poly(ethylene glycol); PLA—poly(lactic acid): PLGA—poly(lactic-co-glycolic acid); TNF-α—tumor necrosis factor alpha; WBC—white blood cells.
Table 2. Synopsis of studies employing microparticles (MPs). Legend: ADAMTS-5—A disintegrin and metalloproteinase with thrombospondin motifs 5 2; CrmA—cytokine response modifier A; DPPC—1,2-dipalmitoyl-sn-glycero-3-phosphocholine; FBR—foreign-body responses; HA—hyaluronic acid; HDI—hexamethylene diisocyanate; IL—interleukin; MMP—matrix metalloproteinase; NGF—nerve growth factor; OARSI—Osteoarthritis Research Society International; PAE—poly(β-amino ester); PEAs—poly(ester amide)s; PEG—poly(ethylene glycol); PLA—poly(lactic acid): PLGA—poly(lactic-co-glycolic acid); TNF-α—tumor necrosis factor alpha; WBC—white blood cells.
ArticlesDrugMaterialAnimal ModelResultsRef
Abou Elnour M 2019Triamcinolone acetonide (TA)PLGARatMP performed higher in inflammation suppression, compared to the free drug suspension[46]
Allah HA 2016LornoxicamChitosanRatPersistent inhibition of knee swelling and lower IL-6 levels until 14 days[35]
Aydin O 2015Doxycycline (D) and doxycycline-chondroitin sulfate (D-CS)poly-ɛ-caprolactone (PCL)RabbitRadiographic scores of D MS and D-CS MS groups improved after 8 weeks when compared to OA groups[58]
Bodick N 2018Triamcinolone acetonidePLGADogToxicity study, the synovial FBR to PLGA microspheres was focal and transient[47]
Chen H 2019HydroxychloroquinePLGARatDrug detectable inside the joint up to week 2[50]
Conaghan GP 2018Triamcinolone acetonidePLGAHumanDrug detectable inside the joint up to week 12[48]
Conaghan GP 2018Triamcinolone acetonidePLGAHumanDrug detectable inside the joint up to week 13[49]
Ho Jin M 2019Triamcinolone Acetonide MicrocrystalsPLGARatThe novel MS was physicochemically stable, with no changes in drug crystallinity and release profile over 12 months[52]
Kraus VB 2017Triamcinolone acetonidePLGAHumanDrug detectable inside the joint up to week 12[51]
Kumar A 2015Triamcinolone acetonidePLGA FX006RatImproved histological joint scores and better gait (pain) scores[53]
Park JW 2016IbuprofenPLGA, PVARatReduction in IL-1β, IL-6, IL-17, TNF-α, ADAMTS-5 and MMP3[60]
Rudnik-Jansen I 2019triamcinolone acetonidePLGA-PEARatA single intra-articular injection of TAA-loaded PEA microspheres reduced joint swelling and induced longer pain relief compared to bolus injection.[55]
Russel SJ 2018Triamcinolone acetonidePLGAHumanIn diabetic patients, who represented the study population, intra-articular PLGA-TA provided better post-injective glycemic control than standard TA[54]
Tellier EL 2018TNF-α stimulated gene-6 (TSG-6)Heparin based MPRatAfter 21 days, cartilage thickness, volume, and attenuation were significantly increased[59]
Zhang Z 2011LornoxicamPLGARatLonger retention in the joint with MP system[56]
Zhang Z 2012LornoxicamPLGARatRetention of drug in the joint for >96 h[57]
Table 3. Synopsis of studies employing hydrogels. Legend: ADAMTS-5A—disintegrin and metalloproteinase with thrombospondin motifs 5 2; ADH—adipic acid dihydrazide; CrmA—cytokine response modifier A; DPPC—1,2-dipalmitoyl-sn-glycero-3-phosphocholine; EG—ethyl-glycol; FBR—foreign-body responses; HA—hyaluronic acid; HDI—hexamethylene diisocyanate; IL—interleukin; MMP—matrix metalloproteinase; NGF—nerve growth factor; OARSI—Osteoarthritis Research Society International; PAE—poly(β-amino ester); PEAs—poly(ester amide)s; PEG—poly(ethylene glycol); PLA—poly(lactic acid): PLGA—poly(lactic-co-glycolic acid); TNF-α—tumor necrosis factor alpha; WBC—white blood cells.
Table 3. Synopsis of studies employing hydrogels. Legend: ADAMTS-5A—disintegrin and metalloproteinase with thrombospondin motifs 5 2; ADH—adipic acid dihydrazide; CrmA—cytokine response modifier A; DPPC—1,2-dipalmitoyl-sn-glycero-3-phosphocholine; EG—ethyl-glycol; FBR—foreign-body responses; HA—hyaluronic acid; HDI—hexamethylene diisocyanate; IL—interleukin; MMP—matrix metalloproteinase; NGF—nerve growth factor; OARSI—Osteoarthritis Research Society International; PAE—poly(β-amino ester); PEAs—poly(ester amide)s; PEG—poly(ethylene glycol); PLA—poly(lactic acid): PLGA—poly(lactic-co-glycolic acid); TNF-α—tumor necrosis factor alpha; WBC—white blood cells.
ArticlesDrugMaterialAnimal ModelResultsRef
Garcia-Fernandez L 2020Naproxen or DexamethasoneDextran and HARabbitDexamethasone group showed higher collagen type II levels and better recovery[65]
Hui JH 2007Chondroitin-sulfateα-Chondroitin Sulfate-EGRabbitThicker layer composed of hyaline and fibrocartilage compared to control (saline)[67]
Lu HT 2013DoxycyclineHA hydrogelRabbitLower grade of OA in the study group compared to control (saline)[64]
Matsuzaki T 2014RapamycinPolylactic acid, gelatinRatDelayed OA progression was maintained even at 16 weeks[61]
Mok SW 2020QuercetinPEG hydrogelRatReleased of Que could be sustained for >28 days. Higher OARSI score than control[68]
Stefani RM 2020DexamethasoneAgarose hydrogel loaded with MPDogsImproved OARSI scores for proteoglycan, chondrocyte, and collagen pathology[69]
Tanaka T 2019SimvastatitnPolylactic acid gelatinRatDecreased level cartilage-degrading enzymes and IL-1β and increased level type II collagen[62]
Tsubosaka T 2020Eicosapentanoic acidPolylactic acid gelatinRatMMP-3-, MMP-13-, IL-1β-, and p-IKK α/β-positive cell ratio were significantly lower[63]
Wang QS 2020DexamethasoneChitosan–glycerin-borax–hydrogelRatDecreased OA scores and joint inflammation compared to control[70]
Zhang T 2020GlucosaminePoloxamer 407 and 188 hydrogelRabbitDecreased swelling and inflammatory factors compared to control[66]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gambaro, F.M.; Ummarino, A.; Torres Andón, F.; Ronzoni, F.; Di Matteo, B.; Kon, E. Drug Delivery Systems for the Treatment of Knee Osteoarthritis: A Systematic Review of In Vivo Studies. Int. J. Mol. Sci. 2021, 22, 9137. https://doi.org/10.3390/ijms22179137

AMA Style

Gambaro FM, Ummarino A, Torres Andón F, Ronzoni F, Di Matteo B, Kon E. Drug Delivery Systems for the Treatment of Knee Osteoarthritis: A Systematic Review of In Vivo Studies. International Journal of Molecular Sciences. 2021; 22(17):9137. https://doi.org/10.3390/ijms22179137

Chicago/Turabian Style

Gambaro, Francesco Manlio, Aldo Ummarino, Fernando Torres Andón, Flavio Ronzoni, Berardo Di Matteo, and Elizaveta Kon. 2021. "Drug Delivery Systems for the Treatment of Knee Osteoarthritis: A Systematic Review of In Vivo Studies" International Journal of Molecular Sciences 22, no. 17: 9137. https://doi.org/10.3390/ijms22179137

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