371
Views
15
CrossRef citations to date
0
Altmetric
Review

Towards Precision Medicine in Systemic Lupus Erythematosus

ORCID Icon, &
Pages 39-49 | Published online: 04 Feb 2020

References

  • Lisnevskaia L, Murphy G, Isenberg DA. Systemic lupus erythematosus. Lancet. 2014;384:1878–1888. doi:10.1016/S0140-6736(14)60128-824881804
  • Moulton VR. Sex hormones in acquired immunity and autoimmune disease. Front Immunol. 2018;9:2279. doi:10.3389/fimmu.2018.0227930337927
  • Sharif K, Watad A, Coplan L, et al. The role of stress in the mosaic of autoimmunity: an overlooked association. Autoimmun Rev. 2018;17:967–983. doi:10.1016/j.autrev.2018.04.00530118900
  • Smatti MK, Cyprian FS, Nasrallah GK, et al. Viruses and autoimmunity: a review on the potential interaction and molecular mechanisms. Viruses. 2019;11(8):762. doi:10.3390/v11080762
  • Barbhaiya M, Tedeschi SK, Lu B, et al. Cigarette smoking and the risk of systemic lupus erythematosus, overall and by anti-double stranded DNA antibody subtype, in the nurses’ health study cohorts. Ann Rheum Dis. 2018;77:196–202. doi:10.1136/annrheumdis-2017-21167528988206
  • Mak A, Tay SH. Environmental factors, toxicants and systemic lupus erythematosus. Int J Mol Sci. 2014;15(9):16043–16056. doi:10.3390/ijms15091604325216337
  • Mu Q, Zhang H, Luo XM. SLE: another autoimmune disorder influenced by microbes and diet? Front Immunol. 2015;6:608. doi:10.3389/fimmu.2015.0060826648937
  • Petri M, Orbai AM, Alarcon GS, et al. Derivation and validation of the systemic lupus erythematosus international collaborating clinics classification criteria for systemic lupus erythematosus. Arthritis Rheum. 2012;64:2677–2686. doi:10.1002/art.3447322553077
  • Hochberg MC. Updating the American college of rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40:1725. doi:10.1002/(ISSN)1529-0131
  • Aringer M, Costenbader KH, Daikh D, et al. European league against rheumatism/American college of rheumatology classification criteria for systemic lupus erythematosus. Ann Rheum Dis. 2019;78:1151–1159. doi:10.1136/annrheumdis-2018-21481931383717
  • Aringer M, Costenbader KH, Brinks R, et al. Validation of new systemic lupus erythematosus classification criteria. Ann Rheum Dis. 2018;77:60–62. doi:10.1136/annrheumdis-2017-212463
  • Gegenava M, Beaart HJL, Monahan RC, et al. Performance of the proposed ACR-EULAR classification criteria for systemic lupus erythematosus (SLE) in a cohort of patients with SLE with neuropsychiatric symptoms. RMD Open. 2019;5:e000895. doi:10.1136/rmdopen-2019-00089531245049
  • Fanouriakis A, Kostopoulou M, Alunno A, et al. update of the EULAR recommendations for the management of systemic lupus erythematosus. Ann Rheum Dis. 2019;78:736–745. doi:10.1136/annrheumdis-2019-21508930926722
  • Medina-Quinones CV, Ramos-Merino L, Ruiz-Sada P, Isenberg DA. Analysis of complete remission in systemic lupus erythematosus patients over a 32-year period. Arthritis Care Res. 2016;68:981–987. doi:10.1002/acr.v68.7
  • Nagata S. Apoptosis and clearance of apoptotic cells. Annu Rev Immunol. 2018;36:489–517. doi:10.1146/annurev-immunol-042617-05301029400998
  • Smith CK, Kaplan MJ. The role of neutrophils in the pathogenesis of systemic lupus erythematosus. Curr Opinion Rheum. 2015;5:448–453. doi:10.1097/BOR.0000000000000197
  • Urbonaviciute V, Luo H, Sjöwall C, Bengtsson A, Holmdahl R. Low production of reactive oxygen species drives systemic lupus erythematosus. Trends Mol Med. 2019;25:826–835. doi:10.1016/j.molmed.2019.06.00131303528
  • Kaplan MJ. Neutrophils in the pathogenesis and manifestations of SLE. Nat Rev Rheumatol. 2011;7:691–699. doi:10.1038/nrrheum.2011.13221947176
  • Frangou E, Vassilopoulos D, Boletis J, Boumpas DT, Boletis J. An emerging role of neutrophils and NETosis in chronic inflammation and fibrosis in systemic lupus erythematosus (SLE) and ANCA-associated vasculitides (AAV): implications for the pathogenesis and treatment. Autoimmun Rev. 2019;18:751–760. doi:10.1016/j.autrev.2019.06.01131181324
  • Delgado-Rizo V, Martínez-Guzmán MA, Iñiguez-Gutierrez L, et al. Neutrophil extracellular traps and its implications in inflammation: an overview. Front Immunol. 2017;8(1–20):81. doi:10.3389/fimmu.2017.0008128220120
  • Herrada AA, Escobedo N, Iruretagoyena M, et al. Innate immune cells’ contribution to systemic lupus erythematosus. Front Immunol. 2019;10. doi:10.3389/fimmu.2019.00772
  • Shin JI, Lee KH, Joo YH, et al. Inflammasomes and autoimmune and rheumatic diseases: A comprehensive review. J Autoimmun. 2019;103:102299. doi:10.1016/j.jaut.2019.06.01031326231
  • Ma C, Xia Y, Yang Q, et al. The contribution of macrophages to systemic lupus erythematosus. Clin Immunol. 2019;207:1–9. doi:10.1016/j.clim.2019.06.00931255802
  • Thorlacius GE, Wahren-Herlenius M, Rönnblom L. An update on the role of type I interferons in systemic lupus erythematosus and Sjögren’s syndrome. Curr Opin Rheumatol. 2018;30:471–478. doi:10.1097/BOR.000000000000052429889694
  • Celhar T, Fairhurst A. Toll-like receptors in systemic lupus erythematosus: potential for personalized treatment. Frontiers Pharmacol. 2014;5(265):1–8. doi:10.3389/fphar.2014.00265
  • Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration in health and disease. Nat Rev Immunol. 2017;17:30–48. doi:10.1038/nri.2016.11627890914
  • Klarquist J, Zhou Z, Shen N, Janssen EM. Dendritic cells in systemic lupus erythematosus: from pathogenic players to therapeutic tools. Mediators Inflamm. 2016;2016:5045248. doi:10.1155/2016/5045248
  • Sandhu V, Quan M. SLE and serum complement: causative, concomitant or coincidental. Open Rheumatol. 2017;11:113–122. doi:10.2174/1874312901711010113
  • Zhou H, Li B, Li J, et al. Dysregulated T-cell activation and aberrant cytokine expression profile in systemic lupus erythematosus. Mediators of Inflamm. 2019;2019. doi:10.1155/2019/8450947
  • SL P. Altered T and B lymphocyte signaling pathways in lupus. Autoimmun Rev. 2009;8:179–183. doi:10.1016/j.autrev.2008.07.04018721908
  • Stadhouders R, Lubberts E, Hendriks RW. A cellular and molecular view of T helper 17 cell plasticity in autoimmunity. J Autoimmun. 2018;87:1–15. doi:10.1016/j.jaut.2017.12.00729275836
  • Ohl K, Tenbrock K. Regulatory T cells in systemic lupus erythematosus. Eur J Immunol. 2015;45:344–355. doi:10.1002/eji.20134428025378177
  • Blanco P, Ueno H, Schmitt N. T follicular helper (Tfh) cells in lupus: activation and involvement in SLE pathogenesis. Eur J Immunol. 2016;46:281–290. doi:10.1002/eji.20154576026614103
  • Bombardieri M, Lewis M, Pitzalis C. Ectopic lymphoid neogenesis in rheumatic autoimmune diseases. Nat Rev Rheumatol. 2017;13:141–154. doi:10.1038/nrrheum.2016.21728202919
  • Costa S, Bevilacqua D, Cassatella MA. Recent advances on the crosstalk between neutrophils and B or T lymphocytes. Immunology. 2019;156:23–32. doi:10.1111/imm.1300530259972
  • Karrar S, Cunningham-Graham D. Abnormal B-cell development in systemic lupus erythematosus. Arthritis Rheumatol. 2018;70:496–502. doi:10.1002/art.4039629207444
  • Murphy G, Isenberg DA. New therapies for systemic lupus erythematosus: past, imperfect, future tense? Nat Rev Rheum. 2019;15:403–412. doi:10.1038/s41584-019-0235-5
  • Arbuckle MR, McClain MT, Rubertone MV, et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med. 2003;349:1526–1533. doi:10.1056/NEJMoa02193314561795
  • Flower C, Hennis A, Hambleton IR, Nicholson G. Lupus nephritis in an Afro-Caribbean population: renal indices and clinical outcomes. Lupus. 2006;15(10):689–694. doi:10.1177/096120330607241517120598
  • Mavragani CP, Fragoulis GE, Somarakis G, Drosos A, Tzioufas AG, Moutsopoulos HM. Clinical and laboratory predictors of distinct histopathological features of lupus nephritis. Medicine. 2015;94:1–8. doi:10.1097/MD.0000000000000829
  • Yu F, Haas M, Glassock R, et al. Redefining lupus nephritis: clinical implications of pathophysiologic subtypes. Nat Rev Nephrol. 2017;13:483–495. doi:10.1038/nrneph.2017.8528669995
  • Londoño Jiminez A, Mowrey WB, Putternam C, Buyon J, Goilav B, Broder A. Brief report: tubulointerstitial damage in lupus nephritis: A comparison of the factors associated with tubulointerstitial inflammation and renal scarring. Arthritis Rheumatol. 2018;70:1801–1806. doi:10.1002/art.4057529851285
  • Arazi A, Rao DA, Berthier CC etal,. The immune cell landscape in kidneys of patients with lupus nephritis. Nat Immunol. 2019;20:902–914. doi:10.1038/s41590-019-0398-x31209404
  • Yung S, Chan TM. Anti-dsDNA antibodies and resident renal cells - Their putative roles in pathogenesis of renal lesions in lupus nephritis. Clin Immunol. 2017;185:40–50. doi:10.1016/j.clim.2016.09.00227612436
  • Nalewajska M, Gurazda K, Styczyńska-Kowalska E, et al. The role of MicroRNAs in selected forms of glomerulonephritis. Int J Mol Sci. 2019;20:1–9. doi:10.3390/ijms20205050
  • Sakhi H, Moktefi A, Bouachi K, et al. Podocyte injury in lupus nephritis. J Clin Med. 2019;8:1340. doi:10.3390/jcm8091340
  • Mejía-Vilet JM, Córdova-Sánchez BM, Uribe-Uribe NO, Correa-Rotter R, Morales-Buenrostro LE. Prognostic significance of renal vascular pathology in lupus nephritis. Lupus. 2017;26:1042–1050. doi:10.1177/096120331769241928178879
  • Jafri K, Patterson SL, Lanata C. Central nervous system manifestations of systemic lupus erythematosus. Rheumatic Dis Clin N Am. 2017;43:531–548. doi:10.1016/j.rdc.2017.06.003
  • Hanly J, Urowitz MB, Su L, et al. Autoantibodies as biomarkers for the prediction of neuropsychiatric events in systemic lupus erythematosus. Ann Rheum Dis. 2011;10:1726–1732. doi:10.1136/ard.2010.148502
  • McGlasson S, Wiseman S, Wardlaw J, Dhaun N, Hunt DPJ. Neurological disease in lupus: toward a personalized medicine approach. Front Immunol. 2018;9(1146):1–12. doi:10.3389/fimmu.2018.0114629403488
  • Tchessalova D, Posillico CK, Tronson NC. Neuroimmune activation drives multiple brain states. Front Syst Neurosci. 2018;12:39. doi:10.3389/fnsys.2018.00039
  • Schwartz N, Stock AD, Putterman C. Neuropsychiatric lupus: new mechanistic insights and future treatment directions. Nat Rev Rheumatol. 2019;15:137–152. doi:10.1038/s41584-018-0156-830659245
  • Stock AD, Ben-Zui A, Putterman C. Pathogenesis of neuropsychiatric lupus In: Wallace DJ, Hahn BH, editors. Dubois Lupus Erythematosus and Related Symptoms. 9th ed. Elsevier; 317.23. doi:10.1186/s13075-018-1604-1
  • Julià A, López-Longo FJ, Pérez Venegas JJ, et al. Genome-wide association study meta-analysis identifies five new loci for systemic lupus erythematosus. Arthritis Ther Res. 2018;1(100):1–10. doi:10.1186/s13075-018-1604-1
  • Amlöf JC, Alexsson A, Imgenberg-Kreuz J, et al. Novel risk genes for systemic lupus erythematosus predicted by random forest classification. Sci Rep. 2017;1(6236):1–11. doi:10.1038/s41598-017-06516-1
  • Catalina MD, Owen KA, Labonte AC, Grammer AC, Lipsky PE, Labonte AC. The pathogenesis of systemic lupus erythematosus: harnessing big data to understand the molecular basis of lupus. J Autoimmun. 2019;2:102359. doi:10.1016/j.jaut.2019.102359
  • Surace AEA, Hedrich CM. The role of epigenetics in autoimmune/inflammatory disease. Front Immunol. 2019. doi:10.3389/fimmu.2019:01525
  • Imgenberg-Kreuz J, Carlsson Almlöf J, Leonard D, et al. DNA methylation mapping identifies gene regulatory effects in patients with systemic lupus erythematosus. Ann Rheum Dis. 2018;77(5):736–743. doi:10.1136/annrheumdis-2017-21237929437559
  • Hedrich CM. Epigenetics in SLE. Curr Rheumatol Rep. 2017;19(58):1–13. doi:10.1007/s11926-017-0685-128116577
  • Wang Z, Yin H, Lau CS, Lu Q. Histone posttranslational modifications of CD4+ T cell in autoimmune diseases. Int J Mol Sci. 2016;17(1547):1–16. doi:10.3390/ijms18010001
  • Wu Z, Mei X, Ying Z, Sun Y, Song J, Shi W. Ultraviolet B inhibition of DNMT1 activity via AhR activation dependent SIRT1 suppression in CD4+ T cells from systemic lupus erythematosus patients. J Dermatol Sci. 2017;86:230–237. doi:10.1016/j.jdermsci.2017.03.00628336124
  • Mak A. The impact of vitamin D on the immunopathophysiology, disease activity, and extra-musculoskeletal manifestations of systemic lupus erythematosus. Int J Mol Sci. 2018;19:2355; 1–14. doi:10.3390/ijms19082355
  • Draborg A, Izarzugaza JM, Houen G. How compelling are the data for Epstein-Barr virus being a trigger for systemic lupus and other autoimmune diseases? Curr Opin Rheumatol. 2016;28:398–404. doi:10.1097/BOR.000000000000028926986247
  • Myers DR, Wheeler B, Roose JP. mTOR and other effector kinase signals that impact T cell function and activity. Immunol Rev. 2019;291:134–153. doi:10.1111/imr.v291.131402496
  • Zhang J1, Yuan B, Zhang F, et al. Cyclophosphamide perturbs cytosine methylation in Jurkat-T cells through LSD1-mediated stabilization of DNMT1 protein. Chem Res Toxicol. 2011;24:2040–2043. doi:10.1021/tx200384922007908
  • Silverman GJ. The microbiome in SLE pathogenesis. Nat Rev Rheumatol. 2019;15(2):72–74.30607012
  • Kim JW, Kwok SK, Choe JY, et al. Recent advances in our understanding of the link between the intestinal microbiota and systemic lupus erythematosus. Int J Mol Sci. 2019;20(19). doi:10.3390/ijms20194871.
  • Manfredo Vieira S, Hiltensperger M, Kumar V, et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science. 2018;359(6380):1156–1161. doi:10.1126/science.aar720129590047
  • Mu Q, Zhang H, Liao X, et al. Control of lupus nephritis by changes of gut microbiota. Microbiome. 2017;5(1):73. doi:10.1186/s40168-017-0300-828697806
  • Greiling TM, Dehner C, Chen X, et al. Commensal orthologs of the human autoantigen Ro60 as triggers of autoimmunity in lupus. Sci Transl Med. 2018;10(434). doi:10.1126/scitranslmed.aan2306.
  • Cuervo A, Hevia A, López P, et al. Association of polyphenols from oranges and apples with specific intestinal microorganisms in systemic lupus erythematosus patients. Nutrients. 2015;7(2):1301–1317. doi:10.3390/nu702130125690419
  • Gordon C, Amissah-Arthur MB, Gayed M, et al. The British Society for Rheumatology guideline for the management of systemic lupus erythematosus in adults. Rheumatology (Oxford). 2018;57:e1–45. doi:10.1093/rheumatology/kex28629029350
  • Da Costa R, Aguirre-Alastuey E, Isenberg DA, Saracino A. Assessment of response to B-cell depletion using Rituximab in cutaneous lupus erythematosus. JAMA Dermatol. 2018;154(12):1432–1440. doi:10.1001/jamadermatol.2018.37930383114
  • Romero-Diaz J, Isenberg D, Ramsey-Goldman R. Measures of adult systemic lupus erythematosus. Arthritis Care Res (Hoboken). 2011;63(Suppl 11):S37–46. doi:10.1002/acr.2057222588757
  • Yee CS, Isenberg DA, Prabu A, et al. BILAG 2004 index captures systemic lupus erythematosus disease activity better than SLEDAI-2000. Ann Rheum Dis. 2008;67(21):873–876. doi:10.1136/ard.2007.07084717519277
  • Freitas S, Mozo Ruiz M, Costa Carneiro A, Isenberg DA. Why do some patients with systemic lupus erythematosus fail to respond to B-cell depletion using rituximab?Clin Exp Rheumatol. 2019;58(Supplement_3):kez107–055 In Press.
  • Plant D, Maciejewski M, Smith S, Nair N. Maximising therapeutic utility in rheumatoid arthritis consortium, the RAMS study group, Hyrich K1, Ziemek D2, Barton A1, Verstappen S1 Profiling of gene expression biomarkers as a classifier of methotrexate nonresponse in patients with rheumatoid arthritis. Arthritis Rheumatol. 2019;71:678–684. doi:10.1002/art.4081030615300
  • Merrell M, Shulman LE. Determination of prognosis in chronic disease illustrated by systemic lupus erythematosus. J Chronic Dis. 1955;1:12–32. doi:10.1016/0021-9681(55)90018-713233308
  • Mak A, Cheung MW-L, Chiew KJ, et al. Global trend of survival and damage of systemic lupus erythematosus: meta-analysis and meta-regression of observational studies from the 1950s to 2000s. Semin Arthritis Rheum. 2012;41:830–839. doi:10.1016/j.semarthrit.2011.11.00222257558
  • Dooley MA, Hogan S, Falk R. Cyclophosphamide therapy for lupus nephritis: poor survival in black Americans. glomerular disease collaborative network. Kidn Int. 1997;51:1188–1195. doi:10.1038/ki.1997.162
  • Ciurtin C, Isenberg DA, Eds. Biologics in Rheumatology. New York: Nova Biomedical; 2016.
  • Van Vollenhoven RF, Petri MA, Cervera R, et al. Belimumab in the treatment of systemic lupus erythematosus: high disease activity predictions of response. Ann Rheum Dis. 2012;71:1343–1349. doi:10.1136/annrheumdis-2011-20093722337213
  • Hahn BH, McMahon MA, Wilkinson A, et al. American college of rheumatology guidelines for screening, treatment and management of lupus nephritis. Arthritis Care Res. 2012;64:797–808. doi:10.1002/acr.21664
  • Bertsios G, Tektonidou M, Amura Z, et al. Joint European League Against Rheumatism and European Renal Association – European Dialysis and Transplant Association [EULAR/ERA-EDTA] recommendations for the management of adult and paediatric lupus nephritis. Ann Rheum Dis. 2012;71:177–182.
  • Felten R, Scher F, Sagez F, Chasset F, Arnaud L. Spotlight on anifrolumab and its potential for the treatment of moderate-to-severe systemic lupus erythematosus: evidence to date. Drug Des Devel Ther. 2019;13:1535–1543. doi:10.2147/DDDT.S170969
  • Merrill JT, Wallace DJ, Wax S, et al. Efficacy and safety of Atacicept in patients with systemic lupus erythematosus. Arthritis Rheum. 2018;70:266–276. doi:10.1002/art.40360
  • Laccarino L, Andreoli L, Bocci EB, et al. Clinical predictors of response and discontinuation of belimumab in patients with systemic lupus erythematosus in real life setting. Results of a large, multicentric, nationwide study. J Autoimmun. 2018;86:1–8. doi:10.1016/j.jaut.2017.09.00428935492
  • Piantoni S, Andreoli L, Lowin T. Baseline serum levels of baff or april are independent predictors of sledai response after 12 months of treatment with belimumab in patients with refractory systemic lupus erythematosus. doi:10.1136/lupus-2018-abstract.43
  • Parodis I, Johansson P, Gomez A, Soukka S, Emamikia S, Chatzidionysiou K, Gomez A. Predictors of low disease activity and clinical remission following belimumab treatment in systemic lupus erythematosus. Rheumatology (Oxford). 2019;58(12):2170–2176. doi:10.1093/rheumatology/kez19131157891
  • Alase A, Wigston Z, Burska A. Prediction of response to rituximab in SLE using a validated two-score system for interferon status. doi:10.1136/lupus-2019-lsm.71
  • Suttichet TB, Kittanamongkolchai W, Phromjeen C. Urine TWEAK level as a biomarker for early response to treatment in active lupus nephritis: a prospective multicentre study. Lupus Sci Med. 2019 eCollection 2019. doi:10.1136/lupus-2018-000298
  • Mendoza-Pinto C, Pirone C, van der Windt DA. Can we identify who gets benefit or harm from mycophenolate mofetil in systemic lupus erythematosus? Syst Rev Semin Arthritis Rheum. 2017;47:65–76.