3,497
Views
5
CrossRef citations to date
0
Altmetric
Research Article

Immunomodulatory nano-preparations for rheumatoid arthritis

ORCID Icon, , , , , & show all
Pages 9-19 | Received 12 Sep 2022, Accepted 21 Nov 2022, Published online: 08 Dec 2022

References

  • Abdolmaleki F, Farahani N, Gheibi Hayat SM, et al. (2018). The role of efferocytosis in autoimmune diseases. Front Immunol 9:1645.
  • Abualrous ET, Sticht J, Freund C. (2021). Major histocompatibility complex (MHC) class I and class II proteins: impact of polymorphism on antigen presentation. Curr Opin Immunol 70:95–104.
  • Adorini L, Giarratana N, Penna G. (2004). Pharmacological induction of tolerogenic dendritic cells and regulatory T cells. Semin Immunol 16:127–34.
  • Adu-Berchie K, Mooney DJ. (2020). Biomaterials as local niches for immunomodulation. Acc Chem Res 53:1749–60.
  • Ali OA, Huebsch N, Cao L, et al. (2009). Infection-mimicking materials to program dendritic cells in situ. Nat Mater 8:151–8.
  • Awate S, Babiuk LA, Mutwiri G. (2013). Mechanisms of action of adjuvants. Front Immunol 4:114.
  • Bahmani B, Uehara M, Jiang L, et al. (2018). Targeted delivery of immune therapeutics to lymph nodes prolongs cardiac allograft survival. J Clin Invest 128:4770–86.
  • Barnabei L, Laplantine E, Mbongo W, et al. (2021). NF-kappaB: at the borders of autoimmunity and inflammation. Front Immunol 12:716469.
  • Baxter AG, Hodgkin PD. (2002). Activation rules: the two-signal theories of immune activation. Nat Rev Immunol 2:439–46.
  • Bell GM, Anderson AE, Diboll J, et al. (2017). Autologous tolerogenic dendritic cells for rheumatoid and inflammatory arthritis. Ann Rheum Dis 76:227–34.
  • Benham H, Nel HJ, Law SC, et al. (2015). Citrullinated peptide dendritic cell immunotherapy in HLA risk genotype-positive rheumatoid arthritis patients. Sci Transl Med 7:290ra287.
  • Benjamin D, Colombi M, Moroni C, et al. (2011). Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov 10:868–80.
  • Bivona G, Agnello L, Ciaccio M. (2018). The immunological implication of the new vitamin D metabolism. Cent Eur J Immunol 43:331–4.
  • Bluestone JA, Anderson M. (2020). Tolerance in the age of immunotherapy. N Engl J Med 383:1156–66.
  • Boggiatto PM, Schaut RG, Kanipe C, et al. (2019). Sustained antigen release polyanhydride-based vaccine platform for immunization against bovine brucellosis. Heliyon 5:e02370.
  • Bonnefoy F, Daoui A, Valmary-Degano S, et al. (2016). Apoptotic cell infusion treats ongoing collagen-induced arthritis, even in the presence of methotrexate, and is synergic with anti-TNF therapy. Arthritis Res Ther 18:184.
  • Bonnefoy F, Gauthier T, Vallion R, et al. (2018). Factors produced by macrophages eliminating apoptotic cells demonstrate pro-resolutive properties and terminate ongoing inflammation. Front Immunol 9:2586.
  • Brusko MA, Stewart JM, Posgai AL, et al. (2020). Immunomodulatory dual-sized microparticle system conditions human antigen presenting cells into a tolerogenic phenotype in vitro and inhibits type 1 diabetes-specific autoreactive T cell responses. Front Immunol 11:574447.
  • Capini C, Jaturanpinyo M, Chang HI, et al. (2009). Antigen-specific suppression of inflammatory arthritis using liposomes. J Immunol 182:3556–65.
  • Casella G, Rasouli J, Boehm A, et al. (2020). Oligodendrocyte-derived extracellular vesicles as antigen-specific therapy for autoimmune neuroinflammation in mice. Sci Transl Med 12:eaba0599.
  • Chapman NM, Chi H. (2014). mTOR signaling, Tregs and immune modulation. Immunotherapy 6:1295–311.
  • Charbonnier LM, Wang S, Georgiev P, et al. (2015). Control of peripheral tolerance by regulatory T cell-intrinsic Notch signaling. Nat Immunol 16:1162–73.
  • Chen W, Jin W, Hardegen N, et al. (2003). Conversion of peripheral CD4 + CD25– naive T cells to CD4 + CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med 198:1875–86.
  • Chen X, Du G, Bai S, et al. (2021a). Restoring immunological tolerance in established experimental arthritis by combinatorial citrullinated peptides and immunomodulatory signals. Nano Today 41:101307.
  • Chen X, Du G, Sun X. (2021b). Targeting lymphoid tissues to promote immune tolerance. Adv Therapeutics 4:2100056.
  • Chen Y, Guan SY, Deng J, et al. (2020). B7-H3: a promising therapeutic target for autoimmune diseases. Cell Immunol 352:104077.
  • Choi HS, Liu W, Misra P, et al. (2007). Renal clearance of quantum dots. Nat Biotechnol 25:1165–70.
  • Cifuentes-Rius A, Desai A, Yuen D, et al. (2021). Inducing immune tolerance with dendritic cell-targeting nanomedicines. Nat Nanotechnol 16:37–46.
  • Clemente-Casares X, Blanco J, Ambalavanan P, et al. (2016). Expanding antigen-specific regulatory networks to treat autoimmunity. Nature 530:434–40.
  • Cooper GS, Bynum ML, Somers EC. (2009). Recent insights in the epidemiology of autoimmune diseases: improved prevalence estimates and understanding of clustering of diseases. J Autoimmun 33:197–207.
  • Demento SL, Cui W, Criscione JM, et al. (2012). Role of sustained antigen release from nanoparticle vaccines in shaping the T cell memory phenotype. Biomaterials 33:4957–64.
  • Doran AC, Yurdagul AJr, Tabas I. (2020). Efferocytosis in health and disease. Nat Rev Immunol 20:254–67.
  • Eskandari SK, Allos H, Al Dulaijan BS, et al. (2022). mTORC1 inhibition protects human regulatory T cells from granzyme-B-induced apoptosis. Front Immunol 13:899975.
  • Fadok VA, Voelker DR, Campbell PA, et al. (1992). Exposure of phosphatidylserine on the surface of apoptotic lymphocytes triggers specific recognition and removal by macrophages. J Immunol 148:2207–16.
  • Fischer R, Turnquist HR, Taner T, et al. (2009). Use of rapamycin in the induction of tolerogenic dendritic cells. Handb Exp Pharmacol 188:215–32.
  • Gagliani N, Magnani CF, Huber S, et al. (2013). Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med 19:739–46.
  • Galea R, Nel HJ, Talekar M, et al. (2019). PD-L1- and calcitriol-dependent liposomal antigen-specific regulation of systemic inflammatory autoimmune disease. JCI Insight 4:e126025.
  • Gavrila BI, Ciofu C, Stoica V. (2016). Biomarkers in rheumatoid arthritis, what is new? J Med Life 9:144–8.
  • Getts DR, Martin AJ, McCarthy DP, et al. (2012). Microparticles bearing encephalitogenic peptides induce T-cell tolerance and ameliorate experimental autoimmune encephalomyelitis. Nat Biotechnol 30:1217–24.
  • Getts DR, Shea LD, Miller SD, et al. (2015). Harnessing nanoparticles for immune modulation. Trends Immunol 36:419–27.
  • Getts DR, Turley DM, Smith CE, et al. (2011). Tolerance induced by apoptotic antigen-coupled leukocytes is induced by PD-L1+ and IL-10-producing splenic macrophages and maintained by T regulatory cells. J Immunol 187:2405–17.
  • Harms RZ, Ostlund KR, Cabrera MS, et al. (2020). Confirmation and identification of biomarkers implicating environmental triggers in the pathogenesis of type 1 diabetes. Front Immunol 11:1922.
  • Heo R, Park JS, Jang HJ, et al. (2014). Hyaluronan nanoparticles bearing gamma-secretase inhibitor: in vivo therapeutic effects on rheumatoid arthritis. J Control Release 192:295–300.
  • Herrington FD, Carmody RJ, Goodyear CS. (2016). Modulation of NF-kappaB signaling as a therapeutic target in autoimmunity. J Biomol Screen 21:223–42.
  • Hong X, Zhong X, Du G, et al. (2020). The pore size of mesoporous silica nanoparticles regulates their antigen delivery efficiency. Sci Adv 6:eaaz4462.
  • Hoshii T, Kasada A, Hatakeyama T, et al. (2014). Loss of mTOR complex 1 induces developmental blockage in early T-lymphopoiesis and eradicates T-cell acute lymphoblastic leukemia cells. Proc Natl Acad Sci USA 111:3805–10.
  • Hou X, Zaks T, Langer R, et al. (2021). Lipid nanoparticles for mRNA delivery. Nat Rev Mater 6:1078–94.
  • Huynh ML, Fadok VA, Henson PM. (2002). Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. J Clin Invest 109:41–50.
  • Jiang H, Wang Q, Li L, et al. (2018). Turning the old adjuvant from gel to nanoparticles to amplify CD8(+) T cell responses. Adv Sci (Weinh) 5:1700426.
  • Jiang H, Wang Q, Sun X. (2017). Lymph node targeting strategies to improve vaccination efficacy. J Control Release 267:47–56.
  • Kawane K, Tanaka H, Kitahara Y, et al. (2010). Cytokine-dependent but acquired immunity-independent arthritis caused by DNA escaped from degradation. Proc Natl Acad Sci USA 107:19432–7.
  • Kim WU, Lee WK, Ryoo JW, et al. (2002). Suppression of collagen-induced arthritis by single administration of poly(lactic-co-glycolic acid) nanoparticles entrapping type II collagen: a novel treatment strategy for induction of oral tolerance. Arthritis Rheum 46:1109–20.
  • Kishimoto TK, Ferrari JD, LaMothe RA, et al. (2016). Improving the efficacy and safety of biologic drugs with tolerogenic nanoparticles. Nat Nanotechnol 11:890–9.
  • Kishimoto TK, Maldonado RA. (2018). Nanoparticles for the induction of antigen-specific immunological tolerance. Front Immunol 9:230.
  • Koide N, Kaneda A, Yokochi T, et al. (2015). Inhibition of RANKL- and LPS-induced osteoclast differentiations by novel NF-kappaB inhibitor DTCM-glutarimide. Int Immunopharmacol 25:162–8.
  • Krienke C, Kolb L, Diken E, et al. (2021). A noninflammatory mRNA vaccine for treatment of experimental autoimmune encephalomyelitis. Science 371:145–53.
  • Lee HJ, Park SY, Jeong HJ, et al. (2018). Glucocorticoids induce corneal allograft tolerance through expansion of monocytic myeloid-derived suppressor cells. Am J Transplant 18:3029–37.
  • Lee WK, Park JY, Jung S, et al. (2005). Preparation and characterization of biodegradable nanoparticles entrapping immunodominant peptide conjugated with PEG for oral tolerance induction. J Control Release 105:77–88.
  • Li C, Chen X, Luo X, et al. (2021). Nanoemulsions target to ectopic lymphoids in inflamed joints to restore immune tolerance in rheumatoid arthritis. Nano Lett 21:2551–61.
  • Li C, Zheng X, Hu M, et al. (2022a). Recent progress in therapeutic strategies and biomimetic nanomedicines for rheumatoid arthritis treatment. Expert Opin Drug Deliv 19: 883–98.
  • Li Z, Li G, Xu J, et al. (2022b). Hydrogel transformed from nanoparticles for prevention of tissue injury and treatment of inflammatory diseases. Adv Mater 34:e2109178.
  • Ma HM, Wu Z, Nakanishi H. (2011). Phosphatidylserine-containing liposomes suppress inflammatory bone loss by ameliorating the cytokine imbalance provoked by infiltrated macrophages. Lab Invest 91:921–31.
  • Ma Q, Bai J, Xu J, et al. (2021). Reshaping the inflammatory environment in rheumatoid arthritis joints by targeting delivery of berberine with platelet-derived extracellular vesicles. Adv NanoBio Res 1:2100115.
  • Maldonado RA, LaMothe RA, Ferrari JD, et al. (2015). Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc Natl Acad Sci USA 112:E156–E165.
  • Mann EH, Chambers ES, Chen YH, et al. (2015). 1alpha, 25-dihydroxyvitamin D3 acts via transforming growth factor-beta to up-regulate expression of immunosuppressive CD73 on human CD4+ Foxp3- T cells. Immunology 146:423–31.
  • Manolova V, Flace A, Bauer M, et al. (2008). Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol 38:1404–13.
  • McInnes IB, Schett G. (2011). The pathogenesis of rheumatoid arthritis. N Engl J Med 365:2205–19.
  • McKarns SC, Schwartz RH. (2005). Distinct effects of TGF-beta 1 on CD4+ and CD8+ T cell survival, division, and IL-2 production: a role for T cell intrinsic Smad3. J Immunol 174:2071–83.
  • Medina CB, Mehrotra P, Arandjelovic S, et al. (2020). Metabolites released from apoptotic cells act as tissue messengers. Nature 580:130–5.
  • Mir M, Ahmed N, Rehman AU. (2017). Recent applications of PLGA based nanostructures in drug delivery. Colloids Surf B Biointerfaces 159:217–31.
  • Moulis G, Pugnet G, Costedoat-Chalumeau N, et al. (2018). Efficacy and safety of biologics in relapsing polychondritis: a French national multicentre study. Ann Rheum Dis 77:1172–8.
  • Mullazehi M, Wick MC, Klareskog L, et al. (2012). Anti-type II collagen antibodies are associated with early radiographic destruction in rheumatoid arthritis. Arthritis Res Ther 14:R100.
  • Noon L. (1911). Prophylactic inoculation against hay fever. Lancet 177:1572–3.
  • Okada Y, Kim K, Han B, et al. (2014). Risk for ACPA-positive rheumatoid arthritis is driven by shared HLA amino acid polymorphisms in Asian and European populations. Hum Mol Genet 23:6916–26.
  • Phelps EA, Enemchukwu NO, Fiore VF, et al. (2012). Maleimide cross-linked bioactive PEG hydrogel exhibits improved reaction kinetics and cross-linking for cell encapsulation and in situ delivery. Adv Mater 24:64–70.
  • Poon IK, Lucas CD, Rossi AG, et al. (2014). Apoptotic cell clearance: basic biology and therapeutic potential. Nat Rev Immunol 14:166–80.
  • Radwan J, Babik W, Kaufman J, et al. (2020). Advances in the evolutionary understanding of MHC polymorphism. Trends Genet 36:298–311.
  • Rambhia KJ, Ma PX. (2015). Controlled drug release for tissue engineering. J Control Release 219:119–28.
  • Raychaudhuri SP, Wilken R, Sukhov AC, et al. (2017). Management of psoriatic arthritis: early diagnosis, monitoring of disease severity and cutting edge therapies. J Autoimmun 76:21–37.
  • Riemann M, Andreas N, Fedoseeva M, et al. (2017). Central immune tolerance depends on crosstalk between the classical and alternative NF-kappaB pathways in medullary thymic epithelial cells. J Autoimmun 81:56–67.
  • Roodenrijs NMT, Welsing PMJ, van der Goes MC, et al. (2021). Healthcare utilization and economic burden of difficult-to-treat rheumatoid arthritis: a cost-of-illness study. Rheumatology (Oxford) 60:4681–90.
  • Samaridou E, Heyes J, Lutwyche P. (2020). Lipid nanoparticles for nucleic acid delivery: current perspectives. Adv Drug Deliv Rev 154–155:37–63.
  • Sands E, Kivitz A, DeHaan W, et al. (2022). Tolerogenic nanoparticles mitigate the formation of anti-drug antibodies against pegylated uricase in patients with hyperuricemia. Nat Commun 13:272.
  • Schinnerling K, Rosas C, Soto L, et al. (2019). Humanized mouse models of rheumatoid arthritis for studies on immunopathogenesis and preclinical testing of cell-based therapies. Front Immunol 10:203.
  • Serra P, Santamaria P. (2019). Antigen-specific therapeutic approaches for autoimmunity. Nat Biotechnol 37:238–51.
  • Shakya AK, Nandakumar KS. (2018). Antigen-specific tolerization and targeted delivery as therapeutic strategies for autoimmune diseases. Trends Biotechnol 36:686–99.
  • Shao K, Singha S, Clemente-Casares X, et al. (2015). Nanoparticle-based immunotherapy for cancer. ACS Nano 9:16–30.
  • Singh A, Peppas NA. (2014). Hydrogels and scaffolds for immunomodulation. Adv Mater 26:6530–41.
  • Singh A, Qin H, Fernandez I, et al. (2011). An injectable synthetic immune-priming center mediates efficient T-cell class switching and T-helper 1 response against B cell lymphoma. J Control Release 155:184–92.
  • Singh A, Suri S, Roy K. (2009). In-situ crosslinking hydrogels for combinatorial delivery of chemokines and siRNA-DNA carrying microparticles to dendritic cells. Biomaterials 30:5187–200.
  • Smolen JS, Aletaha D, Barton A, et al. (2018). Rheumatoid arthritis. Nat Rev Dis Primers 4:18001.
  • Smolen JS, Aletaha D, McInnes IB. (2016). Rheumatoid arthritis. Lancet 388:2023–38.
  • Stabler CL, Li Y, Stewart JM, et al. (2019). Engineering immunomodulatory biomaterials for type 1 diabetes. Nat Rev Mater 4:429–50.
  • Stead SO, Kireta S, McInnes SJP, et al. (2018). Murine and non-human primate dendritic cell targeting nanoparticles for in vivo generation of regulatory T-cells. ACS Nano 12:6637–47.
  • Su YJ. (2020). Early diagnosis of psoriatic arthritis among psoriasis patients: clinical experience sharing. Clin Rheumatol 39:3677–84.
  • Suri S, Han LH, Zhang W, et al. (2011). Solid freeform fabrication of designer scaffolds of hyaluronic acid for nerve tissue engineering. Biomed Microdevices 13:983–93.
  • Svajger U, Obermajer N, Jeras M. (2010). Novel findings in drug-induced dendritic cell tolerogenicity. Int Rev Immunol 29:574–607.
  • Szodoray P, Nakken B, Gaal J, et al. (2008). The complex role of vitamin D in autoimmune diseases. Scand J Immunol 68:261–9.
  • Szostak B, Machaj F, Rosik J, et al. (2020). Using pharmacogenetics to predict methotrexate response in rheumatoid arthritis patients. Expert Opin Drug Metab Toxicol 16:617–26.
  • Thompson MG, Burgess JL, Naleway AL, et al. (2021). Prevention and attenuation of Covid-19 with the BNT162b2 and mRNA-1273 vaccines. N Engl J Med 385:320–9.
  • Thomson AW, Turnquist HR, Raimondi G. (2009). Immunoregulatory functions of mTOR inhibition. Nat Rev Immunol 9:324–37.
  • Tostanoski LH, Chiu YC, Gammon JM, et al. (2016). Reprogramming the local lymph node microenvironment promotes tolerance that is systemic and antigen specific. Cell Rep 16:2940–52.
  • Toussirot E, Bonnefoy F, Vauchy C, et al. (2021). Mini-review: the administration of apoptotic cells for treating rheumatoid arthritis: current knowledge and clinical perspectives. Front Immunol 12:630170.
  • Tsai S, Shameli A, Yamanouchi J, et al. (2010). Reversal of autoimmunity by boosting memory-like autoregulatory T cells. Immunity 32:568–80.
  • Umeshappa CS, Singha S, Blanco J, et al. (2019). Suppression of a broad spectrum of liver autoimmune pathologies by single peptide-MHC-based nanomedicines. Nat Commun 10:2150.
  • Umeshappa CS, Sole P, Surewaard BGJ, et al. (2021). Liver-specific T regulatory type-1 cells program local neutrophils to suppress hepatic autoimmunity via CRAMP. Cell Rep 34:108919.
  • Verbeke CS, Gordo S, Schubert DA, et al. (2017). Multicomponent injectable hydrogels for antigen-specific tolerogenic immune modulation. Adv Healthc Mater 6:1600773.
  • Wraith D. (2016). Autoimmunity: antigen-specific immunotherapy. Nature 530:422–3.
  • Wu Z, Ma HM, Kukita T, et al. (2010). Phosphatidylserine-containing liposomes inhibit the differentiation of osteoclasts and trabecular bone loss. J Immunol 184:3191–201.
  • Xing Y, Hogquist KA. (2012). T-cell tolerance: central and peripheral. Cold Spring Harb Perspect Biol 4:a006957.
  • Yang M, Ding J, Feng X, et al. (2017). Scavenger receptor-mediated targeted treatment of collagen-induced arthritis by dextran sulfate-methotrexate prodrug. Theranostics 7:97–105.
  • Yi L, Swensen AC, Qian WJ. (2018). Serum biomarkers for diagnosis and prediction of type 1 diabetes. Transl Res 201:13–25.
  • Zhang FX, Liu P, Ding W, et al. (2021). Injectable mussel-inspired highly adhesive hydrogel with exosomes for endogenous cell recruitment and cartilage defect regeneration. Biomaterials 278:121169.
  • Zhang L, Xiao X, Arnold PR, et al. (2019). Transcriptional and epigenetic regulation of immune tolerance: roles of the NF-kappaB family members. Cell Mol Immunol 16:315–23.
  • Zhang Q, Dehaini D, Zhang Y, et al. (2018). Neutrophil membrane-coated nanoparticles inhibit synovial inflammation and alleviate joint damage in inflammatory arthritis. Nat Nanotechnol 13:1182–90.
  • Zhao G, Ren R, Wei X, et al. (2021). Thermoresponsive polymeric dexamethasone prodrug for arthritis pain. J Control Release 339:484–97.
  • Zhou A, Chen S, He B, et al. (2016). Controlled release of TGF-beta 1 from RADA self-assembling peptide hydrogel scaffolds. Drug Des Devel Ther 10:3043–51.