376
Views
15
CrossRef citations to date
0
Altmetric
Review

Advances in nanomedicine for the treatment of ankylosing spondylitis

ORCID Icon, , ORCID Icon, , , , , & ORCID Icon show all
Pages 8521-8542 | Published online: 29 Oct 2019

References

  • Shaikh SA. Ankylosing spondylitis: recent breakthroughs in diagnosis and treatment. J Can Chiropr Assoc. 2007;51(4):249–260.18060011
  • Braun J, Sieper J. Ankylosing spondylitis. Lancet. 2007;369(9570):1379–1390. doi:10.1016/S0140-6736(07)60635-717448825
  • Dakwar E,  Reddy J, Vale FL, Uribe JS. A review of the pathogenesis of ankylosing spondylitis. Neurosurg Focus 2008;24(1):E2.
  • Gran JT, Husby G, Hordvik M. Prevalence of ankylosing spondylitis in males and females in a young middle-aged population of Tromsø, northern Norway. Ann Rheum Dis. 1985;44(6):359–367. doi:10.1136/ard.44.6.3593160312
  • Feldtkeller E, Khan MA, van der Heijde D, van der Linden S, Braun J. Age at disease onset and diagnosis delay in HLA-B27 negative vs. positive patients with ankylosing spondylitis. Rheumatol Int. 2003;23(2):61–66. doi:10.1007/s00296-002-0237-412634937
  • Colbert RA, Tran TM, Layh-Schmitt G. HLA-B27 misfolding and ankylosing spondylitis. Mol Immunol. 2014;57(1):44–51. doi:10.1016/j.molimm.2013.07.01323993278
  • Keidel S, Chen L, Pointon J, Wordsworth P. ERAP1 and ankylosing spondylitis. Curr Opin Immunol. 2013;25(1):97–102. doi:10.1016/j.coi.2012.11.00223452840
  • Rashid T, Ebringer A. Ankylosing spondylitis is linked to Klebsiella – the evidence. Clin Rheumatol. 2007;26(6):858–864. doi:10.1007/s10067-006-0488-717186116
  • Li Y, Wang P, Xie Z, et al. Whole genome expression profiling and signal pathway screening of MSCs in ankylosing spondylitis. Stem Cells Int. 2014;2014.
  • Sheehan NJ. The ramifications of HLA-B27. J R Soc Med. 2004;97(1):10–14. doi:10.1258/jrsm.97.1.1014702356
  • Benjamin R, Parham P. Guilt by association: HLA-B27 and ankylosing spondylitis. Immunol Today. 1990;11:137–142.2187471
  • Ebringer R, Cooke D, Cawdell DR, Cowling P, Ebringer A. Ankylosing Spondylitis: Klebsiella and HL-A B27. Rheumatol Rehabil 1977;16(3):190–196.
  • Eastmond C, Willshaw HE, Burgess SE, Shinebaum R, Cooke EM, Wright V. Frequency of faecal Klebsiella aerogenes in patients with ankylosing spondylitis and controls with respect to individual features of the disease. Ann Rheum Dis. 1980;39(2):118–123. doi:10.1136/ard.39.2.1186992718
  • Hunter T, Harding GKM, Kaprove RE, Schroeder M-L. Fecal carriage of various Klebsiella and Enterobacter species in patients with active ankylosing spondylitis. Arthritis Rheum. 1981;24(1):106–108. doi:10.1002/(ISSN)1529-01317470165
  • Morse HG, Morse HG, Rate RG, Bonnell MD. Increased recovery of Klebsiella from the gastrointestinal tract of Reiter’s syndrome and ankylosing spondylitis patients. Rheumatology. 1983;XXII(suppl_2):85–90. doi:10.1093/rheumatology/XXII.suppl_2.85
  • Cuvelier C, Veys EM, Cuvelier C, de Vos M. Ileocolonoscopic findings in seronegative spondylarthropathies. Rheumatology. 1988;XXVII(suppl_2):95–105. doi:10.1093/rheumatology/XXVII.suppl_2.95
  • Martínez-González O, Cantero-Hinojosa J, Paule-Sastre P, Gómez-Magán JC, Salvatierra-Ríos D.> Intestinal permeability in patients with ankylosing spondylitis and their healthy relatives. Rheumatology. 1994;33(7):644–647. doi:10.1093/rheumatology/33.7.644
  • Tam L-S, Gu J, Yu D. Pathogenesis of ankylosing spondylitis. Nature Rev Rheumatol. 2010;6:399. doi:10.1038/nrrheum.2010.7920517295
  • McGonagle D, Gibbon W, Emery P. Classification of inflammatory arthritis by enthesitis. Lancet. 1998;352(9134):1137–1140. doi:10.1016/S0140-6736(97)12004-99798608
  • Martin TM, Smith JR, Rosenbaum JT. Anterior uveitis: current concepts of pathogenesis and interactions with the spondyloarthropathies. Curr Opin Rheumatol. 2002;14(4):337–341.12118164
  • van der Heijde D, Dijkmans B, Geusens P, et al. Efficacy and safety of infliximab in patients with ankylosing spondylitis: results of a randomized, placebo‐controlled trial (ASSERT). Arthritis Rheum. 2005;52(2):582–591. doi:10.1002/(ISSN)1529-013115692973
  • Fleischmann RM, Iqbal I, Stern RL. Considerations with the use of biological therapy in the treatment of rheumatoid arthritis. Expert Opin Drug Saf. 2004;3(5):391–403. doi:10.1517/14740338.3.5.39115335295
  • Merkesdal S, Ruof J, Mittendorf T, Zeidler H. Cost-effectiveness of TNF-α-blocking agents in the treatment of rheumatoid arthritis. Expert Opin Pharmacother. 2004;5(9):1881–1886. doi:10.1517/14656566.5.9.188115330726
  • Reveille JD. 57 – Spondyloarthritis, in Clinical Immunology. Fifth Rich RR, et al., Editors, London: Content Repository Only; 2019:769–787.
  • Skalska U, Kozakiewicz A, Maśliński W, Jurkowska M. HLA-B27 detection – comparison of genetic sequence-based method and flow cytometry assay. Reumatologia. 2015;53(2):74–78. doi:10.5114/reum.2015.5150627407231
  • Khan MA. Polymorphism of HLA-B27: 105 subtypes currently known. Curr Rheumatol Rep. 2013;15(10):362. doi:10.1007/s11926-013-0362-y23990399
  • Rysnik O, McHugh K, van Duivenvoorde L, et al. Non-conventional forms of HLA-B27 are expressed in spondyloarthritis joints and gut tissue. J Autoimmun. 2016;70:12–21. doi:10.1016/j.jaut.2016.03.00927036372
  • Colbert RA, DeLay ML, Klenk EI, Layh-Schmitt G. From HLA-B27 to spondyloarthritis: a journey through the ER. Immunol Rev. 2010;233(1):181–202. doi:10.1111/j.0105-2896.2009.00865.x20193000
  • Guiliano DB, Fussell H, Lenart I, et al. Endoplasmic reticulum degradation-enhancing α-mannosidase-like protein 1 targets misfolded HLA–B27 dimers for endoplasmic reticulum-associated degradation. Arthritis Rheumatol. 2014;66(11):2976–2988. doi:10.1002/art.3880925132672
  • Bowness P, Ridley A, Shaw J, et al. Th17 cells expressing KIR3DL2+ and responsive to HLA-B27 homodimers are increased in ankylosing spondylitis. J Immunol. 2011;186(4):2672–2680. doi:10.4049/jimmunol.100265321248258
  • Ridley A, Hatano H, Wong-Baeza I, et al. Activation-induced killer cell immunoglobulin-like receptor 3DL2 binding to HLA–B27 licenses pathogenic T cell differentiation in spondyloarthritis. Arthritis Rheumatol. 2016;68(4):901–914. doi:10.1002/art.3951526841353
  • The Australo-Anglo-American Spondyloarthritis, C., et al.Genome-wide association study of ankylosing spondylitis identifies non-MHC susceptibility loci.Nat Genet.2010;42(p):123. doi:10.1038/ng.51320062062
  • Saveanu L, Carroll O, Lindo V, et al. Concerted peptide trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic reticulum. Nat Immunol. 2005;6:689. doi:10.1038/ni120815908954
  • Chang S-C, Momburg F, Bhutani N, Goldberg AL. The ER aminopeptidase, ERAP1, trims precursors to lengths of MHC class I peptides by a “molecular ruler” mechanism. Proc Natl Acad Sci U S A. 2005;102(47):17107–17112. doi:10.1073/pnas.050072110216286653
  • Chen L, Ridley A, Hammitzsch A, et al. Silencing or inhibition of endoplasmic reticulum aminopeptidase 1 (ERAP1) suppresses free heavy chain expression and Th17 responses in ankylosing spondylitis. Ann Rheum Dis. 2016;75(5):916–923. doi:10.1136/annrheumdis-2014-20699626130142
  • García-Medel N, Sanz-Bravo A, Van Nguyen D, et al. Functional interaction of the ankylosing spondylitis-associated endoplasmic reticulum aminopeptidase 1 polymorphism and HLA-B27 in vivo. Mol Cell Proteomics. 2012;11(11):1416–1429. doi:10.1074/mcp.M112.01958822918227
  • Wendling D, Cedoz J-P, Racadot E, Dumoulin G. Serum IL-17, BMP-7, and bone turnover markers in patients with ankylosing spondylitis. Joint Bone Spine. 2007;74:304–305. doi:10.1016/j.jbspin.2006.11.00517369068
  • Mei Y, Pan F, Gao J, et al. Increased serum IL-17 and IL-23 in the patient with ankylosing spondylitis. Clin Rheumatol. 2011;30(2):269–273. doi:10.1007/s10067-010-1647-421161669
  • Gracey E, Yao Y, Green B, et al. Sexual dimorphism in the Th17 signature of ankylosing spondylitis. Arthritis Rheumatol. 2016;68(3):679–689. doi:10.1002/art.3946426473967
  • Turvey SE, Broide DH. Innate immunity. J Allergy Clin Immunol. 2010;125(2, Supplement 2)):S24–S32. doi:10.1016/j.jaci.2009.07.01619932920
  • Robinson PC, Brown MA. Genetics of ankylosing spondylitis. Mol Immunol. 2014;57(1):2–11. doi:10.1016/j.molimm.2013.06.01323916070
  • Palomo J, Dietrich D, Martin P, Palmer G, Gabay C. The interleukin (IL)-1 cytokine family – balance between agonists and antagonists in inflammatory diseases. Cytokine. 2015;76(1):25–37. doi:10.1016/j.cyto.2015.06.01726185894
  • Loo FAJVD, Joosten LAB, Van Lent PLEM, Arntz OJ, Van Den Berg WB. Role of interleukin-1, tumor necrosis factor α, and interleukin-6 in cartilage proteoglycan metabolism and destruction effect of in situ blocking in murine antigen- and zymosan-induced arthritis. Arthritis Rheum. 1995;38(2):164–172. doi:10.1002/(ISSN)1529-01317848306
  • DANIS VA, MILLINGTON M, HYLAND VJ, GRENNAN D. Cytokine production by normal human monocytes: inter-subject variation and relationship to an IL-1 receptor antagonist (IL-1Ra) gene polymorphism. Clinical & Experimental Immunology. 1995;99(2):303–310. doi:10.1111/(ISSN)1365-22497851026
  • Vassalli P. The pathophysiology of tumor necrosis factors. Annu Rev Immunol. 1992;10(1):411–452. doi:10.1146/annurev.iy.10.040192.0022111590993
  • Braun J, Bollow M, Neure L, et al. Use of immunohistologic and in situ hybridization techniques in the examination of sacroiliac joint biopsy specimens from patients with ankylosing spondylitis. Arthritis Rheum. 1995;38(4):499–505. doi:10.1002/(ISSN)1529-01317718003
  • Duivenvoorde LMV, Ambarus CA, Masdar H, et al. A2.15 relative overexpression of transmembrane versus soluble TNF in human and experimental spondyloarthritis. Ann Rheum Dis. 2013;72(Suppl 1):A9–A10. doi:10.1136/annrheumdis-2013-203215.15
  • Campbell RD, Trowsdale J. Map of the human MHC. Immunol Today. 1993;14(7):349–352. doi:10.1016/0167-5699(93)90234-C8363724
  • Wilson AG, Symons JA, McDowell TL, McDevitt HO, Duff GW. Effects of a polymorphism in the human tumor necrosis factor alpha promoter on transcriptional activation. Proc Natl Acad Sci U S A. 1997;94(7):3195–3199. doi:10.1073/pnas.94.7.31959096369
  • El-Tahan RR, Ghoneim AM, El-Mashad N. TNF-α gene polymorphisms and expression. SpringerPlus. 2016;5(1):1508. doi:10.1186/s40064-016-3197-y27652081
  • Mahmoudi M, Jamshidi AR, Amirzargar AA, et al. Association between endoplasmic reticulum aminopeptidase-1 (ERAP-1) and susceptibility to ankylosing spondylitis in Iran. Iran J Allergy Asthma Immunol. 2012;11(4);294–300.23264405
  • Bettencourt BF, Rocha FL, Alves H, et al. Protective effect of an ERAP1 haplotype in ankylosing spondylitis: investigating non-MHC genes in HLA-B27-positive individuals. Rheumatology. 2013;52(12):2168–2176. doi:10.1093/rheumatology/ket26924046467
  • Haroon N, Tsui FWL, Chiu B, Tsui HW, Inman RD. Serum cytokine receptors in ankylosing spondylitis: relationship to inflammatory markers and endoplasmic reticulum aminopeptidase polymorphisms. J Rheumatol. 2010;37(9):1907–1910. doi:10.3899/jrheum.10001920595269
  • Hacquard‐Bouder C, Falgarone G, Bosquet A, et al. Defective costimulatory function is a striking feature of antigen‐presenting cells in an HLA-B27-transgenic rat model of spondylarthropathy. Arthritis Rheum. 2004;50(5):1624–1635. doi:10.1002/(ISSN)1529-013115146433
  • Hacquard‐Bouder C, Chimenti M-S, Giquel B, et al. Alteration of antigen‐independent immunologic synapse formation between dendritic cells from HLA-B27-transgenic rats and CD4+ T cells: selective impairment of costimulatory molecule engagement by mature HLA–B27. Arthritis Rheum. 2007;56(5):1478–1489. doi:10.1002/(ISSN)1529-013117469106
  • Glatigny S, Fert I, Blaton MA, et al. Proinflammatory Th17 cells are expanded and induced by dendritic cells in spondylarthritis‐prone HLA-B27-transgenic rats. Arthritis Rheum. 2012;64(1):110–120. doi:10.1002/art.3332121905004
  • Pan F, Barbi J, Pardoll DM. Hypoxia-inducible factor 1: a link between metabolism and T cell differentiation and a potential therapeutic target. Oncoimmunology. 2012;1(4):510–515. doi:10.4161/onci.1945722754770
  • Veldhoen M, Hirota K, Westendorf AM, et al. The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature. 2008;453:106. doi:10.1038/nature0688118362914
  • Bluestone JA, Mackay CR, O’Shea JJ, Stockinger B. The functional plasticity of T cell subsets. Nature Rev Immunol. 2009;9:811. doi:10.1038/nri265419809471
  • El-Behi M, Ciric B, Dai H, et al. The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol. 2011;12(6):568–575. doi:10.1038/ni.203121516111
  • Rudwaleit M, Höhler T. Cytokine gene polymorphisms relevant for the spondyloarthropathies. Curr Opin Rheumatol. 2001;13(4):250–254.11555724
  • Benjamin M, McGONAGLE D. Basic concepts of enthesis biology and immunology. J Rheumatol Suppl. 2009;83:12–13. doi:10.3899/jrheum.09021119661528
  • Benjamin M, Toumi H, Suzuki D, Redman S, Emery P, McGonagle D. Microdamage and altered vascularity at the enthesis–bone interface provides an anatomic explanation for bone involvement in the HLA-B27-associated spondylarthritides and allied disorders. Arthritis Rheum. 2007;56(1):224–233. doi:10.1002/(ISSN)1529-013117195226
  • Costello M-E, Robinson PC, Benham H, Brown MA. The intestinal microbiome in human disease and how it relates to arthritis and spondyloarthritis. Best Pract Res Clin Rheumatol. 2015;29(2):202–212. doi:10.1016/j.berh.2015.08.00126362739
  • Khan MA, van Der Linden SM, Kushner I, Valkenburg HA, Cats A. Spondylitic disease without radiologic evidence of sacroiliitis in relatives of HLA‐B27 positive ankylosing spondylitis patients. Arthritis Rheum. 1985;28(1):40–43. doi:10.1002/(ISSN)1529-01313871329
  • McVeigh CM, Cairns AP. Diagnosis and management of ankylosing spondylitis. BMJ. 2006;333(7568):581–585. doi:10.1136/bmj.38954.689583.DE16974012
  • Sieper J, Braun J, Rudwaleit M, Boonen A, Zink A. Ankylosing spondylitis: an overview. Ann Rheum Dis. 2002;61(suppl 3):iii8–iii18. doi:10.1136/ard.61.suppl_3.iii812381506
  • Spoorenberg A, Dougados M, Mielants H. Relative value of erythrocyte sedimentation rate and C-reactive protein in assessment of disease activity in ankylosing spondylitis. J Rheumatol. 1999;26(4):980–984.10229432
  • Ruof J, Stucki G. Validity aspects of erythrocyte sedimentation rate and C-reactive protein in ankylosing spondylitis: a literature review. J Rheumatol. 1999;26(4):966–970.10229429
  • Underwood M, Dawes P. Inflammatory back pain in primary care. Rheumatology. 1995;34(11):1074–1077. doi:10.1093/rheumatology/34.11.1074
  • Dougados M, Linden SVD, Juhlin R, et al. The European spondylarthropathy study group preliminary criteria for the classification of spondylarthropathy. Arthritis Rheum. 1991;34(10):1218–1227. doi:10.1002/(ISSN)1529-01311930310
  • Wong RS. Disease-modifying effects of long-term and continuous use of nonsteroidal anti-inflammatory drugs (NSAIDs) in Spondyloarthritis. Adv Pharmacol Sci. 2019;2019.
  • Nunes GPS,  Cunha PDS, Bosco DPD, Ribeiro SLE. Challenging management of hepatitis B infection in ankylosing spondylitis patients in an endemic area during immunosuppressive therapy. Rev Soc Bras Med Trop. 2019;52.
  • Kroon FP, van der Burg LRA, Ramiro S, et al. Nonsteroidal antiinflammatory drugs for axial spondyloarthritis: a cochrane review. J Rheumatol. 2016;43(3):607–617. doi:10.3899/jrheum.15072126834216
  • Evans DM, Spencer CCA, Pointon JJ, et al. Interaction between ERAP1 and HLA-B27 in ankylosing spondylitis implicates peptide handling in the mechanism for HLA-B27 in disease susceptibility. Nat Genet. 2011;43(8):761. doi:10.1038/ng.87321743469
  • Dougados M, Béhier J-M, Jolchine I, et al. Efficacy of celecoxib, a cyclooxygenase 2–specific inhibitor, in the treatment of ankylosing spondylitis: a six‐week controlled study with comparison against placebo and against a conventional nonsteroidal antiinflammatory drug. Arthritis Rheum. 2001;44(1):180–185. doi:10.1002/(ISSN)1529-013111212158
  • Coxib and traditional NSAID Trialists (CNT) Collaboration, Bhala N, et al. Vascular and Upper Gastrointestinal Effects of Non-steroidal Anti-inflammatory Drugs: Meta-analyses of Individual Participant Data from Randomised Trials. Lancet 2013 Aug 31;382(9894):769–779. doi:10.1016/S0140-6736(13)60900-9.
  • Dougados M, Gueguen A, Nakache JP, et al. Ankylosing spondylitis: what is the optimum duration of a clinical study? A one year versus a 6 weeks non-steroidal anti-inflammatory drug trial. Rheumatology (Oxford). 1999;38(3):235–244. doi:10.1093/rheumatology/38.3.23510325662
  • Wanders A, Heijde DVD, Landewé R, et al. Nonsteroidal antiinflammatory drugs reduce radiographic progression in patients with ankylosing spondylitis: a randomized clinical trial. Arthritis Rheum. 2005;52(6):1756–1765. doi:10.1002/(ISSN)1529-013115934081
  • Sieper J, Listing J, Poddubnyy D, et al. Effect of continuous versus on-demand treatment of ankylosing spondylitis with diclofenac over 2 years on radiographic progression of the spine: results from a randomised multicentre trial (ENRADAS). Ann Rheum Dis. 2016;75(8):1438–1443. doi:10.1136/annrheumdis-2015-20789726242443
  • Moon K-H, Kim Y-T. Medical treatment of ankylosing spondylitis. Hip Pelvis. 2014;26(3):129–135. doi:10.5371/hp.2014.26.3.12927536570
  • Haroon NN, Paterson JM, Li P, Inman RD, Haroon N. Patients with ankylosing spondylitis have increased cardiovascular and cerebrovascular mortality: a population-based study. Ann Intern Med. 2015;163(6):409–416. doi:10.7326/M14-247026258401
  • Rademacher J, Poddubnyy D. Emerging drugs for the treatment of axial spondyloarthritis. Expert Opin Emerg Drugs. 2018;23(1):83–96. doi:10.1080/14728214.2018.144571929475394
  • Fattahi MJ, Jamshidi AR, Mahmoudi M, et al. Evaluation of the efficacy and safety of β-d-mannuronic acid in patients with ankylosing spondylitis: a 12-week randomized, placebo-controlled, phase I/II clinical trial. Int Immunopharmacol. 2018;54:112–117. doi:10.1016/j.intimp.2017.11.00329127910
  • Emkey R, Rosenthal N, Wu S-C, Jordan D, Kamin M. Efficacy and safety of tramadol/acetaminophen tablets (Ultracet) as add-on therapy for osteoarthritis pain in subjects receiving a COX-2 nonsteroidal antiinflammatory drug: a multicenter, randomized, double-blind, placebo-controlled trial. J Rheumatol. 2004;31(1):150–156.14705234
  • Peloso PM, Fortin L, Beaulieu A, Kamin M, Rosenthal N. Analgesic efficacy and safety of tramadol/acetaminophen combination tablets (Ultracet) in treatment of chronic low back pain: a multicenter, outpatient, randomized, double blind, placebo controlled trial. J Rheumatol. 2004;31(12):2454–2463.15570651
  • Bennett RM, Kamin M, Karim R, Rosenthal N. Tramadol and acetaminophen combination tablets in the treatment of fibromyalgia pain: a double-blind, randomized, placebo-controlled study. Am J Med. 2003;114(7):537–545. doi:10.1016/s0002-9343(03)00116-512753877
  • Chang J-K, Yu C-T, Lee M-Y, et al. Tramadol/acetaminophen combination as add-on therapy in the treatment of patients with ankylosing spondylitis. Clin Rheumatol. 2013;32(3):341–347. doi:10.1007/s10067-012-2125-y23192419
  • Farrouq Mahmood PH. Ankylosing spondylitis: a review. Eur Med J. 2017;2(4):134–139.
  • Ryall NH, Helliwell P. A critical review of ankylosing spondylitis. Crit Rev™ Phys Rehabil Med. 1998;10:3.
  • sailaja AK. An overall review on ankylosing spondylitis. Int J Current Pharm Sci. 2014;1(1):1–5.
  • Xie J, Fan Z, Li Y, et al. Design of pH-sensitive methotrexate prodrug-targeted curcumin nanoparticles for efficient dual-drug delivery and combination cancer therapy. Int J Nanomedicine. 2018;13:1381–1398. doi:10.2147/IJN.S15231229563794
  • Nosrati H, Salehiabar M, Davaran S, Danafar H, Manjili HK. Methotrexate-conjugated L-lysine coated iron oxide magnetic nanoparticles for inhibition of MCF-7 breast cancer cells. Drug Dev Ind Pharm. 2018;44(6):886–894. doi:10.1080/03639045.2017.1417422
  • Abdelrady H, Hathout RM, Osman R, Saleem I, Mortada ND. Exploiting gelatin nanocarriers in the pulmonary delivery of methotrexate for lung cancer therapy. EurJ Pharm Sci. 2019;133:115–126. doi:10.1016/j.ejps.2019.03.01630905615
  • Haibel H, Brandt HC, Song IH, et al. No efficacy of subcutaneous methotrexate in active ankylosing spondylitis: a 16-week open-label trial. Ann Rheum Dis. 2007;66(3):419–421. doi:10.1136/ard.2006.05409816901959
  • Chen J, Lin S, Liu C. Sulfasalazine for ankylosing spondylitis. Cochrane Database Syst Rev. 2014;11.
  • Xue H-X, Fu W-Y, Cui H-D, Yang -L-L, Zhang N, Zhao L-J. High-dose thalidomide increases the risk of peripheral neuropathy in the treatment of ankylosing spondylitis. Neural Regener Res. 2015;10(5):814–818. doi:10.4103/1673-5374.156988
  • Wei J-C-C, Chan TW, Lin H-S, Huang F, Chou C-T. Thalidomide for severe refractory ankylosing spondylitis: a 6-month open-label trial. J Rheumatol. 2003;30(12):2627–2631.14719205
  • Wilsdon TD, Whittle SL, Thynne TRJ, Mangoni AA. Methotrexate for psoriatic arthritis. Cochrane Database Syst Rev. 2019;1.
  • Huang F, Gu J, Zhao W, Zhu J, Zhang J, Yu DTY. One-year open-label trial of thalidomide in ankylosing spondylitis. Arthritis Care Res (Hoboken). 2002;47(3):249–254. doi:10.1002/(ISSN)1529-0131
  • Nabeel AI, Moawed FSM, Hassan H. Immunomodulatory effect of new quinolone derivative against cisplatin/gamma radiation-induced renal and brain toxicity in mice. J Photochem Photobiol B. 2018;184:54–60. doi:10.1016/j.jphotobiol.2018.05.01329803073
  • Öğrendik M. Treatment of Ankylosing Spondylitis with Moxifloxacin. South Med Assoc. 2007;100(4):366–370. doi:10.1097/SMJ.0b013e31802fa2a8
  • Öğrendik M. Treatment of ankylosing spondylitis with co-amoxiclav. Clin Exp Pharmacol Physiol. 2018;45(7):742–744. doi:10.1111/1440-1681.1295229676028
  • Taurog JD, Chhabra A, Colbert RA. Ankylosing spondylitis and axial spondyloarthritis. N Engl J Med. 2016;374(26):2563–2574. doi:10.1056/NEJMra140618227355535
  • Gratacós J, Collado A, Filella X, et al.. Serum cytokines (IL-6, TNF-α, IL-1β and IFN-γ) in ankylosing spondylitis: a close correlation between serum IL-6 and disease activity and severity. Rheumatology. 1994;33(10):927–931. doi:10.1093/rheumatology/33.10.927
  • Baraliakos X, et al. Persistent clinical efficacy and excellent safety over 5 years of treatment with the anti-TNF alpha agent infliximab in patients with ankylosing spondylitis In: Arthritis and Rheumatism. Hoboken, NJ: Wiley-Liss Div John Wiley & Sons Inc; 2006. doi:10.1136/ard.2007.075879
  • Braun J, Deodhar A, Dijkmans B, et al. Efficacy and safety of infliximab in patients with ankylosing spondylitis over a two‐year period. Arthritis Care Res. 2008;59(9):1270–1278. doi:10.1002/art.v59:9
  • Van der Heijde D, et al. Long-term adalimumab treatment reduces signs and symptoms in ankylosing spondylitis (AS) patients – results from the ATLAS trial In: Leiden University Medical Center, editor. Annals of the Rheumatic Diseases. London, UK: BMJ Publishing Group British Med Assoc House; 2006.
  • Braun J, Landewé R, Hermann K-GA, et al. Major reduction in spinal inflammation in patients with ankylosing spondylitis after treatment with infliximab: results of a multicenter, randomized, double-blind, placebo-controlled magnetic resonance imaging study. Arthritis Rheum. 2006;54(5):1646–1652. doi:10.1002/(ISSN)1529-013116646033
  • Baraliakos X, Listing J, Rudwaleit M, Brandt J, Sieper J, Braun J. Radiographic progression in patients with ankylosing spondylitis after 2 years of treatment with the tumour necrosis factor α antibody infliximab. Ann Rheum Dis. 2005;64(10):1462–1466. doi:10.1136/ard.2004.03347215778240
  • Haibel H, Baraliakos X, Listing J, et al. Persistent reduction of spinal inflammation as assessed by magnetic resonance imaging in patients with ankylosing spondylitis after 2 yrs of treatment with the anti-tumour necrosis factor agent infliximab. Rheumatology. 2005;44(12):1525–1530. doi:10.1093/rheumatology/kei04616091396
  • Davis JC Jr, Van Der Heijde D, Braun J, et al. Recombinant human tumor necrosis factor receptor (etanercept) for treating ankylosing spondylitis: a randomized, controlled trial. Arthritis Rheum. 2003;48(11):3230–3236. doi:10.1002/(ISSN)1529-013114613288
  • Rudwaleit M, Baeten D. Ankylosing spondylitis and bowel disease. Best Pract Res Clin Rheumatol. 2006;20(3):451–471. doi:10.1016/j.berh.2006.03.01016777576
  • Voulgari PV, Venetsanopoulou AI, Epagelis EK, Alamanos Y, Takalou I, Drosos AA. Infliximab in refractory psoriatic arthritis with severe psoriasis: a 2-year experience. Ann Rheum Dis. 2007;66(2):270–271. doi:10.1136/ard.2006.05873517068063
  • Hanauer SB, Sandborn WJ, Rutgeerts P, et al. Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn’s disease: the CLASSIC-I trial. Gastroenterology. 2006;130(2):323–333. doi:10.1053/j.gastro.2005.11.03016472588
  • Markham A, Lamb HM. Infliximab. Drugs. 2000;59(6):1341–1359. doi:10.2165/00003495-200059060-0001010882166
  • Cook DA. J.B.D., D.C. Gallagher. Etanercept Rheumatoid Arthritis Drugs R & D. 1999;1(1):75–77.
  • Toussirot É, Wendling D. Recent progress in ankylosing spondylitis treatment. Expert Opin Pharmacother. 2003;4(1):1–12. doi:10.1517/14656566.4.1.112517239
  • Park W, Yoo DH, Miranda P, et al. Efficacy and safety of switching from reference infliximab to CT-P13 compared with maintenance of CT-P13 in ankylosing spondylitis: 102-week data from the PLANETAS extension study. Ann Rheum Dis. 2017;76(2):346–354. doi:10.1136/annrheumdis-2015-20878327117698
  • Yoo DH, Prodanovic N, Jaworski J, et al. Efficacy and safety of CT-P13 (biosimilar infliximab) in patients with rheumatoid arthritis: comparison between switching from reference infliximab to CT-P13 and continuing CT-P13 in the PLANETRA extension study. Ann Rheum Dis. 2017;76(2):355–363. doi:10.1136/annrheumdis-2015-20878627130908
  • Rudwaleit M, Listing J, Brandt J, Braun J, Sieper J. Prediction of a major clinical response (BASDAI 50) to tumour necrosis factor α blockers in ankylosing spondylitis. Ann Rheum Dis. 2004;63(6):665–670. doi:10.1136/ard.2003.01638615037444
  • Liu W, Wu Y-H, Zhang L, et al. Elevated serum levels of IL-6 and IL-17 may associate with the development of ankylosing spondylitis. Int J Clin Exp Med. 2015;8(10):17362–17376.26770328
  • Braun J, Baraliakos X, Deodhar A, et al. Effect of secukinumab on clinical and radiographic outcomes in ankylosing spondylitis: 2-year results from the randomised phase III MEASURE 1 study. Ann Rheum Dis. 2017;76(6):1070–1077. doi:10.1136/annrheumdis-2016-20973027965257
  • Yago T, Nanke Y, Kawamoto M, Kobashigawa T, Yamanaka H, Kotake S. IL-23 and Th17 disease in inflammatory arthritis. J Clin Med. 2017;6:9.
  • Baraliakos X, Kivitz AJ, Deodhar AA, et al. Long-term effects of interleukin-17A inhibition with secukinumab in active ankylosing spondylitis: 3-year efficacy and safety results from an extension of the Phase 3 MEASURE 1 trial. Clin Exp Rheumatol. 2018;36(1):50–55.
  • Marzo-Ortega H, Sieper J, Kivitz A, et al. Secukinumab and sustained improvement in signs and symptoms of patients with active Ankylosing Spondylitis through two years: results from a phase III study. Arthritis Care Res (Hoboken). 2017;69(7):1020–1029. doi:10.1002/acr.2323328235249
  • Pavelka K, Kivitz A, Dokoupilova E, et al. Efficacy, safety, and tolerability of secukinumab in patients with active ankylosing spondylitis: a randomized, double-blind phase 3 study, MEASURE 3. Arthritis Res Ther. 2017;19(1):285. doi:10.1186/s13075-017-1490-y29273067
  • El Miedany Y, Youssef S, Ahmed I, El Gaafary M. The gastrointestinal safety and effect on disease activity of etoricoxib, a selective cox-2 inhibitor in inflammatory bowel diseases. Am J Gastroenterol. 2006;101(2):311. doi:10.1111/ajg.2006.101.issue-216454836
  • Sherlock JP, Joyce-Shaikh B, Turner SP, et al. IL-23 induces spondyloarthropathy by acting on ROR-gammat+ CD3+CD4-CD8- entheseal resident T cells. Nat Med. 2012;18(7):1069–1076. doi:10.1038/nm.281722772566
  • McInnes IB, Kavanaugh A, Gottlieb AB, et al. Efficacy and safety of ustekinumab in patients with active psoriatic arthritis: 1 year results of the phase 3, multicentre, double-blind, placebo-controlled PSUMMIT 1 trial. Lancet. 2013;382(9894):780–789. doi:10.1016/S0140-6736(13)60594-223769296
  • Poddubnyy D, Hermann K-GA, Callhoff J, Listing J, Sieper J. Ustekinumab for the treatment of patients with active ankylosing spondylitis: results of a 28-week, prospective, open-label, proof-of-concept study (TOPAS). Ann Rheum Dis. 2014;73(5):817–823. doi:10.1136/annrheumdis-2013-20424824389297
  • Fleischmann RM, Wagner F, Kivitz AJ, et al. Safety, tolerability, and pharmacodynamics of ABT-122, a tumor necrosis factor- and interleukin-17-targeted dual variable domain immunoglobulin, in patients with rheumatoid arthritis. Arthritis Rheumatol. 2017;69(12):2283–2291. doi:10.1002/art.4031928941216
  • Genovese MC, Weinblatt ME, Mease PJ, et al. Dual inhibition of tumour necrosis factor and interleukin-17A with ABT-122: open-label long-term extension studies in rheumatoid arthritis or psoriatic arthritis. Rheumatology (Oxford). 2018;57(11):1972–1981. doi:10.1093/rheumatology/key17330032191
  • Sieper J, Braun J, Kay J, et al. Sarilumab for the treatment of ankylosing spondylitis: results of a Phase II, randomised, double-blind, placebo-controlled study (ALIGN). Ann Rheum Dis. 2015;74(6):1051–1057. doi:10.1136/annrheumdis-2013-20496324550171
  • 6 daily stretches for ankylosing spondylitis that can help ease pain. Available from: https://creakyjoints.org/diet-exercise/ankylosing-spondylitis-stretches-exercises/ Accessed September 4, 2019.
  • American College of Rheumatology Releases New Ankylosing Spondylitis & Non-Radiographic Axial Spondyloarthritis Treatment Recommendations; 2015 Available from: https://www.rheumatology.org/About-Us/Newsroom/Press-Releases/ID/679 Accessed September 4, 2019.
  • Versus Arthritis. What is ankylosing spondylitis (AS)?. 2015 Available from: https://www.versusarthritis.org/about-arthritis/conditions/ankylosing-spondylitis/. Accessed September 4, 2019.
  • Goie The HS, Steven MM, van der Linden SM, Cats A. Evaluation of diagnostic criteria for ankylosing spondylitis: a comparison of the Rome, New York and modified New York criteria in patients with a positive clinical history screening test for ankylosing spondylitis. Br J Rheumatol. 1985;24(3):242–249. doi:10.1093/rheumatology/24.3.2423160423
  • Jacobs WB, Fehlings MG. Ankylosing spondylitis and spinal cord injury: origin, incidence, management, and avoidance. Neurosurg Focus. 2008;24(1):E12. doi:10.3171/FOC/2008/24/1/E12
  • Bron JL, de Vries MK, Snieders MN, van der Horst-Bruinsma IE, van Royen BJ. Discovertebral (Andersson) lesions of the spine in ankylosing spondylitis revisited. Clin Rheumatol. 2009;28(8):883–892. doi:10.1007/s10067-009-1151-x19294478
  • Britto NMF, Renor BS, Ghizoni E, Tedeschi H, Joaquim AF. Spine surgery in patients with ankylosing spondylitis. Rev Assoc Med Bras 2018;64(4):379–383. doi:10.1590/1806-9282.64.04.379
  • Lu MC, Koo M, Lai NS. Incident spine surgery in patients with ankylosing spondylitis: a secondary cohort analysis of a nationwide, population-based health claims database. Arthritis Care Res (Hoboken). 2018;70(9):1416–1420. doi:10.1002/acr.2347829193835
  • Kurucan E, Bernstein DN, Mesfin A. Surgical management of spinal fractures in ankylosing spondylitis. J Spine Surg. 2018;4(3):501–508. doi:10.21037/jss.2018.06.1530547111
  • Mountziaris PM, Kramer PR, Mikos AG. Emerging intra-articular drug delivery systems for the temporomandibular joint. Methods. 2009;47(2):134–140. doi:10.1016/j.ymeth.2008.09.00118835358
  • Leone G, Fini M, Torricelli P, Giardino R, Barbucci R. An amidated carboxymethylcellulose hydrogel for cartilage regeneration. J Mater Sci. 2008;19(8):2873–2880. doi:10.1007/s10856-008-3412-7
  • Huang Y-C, Tsai S-K, Huang C-H, et al. Intravenous tenoxicam reduces uterine cramps after Cesarean delivery. Can J Anesthesia. 2002;49(4):384–387. doi:10.1007/BF03017327
  • Mohammad IS, He W, Yin L. A smart paclitaxel-disulfiram nanococrystals for efficient MDR reversal and enhanced apoptosis. Pharm Res. 2018;35(4):77. doi:10.1007/s11095-018-2370-029488114
  • Mohammad IS, He W, Yin L. Understanding of human ATP binding cassette superfamily and novel multidrug resistance modulators to overcome MDR. Biomed Pharmacother. 2018;100:335–348. doi:10.1016/j.biopha.2018.02.03829453043
  • Mohammad IS, Hu H, Yin L, He W. Drug nanocrystals: fabrication methods and promising therapeutic applications. Int J Pharm. 2019;562:187–202. doi:10.1016/j.ijpharm.2019.02.04530851386
  • Mohammad IS, Teng C, Chaurasiya B, Yin L, Wu C, He W. Drug-delivering-drug approach-based codelivery of paclitaxel and disulfiram for treating multidrug-resistant cancer. Int J Pharm. 2019;557:304–313. doi:10.1016/j.ijpharm.2018.12.06730599232
  • DiSanto RM, Subramanian V, Gu Z. Recent advances in nanotechnology for diabetes treatment. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2015;7(4):548–564. doi:10.1002/wnan.132925641955
  • Johnson BM, Junkins RD, Ting JPY. A spike in inflammation. Nat Nanotechnol. 2018;13(11):980–981. doi:10.1038/s41565-018-0292-y30374158
  • Guan S, Munder A, Hedtfeld S, et al. Self-assembled peptide–poloxamine nanoparticles enable in vitro and in vivo genome restoration for cystic fibrosis. Nat Nanotechnol. 2019;14(3):287–297. doi:10.1038/s41565-018-0358-x30692673
  • Chevalier X. Intraarticular treatments for osteoarthritis: new perspectives. Curr Drug Targets. 2010;11(5):546–560.20199394
  • Bhardwaj U, Burgess DJ. Physicochemical properties of extruded and non-extruded liposomes containing the hydrophobic drug dexamethasone. Int J Pharm. 2010;388(1):181–189. doi:10.1016/j.ijpharm.2010.01.00320079409
  • Zhang Z, Huang G. Micro- and nano-carrier mediated intra-articular drug delivery systems for the treatment of osteoarthritis. J Nanotechnol. 2012;2012.
  • Elron-Gross I, Glucksam Y, Margalit R. Liposomal dexamethasone–diclofenac combinations for local osteoarthritis treatment. Int J Pharm. 2009;376(1):84–91. doi:10.1016/j.ijpharm.2009.04.02519409466
  • Srinath P, Vyas SP, Diwan PV. Preparation and pharmacodynamic evaluation of liposomes of indomethacin. Drug Dev Ind Pharm. 2000;26(3):313–321. doi:10.1081/DDC-10010035910738648
  • Gottschalk O, Metz P, Dao Trong ML, et al. Therapeutic effect of methotrexate encapsulated in cationic liposomes (EndoMTX) in comparison to free methotrexate in an antigen-induced arthritis study in vivo. Scand J Rheumatol. 2015;44(6):456–463. doi:10.3109/03009742.2015.103044826114440
  • Alekseeva AA, Moiseeva E, Onishchenko N, et al. Liposomal formulation of a methotrexate lipophilic prodrug: assessment in tumor cells and mouse T-cell leukemic lymphoma. Int J Nanomedicine. 2017;12:3735. doi:10.2147/IJN28553111
  • Gaidukevich SK, Mikulovich YL, Smirnova TG, et al. Antibacterial effects of liposomes containing phospholipid cardiolipin and fluoroquinolone levofloxacin on mycobacterium tuberculosis with extensive drug resistance. Bull Exp Biol Med. 2016;160(5):675–678. doi:10.1007/s10517-016-3247-z27021087
  • Vázquez JL, Montero MT, Trias J, Hernàndez-Borrell J. 6-Fluoroquinolone–liposome interactions: fluorescence quenching study using iodide. Int J Pharm. 1998;171(1):75–86. doi:10.1016/S0378-5173(98)00154-9
  • Grancelli A, Morros A, Cabañas ME, et al. Interaction of 6-fluoroquinolones with dipalmitoylphosphatidylcholine monolayers and liposomes. Langmuir. 2002;18(24):9177–9182. doi:10.1021/la025837h
  • Furneri PM, Fresta M, Puglisi G, Tempera G. Ofloxacin-loaded liposomes: in vitro activity and drug accumulation in bacteria. Antimicrob Agents Chemother. 2000;44(9):2458–2464. doi:10.1128/aac.44.9.2458-2464.200010952595
  • Ten Hagen TLM, Van Der Veen AH, Nooijen PT, Van Tiel ST, Seynhaeve AL, Eggermont AM. Low-dose tumor necrosis factor-α augments antitumor activity of stealth liposomal doxorubicin (DOXIL®) in soft tissue sarcoma-bearing rats. Int J Cancer. 2000;87(6):829–837. doi:10.1002/1097-0215(20000915)87:6<829::aid-ijc12>3.0.co;2-c10956394
  • Kim DW, Andres ML, Li J, et al. Liposome-encapsulated tumor necrosis factor-α enhances the effects of radiation against human colon tumor xenografts. J Interferon Cytokine Res. 2001;21(11):885–897. doi:10.1089/10799900175328949711747620
  • Kedar E, Palgi O, Golod G, Babai I, Barenholz Y. Delivery of cytokines by liposomes. III. Liposome-encapsulated GM-CSF and TNF-alpha show improved pharmacokinetics and biological activity and reduced toxicity in mice. J Immunother. 1997;20(3):180–193.9181456
  • Meka RR, Venkatesha SH, Moudgil KD. Peptide-directed liposomal delivery improves the therapeutic index of an immunomodulatory cytokine in controlling autoimmune arthritis. J Controlled Release. 2018;286:279–288. doi:10.1016/j.jconrel.2018.08.007
  • Lin S-Y, Chen KS, Teng HH, Li MJ. In vitro degradation and dissolution behaviours of microspheres prepared by three low molecular weight polyesters. J Microencapsul. 2000;17(5):577–586. doi:10.1080/02652040041763011038117
  • Tunçay M, Caliş S, Kaş HS, Ercan MT, Peksoy I, Hincal AA. Diclofenac sodium incorporated PLGA (50:50) microspheres: formulation considerations and in vitro/in vivo evaluation. Int J Pharm. 2000;195(1):179–188.10675695
  • Skoutakis VA, Carter CA, Mickle TR, et al. Review of diclofenac and evaluation of its place in therapy as a nonsteroidal antiinflammatory agent. Drug Intell Clin Pharm. 1988;22(11):850–859. doi:10.1177/1060028088022011023069424
  • Saravanan M, Bhaskar K, Maharajan G, Pillai KS. Development of gelatin microspheres loaded with diclofenac sodium for intra-articular administration. J Drug Target. 2011;19(2):96–103. doi:10.3109/1061186100373397920380621
  • Zhao J, Zhao M, Yu C, et al. Multifunctional folate receptor-targeting and pH-responsive nanocarriers loaded with methotrexate for treatment of rheumatoid arthritis. Int J Nanomedicine. 2017;12:6735. doi:10.2147/IJN28932117
  • Zhang H, Li X, Ding J, et al. Delivery of ursolic acid (UA) in polymeric nanoparticles effectively promotes the apoptosis of gastric cancer cells through enhanced inhibition of cyclooxygenase 2 (COX-2). Int J Pharm. 2013;441(1):261–268. doi:10.1016/j.ijpharm.2012.11.03423194884
  • Iqbal S, Du X, Wang J, Li H, Yuan Y, Wang J. Surface charge tunable nanoparticles for TNF-α siRNA oral delivery for treating ulcerative colitis. Nano Res. 2018;11(5):2872–2884. doi:10.1007/s12274-017-1918-3
  • Davis SM, Reichel D, Bae Y, Pennypacker KR. Leukemia inhibitory factor-loaded nanoparticles with enhanced cytokine metabolic stability and anti-inflammatory activity. Pharm Res. 2018;35(1):6. doi:10.1007/s11095-017-2282-429294201
  • Peppas N, Bures P, Leobandung W, Ichikawa H. Hydrogels in pharmaceutical formulations. Eur J Pharm Biopharm. 2000;50(1):27–46.10840191
  • Barbucci R, Lamponi S, Borzacchiello A, et al. Hyaluronic acid hydrogel in the treatment of osteoarthritis. Biomaterials. 2002;23(23):4503–4513. doi:10.1016/s0142-9612(02)00194-112322970
  • Hu X-B, Kang -R-R, Tang -T-T, et al. Topical delivery of 3,5,4′-trimethoxy-trans-stilbene-loaded microemulsion-based hydrogel for the treatment of osteoarthritis in a rabbit model. Drug Deliv Transl Res. 2019;9(1):357–365. doi:10.1007/s13346-018-00604-z30430453
  • Ahmad U, Sohail M, Ahmad M, et al. Chitosan based thermosensitive injectable hydrogels for controlled delivery of loxoprofen: development, characterization and in-vivo evaluation. Int J Biol Macromol. 2019;129:233–245. doi:10.1016/j.ijbiomac.2019.02.03130738157
  • Dong L, Xia S, Chen H, Chen J, Zhang J. Spleen-specific suppression of TNF-α by cationic hydrogel-delivered antisense nucleotides for the prevention of arthritis in animal models. Biomaterials. 2009;30(26):4416–4426. doi:10.1016/j.biomaterials.2009.04.04519481251
  • Robert M-C, Frenette M, Zhou C, et al. A drug delivery system for administration of anti-TNF-α antibody. Transl Vis Sci Technol. 2016;5(2):11. doi:10.1167/tvst.5.2.11
  • 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(5):255–263. doi:10.3109/10717544.2012.70096222775466
  • Ko J-Y, Choi Y-J, Jeong G-J, Im G-I. Sulforaphane–PLGA microspheres for the intra-articular treatment of osteoarthritis. Biomaterials. 2013;34(21):5359–5368. doi:10.1016/j.biomaterials.2013.03.06623601658
  • Bozdag S, Caliş S, Kaş HS, Ercan MT, Peksoy I, Hincal AA. In vitro evaluation and intra-articular administration of biodegradable microspheres containing naproxen sodium. J Microencapsul. 2001;18(4):443–456. doi:10.1080/0265204001001864111428674
  • Elron-Gross I, Glucksam Y, Biton IE, Margalit R. A novel Diclofenac-carrier for local treatment of osteoarthritis applying live-animal MRI. J Controlled Release. 2009;135(1):65–70. doi:10.1016/j.jconrel.2008.12.005
  • Whitmire RE, Wilson DS, Singh A, Levenston ME, Murthy N, García AJ. Self-assembling nanoparticles for intra-articular delivery of anti-inflammatory proteins. Biomaterials. 2012;33(30):7665–7675. doi:10.1016/j.biomaterials.2012.06.10122818981
  • 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(1):285–290. doi:10.1016/j.ijpharm.2012.11.03123194887
  • Hui JH, Chan S-W, Li J, et al. Intra-articular delivery of chondroitin sulfate for the treatment of joint defects in rabbit model. J Mol Histol. 2007;38(5):483–489. doi:10.1007/s10735-007-9120-717653606