440
Views
5
CrossRef citations to date
0
Altmetric
Review

Recent progress in therapeutic strategies and biomimetic nanomedicines for rheumatoid arthritis treatment

, , , , &
Pages 883-898 | Received 18 Mar 2022, Accepted 22 Jun 2022, Published online: 05 Jul 2022

References

  • Burmester GR, Pope JE. Novel treatment strategies in rheumatoid arthritis. Lancet. 2017;389(10086):2338–2348.
  • Aletaha D, Smolen JS. Diagnosis and management of rheumatoid arthritis: a review. Jama. 2018;320(13):1360–1372.
  • Alam J, Jantan I, Bukhari SNA. Rheumatoid arthritis: recent advances on its etiology, role of cytokines and pharmacotherapy. Biomed Pharmacothe. 2017;92:615–633.
  • Littlejohn EA, Monrad SU. Early diagnosis and treatment of rheumatoid arthritis. Prim Care. 2018;45(2):237–255.
  • Hyndman IJ. Rheumatoid arthritis: past, present and future approaches to treating the disease. Int J Rheum Dis. 2017;20(4):417–419.
  • Jeong M, Park JH. Nanomedicine for the treatment of rheumatoid arthritis. Mol Pharm. 2021;18(2):539–549.
  • McInnes IB, Schett G. Cytokines in the pathogenesis of rheumatoid arthritis. Nat Rev Immunol. 2007;7(6):429–442.
  • Weyand CM, Fujii H, Shao L, et al. Rejuvenating the immune system in rheumatoid arthritis. Nat Rev Rheumatol. 2009;5(10):583–588.
  • Wang Q, Sun X. Recent advances in nanomedicines for the treatment of rheumatoid arthritis. Biomater Sci. 2017;5(8):1407–1420.
  • McInnes IB, Schett G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet. 2017;389(10086):2328–2337.
  • Dale J. Advances in the management of rheumatoid arthritis. Scott Med J. 2015;60(3):108–114.
  • Jin K, Luo Z, Zhang B, et al. Biomimetic nanoparticles for inflammation targeting. Acta Pharm Sin B. 2018;8(1):23–33.
  • Qamar N, Arif A, Bhatti A, et al. Nanomedicine: an emerging era of theranostics and therapeutics for rheumatoid arthritis. Rheumatology (Oxford). 2019;58(10):1715–1721.
  • Belhadj Z, He B, Deng H, et al. A combined ”eat me/don’t eat me” strategy based on extracellular vesicles for anticancer nanomedicine. J Extracell Vesicles. 2020;9(1):1806444.
  • Yang M, Feng X, Ding J, et al. Nanotherapeutics relieve rheumatoid arthritis. J Control Release. 2017;252:108–124.
  • Lundin KE, Simonson OE, Moreno PM, et al. Nanotechnology approaches for gene transfer. Genetica. 2009;137(1):47–56.
  • Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4(2):145–160.
  • Xiao S, Tang Y, Lv Z, et al. Nanomedicine - advantages for their use in rheumatoid arthritis theranostics. J Control Release. 2019;316:302–316.
  • Prasad LK, O’Mary H, Cui Z. Nanomedicine delivers promising treatments for rheumatoid arthritis. Nanomedicine. 2015;10(13):2063–2074.
  • Yuan F, Quan LD, Cui L, et al. Development of macromolecular prodrug for rheumatoid arthritis. Adv Drug Deliv Rev. 2012;64(12):1205–1219.
  • Mirshafiey A, Mohsenzadegan M. The role of reactive oxygen species in immunopathogenesis of rheumatoid arthritis. Iran J Allergy Asthma Immunol. 2008;7(4):195–202.
  • Cheung TT, McInnes IB. Future therapeutic targets in rheumatoid arthritis? Semin Immunopathol. 2017;39(4):487–500.
  • Hua C, Buttgereit F, Combe B. Glucocorticoids in rheumatoid arthritis: current status and future studies. RMD Open. 2020;6(1):e000536.
  • George MD, Baker JF, Winthrop K, et al. Risk for serious infection with low-dose glucocorticoids in patients with rheumatoid arthritis: a cohort study. Ann Intern Med. 2020;173(11):870–878.
  • Kumar LD, Karthik R, Gayathri N, et al. Advancement in contemporary diagnostic and therapeutic approaches for rheumatoid arthritis. Biomed Pharmacothe. 2016;79:52–61.
  • Jorgensen C, Apparailly F. Prospects for gene therapy in inflammatory arthritis. best practice & research. Clin Rheumatol. 2010;24(4):541–552.
  • Smolen JS, Aletaha D. Rheumatoid arthritis therapy reappraisal: strategies, opportunities and challenges. Nat Rev Rheumatol. 2015;11(5):276–289.
  • Wang Q, Qin X, Fang J, et al. Nanomedicines for the treatment of rheumatoid arthritis: state of art and potential therapeutic strategies. Acta Pharm Sin B. 2021;11(5):1158–1174.
  • Pham CT. Nanotherapeutic approaches for the treatment of rheumatoid arthritis. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2011;3(6):607–619.
  • Deng C, Zhang Q, Fu Y, et al. Coadministration of oligomeric hyaluronic acid-modified liposomes with tumor-penetrating peptide-iRGD enhances the antitumor efficacy of doxorubicin against melanoma. ACS Appl Mater Interfaces. 2017;9(2):1280–1292.
  • Zhou M, Hou J, Zhong Z, et al. Targeted delivery of hyaluronic acid-coated solid lipid nanoparticles for rheumatoid arthritis therapy. Drug Deliv. 2018;25(1):716–722.
  • Talelli M, Barz M, Rijcken CJ, et al. Core-Crosslinked polymeric micelles: principles, preparation, biomedical applications and clinical translation. Nano Today. 2015;10(1):93–117.
  • Viatte S, Barton A. Genetics of rheumatoid arthritis susceptibility, severity, and treatment response. Semin Immunopathol. 2017;39(4):395–408.
  • Schett G, Neurath MF. Resolution of chronic inflammatory disease: universal and tissue-specific concepts. Nat Commun. 2018;9(1):3261.
  • Eming SA, Wynn TA, Martin P. Inflammation and metabolism in tissue repair and regeneration science. Science (New York, N.Y.). 2017;356(6342):1026–1030.
  • Wang S, Wang D, Duan Y, et al. Cellular nanosponges for biological neutralization. Adv Mater. 2022;34(13):e2107719.
  • Feldmann M, Williams RO, Paleolog E. What have we learnt from targeted anti-TNF therapy? Ann Rheum Dis. 2010;69(Suppl 1):97–99.
  • Thamphiwatana S, Angsantikul P, Escajadillo T, et al. Macrophage-like nanoparticles concurrently absorbing endotoxins and proinflammatory cytokines for sepsis management. Proc Natl Acad Sci U S A. 2017;114(43):11488–11493.
  • Zhang Q, Dehaini D, Zhang Y, et al. Neutrophil membrane-coated nanoparticles inhibit synovial inflammation and alleviate joint damage in inflammatory arthritis. Nat Nanotechnol. 2018;13(12):1182–1190.
  • Li X, Makarov SS. An essential role of NF-kappaB in the ”tumor-like” phenotype of arthritic synoviocytes. Proc Natl Acad Sci U S A. 2006;103(46):17432–17437.
  • Deng C, Zhang Q, He P, et al. Targeted apoptosis of macrophages and osteoclasts in arthritic joints is effective against advanced inflammatory arthritis. Nat Commun. 2021;12(1):2174.
  • Huang R, Zhang C, Bu Y, et al. A multifunctional nano-therapeutic platform based on octahedral yolk-shell Au NR@CuS: photothermal/photodynamic and targeted drug delivery tri-combined therapy for rheumatoid arthritis. Biomaterials. 2021;277:121088.
  • Li W, Song Y, Liang X, et al. Mutual-reinforcing sonodynamic therapy against rheumatoid arthritis based on sparfloxacin sonosensitizer doped concave-cubic rhodium nanozyme. Biomaterials. 2021;276:121063.
  • Gang F, Zhang Q, Jiang L, et al. Thermochemotherapy meets tissue engineering for rheumatoid arthritis treatment. Adv Funct Mater. 2021;31(40):2104131.
  • He L, He T, Farrar S, et al. Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell Physiol Biochem. 2017;44(2):532–553.
  • Yang B, Chen Y, Shi J. Reactive oxygen species (ROS)-based nanomedicine. Chem Rev. 2019;119(8):4881–4985.
  • Zhang C, Wang X, Du J, et al. Reactive oxygen species-regulating strategies based on nanomaterials for disease treatment. Adv Sci. 2021;8(3):2002797.
  • Kim J, Kim HY, Song SY, et al. Synergistic oxygen generation and reactive oxygen species scavenging by manganese ferrite/ceria co-decorated nanoparticles for rheumatoid arthritis treatment. ACS nano. 2019;13(3):3206–3217.
  • Kalashnikova I, Chung SJ, Nafiujjaman M, et al. Ceria-based nanotheranostic agent for rheumatoid arthritis. Theranostics. 2020;10(26):11863–11880.
  • Yusuf A, Casey A. Liposomal encapsulation of silver nanoparticles (AgNP) improved nanoparticle uptake and induced redox imbalance to activate caspase-dependent apoptosis. Apoptosis. 2020;25(1–2):120–134.
  • Rehman A, John P, Bhatti A. Biogenic selenium nanoparticles: potential solution to oxidative stress mediated inflammation in rheumatoid arthritis and associated complications. Nanomaterials. 2021;11(8):2005.
  • Kumar S, Adjei IM, Brown SB, et al. Manganese dioxide nanoparticles protect cartilage from inflammation-induced oxidative stress. Biomaterials. 2019;224:119467.
  • Sultana F, Neog MK, Rasool M. Withaferin-A, a steroidal lactone encapsulated mannose decorated liposomes ameliorates rheumatoid arthritis by intriguing the macrophage repolarization in adjuvant-induced arthritic rats. Colloids Surf B Biointerfaces. 2017;155:349–365.
  • McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365(23):2205–2219.
  • Ludewig B, Junt T, Hengartner H, et al. Dendritic cells in autoimmune diseases. Curr Opin Immunol. 2001;13(6):657–662.
  • Hu J, Wan Y. Tolerogenic dendritic cells and their potential applications. Immunology. 2011;132(3):307–314.
  • Buckland J. First-in-human phase I trial of DC immunotherapy for early RA. Nat Rev Rheumatol. 2015;11(8):443.
  • Lee ES, Sul JH, Shin JM, et al. Reactive oxygen species-responsive dendritic cell-derived exosomes for rheumatoid arthritis. Acta Biomater. 2021;128:462–473.
  • Fontana F, Albertini S, Correia A, et al. Bioengineered porous silicon Nanoparticles@Macrophages cell membrane as composite platforms for rheumatoid arthritis. Adv Funct Mater. 2018;28(22):1801355.
  • Huang W-C, Ong XC, Kwon IS, et al. Ultracompliant hydrogel‐based neural interfaces fabricated by aqueous‐phase microtransfer printing. Adv Funct Mater. 2018;28(29):1801059.
  • Hoare TR, Kohane DS. Hydrogels in drug delivery: progress and challenges. Polymer. 2008;49(8):1993–2007.
  • Chou CL, Li HW, Lee SH, et al. Effect of intra-articular injection of hyaluronic acid in rheumatoid arthritis patients with knee osteoarthritis. J Chin Med Assoc. 2008;71(8):411–415.
  • Highley CB, Prestwich GD, Burdick JA. Recent advances in hyaluronic acid hydrogels for biomedical applications. Curr Opin Biotechnol. 2016;40:35–40.
  • Guidolin D, Franceschi F. Viscosupplementation with high molecular weight native hyaluronan. focus on a 1500-2000 KDa fraction (Hyalubrix®). Eur Rev Med Pharmacol Sci. 2014;18(21):3326–3338.
  • Barbucci R, Lamponi S, Borzacchiello A, et al. Hyaluronic acid hydrogel in the treatment of osteoarthritis. Biomaterials. 2002;23(23):4503–4513.
  • Seo J, Park SH, Kim MJ, 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.
  • Bilthariya U, Jain N, Rajoriya V, et al. Folate-conjugated albumin nanoparticles for rheumatoid arthritis-targeted delivery of etoricoxib. Drug Dev Ind Pharm. 2015;41(1):95–104.
  • Thomas TP, Goonewardena SN, Majoros IJ, et al. Folate-targeted nanoparticles show efficacy in the treatment of inflammatory arthritis. Arthritis Rheumatism. 2011;63(9):2671–2680.
  • Shin JM, Kim SH, Thambi T, et al. A hyaluronic acid-methotrexate conjugate for targeted therapy of rheumatoid arthritis. Chem Comm. 2014;50(57):7632–7635.
  • Kim YJ, Chae SY, Jin CH, 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.
  • Heo R, You DG, Um W, et al. Dextran sulfate nanoparticles as a theranostic nanomedicine for rheumatoid arthritis. Biomaterials. 2017;131:15–26.
  • Kim SH, Kim JH, You DG, et al. Self-assembled dextran sulphate nanoparticles for targeting rheumatoid arthritis. Chem Comm. 2013;49(88):10349–10351.
  • Lyu J, Wang L, Bai X, et al. Treatment of rheumatoid arthritis by serum albumin nanoparticles coated with mannose to target neutrophils. ACS Appl Mater Interfaces. 2021;13(1):266–276.
  • Yang X, Chang Y, Wei W. Emerging role of targeting macrophages in rheumatoid arthritis: focus on polarization. metabolism and apoptosis. Cell Prolif. 2020;53(7):e12854.
  • 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;12:6735–6746.
  • Verma A, Jain A, Tiwari A, et al. Folate conjugated double liposomes bearing prednisolone and methotrexate for targeting rheumatoid arthritis. Pharm Res. 2019;36(8):123.
  • Alam MM, Han HS, Sung S, et al. Endogenous inspired biomineral-installed hyaluronan nanoparticles as pH-responsive carrier of methotrexate for rheumatoid arthritis. J Control Release. 2017;252:62–72.
  • Aldayel AM, O’Mary HL, Valdes SA, et al. Lipid nanoparticles with minimum burst release of TNF-α siRNA show strong activity against rheumatoid arthritis unresponsive to methotrexate. J Control Release. 2018;283:280–289.
  • Xu XL, Li WS, Wang XJ, et al. Endogenous sialic acid-engineered micelles: a multifunctional platform for on-demand methotrexate delivery and bone repair of rheumatoid arthritis. Nanoscale. 2018;10(6):2923–2935.
  • Wang Y, Han CC, Cui D, et al. Is macrophage polarization important in rheumatoid arthritis? Int Immunopharmacol. 2017;50:345–352.
  • Jain S, Tran TH, Amiji M. Macrophage repolarization with targeted alginate nanoparticles containing IL-10 plasmid DNA for the treatment of experimental arthritis. Biomaterials. 2015;61:162–177.
  • Duan W, Li H. Combination of NF-kB targeted siRNA and methotrexate in a hybrid nanocarrier towards the effective treatment in rheumatoid arthritis. J Nanobiotechnology. 2018;16(1):58.
  • Yu C, Li X, Hou Y, et al. Hyaluronic acid coated acid-sensitive nanoparticles for targeted therapy of adjuvant-induced arthritis in rats. Molecules. 2019;24(1):146.
  • Dou Y, Li C, Li L, et al. Bioresponsive drug delivery systems for the treatment of inflammatory diseases. J Control Release. 2020;327:641–666.
  • Nogueira E, Gomes AC, Preto A, et al. Folate-targeted nanoparticles for rheumatoid arthritis therapy. Nanomedicine. 2016;12(4):1113–1126.
  • Gonçalves RM, Pereira AC, Pereira IO, et al. Macrophage response to chitosan/poly-(γ-glutamic acid) nanoparticles carrying an anti-inflammatory drug. J Mater Sci Mater Med. 2015;26(4):167.
  • Lee YK, Choi JK, Kang YJ, et al. Triamcinolone-carbon nanotube conjugation inhibits inflammation of human arthritis synovial fibroblasts. J Mater Chem B. 2016;4(9):1660–1671.
  • Li C, Li H, Wang Q, et al. pH-sensitive polymeric micelles for targeted delivery to inflamed joints. J Control Release. 2017;246:133–141.
  • Rodriguez PL, Harada T, Christian DA, et al. Minimal ”Self” peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Science. 2013;339(6122):971–975.
  • Kaur S, Singh SP, Elkahloun AG, et al. CD47-dependent immunomodulatory and angiogenic activities of extracellular vesicles produced by T cells. Matrix Biol. 2014;37:49–59.
  • Li P, Yang X, Yang Y, et al. Synergistic effect of all-trans-retinal and triptolide encapsulated in an inflammation-targeted nanoparticle on collagen-induced arthritis in mice. J Control Release. 2020;319:87–103.
  • He L, Qin X, Fan D, et al. Dual-stimuli responsive polymeric micelles for the effective treatment of rheumatoid arthritis. ACS Appl Mater Interfaces. 2021;13(18):21076–21086.
  • Wang Q, Li Y, Chen X, et al. Optimized in vivo performance of acid-liable micelles for the treatment of rheumatoid arthritis by one single injection. Nano Res. 2019;12(2):421–428.
  • Xu XL, Shu GF, Wang XJ, et al. Sialic acid-modified chitosan oligosaccharide-based biphasic calcium phosphate promote synergetic bone formation in rheumatoid arthritis therapy. J Control Release. 2020;323:578–590.
  • Wang L, Zhu B, Huang J, et al. Ultrasound-targeted microbubble destruction augmented synergistic therapy of rheumatoid arthritis via targeted liposomes. J Mat Chem B. 2020;8(24):5245–5256.
  • Hu L, Luo X, Zhou S, et al. Neutrophil-mediated delivery of dexamethasone palmitate-loaded liposomes decorated with a sialic acid conjugate for rheumatoid arthritis treatment. Pharm Res. 2019;36(7):97.
  • Yang Y, Guo L, Wang Z, et al. Targeted silver nanoparticles for rheumatoid arthritis therapy via macrophage apoptosis and Re-polarization. Biomaterials. 2021;264:120390.
  • Colombo F, Durigutto P, De Maso L, et al. Targeting CD34(+) cells of the inflamed synovial endothelium by guided nanoparticles for the treatment of rheumatoid arthritis. J Autoimmun. 2019;103:102288.
  • Liu L, Hu F, Wang H, et al. Secreted protein acidic and rich in cysteine mediated biomimetic delivery of methotrexate by albumin-based nanomedicines for rheumatoid arthritis therapy. ACS nano. 2019;13(5):5036–5048.
  • Byeon HJ, Min SY, Kim I, et al. Human serum albumin-TRAIL conjugate for the treatment of rheumatoid arthritis. Bioconjug Chem. 2014;25(12):2212–2221.
  • Gong T, Tan T, Zhang P, et al. Palmitic acid-modified bovine serum albumin nanoparticles target scavenger receptor-A on activated macrophages to treat rheumatoid arthritis. Biomaterials. 2020;258:120296.
  • Yan F, Li H, Zhong Z, et al. Co-delivery of prednisolone and curcumin in human serum albumin nanoparticles for effective treatment of rheumatoid arthritis. Int J Nanomedicine. 2019;14:9113–9125.
  • Zhong J, Zhang Q, Zhang Z, et al. Albumin mediated reactive oxygen species scavenging and targeted delivery of methotrexate for rheumatoid arthritis therapy. Nano Res. 2022;15(1):153–161.
  • Thao le Q, Byeon HJ, Lee C, et al. Pharmaceutical potential of tacrolimus-loaded albumin nanoparticles having targetability to rheumatoid arthritis tissues. Int J Pharm. 2016;497(1–2):268–276.
  • Zheng X, Yu X, Wang C, et al. Targeted co-delivery biomimetic nanoparticles reverse macrophage polarization for enhanced rheumatoid arthritis therapy. Drug Deliv. 2022;29(1):1025–1037.
  • He Y, Li R, Liang J, et al. Drug targeting through platelet membrane-coated nanoparticles for the treatment of rheumatoid arthritis. Nano Res. 2018;11(11):6086–6101.
  • Hou M, Wei Y, Zhao Z, et al. Immuno-Engineered nanodecoys for the multi-target anti-inflammatory treatment of autoimmune diseases. Adv Mater. 2022;34(12):e2108817.
  • Fan XX, Xu MZ, Leung EL, et al. ROS-responsive berberine polymeric micelles effectively suppressed the inflammation of rheumatoid arthritis by targeting mitochondria. Nano-Micro Lett. 2020;12(1):76.
  • Ni R, Song G, Fu X, et al. Reactive oxygen species-responsive dexamethasone-loaded nanoparticles for targeted treatment of rheumatoid arthritis via suppressing the iRhom2/TNF-α/BAFF signaling pathway. Biomaterials. 2020;232:119730.
  • Ma B, Xu H, Zhuang W, et al. Reactive oxygen species responsive theranostic nanoplatform for two-photon aggregation-induced emission imaging and therapy of acute and chronic inflammation. ACS nano. 2020;14(5):5862–5873.
  • He L, Fan D, Liang W, et al. Matrix metalloproteinase-responsive pegylated lipid nanoparticles for controlled drug delivery in the treatment of rheumatoid arthritis. ACS Appl Bio Mater. 2020;3(5):3276–3284.
  • Chu D, Dong X, Shi X, et al. Neutrophil-based drug delivery systems. Adv Mater. 2018;30(22):e1706245.
  • Qu M, Lin Q, Huang L, et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J Control Release. 2018;287:156–166.
  • Chen HY, Deng J, Wang Y, et al. Hybrid cell membrane-coated nanoparticles: a multifunctional biomimetic platform for cancer diagnosis and therapy. Acta Biomater. 2020;112:1–13.
  • Wunder A, Müller-Ladner U, Stelzer EH, et al. Albumin-based drug delivery as novel therapeutic approach for rheumatoid arthritis. J Iimmunol. 2003;170(9):4793–4801.
  • Fiehn C, Kratz F, Sass G, et al. Targeted drug delivery 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):1188–1191.
  • Gholijani N, Abolmaali SS, Kalantar K, et al. Therapeutic effect of carvacrol-loaded albumin nanoparticles on arthritic rats. Iran J Pharm Res. 2020;19(1):312–320.
  • Zhao L, Gu C, Gan Y, et al. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. J Control Release. 2020;318:1–15.
  • Kanada M, Bachmann MH, Hardy JW, et al. Differential fates of biomolecules delivered to target cells via extracellular vesicles. Proc Natl Acad Sci U S A. 2015;112(12):E1433–42.
  • Wu G, Zhang J, Zhao Q, et al. Molecularly engineered macrophage-derived exosomes with inflammation tropism and intrinsic heme biosynthesis for atherosclerosis treatment. Angew Chem. 2020;59(10):4068–4074.
  • Kim SH, Lechman ER, Bianco N, et al. Exosomes derived from IL-10-treated dendritic cells can suppress inflammation and collagen-induced arthritis. J Iimmunol. 2005;174(10):6440–6448.
  • Tang TT, Lv LL, Wang B, et al. Employing macrophage-derived microvesicle for kidney-targeted delivery of dexamethasone: an efficient therapeutic strategy against renal inflammation and fibrosis. Theranostics. 2019;9(16):4740–4755.
  • Kamerkar S, LeBleu VS, Sugimoto H, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546(7659):498–503.
  • Li H, Feng Y, Zheng X, et al. M2-type exosomes nanoparticles for rheumatoid arthritis therapy via macrophage re-polarization. J Control Release. 2021;341:16–30.
  • Yan F, Zhong Z, Wang Y, et al. Exosome-based biomimetic nanoparticles targeted to inflamed joints for enhanced treatment of rheumatoid arthritis. J Nanobiotechnology. 2020;18(1):115.
  • Li R, He Y, Zhu Y, et al. Route to rheumatoid arthritis by macrophage-derived microvesicle-coated nanoparticles. Nano Lett. 2019;19(1):124–134.
  • Robbins PD, Dorronsoro A, Booker CN. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J Clin Invest. 2016;126(4):1173–1180.
  • You DG, Lim GT, Kwon S, et al. Metabolically engineered stem cell-derived exosomes to regulate macrophage heterogeneity in rheumatoid arthritis. Sci Adv. 2021;7(23):eabe0083.
  • Li SY, Cheng H, Xie BR, et al. Cancer cell membrane camouflaged cascade bioreactor for cancer targeted starvation and photodynamic therapy. ACS nano. 2017;11(7):7006–7018.
  • Hu CM, Zhang L, Aryal S, et al. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc Natl Acad Sci U S A. 2011;108(27):10980–10985.
  • Xuan M, Shao J, Dai L, et al. Macrophage cell membrane camouflaged mesoporous silica nanocapsules for in vivo cancer therapy. Adv Healthc Mater. 2015;4(11):1645–1652.
  • Xuan M, Shao J, Dai L, et al. Macrophage cell membrane camouflaged au nanoshells for in vivo prolonged circulation life and enhanced cancer photothermal therapy. ACS Appl Mater Interfaces. 2016;8(15):9610–9618.
  • Li SY, Cheng H, Qiu WX, et al. Cancer cell membrane-coated biomimetic platform for tumor targeted photodynamic therapy and hypoxia-amplified bioreductive therapy. Biomaterials. 2017;142:149–161.
  • Gao W, Hu CM, Fang RH, et al. Surface functionalization of gold nanoparticles with red blood cell membranes. Adv Mater. 2013;25(26):3549–3553.
  • Fang RH, Hu CM, Zhang L. Nanoparticles disguised as red blood cells to evade the immune system. Expert Opin Biol Ther. 2012;12(4):385–389.
  • Ding H, Lv Y, Ni D, et al. Erythrocyte membrane-coated NIR-triggered biomimetic nanovectors with programmed delivery for photodynamic therapy of cancer. Nanoscale. 2015;7(21):9806–9815.
  • Fu Y, Liu W, Wang L-Y, et al. Erythrocyte-membrane-camouflaged nanoplatform for intravenous glucose-responsive insulin delivery. Adv Funct Mater. 2018;28(41):1802250.
  • Li C, Wang X, Li R, et al. Resveratrol-loaded PLGA nanoparticles functionalized with red blood cell membranes as a biomimetic delivery system for prolonged circulation time. J Drug Delivery Sci Technol. 2019;54:101369.
  • Rao L, Bu LL, Xu JH, et al. Red blood cell membrane as a biomimetic nanocoating for prolonged circulation time and reduced accelerated blood clearance. Small. 2015;11(46):6225–6236.
  • Horckmans M, Ring L, Duchene J, et al. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J. 2017;38(3):187–197.
  • Li J, Yang P, Wu Q, et al. Death receptor 5-targeted depletion of interleukin-23-producing macrophages, Th17, and Th1/17 associated with defective tyrosine phosphatase in mice and patients with rheumatoid arthritis. Arthritis Rheum. 2013;65(10):2594–2605.
  • Shi Y, Xie F, Rao P, et al. TRAIL-expressing cell membrane nanovesicles as an anti-inflammatory platform for rheumatoid arthritis therapy. J Control Release. 2020;320:304–313.
  • Dehaini D, Wei X, Fang RH, et al. Erythrocyte-platelet hybrid membrane coating for enhanced nanoparticle functionalization. Adv Mater. 2017;29(16):1606209.
  • Yu H, Fan J, Shehla N, et al. Biomimetic hybrid membrane-coated xuetongsu assisted with laser irradiation for efficient rheumatoid arthritis therapy. ACS nano. 2021;16(1):502–521.
  • Clinicaltrials.gov [internet]. [cited 2022 APR 19]. Available from: https://clinicaltrials.gov/ct2/show/NCT05251870
  • Clinicaltrials.gov [internet]. [cited 2022 APR 19]. Available from: https://clinicaltrials.gov/ct2/show/NCT03798028

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.