Receptor targeting drug delivery strategies and prospects in the treatment of rheumatoid arthritis

Jaber Emami , Zahra Ansarypour

Abstract


Rheumatoid arthritis (RA), a chronic inflammatory disease, is characterized by cartilage damage,bone tissue destruction, morphological changes in synovial fluids, and synovial joint inflammation.The inflamed synovial tissue has potential for passive and active targeting because of enhanced permeability and retention effect and the existence of RA synovial macrophages and fibroblasts that selectively express surface receptors such as folate receptor β, CD44 and integrin αVβ. Although there are numerous interventions in RA treatment, they are not safe and effective. Therefore, it is important to develop new drug or drug delivery systems that specifically targets inflamed/swollen joints but attenuates other possible damages to healthy tissues. Recently some receptors such as toll-like receptors (TLRs),the nucleotide-binding oligomerization domain-like receptors, and Fc-γ receptor have been identified in synovial tissue and immune cells that are involved in induction or suppression of arthritis. Analysis of  the TLR pathway has moreover suggested new insights into the pathogenesis of RA. In the present paper,we have reviewed drug delivery strategies based on receptor targeting with novel ligand-anchored carriers exploiting CD44, folate and integrin αVβ as well as TLRs expressed on synovial monocytes  and macrophages and antigen presenting cells, for possible active targeting in RA. TLRs could not only open a new horizon for developing new drugs but also their antagonists or humanized monoclonal antibodies  that block TLRS specially TLR4 and TLR9 signaling could be used as targeting agents to antigen presenting cells and dendritic cells. As a conclusion, common conventional receptors and multifunctional ligands that arte involved in targeting receptors or developing nanocarriers with appropriate ligands for TLRs can provide profoundly targeting drug delivery systems for the effective treatment of RA.


Keywords


Delivery; Drug; Inflammation; Receptor; Rheumatoid arthritis; Target.

Full Text:

PDF

References


Gabriel SE. The epidemiology of rheumatoid arthritis. Rheum Dis Clin North Am. 2001;27(2):269-281.

Fauci AS, Langford C. Harrison's Rheumatology. 3rd ed. New York: McGraw-Hill Education; 2013. pp. 87-106.

Shah A, Clair EW. Rheumatoid Arthritis. In: Jameson JL, Fauci A, Kasper D, Hauser S, Longo D, Loscalzo J. Harrison's Principles of Internal Medicine. 20th ed. New York: McGraw Hill Professional; 2018. pp. 2527-2541.

Ballara S, Taylor PC, Reusch P, Marmé D, Feldmann M, Maini RN, et al. Raised serum vascular endothelial growth factor levels are associated with destructive change in inflammatory arthritis. Arthritis Rheum. 2001;44(9):2055-2064.

Kumar P, Erroi A, Sattar A, Kumar S. Weibel-Palade bodies as a marker for neovascularization induced by tumor and rheumatoid angiogenesis factors. Cancer Res. 1985;45(9):4339-4348.

Semble E, Turner RA, McCrickard EL. Rheumatoid arthritis and osteoarthritis synovial fluid effects on primary human endothelial cell cultures. J Rheumatol. 1985;12(2):237-241.

Smolen JS, Steiner G. Therapeutic strategies for rheumatoid arthritis. Nat Rev Drug Discov. 2003;2(6):473-488.

Feng X, Chen Y. Drug delivery targets and systems for targeted treatment of rheumatoid arthritis. J Drug Target. 2018;26(10):845-857.

Choy EH. Clinical significance of Janus kinase inhibitor selectivity. Rheumatology (Oxford). 2019;58(6):953-962.

Yuan F, Quan L, Cui L, Goldring SR, Wang D. Development of macromolecular prodrug for rheumatoid arthritis. Adv Drug Deliv Rev. 2012;64(12):1205-1219.

Johnson BA, Haines GK, Harlow LA, Koch AE. Adhesion molecule expression in human synovial tissue. Arthritis Rheum. 1993;36(2):137-146.

Wilder RL. Integrin alpha V beta 3 as a target for treatment of rheumatoid arthritis and related rheumatic diseases. Ann Rheum Dis. 2002;61(Suppl 2):ii96-ii99.

Nagayoshi R, Nagai T, Matsushita K, Sato K, Sunahara N, Matsuda T, et al. Effectiveness of anti-folate receptor beta antibody conjugated with truncated Pseudomonas exotoxin in the targeting of rheumatoid arthritis synovial macrophages. Arthritis Rheum. 2005;52(9):2666-2675.

van der Heijden JW, Oerlemans R, Dijkmans BA, Qi H, van der Laken CJ, Lems WF, et al. Folate receptor beta as a potential delivery route for novel folate antagonists to macrophages in the synovial tissue of rheumatoid arthritis patients. Arthritis Rheum. 2009;60(1):12-21.

Jamar F, Houssiau FA, Devogelaer JP, Chapman PT, Haskard DO, Beaujean V, et al. Scintigraphy using a technetium 99m-labelled anti-E-selectin Fab fragment in rheumatoid arthritis. Rheumatology (Oxford). 2002;41(1):53-61.

Kamen BA, Smith AK, Anderson RG. The folate receptor works in tandem with a probenecid-sensitive carrier in MA104 cells in vitro. J Clin Invest. 1991;87(4):1442-1449.

Antony AC. The biological chemistry of folate receptors. Blood. 1992;79(11):2807-2820.

Ross JF, Chaudhuri PK, Ratnam M. Differential regulation of folate receptor isoforms in normal and malignant tissues in vivo and in established cell lines. Physiologic and clinical implications. Cancer. 1994;73(9):2432-2443.

Nakashima-Matsushita N, Homma T, Yu S, Matsuda T, Sunahara N, Nakamura T, et al. Selective expression of folate receptor beta and its possible role in methotrexate transport in synovial macrophages from patients with rheumatoid arthritis. Arthritis Rheum. 1999;42(8):1609-1616.

Kinne RW, Brauer R, Stuhlmuller B, Palombo-Kinne E, Burmester GR. Macrophages in rheumatoid arthritis. Arthritis Res. 2000;2(3):189-202.

Chandrasekar D, Sistla R, Ahmad FJ, Khar RK, Diwan PV. The development of folate-PAMAM dendrimer conjugates for targeted delivery of anti-arthritic drugs and their pharmacokinetics and biodistribution in arthritic rats. Biomaterials. 2007;28(3):504-512.

Chandrasekar D, Sistla R, Ahmad FJ, Khar RK, Diwan PV. Folate coupled poly(ethyleneglycol) conjugates of anionic poly(amidoamine) dendrimer for inflammatory tissue specific drug delivery. J Biomed Mater Res A. 2007;82(1):92-103.

Thomas TP, Goonewardena SN, Majoros IJ, Kotlyar A, Cao Z, Leroueil PR, et al. Folate-targeted nanoparticles show efficacy in the treatment of inflammatory arthritis. Arthritis Rheum. 2011;63(9):2671-2680.

Mansouri S, Cuie Y, Winnik F, Shi Q, Lavigne P, Benderdour M, et al. Characterization of folate-chitosan-DNA nanoparticles for gene therapy. Biomaterials. 2006;27(9):2060-2065.

Hattori Y, Sakaguchi M, Maitani Y. Folate-linked lipid-based nanoparticles deliver a NFkappaB decoy into activated murine macrophage-like RAW264.7 cells. Biol Pharm Bull. 2006;29(7):1516-1520.

Duan W, Li H. Combination of NF-kB targeted siRNA and methotrexate in a hybrid nanocarrier towards the effective treatment in rheumatoid arthritis. J Nanobiotechnol. 2018;16(1):58-66.

Sun X, Dong S, Li X, yu K, Sun F, Lee R, et al. Delivery of siRNA using folate receptor-targeted pH-sensitive polymeric nanoparticles for rheumatoid arthritis therapy. Nanomed Nanotechnol. 2019;20:1-11.

Zhao J, Zhang X, Sun X, Zhao M, Yu C, Lee RJ, et al. Dual-functional lipid polymeric hybrid pH-responsive nanoparticles decorated with cell penetrating peptide and folate for therapy against rheumatoid arthritis. Eur J Pharm Biopharm. 2018;130:39-47.

Zhang N, Xu C, Li N, Zhang S, Fu L, Chu X, et al. Folate receptor-targeted mixed polysialic acid micelles for combating rheumatoid arthritis: in vitro and in vivo evaluation. Drug Deliv. 2018;25(1):1182-1191.

Verma A, Jain A, Tiwari A, Saraf S, Panda PK, Agrawal GP, et al. Folate conjugated double liposomes bearing prednisolone and methotrexate for targeting rheumatoid arthritis. Pharm Res. 2019;36(8):123-135.

Fischer C, Thiele HG, Hamann A. Adhesion molecules and homing in inflamed synovial. Immun Infekt. 1993;1:12-3.

Haynes BF, Hale LP, Patton KL, Martin ME, McCallum RM. Measurement of an adhesion molecule as an indicator of inflammatory disease activity. Up-regulation of the receptor for hyaluronate (CD44) in rheumatoid arthritis. Arthritis Rheum. 1991;34(11):1434-1443.

Naor D, Nedvetzki S. CD44 in rheumatoid arthritis. Arthritis Res Ther. 2003;5(3):105-115.

Heo R, Park JS, Jang HJ, Kim SH, Shin JM, Suh YD, et al. Hyaluronan nanoparticles bearing gamma-secretase inhibitor: in vivo therapeutic effects on rheumatoid arthritis. J Control Release. 2014;192:295-300.

Yu C, Li X, Hou Y, Meng X, Wang D, Liu J, et al. Hyaluronic acid coated acid-sensitive nanoparticles for targeted therapy of adjuvant-induced arthritis in rats. Molecules. 2019;24(1):E146-E161.

Zhou M, Hou J, Zhong Z, Hao N, Lin Y, Li C. Targeted delivery of hyaluronic acid-coated solid lipid nanoparticles for rheumatoid arthritis therapy. Drug Deliv. 2018;25(1):716-722.

Lee H, Lee MY, Bhang SH, Kim BS, Kim YS, Ju JH, et al. Hyaluronate-gold nanoparticle/tocilizumab complex for the treatment of rheumatoid arthritis. ACS Nano. 2014;8(5):4790-4798.

Shin JM, Kim SH, Thambi T, You DG, Jeon J, Lee JO, et al. A hyaluronic acid-methotrexate conjugate for targeted therapy of rheumatoid arthritis. Chem Commun (Camb). 2014;50(57):7632-7635.

Kim YJ, Chae SY, Jin CH, Sivasubramanian M, Son S, Choi KY, et al. Ionic complex systems based on hyaluronic acid and PEGylated TNF-related apoptosis-inducing ligand for treatment of rheumatoid arthritis. Biomaterials. 2010;31(34):9057-9064.

Tamura T, Higuchi Y, Kitamura H, Murao N, Saitoh R, Morikawa T, et al. Novel hyaluronic acid-methotrexate conjugate suppresses joint inflammation in the rat knee: efficacy and safety evaluation in two rat arthritis models. Arthritis Res Ther. 2016;18:79-89.

Jeon J, Rao NV, Byun JH, Heo R, Han HS, Park JH. pH-Responsive hyaluronic acid-based nanocarrier for treatment of rheumatoid arthritis. J Nanosci Nanotechnol. 2016;16(11):11849-11856.

Seo J, Park SH, Kim MJ, Ju HJ, Yin XY, Min BH, et al. Injectable click-crosslinked hyaluronic acid depot to prolong therapeutic activity in articular joints affected by rheumatoid arthritis. ACS Appl Mater Interfaces. 2019;11(28):24984-24998.

Jung YS, Park W, Park H, Lee DK, Na K. Thermo-sensitive injectable hydrogel based on the physical mixing of hyaluronic acid and Pluronic F-127 for sustained NSAID delivery. Carbohyd Polym. 2017;156:403-408.

Gouveia VM, Lopes-de-Araujo J, Costa Lima SA, Nunes C, Reis S. Hyaluronic acid-conjugated pH-sensitive liposomes for targeted delivery of prednisolone on rheumatoid arthritis therapy. Nanomedicine (Lond). 2018;13(9):1037-1049.

Hsu AR, Veeravagu A, Cai W, Hou LC, Tse V, Chen X. Integrin alpha v beta 3 antagonists for anti-angiogenic cancer treatment. Recent Pat Anticancer Drug Discov. 2007;2(2):143-158.

Brooks PC, Clark RA, Cheresh DA. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science. 1994;264(5158):569-571.

Tsou R, Isik FF. Integrin activation is required for VEGF and FGF receptor protein presence on human microvascular endothelial cells. Mol Cell Biochem. 2001;224(1-2):81-89.

Weber AJ, De Bandt M, Gaudry M. Immunohistochemical analysis of vascular endothelial growth factor expression in severe and destructive rheumatoid arthritis. J Rheumatol. 2000;27(9):2284-2286.

De Bandt M, Mahdi MHB, Ollivier V, Grossin M, Dupuis M, Gaudry M, et al. Blockade of vascular endothelial growth factor receptor I (VEGF-RI), but not VEGF-RII, suppresses joint destruction in the K/BxN model of rheumatoid arthritis. J Immunol. 2003;171(9):4853-4859.

Lainer-Carr D, Brahn E. Angiogenesis inhibition as a therapeutic approach for inflammatory synovitis. Nat Clin Pract Rheumatol. 2007;3(8):434-442.

Yumoto K, Ishijima M, Rittling SR, Tsuji K, Tsuchiya Y, Kon S, et al. Osteopontin deficiency protects joints against destruction in anti-type II collagen antibody-induced arthritis in mice. Proc Natl Acad Sci USA. 2002;99(7):4556-4561.

Scheinman RI, Trivedi R, Vermillion S, Kompella UB. Functionalized STAT1 siRNA nanoparticles regress rheumatoid arthritis in a mouse model. Nanomedicine (Lond). 2011;6(10):1669-1682.

Lee SM, Kim HJ, Ha YJ, Park YN, Lee SK, Park YB, et al. Targeted chemo-photothermal treatments of rheumatoid arthritis using gold half-shell multifunctional nanoparticles. ACS Nano. 2013;7(1):50-57.

Koning GA, Schiffelers RM, Wauben MH, Kok RJ, Mastrobattista E, Molema G, et al. Targeting of angiogenic endothelial cells at sites of inflammation by dexamethasone phosphate-containing RGD peptide liposomes inhibits experimental arthritis. Arthritis Rheum. 2006;54(4):1198-1208.

Wang Y, Liu Z, Li T, Chen L, Lyu J, Li C, et al. Enhanced therapeutic effect of RGD-modified polymeric micelles loaded with low-dose methotrexate and nimesulide on Rheumatoid Arthritis. Theranostics. 2019;9(3):708-720.

Ye C, Kiriyama K, Mistuoka C, Kannagi R, Ito K, Watanabe T, et al. Expression of E-selectin on endothelial cells of small veins in human colorectal cancer. Int J Cancer. 1995;61(4):455-460.

Vallien G, Langley R, Jennings S, Specian R, Granger DN. Expression of endothelial cell adhesion molecules in neovascularized tissue. Microcirculation. 2000;7(4):249-258.

Bhaskar V, Law DA, Ibsen E, Breinberg D, Cass KM, DuBridge RB, et al. E-selectin up-regulation allows for targeted drug delivery in prostate cancer. Cancer Res. 2003;63(19):6387-6394.

Ehrhardt C, Kneuer C, Bakowsky U. Selectins-an emerging target for drug delivery. Adv Drug Deliv Rev. 2004;56(4):527-549.

Magnani JL. The discovery, biology, and drug development of sialyl Lea and sialyl Lex. Arch Biochem Biophys. 2004;426(2):122-131.

Simanek EE, McGarvey GJ, Jablonowski JA, Wong CH. Selectin-carbohydrate interactions: from natural ligands to designed mimics. Chem Rev. 1998;98(2):833-862.

Kaila N, Thomas BE 4th. Design and synthesis of sialyl Lewis(x) mimics as E- and P-selectin inhibitors. Med Res Rev. 2002;22(6):566-601.

Everts M, Koning GA, Kok RJ, Asgeirsdottir SA, Vestweber D, Meijer DK, et al. In vitro cellular handling and in vivo targeting of E-selectin-directed immunoconjugates and immunoliposomes used for drug delivery to inflamed endothelium. Pharm Res. 2003;20(1):64-72.

Mitragotri S, Yoo JW. Designing micro- and nano-particles for treating rheumatoid arthritis. Arch Pharm Res. 2011;34(11):1887-1897.

Hirai M, Minematsu H, Kondo N, Oie K, Igarashi K, Yamazaki N. Accumulation of liposome with Sialyl Lewis X to inflammation and tumor region: application to in vivo bio-imaging. Biochem Biophys Res Commun. 2007;353(3):553-558.

Patra MC, Choi S. Recent progress in the development of toll-like receptor (TLR) antagonists. Expert Opin Ther Pat. 2016;26(6):719-730.

McCormack WJ, Parker AE, O'Neill LA. Toll-like receptors and NOD-like receptors in rheumatic diseases. Arthritis Res Ther. 2009;11(5):243-250.

Mayadas TN, Tsokos GC, Tsuboi N. Mechanisms of immune complex-mediated neutrophil recruitment and tissue injury. Circulation. 2009;120(20):2012-2024.

Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol. 2008;8(1):34-47.

Silman AJ, Pearson JE. Epidemiology and genetics of rheumatoid arthritis. Arthritis Res. 2002;4(Suppl 3):S265-S272.

Andreakos E, Sacre S, Foxwell BM, Feldmann M. The toll-like receptor-nuclear factor kappaB pathway in rheumatoid arthritis. Front Biosci. 2005;10:2478-2488.

Imler JL, Hoffmann JA. Toll receptors in innate immunity. Trends Cell Biol. 2001;11(7):304-311.

Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immuno. 2003;21(1):335-376.

Kempsell KE, Cox CJ, Hurle M, Wong A, Wilkie S, Zanders ED, et al. Reverse transcriptase-PCR analysis of bacterial rRNA for detection and characterization of bacterial species in arthritis synovial tissue. Infect Immun. 2000;68(10):6012-6026.

van der Heijden IM, Wilbrink B, Tchetverikov I, Schrijver IA, Schouls LM, Hazenberg MP, et al. Presence of bacterial DNA and bacterial peptidoglycans in joints of patients with rheumatoid arthritis and other arthritides. Arthritis Rheum. 2000;43(3):593-598.

Roelofs MF, Joosten LA, Abdollahi-Roodsaz S, van Lieshout AW, Sprong T, van den Hoogen FH, et al. The expression of toll-like receptors 3 and 7 in rheumatoid arthritis synovium is increased and costimulation of toll-like receptors 3, 4, and 7/8 results in synergistic cytokine production by dendritic cells. Arthritis Rheum. 2005;52(8):2313-2322.

Iwahashi M, Yamamura M, Aita T, Okamoto A, Ueno A, Ogawa N, et al. Expression of toll-like receptor 2 on CD16+ blood monocytes and synovial tissue macrophages in rheumatoid arthritis. Arthritis Rheum. 2004;50(5):1457-1467.

Murata M. Activation of Toll-like receptor 2 by a novel preparation of cell wall skeleton from Mycobacterium bovis BCG Tokyo (SMP-105) sufficiently enhances immune responses against tumors. Cancer Sci. 2008;99(7):1435-1440.

Ultaigh SN, Saber TP, McCormick J, Connolly M, Dellacasagrande J, Keogh B, et al. Blockade of Toll-like receptor 2 prevents spontaneous cytokine release from rheumatoid arthritis ex vivo synovial explant cultures. Arthritis Res Ther. 2011;13(1):R33-R41.

Elshabrawy HA, Essani AE, Szekanecz Z, Fox DA, Shahrara S. TLRs, future potential therapeutic targets for RA. Autoimmun Rev. 2017;16(2):103-113.

Wu YW, Tang W, Zuo JP. Toll-like receptors: potential targets for lupus treatment. Acta Pharmacol Sin. 2015;36(12):1395-1407.

Thwaites R, Chamberlain G, Sacre S. Emerging role of endosomal toll-like receptors in rheumatoid arthritis. Front Immunol. 2014;5(1):1-8.

Muzio M, Bosisio D, Polentarutti N, D’amico G, Stoppacciaro A, Mancinelli R, et al. Differential expression and regulation of toll-like receptors (TLR) in human leukocytes: selective expression of TLR3 in dendritic cells. J Immunol. 2000;164(11):5998-6004.

Brentano F, Schorr O, Gay RE, Gay S, Kyburz D. RNA released from necrotic synovial fluid cells activates rheumatoid arthritis synovial fibroblasts via Toll‐like receptor 3. Arthritis Rheum. 2005;52(9):2656-2665.

Paulsen F, Pufe T, Conradi L, Varoga D, Tsokos M, Papendieck J, et al. Antimicrobial peptides are expressed and produced in healthy and inflamed human synovial membranes. J Pathol. 2002;198(3):369-377.

Lai Y, Adhikarakunnathu S, Bhardwaj K, Ranjith-Kumar C, Wen Y, Jordan JL, et al. LL37 and cationic peptides enhance TLR3 signaling by viral double-stranded RNAs. PLoS One. 2011;6(10):e26632.

Hu F, Mu R, Zhu J, Shi L, Li Y, Liu X, et al. Hypoxia and hypoxia-inducible factor-1α provoke toll-like receptor signalling-induced inflammation in rheumatoid arthritis. Ann Rheum Dis. 2014;73(5):928-936.

Moon SJ, Park MK, Oh HJ, Lee SY, Kwok SK, Cho ML, et al. Engagement of toll-like receptor 3 induces vascular endothelial growth factor and interleukin-8 in human rheumatoid synovial fibroblasts. Korean J Intern Med. 2010;25(4):429-435.

Meng L, Zhu W, Jiang C, He X, Hou W, Zheng F, et al. Toll-like receptor 3 upregulation in macrophages participates in the initiation and maintenance of pristane-induced arthritis in rats. Arthritis Res Ther. 2010;12(3):R103-R114.

Zhu W, Meng L, Jiang C, Xu J, Wang B, Han Y, et al. Overexpression of toll-like receptor 3 in spleen is associated with experimental arthritis in rats. Scand J Immunol. 2012;76(3):263-270.

O'Neill LA, Bryant CE, Doyle SL. Therapeutic targeting of toll-like receptors for infectious and inflammatory diseases and cancer. Pharmacol Rev. 2009;61(2):177-197.

Roelofs MF, Boelens WC, Joosten LA, Abdollahi-Roodsaz S, Geurts J, Wunderink LU, et al. Identification of small heat shock protein B8 (HSP22) as a novel TLR4 ligand and potential involvement in the pathogenesis of rheumatoid arthritis. J Immunol. 2006;176(11):7021-7027.

Midwood K, Sacre S, Piccinini AM, Inglis J, Trebaul A, Chan E, et al. Tenascin-C is an endogenous activator of toll-like receptor 4 that is essential for maintaining inflammation in arthritic joint disease. Nat Med. 2009;15(7):774-780.

Pierer M, Wagner U, Rossol M, Ibrahim S. Toll-like receptor 4 is involved in inflammatory and joint destructive pathways in collagen-induced arthritis in DBA1J mice. PLoS One. 2011;6(8):e23539.

Campo GM, Avenoso A, Campo S, D'Ascola A, Nastasi G, Calatroni A. Molecular size hyaluronan differently modulates toll-like receptor-4 in LPS-induced inflammation in mouse chondrocytes. Biochimie. 2010;92(2):204-215.

Campo GM, Avenoso A, D'Ascola A, Scuruchi M, Prestipino V, Nastasi G, et al. The inhibition of hyaluronan degradation reduced pro-inflammatory cytokines in mouse synovial fibroblasts subjected to collagen-induced arthritis. J Cell Biochem. 2012;113(6):1852-1867.

Noble PW, McKee CM, Cowman M, Shin HS. Hyaluronan fragments activate an NF-kappa B/I-kappa B alpha autoregulatory loop in murine macrophages. J Exp Med. 1996;183(5):2373-2378.

Termeer CC, Hennies J, Voith U, Ahrens T, Weiss JM, Prehm P, et al. Oligosaccharides of hyaluronan are potent activators of dendritic cells. J Immunol. 2000;165(4):1863-1870.

Campo GM, Avenoso A, D'Ascola A, Prestipino V, Scuruchi M, Nastasi G, et al. Hyaluronan differently modulates TLR-4 and the inflammatory response in mouse chondrocytes. Biofactors. 2012;38(1):69-76.

Monnet E, Shang L, Lapeyre G, deGraaf K, Hatterer E, Buatois V, et al. AB0451 NI-0101, a monoclonal antibody targeting toll like receptor 4 (TLR4) being developed for rheumatoid arthritis (RA) treatment with a potential for personalized medicine. Ann Rheum Dis. 2015;74(2):1046.

Park S, Shin HJ, Shah M, Cho HY, Anwar MA, Achek A, et al. TLR4/MD2 specific peptides stalled in vivo LPS-induced immune exacerbation. Biomaterials. 2017;126:49-60.

Babazada H, Yamashita F, Hashida M. Suppression of experimental arthritis with self-assembling glycol-split heparin nanoparticles via inhibition of TLR4-NF-kappaB signaling. J Control Release. 2014;194:295-300.

Rao K, Roome T, Aziz S, Razzak A, Abbas G, Imran M, et al. Bergenin loaded gum xanthan stabilized silver nanoparticles suppress synovial inflammation through modulation of the immune response and oxidative stress in adjuvant induced arthritic rats. J Mater Chem B. 2018;6(27):4486-4501.

Roome T, Aziz S, Razzak A, Aslam Z, Lubna, Jamali KS, et al. Opuntioside, opuntiol and its metallic nanoparticles attenuate adjuvant-induced arthritis: novel suppressors of toll-like receptors -2 and -4. Biomed Pharmacother. 2019;112:1-15.

Chamberlain ND, Vila OM, Volin MV, Volkov S, Pope RM, Swedler W, et al. TLR5, a novel and unidentified inflammatory mediator in rheumatoid arthritis that correlates with disease activity score and joint TNF-α levels. J Immunol. 2012;189(1):475-483.

Alzabin S, Kong P, Medghalchi M, Palfreeman A, Williams R, Sacre S. Investigation of the role of endosomal toll-like receptors in murine collagen-induced arthritis reveals a potential role for TLR7 in disease maintenance. Arthritis Res Ther. 2012;14(3):R142-R152.

Alzabin S, Williams RO. Effector T cells in rheumatoid arthritis: lessons from animal models. FEBS Lett. 2011;585(23):3649-3659.

Hayashi T, Gray CS, Chan M, Tawatao RI, Ronacher L, McGargill MA, et al. Prevention of autoimmune disease by induction of tolerance to Toll-like receptor 7. Proc Natl Acad Sci USA. 2009;106(8):2764-2769.

Demaria O, Pagni PP, Traub S, de Gassart A, Branzk N, Murphy AJ, et al. TLR8 deficiency leads to autoimmunity in mice. J Clin Invest. 2010;120(10):3651-3662.

Li Y, Berke IC, Modis Y. DNA binding to proteolytically activated TLR9 is sequence-independent and enhanced by DNA curvature. EMBO J. 2012;31(4):919-931.

Viglianti GA, Lau CM, Hanley TM, Miko BA, Shlomchik MJ, Marshak-Rothstein A. Activation of autoreactive B cells by CpG dsDNA. Immunity. 2003;19(6):837-847.

Fischer A, Abdollahi-Roodsaz S, Bohm C, Niederreiter B, Meyer B, Yau ACY, et al. The involvement of Toll-like receptor 9 in the pathogenesis of erosive autoimmune arthritis. J Cell Mol Med. 2018;22(9):4399-4409.

Torigoe M, Sakata K, Ishii A, Iwata S, Nakayamada S, Tanaka Y. Hydroxychloroquine efficiently suppresses inflammatory responses of human class-switched memory B cells via Toll-like receptor 9 inhibition. Clin Immunol. 2018;195:1-7.

Sacre S, Medghalchi M, Gregory B, Brennan F, Williams R. Fluoxetine and citalopram exhibit potent antiinflammatory activity in human and murine models of rheumatoid arthritis and inhibit toll‐like receptors. Arthritis Rheum. 2010;62(3):683-693.

Kandimalla ER, Bhagat L, Wang D, Yu D, Sullivan T, La Monica N, et al. Design, synthesis and biological evaluation of novel antagonist compounds of toll-like receptors 7, 8 and 9. Nucleic Acids Res. 2013;41(6):3947-3961.

Tsai CY, Lu SL, Hu CW, Yeh CS, Lee GB, Lei HY. Size-dependent attenuation of TLR9 signaling by gold nanoparticles in macrophages. J Immunol. 2012;188(1):68-76.

Peng B, Liang H, Li Y, Dong C, Shen J, Mao HQ, et al. Tuned cationic dendronized polymer: molecular scavenger for rheumatoid arthritis treatment. Angew Chem. 2019;131(13):4298-4302.

Fukata M, Vamadevan AS, Abreu MT. Toll-like receptors (TLRs) and Nod-like receptors (NLRs) in inflammatory disorders. Semin Immunol. 2009;21(4):242-253.

Guo C, Fu R, Wang S, Huang Y, Li X, Zhou M, et al. NLRP3 inflammasome activation contributes to the pathogenesis of rheumatoid arthritis. Clin Exp Immunol. 2018;194(2):231-243.

Yang Y, Zhang X, Xu M, Wu X, Zhao F, Zhao C. Quercetin attenuates collagen-induced arthritis by restoration of Th17/Treg balance and activation of heme oxygenase 1-mediated anti-inflammatory effect. Int Immunopharmacol. 2018;54:153-162.

Chen B, Vousden KA, Naiman B, Turman S, Sun H, Wang S, et al. Humanised effector-null FcγRIIA antibody inhibits immune complex-mediated proinflammatory responses. Ann Rheum Dis. 2019;78(2):228-237.

Church LD, Cook GP, McDermott MF. Primer: inflammasomes and interleukin 1beta in inflammatory disorders. Nat Clin Pract Rheumatol. 2008;4(1):34-42.

Kanneganti TD, Lamkanfi M, Kim YG, Chen G, Park JH, Franchi L, et al. Pannexin-1-mediated recognition of bacterial molecules activates the cryopyrin inflammasome independent of toll-like receptor signaling. Immunity. 2007;26(4):433-443.

Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1(6):a001651,1-10.

van Egmond M, Vidarsson G, Bakema JE. Cross-talk between pathogen recognizing Toll-like receptors and immunoglobulin Fc receptors in immunity. Immunol Rev. 2015;268(1):311-327.


Refbacks

  • There are currently no refbacks.


Creative Commons LicenseThis work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License which allows users to read, copy, distribute and make derivative works for non-commercial purposes from the material, as long as the author of the original work is cited properly.