Suscripción a Biblioteca: Guest
Critical Reviews™ in Therapeutic Drug Carrier Systems

Publicado 6 números por año

ISSN Imprimir: 0743-4863

ISSN En Línea: 2162-660X

The Impact Factor measures the average number of citations received in a particular year by papers published in the journal during the two preceding years. 2017 Journal Citation Reports (Clarivate Analytics, 2018) IF: 2.7 To calculate the five year Impact Factor, citations are counted in 2017 to the previous five years and divided by the source items published in the previous five years. 2017 Journal Citation Reports (Clarivate Analytics, 2018) 5-Year IF: 3.6 The Immediacy Index is the average number of times an article is cited in the year it is published. The journal Immediacy Index indicates how quickly articles in a journal are cited. Immediacy Index: 0.8 The Eigenfactor score, developed by Jevin West and Carl Bergstrom at the University of Washington, is a rating of the total importance of a scientific journal. Journals are rated according to the number of incoming citations, with citations from highly ranked journals weighted to make a larger contribution to the eigenfactor than those from poorly ranked journals. Eigenfactor: 0.00023 The Journal Citation Indicator (JCI) is a single measurement of the field-normalized citation impact of journals in the Web of Science Core Collection across disciplines. The key words here are that the metric is normalized and cross-disciplinary. JCI: 0.39 SJR: 0.42 SNIP: 0.89 CiteScore™:: 5.5 H-Index: 79

Indexed in

Current Developments in Therapeutic Drug Targeting for the Management of Rheumatoid Arthritis: An Emerging Paradigm

Volumen 36, Edición 6, 2019, pp. 485-536
DOI: 10.1615/CritRevTherDrugCarrierSyst.2019025729
Get accessGet access

SINOPSIS

Rheumatoid arthritis (RA) is a debilitating condition that results in impairment of joints and ligaments and thus constrained mobility and decreased array of movement. It is a broad expression that encompasses additional 100 very diverse disorders mainly affecting joints. In the field of drug discovery, there is no well-known treatment for RA that can eradicate the disease permanently and alleviate the pain. The common non-targeted treatment approaches leads to serious side effects and systemic complications for RA patients. Therefore, targeted drug delivery systems, strategies, and diverse therapeutic approach for treatment of RA have gained increasing attention in the past few years. However, with the current understandings, researchers aim at accomplishing complete and long-lasting remission by the development of smart formulations/smart drug-delivery systems. Treatment for RA patients can be more efficient and effective utilizing these smart approaches. The present review focuses on the existing novel drug-delivery systems, strategies, and current trends in the treatment of RA.

REFERENCIAS
  1. Chopra A. Ayurvedic medicine and arthritis. Rheum Dis Clin North Am. 2000;26(1):133-44.

  2. Lee SJ, Kavanaugh A. Pharmacological treatment of established rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2003;17(5):811-29.

  3. American College of Rheumatology. Rheumatoid arthritis [cited 2019 July 5]. Available from: htt-ps://www.rheumatology.org/Search-Results?search=rheumatoid%20arthritis.

  4. Lipsky PE, Kavanaugh A. The impact of pharmaco-economic considerations on the utilization of novel anti-rheumatic therapies. Rheumatology. 1999;38 (Suppl 2):41-4.

  5. Misra R, Sharma BL, Gupta R, Pandya S, Agarwal S, Agarwal P, Grover S, Sarma P, Wangjam K. Indian Rheumatology Association consensus statement on the management of adults with rheumatoid arthritis. Indian J Rheumatol. 2008;3:S1-S16.

  6. Herman CJ, Allen P, Hunt WC, Prasad A, Brady TJ. Use of complementary therapies among primary care clinic patients with arthritis. Prev Chronic Dis. 2004;1(4):1-12.

  7. Kapoor B, Singh S, Gulati M, Gupta R, Vaidya Y. Application of liposomes in treatment of rheumatoid arthritis: quo vadis. Sci World J. 2014;1-17.

  8. Huber L, Distler O, Tarner, Gay R, Gay S, Pap T. Synovial fibroblasts: key players in rheumatoid arthritis. Rheumatology. 2006;45(6):669-75.

  9. Gravallese EM, Harada Y, Wang JT, Gorn AH, Thornhill TS, Goldring SR. Identification of cell types responsible for bone resorption in rheumatoid arthritis and juvenile rheumatoid arthritis. Am J Clin Pathol. 1998;152(4):943-51.

  10. R.A.A.I.D. Raising awareness of auto-immune diseases [cited 2018 Feb 7]. Available from: http:// www.weishendopublications.com/#gsc.tab=0.

  11. Singh JA, Saag KG, Bridges SL, Akl EA, Bannuru RR, Sullivan MC, Vaysbrot E, McNaughton C, Osani M, Shmerling RH, Curtis JR. 2015 American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 2016;68(1):1-26.

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

  13. Mclnnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365(23):2205-19.

  14. Van Hoovels L, Jacobs J, Vander Cruyssen B, Van den Bremt S, Verschueren P, Bossuyt X. Performance characteristics of rheumatoid factor and anti-cyclic citrullinated peptide antibody assays may impact ACR/EULAR classification of rheumatoid arthritis. Ann Rheum Dis. 2018;77:5):667-77.

  15. Koppejan H, Trouw LA, Sokolove J, Lahey LJ, Huizinga TJ, Smolik IA, Robinson DB, El-Gabalawy HS, Toes RE, Hitchon CA. Role of Anti-carbamylated protein antibodies compared to anti-citrullinated protein antibodies in indigenous North Americans with rheumatoid arthritis, their first-degree relatives, and healthy controls. Arthritis Rheumatol. 2016;68(9):2090-8.

  16. Dellavance A, de Melo Cruvinel W, Francescantonio PL, Andrade LE. Antinuclear antibody tests. In: Detrick B, Hamilton RG, Schmitz JL, editors. Manual of molecular and clinical laboratory immunology, 8th ed. Washington, DC: American Society of Microbiology; 2016, p. 843-8.

  17. Isiksacan Z, Erel O, Elbuken C. A portable microfluidic system for rapid measurement of the erythrocyte sedimentation rate. Lab Chip. 2016;16(24):4682-90.

  18. Ridker PM. A test in context: high-sensitivity C-reactive protein. J Am Coll Cardiol. 2016;67(6): 712-23.

  19. Olivares-Martinez E, Hernandez-Ramirez DF, Nunez-Alvarez CA, Cabral AR, Llorente L. The amount of citrullinated proteins in synovial tissue is related to serum anti-cyclic citrullinated peptide (anti-CCP) antibody levels. Clin Rheumatol. 2016;35(1):55-61.

  20. RheumatoidArthritis.net [cited 2018 Feb 07]. Available from: https://rheumatoidarthritis.net/.

  21. Shinde CG, Venkatesh MP, Kumar TP, Shivakumar HG. Methotrexate: a gold standard for treatment of rheumatoid arthritis. J Pain Palliat Care Pharmacother. 2014;28(4):351-8.

  22. Eve RG, Jean CL. In situ-forming hydrogels: review of temperature-sensitive systems. Eur J Pharm Biopharm. 2004;58:409-26.

  23. AVN Arogya Ayurvedic Hospital. Ayurvedic treatments in India: Rheumatoid arthritis [cited 2019 Juy 5]. Available from: https://www.avnarogya.in/rheumatology.

  24. Shaul MP. From early twinges to mastery: the process of adjustment in living with rheumatoid arthritis. Arthritis Rheumatol. 1995;8(4):290-7.

  25. Ody P. The complete medicinal herbal: a practical guide to the healing properties of herbs. New York: Skyhorse Publishing, Inc.; 2017.

  26. Tedeschi SK, Frits M, Cui J, Zhang ZZ, Mahmoud T, Iannaccone C, Lin TC, Yoshida K, Weinblatt ME, Shadick NA, Solomon DH. Diet and rheumatoid arthritis symptoms: survey results from a rheumatoid arthritis registry. Arthritis Care Res. 2017;69(12):1920-5.

  27. Zintzaras E, Dahabreh IJ, Giannouli S, Voulgarelis M, Moutsopoulos HM. Infliximab and metho-trexate in the treatment of rheumatoid arthritis: a systematic review and meta-analysis of dosage regimens. Clin Ther. 2008;30(11):1939-55.

  28. Konigsberg PJ, Debrick JE, Pawlowski TJ, Staerz UD. Liposome encapsulated aurothiomalate reduces collagen-induced arthritis in DBA/1J mice. Biochim Biophys Acta Biomembr. 1999;1421(1):149-62.

  29. Herrmann ML, Schleyerbach R, Kirschbaum BJ. Leflunomide: an immunomodulatory drug for the treatment of rheumatoid arthritis and other autoimmune diseases. Immunopharmacology. 2000;47(2-3):273-89.

  30. Lao M, Shi M, Zou Y, Huang M, Ye Y, Qiu Q, Xiao Y, Zeng S, Liang L, Yang X, Xu H. Protein inhibitor of activated STAT3 regulates migration, invasion, and activation of fibroblast-like synoviocytes in rheumatoid arthritis. J Immunol. 2016;196(2):596-606.

  31. Brasington RD, Kahl LE, Ranganathan P, Latinis KM, Velazquez C, Atkinson JP. 14. Immunologic rheumatic disorders. J Allergy Clin Immunol. 2003;111(2):S593-601.

  32. Crielaard BJ, Lammers T, Schiffelers RM, Storm G. Drug targeting systems for inflammatory disease: one for all, all for one. J Control Release. 2012;161(2):225-34.

  33. Elshabrawy HA, Chen Z, Volin MV, Ravella S, Virupannavar S, Shahrara S. The pathogenic role of angiogenesis in rheumatoid arthritis. Angiogenesis. 2015;18(4):433-48.

  34. Paradowska-Gorycka A, Pawlik A, Romano wska-Prochnicka K, Haladyj E, Malinowski D, Stypinska B, Manczak M, Olesinska M. Relationship between VEGF gene polymorphisms and serum VEGF protein levels in patients with rheumatoid arthritis. PLoS One. 2016;11(8):e0160769.

  35. Strand V, Kimberly R, Isaacs JD. Biologic therapies in rheumatology: lessons learned future directions. Nat Rev Drug Discov. 2007;6(1):75-92.

  36. Revell PA, Mayston V, Lalor P, Mapp PA. The synovial membrane in osteoarthritis: a histological study including the characterisation of the cellular infiltrate present in inflammatory osteoarthritis using monoclonal antibodies. Ann Rheum Dis. 1988;47(4):300-7.

  37. Mankin HJ, Mow VC, Buckwalter JA, Iannotti JP, Ratcliffe A. Form and function of articular cartilage. Orthop Bas Sci. 1994:1-44.

  38. Butoescu N, Jordan O, Doelker E. Intra-articular drug delivery systems for the treatment of rheumatic diseases: a review of the factors influencing their performance. Eur J Pharm Biopharm. 2009;73(2):205-18.

  39. Sweeney SE, Firestein GS. Rheumatoid arthritis: regulation of synovial inflammation. Int J Biochem Cell Biol. 2004;36(3):372-8.

  40. Feldmann M, Brennan FM, Maini RN. Role of cytokines in rheumatoid arthritis. Annu Rev Immunol. 1996;14:397-440.

  41. Yuan F, Quan LD, Cui L, Goldring SR, Wang D. Development of macromolecular prodrug for rheumatoid arthritis. Adv Drug Deliv Rev. 2012;64(12):1205-19.

  42. Yokoyama M. Drug targeting with nano-sized carrier systems. Int J Artif Organs. 2005;8(2):77-84.

  43. Marcucci F, Lefoulon F. Active targeting with particulate drug carriers in tumor therapy: fundamentals and recent progress. Drug Discov Today. 20041;9(5):219-28.

  44. Pirollo KF, Chang EH. Does a targeting ligand influence nanoparticle tumor localization or uptake? Trends Biotechnol. 2008;26(10):552-8.

  45. Angelotti F, Parma A, Cafaro G, Capecchi R, Alunno A, Puxeddu I. One year in review 2017: pathogenesis of rheumatoid arthritis. Clin Exp Rheumatol. 2017; 35(3):368-78.

  46. Danhier F, Feron O, Preat V. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anticancer drug delivery. J Control Release. 2010;148(2):135-46.

  47. Tinkle M. Folic acid and food fortification: implications for the primary care practitioner. Nurse Practitioner. 1997;22(3):105-6.

  48. Nogueira E, Gomes AC, Preto A, Cavaco-Paulo A. Folate-targeted nanoparticles for rheumatoid arthritis therapy. Nanomed Nanotechnol Bio Med. 2016;12(4):1113-26.

  49. Paulos CM, Turk MJ, Breur GJ, Low PS. Folate receptor-mediated targeting of therapeutic and imaging agents to activated macrophages in rheumatoid arthritis. Adv Drug Deliv Rev. 2004;56(8):1205-17.

  50. Van Der Heijden JW, Oerlemans R, Dijkmans BA, Qi H, Laken CJ, Lems WF, Jackman AL, Kraan MC, Tak PP, Ratnam M, Jansen G. Folate receptor p as a potential delivery route for novel folate antagonists to macrophages in the synovial tissue of rheumatoid arthritis patients. Arthritis Rheumatol. 2009;60(1):12-21.

  51. Thomas TP, Goonewardena SN, Majoros IJ, Kotlyar A, Cao Z, Leroueil PR, Baker JR. Folate targeted nanoparticles show efficacy in the treatment of inflammatory arthritis. Arthritis Rheumatol. 2011;63(9):2671-80.

  52. Azizi G, Boghozian R, Mirshafiey A. The potential role of angiogenic factors in rheumatoid arthritis. Int J Rheum Dis. 2014;17(4):369-83.

  53. 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):ii96-9.

  54. Stromblad S, Cheresh DA. Integrins, angiogenesis and vascular cell survival. Chem Biol. 1996;3(11):881-5.

  55. Byrne AM, Bouchier-Hayes DJ, Harmey JH. Angiogenic and cell survival functions of vascular endothelial growth factor (VEGF). J Cell Mol Med. 2005;9(4):777-94.

  56. Kiselyov A, Balakin KV, Tkachenko SE. VEGF/VEGFR signaling as a target for inhibiting angiogenesis. Expert Opin Investig Drugs. 2007;16(1):83-107.

  57. Martel-Pelletier J, Welsch DJ, Pelletier JP. Metalloproteases and inhibitors in arthritic diseases. Best Pract Res Clin Rheumatol. 2001;15(5):805-29.

  58. Burrage PS, Mix KS, Brinckerhoff CE. Matrix metalloproteinases: role in arthritis. Front Biosci. 2006;11(1):529-43.

  59. Mengshol JA, Mix KS, Brinckerhoff CE. Matrix metalloproteinases as therapeutic targets in arthritic diseases: bull'seye or missing the mark? Arthritis Rheumatol. 2002;46(1):13-20.

  60. Tomita T, Nakase T, Kaneko M, Shi K, Takahi K, Ochi T, Yoshikawa H. Expression of extracellular matrix metalloproteinase inducer and enhancement of the production of matrix metalloproteinases in rheumatoid arthritis. Arthritis Rheumatol. 2002;46(2):373-8.

  61. Vartak DG, Gemeinhart RA. Matrix metalloproteases: underutilized targets for drug delivery. J Drug Target. 2007;15(1):1-20.

  62. Bigg HF, Rowan AD. The inhibition of metalloproteinases as a therapeutic target in rheumatoid arthritis and osteoarthritis. Curr Opin Pharmacol. 2001; 1(3):314-20.

  63. Cronstein BN, Weissmann G. The adhesion molecules of inflammation. Arthritis Rheumatol. 1993;36(2):147-57.

  64. Ehrhardt C, Kneuer C, Bakowsky U. Selectinsan emerging target for drug delivery. Adv Drug Deliv Rev. 2004;56(4):527-49.

  65. Jubeli E, Moine L, Vergnaud Gauduchon J, Barratt G. E-selectin as a target for drug delivery and molecular imaging. J Control Release. 2012;158(2):194-206.

  66. Kessner S, Krause A, Rothe U, Bendas G. Investigation of the cellular uptake of E-selectin-targeted immunoliposomes by activated human endothelial cells. Biochim Biophys Acta Biomembr. 2001;1514(2):177-90.

  67. Gunawan RC, Auguste DT. The role of antibody synergy and membrane fluidity in the vascular targeting of immunoliposomes. Biomaterials. 2010;31(5):900-7.

  68. Bendas G, Krause A, Bakowsky U, Vogel J, Rothe U. Targetability of novel immunoliposomes prepared by a new antibody conjugation technique. Int J Pharm. 1999;181(1):79-93.

  69. Kok RJ, Everts M, Asgeirsdottir SA, Meijer DK, Molema G. Cellular handling of a dexamethasone-anti-E-selectin immunoconjugate by activated endothelial cells: comparison with free dexamethasone. Pharm Res. 2002;19(11):1730-5.

  70. Chorny A, Gonzalez-Rey E, Varela N, Robledo G, Delgado M. Signaling mechanisms of vasoactive intestinal peptide in inflammatory conditions. Regul Pept. 2006;137(1-2):67-74.

  71. El Bannoudi H, Ioan Facsinay A, Toes RE. Bridging autoantibodies and arthritis: the role of Fc receptors. Curr Top Microbiol Immunol. 2014;382:303-19.

  72. Magnusson SE, Wennerberg E, Matt P, Lindqvist U, Kleinau S. Dysregulated Fc receptor function in active rheumatoid arthritis. Immunol Lett. 2014;162(1):200-6.

  73. Mancardi DA, Albanesi M, Jonsson F, Iannascoli B, Van Rooijen N, Kang X, England P, Daeron M, Bruhns P. The high-affinity human IgG receptor FcyRI (CD64) promotes IgG-mediated inflammation, anaphylaxis, and antitumor immunotherapy. Blood. 2013;121(9):1563-73.

  74. Vergunst CE, Tak PP. Chemokines: their role in rheumatoid arthritis. Curr Rheumatol Rep. 2005;7(5):382-8.

  75. Yang L, Zhang J, Wang G. The effect of sodium hyaluronate treating knee osteoarthritis on synovial fluid interleukin-1p and clinical treatment mechanism. Pak J Pharm Sci. 2015;28(1 Suppl):407-10.

  76. Malemud CJ. Matrix metalloproteinases and synovial joint pathology. In: Progress in molecular biology and translational science. Academic Press; 2017. p. 305-325.

  77. Kikuchi J, Hashizume M, Kaneko Y, Yoshimoto K, Nishina N, Takeuchi T. Peripheral blood CD4+ CD25+ CD127 low regulatory T cells are significantly increased by tocilizumab treatment in patients with rheumatoid arthritis: increase in regulatory T cells correlates with clinical response. Arthritis Res Ther. 2015;17(1):10.

  78. Paulissen SM, Van Hamburg JP, Dankers W, Lubberts E. The role and modulation of CCR6+ Th17 cell populations in rheumatoid arthritis. Cytokine. 2015;74(1):43-53.

  79. Fischer JA, Hueber AJ, Wilson S, Galm M, Baum W, Kitson C, Auer J, Lorenz SH, Moelleken J, Bader M, Tissot AC. Combined inhibition of tumor necrosis factor a and interleukin 17 as a therapeutic opportunity in rheumatoid arthritis: development and characterization of a novel bispecific antibody. Arthritis Rheumatol. 2015;67(1):51-62.

  80. Thornton S, Raghu H, Jone A, Cruz C, Rewerts CL, Frederick MD, Degen JL, Flick MJ. Mice deficient in urokinase-type plasminogen activator (uPA) or uPA receptor develop significantly diminished collagen-induced arthritis. Blood. 2014;124:580.

  81. Rhodes A, Deakin A, Spaull J, Coomber B, Aitken A, Life P, Rees S. The generation and characterization of antagonist RNA aptamers to human oncostatin M. J Biol Chem. 2000;275(37): 28555-61.

  82. Wang L. Effects of RNAi mediated inhibition of aggrecanase 1 and aggrecanase 2 on rat costochondral chondrocytes in vitro. Acta Pharmacol Sin. 2008;29(10):1215-26.

  83. Monte K, Wilson C, Shih FF. Increased number and function of FoxP3 regulatory T cells during experimental arthritis. Arthritis Rheumatol. 2008;58(12):3730-41.

  84. Ronnelid J, Hansson M, Mathsson-Alm L, Cornillet M, Reed E, Jakobsson PJ, Alfredsson L, Holmdahl R, Skriner K, Serre G, Lundberg K. Anticitrullinated protein/peptide antibody multiplexing defines an extended group of ACPA-positive rheumatoid arthritis patients with distinct genetic and environmental determinants. Ann Rheum Dis. 2018;77(2):203-11.

  85. Hensvold AH, Magnusson PK, Joshua V, Hansson M, Israelsson L, Ferreira R, Jakobsson PJ, Holmdahl R, Hammarstrom L, Malmstrom V, Askling J. Environmental and genetic factors in the development of anti-citrullinated protein antibodies (ACPAs) and ACPA-positive rheumatoid arthritis: an epidemiological investigation in twins. Ann Rheum Dis. 2015; 74(2):375-80.

  86. Ghivizzani SC, Oligino TJ, Glorioso JC, Robbins PD, Evans CH. Direct gene delivery strategies for the treatment of rheumatoid arthritis. Drug Discov Today. 2001;6(5):259-67.

  87. Evans CH, Ghivizzani SC, Kang R, Muzzonigro T, Wasko MC, Herndon JH, Robbins PD. Gene therapy for rheumatic diseases. Arthritis Rheumatol. 1999;42(1):1-6.

  88. Evans CH, Robbins PD. Gene therapy of arthritis. Intern Med. 1999;38(3):233-9.

  89. Glorioso JC, Krisky D, Marconi P, Oligino T, Ghivizzani SC, Robbins PD, Schmidt MC, Goins WF, Evans C. Progress in development of herpes simplex virus gene vectors for treatment of rheumatoid arthritis. Adv Drug Deliv Rev. 1997;27(1):41-57.

  90. Utada AS, Lorenceau E, Link DR, Kaplan PD, Stone HA, Weitz DA. Monodisperse double emulsions generated from a microcapillary device. Science. 2005;308(5721):537-41.

  91. Roh KH, Martin DC, Lahann J. Biphasic Janus particles with nanoscale anisotropy. Nat Mater. 2005;4(10):759-63.

  92. Emery P, Breedveld FC, Hall S, Durez P, Chang DJ, Robertson D, Singh A, Pedersen RD, Koenig AS, Freundlich B. Comparison of methotrexate monotherapy with a combination of methotrexate and etanercept in active, early, moderate to severe rheumatoid arthritis (COMET): a randomised, double-blind, parallel treatment trial. Lancet. 2008;372(9636):375-82.

  93. O'Dell JR, Haire CE, Erikson N, Drymalski W, Palmer W, Eckhoff PJ, Garwood V, Maloley P, Klassen LW, Wees S, Klein H. Treatment of rheumatoid arthritis with methotrexate alone, sulfasalazine and hydroxychloroquine, or a combination of all three medications. N Engl J Med. 1996;334(20):1287-91.

  94. Mitragotri S, Yoo JW. Designing micro-and nano-particles for treating rheumatoid arthritis. Arch Pharmacal Res. 2011;34(11):1887-97.

  95. Chen FH, Tuan RS. Mesenchymal stem cells in arthritic diseases. Arthritis Res Ther. 2008;10(5):223.

  96. Liu Y, Mu R, Wang S, Long L, Liu X, Li R, Sun J, Guo J, Zhang X, Guo J, Yu P. Therapeutic potential of human umbilical cord mesenchymal stem cells in the treatment of rheumatoid arthritis. Arthritis Res Ther. 2010;12(6):R210.

  97. Djouad F, Fritz V, Apparailly F, Louis Plence P, Bony C, Sany J, Jorgensen C, Noel D. Reversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor a in collagen induced arthritis. Arthritis Rheumatol. 2005;52(5):1595-603.

  98. Chopra D, Gulati M, Singh S, Duggal S, Kumar R. Use of liposomal drugs in the treatment of rheumatoid arthritis. Curr Rheumatol Rev. 2008;4(1):63-70.

  99. Jadot G, Michelson AM, Puget K. Anti-inflammatory activity of superoxide dismutases: studies on adjuvant induced polyarthritis in rats. Free Radic Res Commun. 1986;2(1-2):27-42.

  100. Yamane S, Iwasaki N, Majima T, Funakoshi T, Masuko T, Harada K, Minami A, Monde K, Nishimura SI. Feasibility of chitosan-based hyaluronic acid hybrid biomaterial for a novel scaffold in cartilage tissue engineering. Biomaterials. 2005;26(6):611-9.

  101. Grigolo B, De Franceschi L, Roseti L, Cattini L, Facchini A. Down regulation of degenerative cartilage molecules in chondrocytes grown on a hyaluronan-based scaffold. Biomaterials. 2005;26(28):5668-76.

  102. Matt P, Lindqvist U, Kleinau S. Elevated membrane and soluble CD64: a novel marker reflecting altered FcyR function and disease in early rheumatoid arthritis that can be regulated by anti-rheumatic treatment. PLoS One. 2015; 10(9):e0137474.

  103. Rihova B, Kovar M. Immunogenicity and immunomodulatory properties of HPMA-based polymers. Adv Drug Deliv Rev. 2010;62(2):184-91.

  104. Wang D, Miller SC, Sima M, Parker D, Buswell H, Goodrich KC, Kope P. The arthrotropism of macromolecules in adjuvant-induced arthritis rat model: a preliminary study. Pharm Res. 2004;21(10):1741-9.

  105. Wang D, Miller SC, Liu XM, Anderson B, Wang XS, Goldring SR. Novel dexamethasone-HPMA copolymer conjugate and its potential application in treatment of rheumatoid arthritis. Arthritis Res Ther. 2007;9(1):R2.

  106. Liu XM, Quan LD, Tian J, Alnouti Y, Fu K, Thiele GM, Wang D. Synthesis and evaluation of a well-defined HPMA copolymer-dexamethasone conjugate for effective treatment of rheumatoid arthritis. Pharm Res. 2008;25(12):2910-9.

  107. Quan LD, Purdue PE, Liu XM, Boska MD, Lele SM, Thiele GM, Mikuls TR, Dou H, Goldring SR, Wang D. Development of a macromolecular prodrug for the treatment of inflammatory arthritis: mechanisms involved in arthrotropism and sustained therapeutic efficacy. Arthritis Res Ther. 2010;12(5):R170.

  108. Shin JM, Kim SH, Thambi T, You DG, Jeon J, Lee JO, Chung BY, Jo DG, Park JH. A hyaluronic acid-methotrexate conjugate for targeted therapy of rheumatoid arthritis. Chem Commun. 2014;50(57):7632-5.

  109. Crielaard BJ, Rijcken CJ, Quan L, Van Der Wal S, Altintas I, Van Der Pot M, Kruijtzer JA, Liskamp RM, Schiffelers RM, Van Nostrum CF, Hennink WE. Glucocorticoid loaded core cross linked polymeric micelles with tailorable release kinetics for targeted therapy of rheumatoid arthritis. Angewandte Chemie Int Ed. 2012;51(29):7254-8.

  110. Hao F, Lee RJ, Zhong L, Dong S, Yang C, Teng L, Meng Q, Lu J, Xie J, Teng L. Hybrid micelles containing methotrexate-conjugated polymer and co-loaded with microRNA-124 for rheumatoid arthritis therapy. Theranostics. 2019; 9(18):5282.

  111. Qi R, Majoros I, Misra AC, Koch AE, Campbell P, Marotte H, Bergin IL, Cao Z, Goonewardena S, Morry J, Zhang S. Folate receptor-targeted dendrimer-methotrexate conjugate for inflammatory arthritis. J Biomed Nanotechnol. 2015;11(8):1431-41.

  112. Elzoghby AO, Samy WM, Elgindy NA. Albumin-based nanoparticles as potential controlled release drug delivery systems. J Control Release. 2012;157(2):168-82.

  113. Wunder A, Muller-Ladner U, Stelzer EH, Funk J, Neumann E, Stehle G, Pap T, Sinn H, Gay S, Fiehn C. Albumin-based drug delivery as novel therapeutic approach for rheumatoid arthritis. J Immunol. 2003 May 1;170(9):4793-801.

  114. De Keyser F. Choice of biologic therapy for patients with rheumatoid arthritis: the infection perspective. Curr Rheumatol Rev. 2011;7(1):77-87.

  115. Kratz F. A clinical update of using albumin as a drug vehicle: A commentary. J Control Release. 2014;190:331-6.

  116. Fiehn C, Neumann E, Wunder A, Krienke S, Gay S, Muller-Ladner U. Methotrexate (MTX) and albumin coupled with MTX (MTX-HSA) suppress synovial fibroblast invasion and cartilage degradation in vivo. Ann Rheum Dis. 2004;63(7):884-6.

  117. Fiehn C, Kratz F, Sass G, Muller-Ladner U, Neumann E. Targeted Drug Deliv by in vivo coupling to endogenous albumin: an albumin-binding prodrug of methotrexate (MTX) is better than MTX in the treatment of murine collagen-induced arthritis. Ann Rheum Dis. 2008;67(8):1188-91.

  118. Okada Y, Wu D, Trynka G, Raj T, Terao C, Ikari K, Kochi Y, Ohmura K, Suzuki A, Yoshida S, Graham RR. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature. 2014;506(7488):376-81.

  119. Noss EH, Chang SK, Watts GF, Brenner MB. Modulation of matrix metalloproteinase production by rheumatoid arthritis synovial fibroblasts after cadherin 11 engagement. Arthritis Rheumatol. 2011;63(12):3768-78.

  120. Lee DM, Kiener HP, Agarwal SK, Noss EH, Watts GF, Chisaka O, Takeichi M, Brenner MB. Cadherin-11 in synovial lining formation and pathology in arthritis. Science. 2007;315(5814):1006-10.

  121. Brooks PM. Impact of osteoarthritis on individuals and society: how much disability? Social conse-quences and health economic implications. Curr Opin Rheumatol. 2002;14(5):573-7.

  122. Elders MJ. The increasing impact of arthritis on public health. J Rheumatol Suppl. 2000;60:6-8.

  123. Erdemli O, Ozen S, Keskin D, Usanmaz A, Batu ED, Atilla B, Tezcaner A. In vitro evaluation of effects of sustained anti-TNF release from MPEG-PCL-MPEG and PCL microspheres on human rheumatoid arthritis synoviocytes. J Biomater Appl. 2014;29(4):524-42.

  124. Venkatesh MP, Liladhar PK, Kumar TMP, Shivakumar HG. In situ gels based drug delivery systems. Curr Drug Therap. 2011;6(3):213-22.

  125. Venkatesh MP, Anis S, Kumar TP. Design and development of an injectable in situ forming drug delivery system of methotrexate for the treatment of rheumatoid arthritis. J Drug Deliv Sci. 2013;23(5):445-53.

  126. Natarajan V, Krithica N, Madhan B, Sehgal PK. Formulation and evaluation of quercetin polycaprolactone microspheres for the treatment of rheumatoid arthritis. J Pharm Sci. 2011;100(1):195-205.

  127. Shinde CG, Venkatesh MP, Rajesh KS, Srivastava A, Osmani RA, Sonawane YH. Intra-articular delivery of a methotrexate loaded nanostructured lipid carrier based smart gel for effective treatment of rheumatic diseases. RSC Adv. 2016;6(16):12913-24.

  128. He J, Li H, Liu C, Wang G, Ge L, Ma S, Huang L, Yan S, Xu X. Formulation and evaluation of poly (lactic-co-glycolic acid) microspheres loaded with an altered collagen type II peptide for the treatment of rheumatoid arthritis. J Microencapsul. 2015;32(6):608-17.

  129. Ramasamy T, Ruttala HB, Shanmugam S, Umadevi SK. Eudragit-coated aceclofenac-loaded pectin microspheres in chronopharmacological treatment of rheumatoid arthritis. Drug Deliv. 2013;20(2):65-77.

  130. Date AA, Desai N, Dixit R, Nagarsenker M. Self-nanoemulsifying Drug Deliv systems: formulation insights, applications and advances. Nanomedicine. 2010;5(10):1595-616.

  131. Chime SA, Kenechukwu FC, Attama AA. Nanoemulsions-advances in formulation, characterization and applications in drug delivery. Application of nanotechnology in drug delivery. London: Intech Open Limited; 2014; p.78-126.

  132. Hoscheid J, Outuki PM, Kleinubing SA, Silva MF, Bruschi ML, Cardoso ML. Development and characterization of Pterodon pubescens oil nanoemulsions as a possible delivery system for the treatment of rheumatoid arthritis. Colloids Surf A: Physicochem Eng Aspects. 2015;484:19-27.

  133. Parekh VJ, Desai ND, Shaikh MS, Shinde UA. Self-nanoemulsifying granules (SNEGs) of meloxicam: preparation, characterization, molecular modeling and evaluation of in vivo anti-inflammatory activity. Drug Dev Ind Pharm. 2017;43(4):600-10.

  134. Sriamornsak P, Limmatvapirat S, Piriyaprasarth S, Mansukmanee P, Huang Z. A new self-emulsifying formulation of mefenamic acid with enhanced drug dissolution. Asian J Pharm Sci. 2015;10(2): 121-7.

  135. Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4(2):145-60.

  136. Wang EC, Wang AZ. Nanoparticles and their applications in cell and molecular biology. Integr Biol. 2014;6(1):9-26.

  137. Pham CT. Nanotherapeutic approaches for the treatment of rheumatoid arthritis. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2011;3(6):607-19.

  138. Srinath P, Chary MG, Vyas SP, Diwan PV. Long-circulating liposomes of indomethacin in arthritic rats: a biodisposition study. Pharm Acta Helv. 2000;74(4):399-404.

  139. Palakurthi S, Vyas SP, Diwan PV. Biodisposition of PEG-coated lipid microspheres of indomethacin in arthritic rats. Int J Pharm. 2005;290(1-2):55-62.

  140. Bernardi A, Zilberstein AC, Jager E, Campos MM, Morrone FB, Calixto JB, Pohlmann AR, Guterres SS, Battastini AM. Effects of indomethacin loaded nanocapsules in experimental models of inflammation in rats. Br J Pharmacol. 2009;158(4):1104-11.

  141. Metselaar JM, Wauben MH, Wagenaar Hilbers J, Boerman OC, Storm G. Complete remission of experimental arthritis by joint targeting of glucocorticoids with long-circulating liposomes. Arthritis Rheumatol. 2003;48(7):2059-66.

  142. Koo OM, Rubinstein I, Onyuksel H. Actively targeted low-dose camptothecin as a safe, long-acting, disease-modifying nanomedicine for rheumatoid arthritis. Pharm Res. 2011;28(4):776-87.

  143. Shende P, Salunke M. Transepidermal microneedles for co-administration of folic acid with metho-trexate in the treatment of rheumatoid arthritis. Biomed Phys Eng Express. 2019; 5(2):025023.

  144. Khoury M, Louis Plence P, Escriou V, Noel D, Largeau C, Cantos C, Scherman D, Jorgensen C, Apparailly F. Efficient new cationic liposome formulation for systemic delivery of small interfering RNA silencing tumor necrosis factor a in experimental arthritis. Arthritis Rheumatol. 2006;54(6):1867-77.

  145. Ulmansky R, Turjeman K, Baru M, Katzavian G, Harel M, Sigal A, Naparstek Y, Barenholz Y. Glucocorticoids in nano-liposomes administered intravenously and subcutaneously to adjuvant arthritis rats are superior to the free drugs in suppressing arthritis and inflammatory cytokines. J Control Release. 2012; 160(2):299-305.

  146. Arora R, Kuhad A, Kaur IP, Chopra K. Curcumin loaded solid lipid nanoparticles ameliorate adjuvant induced arthritis in rats. Eur J Pain. 2015;19(7):940-52.

  147. Bhalekar MR, Madgulkar AR, Desale PS, Marium G. Formulation of piperine solid lipid nanoparticles (SLN) for treatment of rheumatoid arthritis. Drug Dev Ind Pharm. 2017;43(6):1003-10.

  148. Garg NK, Tyagi RK, Singh B, Sharma G, Nirbhavane P, Kushwah V, Jain S, Katare OP. Nanostructured lipid carrier mediates effective delivery of methotrexate to induce apoptosis of rheumatoid arthritis via NF-KB and FOXO1. Int J Pharm. 2016;499(1-2):301-20.

  149. Shah RM, Eldridge DS, Palombo EA, Harding IH. Microwave-assisted formulation of solid lipid nanoparticles loaded with non-steroidal anti-inflammatory drugs. Int J Pharm. 2016;515(1-2):543-54.

  150. Ye J, Wang Q, Zhou X, Zhang N. Injectable actarit-loaded solid lipid nanoparticles as passive targeting therapeutic agents for rheumatoid arthritis. Int J Pharm. 2008;352(1-2):273-9.

  151. Torchilin VP. Micellar nanocarriers: pharmaceutical perspectives. Pharm Res. 2007;24(1):1-16.

  152. Grobmyer SR, Iwakuma N, Sharma P, Moudgil BM. What is cancer nanotechnology? Methods Mol Biol. 2010;624:1-9.

  153. Hwang J, Rodgers K, Oliver JC, Schluep T. a-Methylprednisolone conjugated cyclodextrin polymer-based nanoparticles for rheumatoid arthritis therapy. Int J Nanomed. 2008;3(3):359-71.

  154. Nagai N, Ito Y. Effect of solid nanoparticle of indomethacin on therapy for rheumatoid arthritis in adjuvant-induced arthritis rat. Biol Pharm Bull. 2014;37(7):1109-18.

  155. Nagai N, Yoshioka C, Ito Y. Topical therapies for rheumatoid arthritis by gel ointments containing indomethacin nanoparticles in adjuvant-induced arthritis rat. J Oleo Sci. 2015;64(3):337-46.

  156. Kim Y, Park EJ, Na DH. Recent progress in dendrimer-based nanomedicine development. Arch Pharmacal Res. 2018; 41(6):571-82.

  157. Yang M, Ding J, Zhang Y, Chang F, Wang J, Gao Z, Zhuang X, Chen X. Activated macrophage-targeted dextran-methotrexate/folate conjugate prevents deterioration of collagen-induced arthritis in mice. J Mater Chem B. 2016; 4(12):2102-13.

  158. Colombo F, Nunez L, Durigutto P, Biffi S, Rampazzo E, Belmonte B, Sblattero D, Gulino A, Meroni PL, Tedesco F, Macor P. OP0023 targeted polymeric nanoparticles as diagnostic and therapeutic tool for rheumatoid arthritis. Ann Rheum Dis. 2016;75:61.

  159. Heo R, You DG, Um W, Choi KY, Jeon S, Park JS, Choi Y, Kwon S, Kim K, Kwon IC, Jo DG. Dextran sulfate nanoparticles as a theranostic nanomedicine for rheumatoid arthritis. Biomaterials. 2017;131:15-26.

  160. Alam MM, Han HS, Sung S, Kang JH, Sa KH, Al Faruque H, Hong J, Nam EJ, San Kim I, Park JH, Kang YM. Endogenous inspired biomineral-installed hyaluronan nanoparticles as pH-responsive carrier of methotrexate for rheumatoid arthritis. J Control Release. 2017;252:62-72.

  161. Dewangan AK, Perumal Y, Pavurala N, Chopra K, Mazumder S. Preparation, characterization and anti-inflammatory effects of curcumin loaded carboxymethyl cellulose acetate butyrate nanoparticles on adjuvant induced arthritis in rats. J Drug Deliv Sci. 2017;41:269-79.

  162. Sun W, Zhang H, Wang H, Chiu YG, Wang M, Ritchlin CT, Kiernan A, Boyce BF, Xing L. Targeting Notch-activated M1 macrophages attenuates joint tissue damage in a mouse model of inflammatory arthritis. J Bone Miner Res. 2017;32(7):1469-80.

  163. Heo R, Park JS, Jang HJ, Kim SH, Shin JM, Suh YD, Jeong JH, Jo DG, Park JH. Hyaluronan nano-particles bearing y-secretase inhibitor: in vivo therapeutic effects on rheumatoid arthritis. J Control Release. 2014;192:295-300.

  164. Zhou HF, Chan HW, Wickline SA, Lanza GM, Pham CT. avp3 Targeted nanotherapy suppresses inflammatory arthritis in mice. FASEB J. 2009;23(9):2978-85.

  165. Ma AC, Fung TK, Lin RH, Chung MI, Yang D, Ekker SC, Leung AY. Methionine aminopeptidase 2 is required for HSC initiation and proliferation. Blood. 2011;118(20):5448-57.

  166. Zhou HF, Hu G, Wickline SA, Lanza GM. CT Pham, Synergistic effect of antiangiogenic nanotherapy combined with methotrexate in the treatment of experimental inflammatory arthritis. Nanomedicine. 2010;5(7):1065-74.

  167. Kim MJ, Park JS, Lee SJ, Jang J, Park JS, Back SH, Bahn G, Park JH, Kang YM, Kim SH, Kwon IC. Notch1 targeting siRNA delivery nanoparticles for rheumatoid arthritis therapy. J Control Release. 2015;216:140-8.

  168. Lee SJ, Lee A, Hwang SR, Park JS, Jang J, Huh MS, Jo DG, Yoon SY, Byun Y, Kim SH, Kwon IC. TNF-a gene silencing using polymerized siRNA/thiolated glycol chitosan nanoparticles for rheumatoid arthritis. Mol Ther. 2014;22(2):397-408.

  169. Fernandes JC, Wang H, Jreyssaty C, Benderdour M, Lavigne P, Qiu X, Winnik FM, Zhang X, Dai K, Shi Q. Bone-protective effects of nonviral gene therapy with folate-chitosan DNA nanoparticle containing interleukin-1 receptor antagonist gene in rats with adjuvant-induced arthritis. Mol Ther. 2008;16(7):1243-51.

  170. Courties G, Baron M, Presumey J, Escriou V, van Lent PL, Scherman D, Cantagrel A, van den Berg WB, Jorgensen C, Apparailly F, Davignon JL. Cytosolic phospholipase A2a gene silencing in the myeloid lineage alters development of Th1 responses and reduces disease severity in collagen induced arthritis. Arthritis Rheumatol. 2011;63(3):681-90.

  171. Samarasinghe RM, Kanwar RK, Kanwar JR. The effect of oral administration of iron saturated- bovine lactoferrin encapsulated chitosan-nanocarriers on osteoarthritis. Biomaterials. 2014;35(26): 7522-34.

  172. Elsadek B, Kratz F. Impact of albumin on drug delivery: new applications on the horizon. J Control Release. 2012;157(1):4-28.

  173. Gradishar WJ, Tjulandin S, Davidson N, Shaw H, Desai N, Bhar P, Hawkins M, O'Shaughnessy J. Phase III trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based paclitaxel in women with breast cancer. J Clin Oncol. 2005;23(31):7794-803.

  174. Hawkins MJ, Soon Shiong P, Desai N. Protein nanoparticles as drug carriers in clinical medicine. Adv Drug Deliv Rev. 2008;60(8):876-85.

  175. Fiehn C, Muller-Ladner U, Gay S, Krienke S, Freudenberg-Konrad S, Funk J, Ho AD, Sinn H, Wun der A. Albumin-coupled methotrexate (MTX-HSA) is a new anti-arthritic drug which acts synergistically to MTX. Rheumatology. 2004; 43(9):1097-105.

  176. Patel P, Patel H, Panchal S, Mehta T. Formulation strategies for drug delivery of tacrolimus: an overview. Int J Pharm Investig. 2012;2(4):169-75.

  177. Byeon HJ, Lee C, Lee S, Lee ES, Choi HG, Park ES, Youn YS. Pharmaceutical potential of tacrolimus-loaded albumin nanoparticles having targetability to rheumatoid arthritis tissues. Int J Pharm. 2016;497(1-2):268-76.

  178. Coppieters K, Dreier T, Silence K, Haard HD, Lauwereys M, Casteels P, Beirnaert E, Jonckheere H, Wiele CV, Staelens L, Hostens J. Formatted antitumor necrosis factor a VHH proteins derived from camelids show superior potency and targeting to inflamed joints in a murine model of collagen induced arthritis. Arthritis Rheumatol. 2006;54(6):1856-66.

  179. Ren K, Dusad A, Dong R, Quan L. Albumin as a delivery carrier for rheumatoid arthritis. Nanome-dicine. 2013;4(176):2.

  180. Liu M, Huang Y, Hu L, Liu G, Hu X, Liu D, Yang X. Selective delivery of interleukine-1 receptor antagonist to inflamed joint by albumin fusion. BMC Biotechnol. 2012;12(1):68.

  181. Hoes JN, Jacobs JW, Buttgereit F, Bijlsma JW. Current view of glucocorticoid co-therapy with DMARDs in rheumatoid arthritis. Nat Rev Rheumatol. 2010;6(12):693-702.

  182. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Preat V. PLGA-based nanoparticles: an overview of biomedical applications. J Control Release. 2012;161(2):505-22.

  183. Weissleder R. Molecular imaging in cancer. Science. 2006;312(5777):1168-71.

  184. Wang YX, Hussain SM, Krestin GP. Superparamagnetic iron oxide contrast agents: physicochemical characteristics and applications in MR imaging. Eur Radiol. 2001;11(11):2319-31.

  185. Zhao M, Beauregard DA, Loizou L, Davletov B, Brindle KM. Non-invasive detection of apoptosis using magnetic resonance imaging and a targeted contrast agent. Nat Med. 2001;7(11):1241-4.

  186. Ringe J, Sittinger M. Tissue engineering in the rheumatic diseases. Arthritis Res Ther. 2009;11(1):211.

  187. Bouffi C, Djouad F, Mathieu M, Noel D, Jorgensen C. Multipotent mesenchymal stromal cells and rheumatoid arthritis: risk or benefit? Rheumatology. 2009;48(10):1185-9.

  188. Chen Y, Shao JZ, Xiang LX, Dong XJ, Zhang GR. Mesenchymal stem cells: a promising candidate in regenerative medicine. Int J Biochem Cell Biol. 2008;40(5):815-20.

  189. Jones BJ, McTaggart SJ. Immunosuppression by mesenchymal stromal cells: from culture to clinic. Exp Hematol. 2008;36(6):733-41.

  190. Markides H, Kehoe O, Morris RH, El Haj AJ. Whole body tracking of superparamagnetic iron oxide nanoparticle-labelled cells-a rheumatoid arthritis mouse model. Stem Cell Res Ther. 2013;4(5):126.

  191. Saldanha KJ, Doan RP, Ainslie KM, Desai TA, Majumdar S. Micrometer-sized iron oxide particle labeling of mesenchymal stem cells for J Magn Reson Imaging-based monitoring of cartilage tissue engineering. J Magn Reson Imaging. 2011;29(1):40-9.

  192. He G, Zhang H, Wei H, Wang Y, Zhang X, Tang Y, Wei Y, Hu S. In vivo imaging of bone marrow mesenchymal stem cells transplanted into myocardium using magnetic resonance imaging: a novel method to trace the transplanted cells. Int J Cardiol. 2007;114(1):4-10.

  193. Kim TH, Kim JK, Shim W, Kim SY, Park TJ, Jung JY. Tracking of transplanted mesenchymal stem cells labeled with fluorescent magnetic nanoparticle in liver cirrhosis rat model with 3-T MRI. J Magn Reson Imaging. 2010;28(7):1004-13.

  194. Kim HS, Oh SY, Joo HJ, Son KR, Song IC, Moon WK. The effects of clinically used MRI contrast agents on the biological properties of human mesenchymal stem cells. NMR Biomed. 2010;23(5):514-22.

  195. Duan J, Dong J, Zhang T, Su Z, Ding J, Zhang Y, Mao X. Polyethyleneimine-functionalized iron oxide nanoparticles for systemic siRNA delivery in experimental arthritis. Nanomedicine. 2014;9(6): 789-801.

  196. Huang X, Jain PK, El-Sayed IH, El-Sayed MA. Gold nanoparticles: interesting optical properties and recent applications in cancer diagnostics and therapy. Nanomedicine (Lond). 2007;2(5):681-93.

  197. Peng G, Tisch U, Adams O, Hakim M, Shehada N, Broza YY, Billan S, Abdah-Bortnyak R, Kuten A, Haick H. Diagnosing lung cancer in exhaled breath using gold nanoparticles. Nat Nanotechnol. 2009;4(10):669-73.

  198. Liu X, Dai Q, Austin L, Coutts J, Knowles G, Zou J, Chen H, Huo Q. A one-step homogeneous immunoassay for cancer biomarker detection using gold nanoparticle probes coupled with dynamic light scattering. J Am Chem Soc. 2008;130(9):2780-2.

  199. Huang X, El-Sayed IH, Qian W, El-Sayed MA. Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. J Am Chem Soc. 2006;128(6):2115-20.

  200. Mukherjee P, Bhattacharya R, Wang P, Wang L, Basu S, Nagy JA, Atala A, Mukhopadhyay D, Soker S. Antiangiogenic properties of gold nanoparticles. Clin Cancer Res. 2005;11(9):3530-4.

  201. Brown CL, Whitehouse MW, Tiekink ER, Bushell GR. Colloidal metallic gold is not bioinert. Inflammopharmacology. 2008;16(3):133-7.

  202. Ma JS, Kim WJ, Kim JJ, Kim TJ, Ye SK, Song MD, Kang H, Kim DW, Moon WK, Lee KH. Gold nanoparticles attenuate LPS-induced NO production through the inhibition of NF-KB and IFN-p/ STAT1 pathways in RAW264. 7 cells. Nitric Oxide. 2010;23(3):214-9.

  203. James LR, Xu ZQ, Sluyter R, Hawksworth EL, Kelso C, Lai B, Paterson DJ, de Jonge MD, Dixon NE, Beck JL, Ralph SF. An investigation into the interactions of gold nanoparticles and anti-arthritic drugs with macrophages, and their reactivity towards thioredoxin reductase. J Inorg Biochem. 2015;142:28-38.

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

  205. Lee SM, Kim HJ, Ha YJ, Park YN, Lee SK, Park YB, Yoo KH. Targeted chemo-photothermal treatments of rheumatoid arthritis using gold half-shell multifunctional nanoparticles. ACS Nano. 2012;7(1):50-7.

  206. Jaikumar P, Le P, Smith AM. Single-cell, single-molecule analysis of TNF-alpha signaling with quantum dots. 2017. Available from: http://hdl.handle.net/2142/98485.

  207. Jeyadevi R, Sivasudha T, Rameshkumar A, Ananth DA, Aseervatham GS, Kumaresan K, Kumar LD, Jagadeeswari S, Renganathan R. Enhancement of anti-arthritic effect of quercetin using thioglycolic acid-capped cadmium telluride quantum dots as nanocarrier in adjuvant induced arthritic Wistar rats. Colloids Surf B. 2013;112:255-63.

  208. Lee ES, Deepagan VG, You DG, Jeon J, Yi GR, Lee JY, Lee DS, Suh YD, Park JH. Nanoparticles based on quantum dots and a luminol derivative: implications for in vivo imaging of hydrogen peroxide by chemiluminescence resonance energy transfer. Chem Commun. 2016;52(22):4132-5.

  209. Wang Y, Zhao X, Ruan J. Transdermal Drug Deliv system of aceclofenac for rheumatoid arthritis and the effect of permeation enhancers: in vitro and in vivo characterization. Int J Pharmacol. 2015;11(5):456-62.

  210. Das T, Prakash VM, Math PK. Injectable in situ gel of methotrexate for rheumatoid arthritis: Development, in vitro and in vivo evaluation. J Appl Pharm Sci. 2019; 9(05):040-8.

  211. 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(8):1132-6.

  212. Ishihara T, Kubota T, Choi T, Higaki M. Treatment of experimental arthritis with stealth-type polymeric nanoparticles encapsulating betamethasone phosphate. J Pharmacol Exp Ther. 2009;329(2):412-7.

  213. Lodzki M, Godin B, Rakou L, Mechoulam R, Gallily R, Touitou E. Cannabidiol-transdermal delivery and anti-inflammatory effect in a murine model. J Control Release. 2003;93(3):377-87.

  214. Dangol M, Yang H, Li CG, Lahiji SF, Kim S, Ma Y, Jung H. Innovative polymeric system (IPS) for solvent-free lipophilic drug transdermal delivery via dissolving microneedles. J Control Release. 2016;223:118-25.

  215. Monkkonen J, Taskinen M, Auriola SO, Urtti A. Growth inhibition of macrophage-like and other cell types by liposome-encapsulated, calcium-bound, and free bisphosphonates in vitro. J Drug Target. 1994;2(4):299-308.

  216. Bader RA, Silvers AL, Zhang N. Polysialic acid-based micelles for encapsulation of hydrophobic drugs. Biomacromolecules. 2011;12(2):314-20.

  217. Wang Q, Jiang J, Chen W, Jiang H, Zhang Z, Sun X. Targeted delivery of low-dose dexamethasone using PCL-PEG micelles for effective treatment of rheumatoid arthritis. J Control Release. 2016;230:64-72.

  218. Chandrashekar G, Udupa N. Biodegradable injectable implant systems for long term drug deliv using poly (lactic co glycolic) acid copolymers. J Pharm Pharmacol. 1996;48(7):669-74.

  219. El-Newehy MH, El-Naggar ME, Alotaiby S, El-Hamshary H, Moydeen M, Al-Deyab S. Preparation of biocompatible system based on electrospun CMC/PVA nanofibers as controlled release carrier of diclofenac sodium. J Macromol Sci A. 2016;53(9):566-73.

  220. Pignatello, Amico D, Chiechio S, Spadaro C, Puglisi G, Giunchedi PR. Preparation and analgesic activity of Eudragit RS100.

  221. Berliner MN, Giesecke T, Bornhovd KD. Impact of transdermal fentanyl on quality of life in rheumatoid arthritis. Clin J Pain. 2007;23(6):530-4.

  222. Brown KE, Leong K, Huang CH, Dalal R, Green GD, Haimes HB, Jimenez PA, Bathon J. Gelatin/chondroitin 6-sulfate microspheres for the delivery of therapeutic proteins to the joint. Arthritis Rheum. 1998;41(12):2185-95.

  223. Li C, Li H, Wang Q, Zhou M, Li M, Gong T, Zhang Z, Sun X. pH-sensitive polymeric micelles for targeted delivery to inflamed joints. J Control Release. 2017;246:133-41.

  224. 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):504-12.

  225. Jessy S, Vishal P. Novel floating pulsatile approach for chronotherapeutic release of indomethacin. Dhaka University J Pharm Sci. 2007;6(1):37-41.

  226. Katare OP, Vyas SP, Dixit VK. Effervescent granule based proliposomes of ibuprofen. J Microen-capsul. 1990;7(4):455-62.

  227. Edwards SH, Cake MA, Spoelstra G, Read RA. Biodistribution and clearance of intra-articular liposomes in a large animal model using a radiographic marker. J Liposome Res. 2007;17(3-4):249-61.

  228. Park JY, Lee IH. Controlled release of ketoprofen from electrospun porous polylactic acid (PLA) nanofibers. J Polymer Res. 2011;18(6):1287-91.

  229. Amodwala S, Kumar P, Thakkar HP. Statistically optimized fast dissolving microneedle transdermal patch of meloxicam: A patient friendly approach to manage arthritis. Eur J Pharm Sci. 2017;104: 114-23.

  230. Sharma S, Pawar A. Low density multiparticulate system for pulsatile release of meloxicam. Int J Pharm. 2006;313(1):150-8.

  231. Albuquerque J, Moura CC, Sarmento B, Reis S. Solid lipid nanoparticles: a potential multifunctional approach towards rheumatoid arthritis theranostics. Molecules. 2015;20(6):11103-18.

  232. Zhao J. Hyaluronic Acid-Modified and TPCA-1-loaded gold nanocages alleviate inflammation. Pharmaceutics. 2019; 11(3):143.

  233. O'Mary H, Cui Z. Nanomedicine for intra-articular drug delivery in rheumatoid arthritis. Curr Med Chem. 2016; 23(23):2490-506.

  234. Metselaar JV, Van den Berg WB, Holthuysen AE, Wauben MH, Storm GV, Van Lent PL. Liposomal targeting of glucocorticoids to synovial lining cells strongly increases therapeutic benefit in collagen type II arthritis. Ann Rheum Dis. 2004; 63(4):348-53.

  235. Jeong JC, Lee J, Cho K. Effects of crystalline microstructure on drug release behavior of poly (s-caprolactone) microspheres. J Control Release. 2003; 92(3):249-58.

  236. Cao Y, Tao Y, Zhou Y, Gui S. Development of sinomenine hydrochloride-loaded polyvinylalcohol/maltose microneedle for transdermal delivery. J Drug Deliv Sci. 2016;35:1-7.

  237. Corvo ML, Boerman OC, Oyen WJ, Van Bloois L, Cruz ME, Crommelin DJ, Storm G. Intravenous administration of superoxide dismutase entrapped in long circulating liposomes. II. In vivo fate in a rat model of adjuvant arthritis. Biochim Biophys Acta Biomembr. 1999;1419(2):325-34.

  238. Jafari S, Maleki-Dizaji N, Barar J, Barzegar-Jalali M, Rameshrad M, Adibkia K. Physicochemical characterization and in vivo evaluation of triamcinolone acetonide-loaded hydroxyapatite nanocomposites for treatment of rheumatoid arthritis. Colloids Surf B Biointerfaces. 2016;140:223-32.

  239. Johns Hopkins Medicine Arthritis Centre. [Home page on the Internet] [cited 2018 Feb 7]. Available from: https://www.hopkinsarthritis.org/.

  240. Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov. 2010;9(8):615-27.

  241. Hong K, Guo LS, Tseng YL, Shih SF, Chang PC, Tsai CC, Lin HH, inventors; Taiwan Liposome Company Ltd., TLC Biopharmaceuticals Inc., assignees. Methods of treating arthritis. US Patent 9,789,062. 2017 Oct 17.

  242. Ping H, Xiuli Y, Waves S, Cheng Le L, Chunhui D, inventors; Huang Ping, assignee. Anti-rheumatoid arthritis drug gel containing paclitaxel liposome and preparation method of gel. China Patent Application CN104688721A. 2015 June 10.

  243. Diorio C, Lokhnauth J, inventors; Capsugel Belgium NV, assignee. Curcumin solid lipid particles and methods for their preparation and use. US Patent 14/771,117. 2016 Jan 7.

  244. ClinicalTrials.gov. Study of KB003 in biologics inadequate rheumatoid arthritis [cited 2018 Feb 7]. Available from: https://clinicaltrials.gov/ct2/results?term=NCT00995449.

  245. Sachdeva MS, Patlolla R, inventors; Florida Agricultural, Mechanical University, assignee. Nanoparticle formulations for skin delivery. US Patent 8,715,736. 2014 May 6.

  246. Jaap Gaillard P, RIP J, inventors; Eyesiu Medicines B.V., assignee. PEGylated lipid nanoparticle with bioactive lipophilic compound. World Intellectual Property Organization Patent Application WO 2017025588 A1. 2017 Feb 16.

  247. Clinical Trials.gov. Safety and tolerability of MORAb-022 in healthy and rheumatoid arthritis subjects [cited 2018 Feb 7]. Available from: https://clinicaltrials.gov/ct2/results?term=NCT01357759.

  248. Behrens F, Tak PP, Ostergaard M, Stoilov R, Wiland P, Huizinga TW, Berenfus VY, Vladeva S, Rech J, Rubbert-Roth A, Korkosz M. MOR103, a human monoclonal antibody to granulocyte-macrophage colony-stimulating factor, in the treatment of patients with moderate rheumatoid arthritis: results of a phase Ib/IIa randomised, double-blind, placebo-controlled, dose-escalation trial. Ann Rheum Dis. 2015;74(6):1058-64.

  249. ClinicalTrials.gov. A phase 1 MT203 single-dose study to evaluate safety, PK and PD [cited 2018 Feb 7]. Available from: https://clinicaltrials.gov/ct2/show/NCT02354599.

  250. Huizinga TW, Batalov A, Yablanski K, Stoilov R, Lloyd E, Wagner T, Saurigny D, Souberbielle B, Esfandiari E. First-in-patient study of namilumab, an anti-GM-CSF monoclonal antibody, in active rheumatoid arthritis: results of the PRIORA phase Ib study. Ann Rheum Dis. 2015;74 (Suppl 2):S733.

  251. ClinicalTrials.gov. Dose finding study of namilumab in combination with methotrexate in participants with moderate to severe rheumatoid arthritis (RA) [cited 2018 Feb 7]. Available from: https:// clinicaltrials.gov/ct2/show/NCT02379091.

  252. ClinicalTrials.gov. namilumab vs. adalimumab in participants with moderate to severe early rheumatoid arthritis inadequately responding to methotrexate (TELLUS) [cited 2018 Feb 7]. Available from: https://clinicaltrials.gov/ct2/show/NCT02393378.

  253. Zhuang Y, de Vries DE, Xu Z, Marciniak SJ, Chen D, Leon F, Davis HM, Zhou H. Evaluation of disease-mediated therapeutic protein-drug interactions between an anti-interleukin-6 monoclonal antibody (sirukumab) and cytochrome P450 activities in a phase 1 study in patients with rheumatoid arthritis using a cocktail approach. J Clin Pharmacol. 2015;55(12):1386-94.

  254. Smolen JS, Weinblatt ME, Sheng S, Zhuang Y, Hsu B. Sirukumab, a human anti-interleukin-6 monoclonal antibody: a randomised, 2-part (proof-of-concept and dose-finding), phase II study in patients with active rheumatoid arthritis despite methotrexate therapy. Ann Rheum Dis. 2014;73(9):1616-25.

  255. Grigor C, Capell H, Stirling A, McMahon AD, Lock P, Vallance R, Porter D, Kincaid W. Effect of a treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet. 2004;364(9430):263-9.

  256. Van Vollenhoven RF, Ernestam S, Geborek P, Petersson IF, Coster L, Waltbrand E, Zickert A, Theander J, Thorner A, Hellstrom H, Teleman A. Addition of infliximab compared with addition of sulfasalazine and hydroxychloroquine to methotrexate in patients with early rheumatoid arthritis (Swefot trial): 1-year results of a randomised trial. Lancet. 2009;374(9688):459-66.

  257. Rantalaiho V, Korpela M, Laasonen L, Kautiainen H, Jarvenpaa S, Hannonen P, Leirisalo-Repo M, Blafield H, Puolakka K, Karjalainen A, Mottonen T. Early combination disease-modifying antirheumatic drug therapy and tight disease control improve long-term radiologic outcome in patients with early rheumatoid arthritis: the 11-year results of the Finnish Rheumatoid Arthritis Combination Therapy trial. Arthritis Res Ther. 2010;12(3):R122.

  258. Klareskog L, van der Heijde D, de Jager JP, Gough A, Kalden J, Malaise M, Mola EM, Pavelka K, Sany J, Settas L, Wajdula J. Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial. Lancet. 2004;363(9410):675-81.

  259. Klarenbeek NB, Guler Yuksel M, Van der Kooij SM, Han KH, Ronday HK, Kerstens PJ, Seys PE, Huizinga TW, Dijkmans BA, Allaart CF. The impact of four dynamic, goal-steered treatment strategies on the 5-year outcomes of rheumatoid arthritis patients in the BeSt study. Ann Rheum Dis. 2011;70(6):1039-46.

  260. Lipsky PE, Van der Heijde DM, St. Clair EW, Furst DE, Breedveld FC, Kalden JR, Smolen JS, Weisman M, Emery P, Feldmann M, Harriman GR. Infliximab and methotrexate in the treatment of rheumatoid arthritis. N Engl J Med. 2000;343(22):1594-602.

  261. Moreland LW, Odell JR, Paulus HE, Curtis JR, Bathon JM, St Clair EW, Bridges SL, Zhang J, McVie T, Howard G, Van der Heijde D, Cofield SS; TEAR Investigators. A randomized comparative effectiveness study of oral triple therapy versus etanercept plus methotrexate in early aggressive rheumatoid arthritis: the Treatment of Early Aggressive Rheumatoid Arthritis Trial. Arthritis Rheumatol. 2012;64(9):2824-35.

  262. Breedveld FC, Weisman MH, Kavanaugh AF, Cohen SB, Pavelka K, Vollenhoven RV, Sharp J, Perez JL, Spencer-Green GT. The PREMIER study: a multicenter, randomized, double-blind clinical trial of combination therapy with adalimumab plus methotrexate versus methotrexate alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis who had not had previous methotrexate treatment. Arthritis Rheum. 2006;54(1):26-37.

CITADO POR
  1. Shinde Chetan, Venkatesh Madhugiri Prakash, Pramod Kumar Tegginmat, Pai Deeksha Ramananda, Nanostructured lipid carrier-based smart gel: a delivery platform for intra-articular therapeutics, Autoimmunity, 54, 1, 2021. Crossref

  2. Venkatesh Madhugiri Prakash, Kumar Tegginmat Pramod, Pai Deeksha Ramananda, Targeted drug delivery of Methotrexate in situ gels for the treatment of Rheumatoid Arthritis, Saudi Pharmaceutical Journal, 28, 12, 2020. Crossref

Portal Digitalde Biblioteca Digital eLibros Revistas Referencias y Libros de Ponencias Colecciones Precios y Políticas de Suscripcione Begell House Contáctenos Language English 中文 Русский Português German French Spain