16th International Congress on Antiphospholipid Antibodies Task Force Report on Antiphospholipid Syndrome Treatment Trends

Hannah Cohen, Maria J Cuadrado, Doruk Erkan, Ali Duarte-Garcia, David A Isenberg, Jason S Knight, Thomas L Ortel, Anisur Rahman, Jane E Salmon, Maria G Tektonidou, David J Williams, Rohan Willis, Scott C Woller, Danieli Andrade, Hannah Cohen, Maria J Cuadrado, Doruk Erkan, Ali Duarte-Garcia, David A Isenberg, Jason S Knight, Thomas L Ortel, Anisur Rahman, Jane E Salmon, Maria G Tektonidou, David J Williams, Rohan Willis, Scott C Woller, Danieli Andrade

Abstract

Antiphospholipid syndrome (APS), an acquired autoimmune thrombophilia, is characterised by thrombosis and/or pregnancy morbidity in association with persistent antiphospholipid antibodies. The 16th International Congress on Antiphospholipid Antibodies Task Force on APS Treatment Trends reviewed the current status with regard to existing and novel treatment trends for APS, which is the focus of this Task Force report. The report addresses current treatments and developments since the last report, on the use of direct oral anticoagulants in patients with APS, antiplatelet agents, adjunctive therapies (hydroxychloroquine, statins and vitamin D), targeted treatment including rituximab, belimumab, and anti-TNF agents, complement inhibition and drugs based on peptides of beta-2-glycoprotein I. In addition, the report summarises potential new players, including coenzyme Q10, adenosine receptor agonists and adenosine potentiation. In each case, the report provides recommendations for clinicians, based on the current state of the art, and suggests a clinical research agenda. The initiation and development of appropriate clinical studies requires a focus on devising suitable outcome measures, including a disease activity index, an optimal damage index, and a specific quality of life index.

Keywords: Antiphospholipid syndrome; anti-β2-glycoprotein I peptides; biologics; complement inhibition; direct oral anticoagulants; potential new players.

Conflict of interest statement

Declaration of conflicting interests: The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: HC reports, outside the submitted work, institutional research support and support to attend scientific meetings from Bayer Healthcare, with honoraria for lectures from Bayer Healthcare and consultancy fees from UCB paid to University College London Hospitals Charity; DE reports, outside the submitted work, consulting fees from GSK, Exagen and UCB, and research support from American College of Rheumatology, European League Against Rheumatism, Lupus Clinical Trials Consortium, National Institute of Allergy and Infectious Disease, and GSK; JSK reports, outside the submitted work, funding from Jazz Pharmaceuticals for preclinical studies of defibrotide; TLO reports, outside the submitted work, consulting fees from Instrumentation Laboratory and research support from Instrumentation Laboratory and Siemens; AR is a co-inventor on a patent to develop PEGylated Domain I of beta-2-glycoprotein I as a novel therapy for APS; JE Salmon reports, outside the submitted work, support of an investigator-initiated grant form UCB and consultancy fees from Admirx, Akari, BMS, Realta, and UCB; MGT reports, outside the submitted work, consultant fees and unrestricted grants from AbbVie, MSD, Novartis, Pfizer, GSK and UCB deposited to the Special Account for Research Funding (ELKE) of the National and Kapodistrian University of Athens Medical School; SW reports, outside the submitted work, grant support from Bristol‐Meyers‐Squibb/Pfizer Alliance paid to Intermountain Healthcare, and service as co-chair for the American College of Chest Physicians panelists guideline update for the treatment of venous thromboembolism; DJW reports, outside the submitted work, being Chief Clinical investigator of the Stamp Trial; and current research funding from the UK Medical Research Council, Wellbeing of Women, Chief Scientist Office Scotland, Rosetrees Trust and EGA Charity. MJC, A D-G, DA, DAI and RW have nothing to disclose.

References

    1. Miyakis S, Lockshin MD, Atsumi T, et al.. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost 2006; 4: 295–306. doi:10.1111/j.1538-7836.2006.01753.x
    1. Duarte‐García A, Pham MM, Crowson CS, et al.. The epidemiology of antiphospholipid syndrome: a population‐based study. Arthritis Rheumatol 2019; 71: 1545–1552. doi:10.1002/art.40901
    1. Cervera R, Piette J-C, Font J, Euro-Phospholipid Project Group et al.. Antiphospholipid syndrome: Clinical and immunologic manifestations and patterns of disease expression in a cohort of 1,000 patients: Clinical and immunologic manifestations of APS. Arthritis Rheum 2002; 46: 1019–1027.
    1. Cervera R, Rodríguez-Pintó I, Espinosa G. The diagnosis and clinical management of the catastrophic antiphospholipid syndrome: a comprehensive review. J Autoimmun 2018; 92: 1–11.
    1. Erkan D, Aguiar CL, Andrade D, et al.. 14th international congress on antiphospholipid antibodies task force report on antiphospholipid syndrome treatment trends. Autoimmun Rev 2014; 13: 685–696.
    1. Andrade D, Cervera R, Cohen H, et al.. 15th International congress on antiphospholipid antibodies task force on antiphospholipid syndrome treatment trends report. In: Erkan D, Lockshin M. (eds) Antiphospholipid syndrome: Current research highlights and clinical insights. New York: Springer, 2017, pp.317–338. ISBN 978-3-319-55442-6.
    1. Schreiber K, Sciascia S, de Groot PG, et al.. Antiphospholipid syndrome. Nat Rev Dis Primer 2018; 4: 17103.
    1. Willis R, Pierangeli SS. Pathophysiology of the antiphospholipid antibody syndrome. Auto Immun Highlights 2011; 2: 35–52.
    1. Cuadrado MJ, López-Pedrera C, Khamashta MA, et al.. Thrombosis in primary antiphospholipid syndrome: a pivotal role for monocyte tissue factor expression. Arthritis Rheum 1997; 40: 834–841.
    1. Cuadrado MJ, Buendía P, Velasco F, et al.. Vascular endothelial growth factor expression in monocytes from patients with primary antiphospholipid syndrome. J Thromb Haemost 2006; 4: 2461–2469.
    1. Breen KA, Seed P, Parmar K, Moore GW, Stuart-Smith SE, Hunt BJ. Complement activation in patients with isolated antiphospholipid antibodies or primary antiphospholipid syndrome. Thromb Haemost 2012; 107: 423–429.
    1. Arachchillage DRJ, Mackie IJ, Efthymiou M, et al.. Rivaroxaban limits complement activation compared with warfarin in antiphospholipid syndrome patients with venous thromboembolism. J Thromb Haemost 2016; 14: 2177–2186.
    1. Meng H, Yalavarthi S, Kanthi Y, et al.. In vivo role of neutrophil extracellular traps in antiphospholipid antibody-mediated venous thrombosis: in vivo role of NETs in APS. Arthritis Rheumatol 2017; 69: 655–667.
    1. Bordin G, Boldorini R, Meroni PL. The two hit hypothesis in the antiphospholipid syndrome: acute ischaemic heart involvement after valvular replacement despite anticoagulation in a patient with secondary APS. Lupus 2003; 12: 851–853.
    1. Keeling D, Baglin T, Tait C, et al.. British Committee for Standards in Haematology. Guidelines on oral anticoagulation with warfarin – fourth edition: Guideline. Br J Haematol 2011; 154: 311–324.
    1. Tektonidou MG, Andreoli L, Limper M, et al.. EULAR recommendations for the management of antiphospholipid syndrome in adults. Ann Rheum Dis 2019; 78: 1296–1304.
    1. Cohen H, Hunt BJ, Efthymiou M, et al.. Rivaroxaban versus warfarin to treat patients with thrombotic antiphospholipid syndrome, with or without systemic lupus erythematosus (RAPS): a randomised, controlled, open-label, phase 2/3, non-inferiority trial. Lancet Haematol 2016; 3: e426–e436.
    1. Pengo V, Denas G, Zoppellaro G, et al.. Rivaroxaban vs warfarin in high-risk patients with antiphospholipid syndrome. Blood 2018; 132: 1365–1371.
    1. Ordi-Ros J, Sáez-Comet L, Pérez-Conesa M, et al.. Rivaroxaban versus vitamin K antagonist in antiphospholipid syndrome: a randomized noninferiority trial. Ann Intern Med 2019; 171: 685–694.
    1. Woller SC, Stevens SM, Kaplan DA, et al.. Apixaban for the secondary prevention of thrombosis among patients with antiphospholipid syndrome: study rationale and design (ASTRO-APS). Clin Appl Thromb Hemost 2016; 22: 239–247.
    1. Woller SC, Stevens SM, Kaplan DA, T, Rondina M. Protocol modification of apixaban for the secondary prevention of thrombosis among patients with antiphospholipid syndrome study. Clin Appl Thromb Hemost 2018; 24: 192–192.
    1. Legault K, Blostein M, Carrier M, et al.. Single-arm pilot feasibility cohort study of rivaroxaban in antiphospholipid syndrome. Res Pract Thromb Haemost 2018; 2: 204–205.
    1. Crowther MA, Ginsberg JS, Julian J, et al. A comparison of two intensities of warfarin for the prevention of recurrent thrombosis in patients with the antiphospholipid antibody syndrome. N Engl J Med 2003; 349: 1133--1138.
    1. Finazzi G, Marchioli R, Brancaccio V, et al.. A randomized clinical trial of high-intensity warfarin vs. conventional antithrombotic therapy for the prevention of recurrent thrombosis in patients with the antiphospholipid syndrome (WAPS)1. J Thromb Haemost 2005; 3: 848–853.
    1. Malec K, Broniatowska E, Undas A. Direct oral anticoagulants in patients with antiphospholipid syndrome: a cohort study. Lupus 2020; 29: 37–44.
    1. Sato T, Nakamura H, Fujieda Y, et al.. Factor Xa inhibitors for preventing recurrent thrombosis in patients with antiphospholipid syndrome: a longitudinal cohort study. Lupus 2019; 28: 1577–1582.
    1. Sanchez-Redondo J, Espinosa G, Varillas Delgado D, Cervera R. Recurrent thrombosis with direct oral anticoagulants in antiphospholipid syndrome: a systematic literature review and Meta-analysis. Clin Ther 2019; 41: 1839–1862.
    1. Dufrost V, Risse J, Reshetnyak T, et al.. Increased risk of thrombosis in antiphospholipid syndrome patients treated with direct oral anticoagulants. Results from an international patient-level data Meta-analysis. Autoimmun Rev 2018; 17: 1011–1021.
    1. van Es N, Coppens M, Schulman S, Middeldorp S, Büller HR. Direct oral anticoagulants compared with vitamin K antagonists for acute venous thromboembolism: evidence from phase 3 trials. Blood 2014; 124: 1968–1975.
    1. Goldhaber SZ, Eriksson H, Kakkar A, et al.. Efficacy of dabigatran versus warfarin in patients with acute venous thromboembolism in the presence of thrombophilia: Findings from RE-COVER®, RE-COVER™ II, and RE-MEDY™. Vasc Med 2016; 21: 506–514.
    1. Kearon C, Parpia S, Spencer FA, et al.. Antiphospholipid antibodies and recurrent thrombosis after a first unprovoked venous thromboembolism. Blood 2018; 131: 2151–2160.
    1. Miranda S, Park J, Le Gal G, et al.. Prevalence of confirmed antiphospholipid syndrome in 18–50 years unselected patients with first unprovoked venous thromboembolism. J Thromb Haemost 2020; 18: 926–930.
    1. ten Boekel E, Bartels P. Abnormally short activated partial thromboplastin times are related to elevated plasma levels of TAT, F1 + 2, D-Dimer and FVIII:C. Pathophysiol Haemost Thromb 2002; 32: 137–142.
    1. Schouwers SM, Delanghe JR, Devreese KM. Lupus anticoagulant (Lac) testing in patients with inflammatory status: does C-reactive protein interfere with Lac test results? Thromb Res 2010; 125: 102–104.
    1. Tripodi A, Cohen H, Devreese KMJ. Lupus anticoagulant detection in anticoagulated patients. Guidance from the scientific and standardization committee for lupus anticoagulant/antiphospholipid antibodies of the international society on thrombosis and haemostasis. J Thromb Haemost 2020; 18: 1569–1575.
    1. Pengo V, Ruffatti A, Del Ross T, et al.. Confirmation of initial antiphospholipid antibody positivity depends on the antiphospholipid antibody profile. J Thromb Haemost 2013; 11: 1527–1531.
    1. Devignes J, Smaïl‐Tabbone M, Hervé A, et al.. Extended persistence of antiphospholipid antibodies beyond the 12‐week time interval: Association with baseline antiphospholipid antibodies titres. Int J Lab Hematol 2019; 41: 726–730.
    1. Stevens SM, Woller SC, Bauer KA, Kasthuri R, et al.. Guidance for the evaluation and treatment of hereditary and acquired thrombophilia. J Thromb Thrombolysis 2016; 41: 154–164.
    1. Cohen H, Efthymiou M, Isenberg DA. Use of direct oral anticoagulants in antiphospholipid syndrome: Reply. J Thromb Haemost 2020; 18: 259–261.
    1. Tektonidou MG, Andreoli L, Limper M, Tincani A, Ward MM. and Management of thrombotic and obstetric antiphospholipid syndrome: a systematic literature review informing the EULAR recommendations for the management of antiphospholipid syndrome in adults. RMD Open 2019; 5: e000924.
    1. Pharmacovigilance Risk Assessment Committee (PRAC). EMA/ PRAC/219985/2019, prac-recommendation/prac-recommendations-signals-adopted-8- 11-april-2019-prac-meeting _en.pdf. 2019 (accessed 11 August 2020).
    1. Eliquis 5 mg film-coated tablets film-coated tablets. Summary of Product Characteristics, .
    1. Lixiana 60 mg Film-Coated Tablets. Summary of Product Characteristics, (accessed 11 August 2020).
    1. Pradaxa 150 mg hard capsules. Summary of Product Characteristics, (accessed 11 August 2020).
    1. Xarelto 15 and 20 mg film-coated tablets. Summary of Product Characteristics, (accessed 11 August 2020).
    1. Bates SM, Greer IA, Middeldorp S, Veenstra DL, Prabulos A-M, Vandvik PO. VTE, thrombophilia, antithrombotic therapy, and pregnancy. Chest 2012; 141: e691S–e736S.
    1. Mak A, Cheung MW-L, Cheak AA.-c, Ho R.-m. Combination of heparin and aspirin is superior to aspirin alone in enhancing live births in patients with recurrent pregnancy loss and positive anti-phospholipid antibodies: a Meta-analysis of randomized controlled trials and meta-regression. Rheumatology (Oxford) 2010; 49: 281–288.
    1. Arnaud L, Mathian A, Ruffatti A, et al.. Efficacy of aspirin for the primary prevention of thrombosis in patients with antiphospholipid antibodies: an international and collaborative meta-analysis. Autoimmun Rev 2014; 13: 281–291.
    1. Arnaud L, Mathian A, Devilliers H, et al.. Patient-level analysis of five international cohorts further confirms the efficacy of aspirin for the primary prevention of thrombosis in patients with antiphospholipid antibodies. Autoimmun Rev 2015; 14: 192–200.
    1. Erkan D, Harrison MJ, Levy R, et al.. Aspirin for primary thrombosis prevention in the antiphospholipid syndrome: a randomized, double-blind, placebo-controlled trial in asymptomatic antiphospholipid antibody–positive individuals. Arthritis Rheum 2007; 56: 2382–2391.
    1. Gris J-C, Bouvier S, Molinari N, et al.. Comparative incidence of a first thrombotic event in purely obstetric antiphospholipid syndrome with pregnancy loss: the NOH-APS observational study. Blood 2012; 119: 2624–2632.
    1. Heit JA, Spencer FA, White RH. The epidemiology of venous thromboembolism. J Thromb Thrombolysis 2016; 41: 3–14.
    1. Finazzi G. Primary antithrombotic prevention in carriers of antiphospholipid antibodies without systemic autoimmune disorders. Lupus 2012; 21: 747–750.
    1. Zuo Y, Barbhaiya M, Erkan D. Primary thrombosis prophylaxis in persistently antiphospholipid antibody-positive individuals: where do We stand in 2018?. Curr Rheumatol Rep 2018; 20: 66.
    1. Whitlock EP, Burda BU, Williams SB, Guirguis-Blake JM, Evans CV. Bleeding risks with aspirin use for primary prevention in adults: a systematic review for the U.S. Preventive services task force. Ann Intern Med 2016; 164: 826–835.
    1. Ramirez GA, Efthymiou M, Isenberg DA, Cohen H. Under crossfire: thromboembolic risk in systemic lupus erythematosus. Rheumatology (Oxford) 2019; 58: 940–952.
    1. Verro P, Levine SR, Tietjen GE. Cerebrovascular ischemic events with high positive anticardiolipin antibodies. Stroke 1998; 29: 2245–2253.
    1. APASS Investigators. Antiphospholipid antibodies and subsequent thrombo-occlusive events in patients with ischemic stroke. JAMA 2004; 291: 576–584.
    1. Boehringer-Ingelheim. Summary of product characteristics Persantin (dipyridamol) 100mg tablets. The electronic medicines compendium.
    1. Espinola RG, Pierangeli SS, Gharavi AE, Harris EN, Ghara AE. Hydroxychloroquine reverses platelet activation induced by human IgG antiphospholipid antibodies. Thromb Haemost 2002; 87: 518–522.
    1. Belizna C. Hydroxychloroquine as an anti-thrombotic in antiphospholipid syndrome. Autoimmun Rev 2015; 14: 358–362.
    1. Rand JH, Wu X-X, Quinn AS, et al.. Hydroxychloroquine protects the annexin A5 anticoagulant shield from disruption by antiphospholipid antibodies: evidence for a novel effect for an old antimalarial drug. Blood 2010; 115: 2292–2299.
    1. Nuri E, Taraborelli M, Andreoli L, et al.. Long-term use of hydroxychloroquine reduces antiphospholipid antibodies levels in patients with primary antiphospholipid syndrome. Immunol Res 2017; 65: 17–24.
    1. Kravvariti E, Koutsogianni A, Samoli E, Sfikakis PP, Tektonidou MG. The effect of hydroxychloroquine on thrombosis prevention and antiphospholipid antibody levels in primary antiphospholipid syndrome: a pilot open label randomized prospective study. Autoimmun Rev 2020; 19: 102491.
    1. Tektonidou MG, Laskari K, Panagiotakos DB, Moutsopoulos HM. Risk factors for thrombosis and primary thrombosis prevention in patients with systemic lupus erythematosus with or without antiphospholipid antibodies. Arthritis & Rheumatism 2008; 61: 29–36.
    1. Erkan D, Unlu O, Sciascia S, APS ACTION et al.. Hydroxychloroquine in the primary thrombosis prophylaxis of antiphospholipid antibody positive patients without systemic autoimmune disease. Lupus 2018; 27: 399–406.
    1. Schmidt-Tanguy A, Voswinkel J, Henrion D, et al.. Anti-thrombotic effects of hydroxychloroquine in primary antiphospholipid syndrome patients. J Thromb Haemost August 2013; : n/a–n/a.
    1. Belizna C, Pregnolato F, Abad S, et al.. HIBISCUS: hydroxychloroquine for the secondary prevention of thrombotic and obstetrical events in primary antiphospholipid syndrome. Autoimmun Rev 2018; 17: 1153–1168.
    1. Sciascia S, Branch DW, Levy RA, et al.. The efficacy of hydroxychloroquine in altering pregnancy outcome in women with antiphospholipid antibodies: Evidence and clinical judgment. Thromb Haemost 2016; 115: 285–290.
    1. Sciascia S, Hunt BJ, Talavera-Garcia E, Lliso G, Khamashta MA, Cuadrado MJ. The impact of hydroxychloroquine treatment on pregnancy outcome in women with antiphospholipid antibodies. Am J Obstet Gynecol 2016; 214: 273.e1-273–e8.
    1. Schreiber K, Schreiber K, Breen K, Cohen H, et al.. HYdroxychloroquine to improve pregnancy outcome in women with antiphospholipid antibodies (HYPATIA) protocol: a multinational randomized controlled trial of hydroxychloroquine versus placebo in addition to standard treatment in pregnant women with antiphospholipid syndrome or antibodies. Semin Thromb Hemost 2017; 43: 562–571.
    1. Oesterle A, Laufs U, Liao JK. Pleiotropic effects of statins on the cardiovascular system. Circ Res 2017; 120: 229–243.
    1. Taylor F, Huffman MD, et al.. Statins for the primary prevention of cardiovascular disease. Cochrane heart group, ed. Cochrane Database Syst Rev 2013; Macedo AF. Published online January 31, doi:10.1002/14651858.CD004816.pub5
    1. Laloux P. Source: Risks and benefits of statins in stroke secondary prevention. Curr Vasc Pharmacol 2013; 11: 812–816.
    1. Glynn RJ, Danielson E, Fonseca FAH, et al.. A randomized trial of rosuvastatin in the prevention of venous thromboembolism. N Engl J Med 2009; 360: 1851–1861.
    1. Ferrara DE, Swerlick R, Casper K, et al.. Fluvastatin inhibits up-regulation of tissue factor expression by antiphospholipid antibodies on endothelial cells: Fluvastatin and antiphospholipid-induced tissue factor. J Thromb Haemost 2004; 2: 1558–1563.
    1. Meroni PL, Raschi E, Testoni C, et al. Statins prevent endothelial cell activation induced by antiphospholipid (anti-beta2-glycoprotein I) antibodies: effect on the proadhesive and proinflammatory phenotype. Arthritis Rheum 2001; 44(12): 2870--2878.
    1. Erkan D, Willis R, Murthy VL, et al.. A prospective open-label pilot study of fluvastatin on proinflammatory and prothrombotic biomarkers in antiphospholipid antibody positive patients. Ann Rheum Dis 2014; 73: 1176–1180.
    1. Sciascia S, Bertolaccini M. Thrombotic risk assessment in APS: the global APS score (GAPSS). Lupus 2014; 23: 1286–1287.
    1. Radin M, Sciascia S, Erkan D, et al.. The adjusted global antiphospholipid syndrome score (aGAPSS) and the risk of recurrent thrombosis: Results from the APS ACTION cohort. Semin Arthritis Rheum 2019; 49: 464–468.
    1. Radin M, Schreiber K, Costanzo P, et al.. The adjusted global AntiphosPholipid syndrome score (aGAPSS) for risk stratification in young APS patients with acute myocardial infarction. Int J Cardiol 2017; 240: 72–77.
    1. Abalos E, Cuesta C, Grosso AL, Chou D, Say L. Global and regional estimates of preeclampsia and eclampsia: a systematic review. Eur J Obstet Gynecol Reprod Biol 2013; 170: 1–7.
    1. Girardi G. Pravastatin to treat and prevent preeclampsia. Preclinical and clinical studies. J Reprod Immunol 2017; 124: 15–20.
    1. Bateman BT, Hernandez-Diaz S, Fischer MA, et al.. Statins and congenital malformations: cohort study. BMJ 2015; 350: h1035–Published 2015 Mar 17.
    1. Lefkou E, Mamopoulos A, Dagklis T, Vosnakis C, Rousso D, Girardi G. Pravastatin improves pregnancy outcomes in obstetric antiphospholipid syndrome refractory to antithrombotic therapy. J Clin Invest 2016; 126: 2933–2940.
    1. Ahmed A, Williams D, Cheed V, StAmP trial Collaborative Group et al.. Pravastatin for early‐onset pre‐eclampsia: a randomised, blinded, placebo‐controlled trial. BJOG Int Bjog 2020; 127: 478–488.
    1. Karalis DG, Hill AN, Clifton S, Wild RA. The risks of statin use in pregnancy: a systematic review. J Clin Lipidol 2016; 10: 1081–1090.
    1. Agmon-Levin N, Blank M, Zandman-Goddard G, et al.. Vitamin D: an instrumental factor in the anti-phospholipid syndrome by inhibition of tissue factor expression. Ann Rheum Dis 2011; 70: 145–150.
    1. Andreoli L, Piantoni S, Dall’Ara F, Allegri F, Meroni P, Tincani A. Vitamin D and antiphospholipid syndrome. Lupus 2012; 21: 736–740.
    1. Piantoni S, Andreoli L, Allegri F, Meroni PL, Tincani A. Low levels of vitamin D are common in primary antiphospholipid syndrome with thrombotic disease. Reumatismo 2012; 64: 307–313.
    1. Riancho-Zarrabeitia L, Cubería M, Muñoz P, et al.. Vitamin D and antiphospholipid syndrome: a retrospective cohort study and Meta-analysis. Semin Arthritis Rheum 2018; 47: 877–882.
    1. García-Carrasco M, Jiménez-Herrera EA, Gálvez-Romero JL, et al.. The anti-thrombotic effects of vitamin D and their possible relationship with antiphospholipid syndrome. Lupus 2018; 27: 2181–2189.
    1. Laplante P, Fuentes R, Salem D, et al.. Anti phospholipid antibody-mediated effects in an arterial model of thrombosis are dependent on toll-like receptor 4. Lupus 2016; 25: 162–176.
    1. Gambhir V, Kim J, Siddiqui S, et al.. Influence of 1,25-dihydroxy vitamin D3 on TLR4-induced activation of antigen presenting cells is dependent on the order of receptor engagement. Immunobiology 2011; 216: 988–996.
    1. Gualtierotti R, Di Giacomo A, Raschi E, Borghi MO, Meroni PL. Vitamin D and anti-Phospholipid antibody syndrome: a comprehensive review. Open Rheumatol J 2018; 12: 248–260.
    1. Willis R, Gonzalez EB, Brasier AR. The journey of antiphospholipid antibodies from cellular activation to antiphospholipid syndrome. Curr Rheumatol Rep 2015; 17: 16.
    1. Martinez-Moreno JM, Herencia C, Montes de Oca A, et al.. Vitamin D modulates tissue factor and protease-activated receptor 2 expression in vascular smooth muscle cells. Faseb J 2016; 30: 1367–1376.
    1. Beltowski J, Atanassova P, Chaldakov GN, Jamroz-Wisniewska A, Kula W, Rusek M. Opposite effects of pravastatin and atorvastatin on insulin sensitivity in the rat: role of vitamin D metabolites. Atherosclerosis 2011 219: 526--531.
    1. Stephanou A, Ross R, Handwerger S. Regulation of human placental lactogen expression by 1,25-dihydroxyvitamin D3. Endocrinology 1994; 135: 2651–2656.
    1. Du H, Daftary GS, Lalwani SI, Taylor HS. Direct regulation of HOXA10 by 1,25-(OH)2D3 in human myelomonocytic cells and human endometrial stromal cells. Mol Endocrinol 2005; 19: 2222–2233.
    1. Bodnar LM, Catov JM, Simhan HN, Holick MF, Powers RW, Roberts JM. Maternal vitamin D deficiency increases the risk of preeclampsia. J Clin Endocrinol Metab 2007; 92: 3517–3522.
    1. Ota K, Dambaeva S, Han AR, Beaman K, Gilman-Sachs A, Kwak-Kim J. Vitamin D deficiency may be a risk factor for recurrent pregnancy losses by increasing cellular immunity and autoimmunity. Hum Reprod 2014; 29: 208–219.
    1. Soroka N, Talako T. AB0415 the effect of vitamin D supplementation on antiphospholipid antibodies level in patients with antiphospholipid syndrome. Ann Rheum Dis 2016; 75: 1048.2–1048.
    1. Scragg R, Stewart AW, Waayer D, et al.. Effect of monthly High-Dose vitamin D supplementation on cardiovascular disease in the vitamin D assessment study : a randomized clinical trial. JAMA Cardiol 2017; 2: 608–616.
    1. Pierangeli SS, Colden-Stanfield M, Liu X, Barker JH, Anderson GL, Harris EN. Antiphospholipid antibodies from antiphospholipid syndrome patients activate endothelial cells in vitro and in vivo. Circulation 1999; 99: 1997–2002.
    1. Merrill JT, Neuwelt CM, Wallace DJ, et al.. Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase ii/iii systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum 2010; 62: 222–233.
    1. Rovin BH, Furie R, Latinis K, LUNAR Investigator Group et al.. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the lupus nephritis assessment with rituximab study. Arthritis Rheum 2012; 64: 1215–1226.
    1. Hahn BH, McMahon MA, Wilkinson A, American College of Rheumatology et al.. American college of rheumatology guidelines for screening, treatment, and management of lupus nephritis. Arthritis Care Res (Hoboken) 2012; 64: 797–808.
    1. Fanouriakis A, Kostopoulou M, Alunno A, et al.. 2019 Update of the EULAR recommendations for the management of systemic lupus erythematosus. Ann Rheum Dis 2019; 78: 736–745.
    1. (accessed 7 March 2020).
    1. Wang CR, Weng CT, Liu MF. Monocentric experience of the rituximab therapy in systemic lupus erythematosus-associated antiphospholipid syndrome with warfarin therapy failure. Semin Arthritis Rheum 2017; 47: e7-e8.
    1. Chalam KV, Gupta SK, Agarwal S. Rituximab effectively reverses papilledema associated with cerebral venous sinus thrombosis in antiphospholipid antibody syndrome. Eur J Ophthalmol 2007; 17: 867–870.
    1. Adamson R, Sangle S, Kaul A, et al.. Clinical improvement in antiphospholipid syndrome after rituximab therapy. J Clin Rheumatol 2008; 14: 359–360. DOI: 10.1097/RHU.0b013e31818f38d4.
    1. Erkan D, Vega J, Ramón G, Kozora E, Lockshin MD. A pilot open-label phase II trial of rituximab for non-criteria manifestations of antiphospholipid syndrome. Arthritis Rheum 2013; 65: 464–471.
    1. Ioannou Y, Lambrianides A, Cambridge G, Leandro MJ, Edwards JCW, Isenberg DA. B cell depletion therapy for patients with systemic lupus erythaematosus results in a significant drop in anticardiolipin antibody titres. Ann Rheum Dis 2007; 67: 425–426.
    1. Gamoudi D, Cutajar M, Gamoudi N, Camilleri DJ, Gatt A. Achieving a satisfactory clinical and biochemical response in antiphospholipid syndrome and severe thrombocytopenia with rituximab: two case reports. Clin Case Rep 2017; 5: 845–848.
    1. Gkogkolou P, Ehrchen J, Goerge T. Severe antiphospholipid antibody syndrome - response to plasmapheresis and rituximab. J Dermatolog Treat 2017; 28: 564–566.
    1. Cartin-Ceba R, Peikert T, Ashrani A, et al.. Primary antiphospholipid Syndrome-Associated diffuse alveolar hemorrhage: Outcomes of primary APS-Associated DAH. Arthritis Care Res (Hoboken) 2014; 66: 301–310.
    1. Yachoui R, Sehgal R, Amlani B, Goldberg JW. Antiphospholipid antibodies-associated diffuse alveolar hemorrhage. Semin Arthritis Rheum 2015; 44: 652–657.
    1. Helgeson SA, Heckman AJ, McCain JD, Cowart JB, Maniaci MJ, Garland JL. A 32-year-old man with hypoxemia and bilateral upper-lobe predominant ground-glass infiltrates on chest imaging. Oxf Med Case Rep 2018; 2018doi:10.1093/omcr/omy072
    1. Aakjær S, Bendstrup E, Ivarsen P, Madsen LB. Continous rituximab treatment for recurrent diffuse alveolar hemorrhage in a patient with systemic lupus erythematosus and antiphosholipid syndrome. Respir Med Case Rep 2017; 22: 263–265.
    1. Berman H, Rodríguez-Pintó I, Cervera R, Catastrophic Antiphospholipid Syndrome (CAPS) Registry Project Group (European Forum on Antiphospholipid Antibodies) et al.. Rituximab use in the catastrophic antiphospholipid syndrome: descriptive analysis of the CAPS registry patients receiving rituximab. Autoimmun Rev 2013; 12: 1085–1090.
    1. Diószegi Á, Tarr T, Nagy-Vincze M, et al.. Microthrombotic renal involvement in an SLE patient with concomitant catastrophic antiphospholipid syndrome: the beneficial effect of rituximab treatment. Lupus 2018; 27: 1552–1558.
    1. Rymarz A, Niemczyk S. The complex treatment including rituximab in the management of catastrophic antiphospholid syndrome with renal involvement. BMC Nephrol 2018; 19: 132.
    1. Abisror N, Mekinian A, Brechignac S, Ruffatti A, Carbillon L, Fain O. Inefficacy of plasma exchanges associated to rituximab in refractory obstetrical antiphospholipid syndrome. Presse Med 2015; 44: 100–102.
    1. Baker KP, Edwards BM, Main SH, et al.. Generation and characterization of LymphoStat-B, a human monoclonal antibody that antagonizes the bioactivities of B lymphocyte stimulator. Arthritis Rheum 2003; 48: 3253–3265.
    1. Sciascia S, Rubini E, Radin M, Cecchi I, Rossi D, Roccatello D. Anticardiolipin and anti-beta 2 glycoprotein-I antibodies disappearance in patients with systemic lupus erythematosus and antiphospholipid syndrome while on belimumab. Ann Rheum Dis 2018; 77: 1694–1695.
    1. Emmi G, Bettiol A, Palterer B, et al.. Belimumab reduces antiphospholipid antibodies in SLE patients independently of hydroxychloroquine treatment. Autoimmun Rev 2019; 18: 312–314.
    1. Chatzidionysiou K, Samoli E, Sfikakis PP, Tektonidou MG. Effect of belimumab treatment on antiphospholipid antibody levels: post-hoc analysis based on two randomised placebo-controlled trials in systemic lupus erythematosus. Ann Rheum Dis 2020; 79: 304–307.
    1. Yazici A, Yazirli B, Erkan D. Belimumab in primary antiphospholipid syndrome. Lupus 2017; 26: 1123–1124.
    1. Lockshin MD, Kim M, Laskin CA et al. Prediction of adverse pregnancy outcome by the presence of lupus anticoagulant, but not anticardiolipin antibody, in patients with antiphospholipid syndrome. Arthritis and Rheumatism 2012; 64: 2311--2318.
    1. Kim MY, Buyon JP, Guerra MM, et al. Angiogenic factor imbalance early in pregnancy predicts adverse outcomes in patients with lupus and antiphospholipid antibodies: results of the PROMISSE study. Am J Obstet Gynecol 2016; 214: 108.e1--108.e14.
    1. Gelber SE, Brent E, Redecha P, et al.. Prevention of defective placentation and pregnancy loss by blocking innate immune pathways in a syngeneic model of placental insufficiency. J Immunol 2015; 195: 1129–1138.
    1. Alijotas-Reig J, Esteve-Valverde E, Llurba E, et al.. Treatment of refractory poor aPL-related obstetric outcomes with TNF-alpha blockers: Maternal-fetal outcomes in a series of 18 cases. Semin Arthritis Rheum 2019; 49: 314–318.
    1. Hemmati I, Kur J. Adalimumab-associated antiphospholipid syndrome: a case report and review of the literature. Clin Rheumatol 2013; 32: 1095–1098.
    1. Cheemalavagu S, McCoy SS, Knight JS. Digital ischaemia secondary to adalimumab-induced antiphospholipid syndrome. BMJ Case Rep 2020; 13: e232907.
    1. Bećarević M. Detrimental roles of TNF-alpha in the antiphospholipid syndrome and de novo synthesis of antiphospholipid antibodies induced by biopharmaceuticals against TNF-alpha. J Thromb Thrombolysis 2017; 44: 565–570.
    1. Dlott JS, Roubey RAS. Drug-Induced lupus anticoagulants and antiphospholipid antibodies. Curr Rheumatol Rep 2012; 14: 71–78. 10.1007/s11926-011-0227-1.
    1. Hennessey A, Lukawska J, Cambridge G, Isenberg D, Leandro M. Adverse infusion reactions to rituximab in systemic lupus erythematosus: a retrospective analysis. BMC Rheumatol 2019; 3: 32.
    1. Pierangeli SS, Vega-Ostertag M, Liu X et al. Complement activation: a novel pathogenic mechanism in the antiphospholipid syndrome. Ann N Y Acad Sci 2005; 1051: 413--420.
    1. Girardi G, Berman J, Redecha P, et al.. Complement C5a receptors and neutrophils mediate fetal injury in the antiphospholipid syndrome. J Clin Invest 2003; 112: 1644–1654.
    1. Hillmen P, Hall C, Marsh JCW, et al.. Effect of eculizumab on hemolysis and transfusion requirements in patients with paroxysmal nocturnal hemoglobinuria. N Engl J Med 2004; 350: 552–559.
    1. Salmon JE, Giradi G, Holers VM. Complement activation as a mediator of antiphospholipid antibody induced pregnancy loss and thrombosis. Ann Rheum 2002; 61(Suppl II): ii46--ii50.
    1. Ritis K, Doumas M, Mastellos D, et al.. A novel C5a Receptor-Tissue factor Cross-Talk in neutrophils links innate immunity to coagulation pathways. J Immunol 2006; 177: 4794–4802.
    1. Redecha P, Tilley R, Tencati M, et al.. Tissue factor: a link between C5a and neutrophil activation in antiphospholipid antibody induced fetal injury. Blood 2007; 110: 2423–2431.
    1. Salmon JE, Girardi G. Antiphospholipid antibodies and pregnancy loss: a disorder of inflammation. J Reprod Immunol 2008; 77: 51–56.
    1. Girardi G, Redecha P, Salmon JE. Heparin prevents antiphospholipid antibody–induced fetal loss by inhibiting complement activation. Nat Med 2004; 10: 1222–1226.
    1. Lonati PA, Scavone M, Gerosa M, et al.. Blood Cell-Bound C4d as a marker of complement activation in patients with the antiphospholipid syndrome. Front Immunol 2019; 10: 773.
    1. Rand JH, Wu XX, Wolgast LR, Lei V, Conway EM. A novel 2-stage approach that detects complement activation in patients with antiphospholipid antibody syndrome. Thromb Res 2017; 156: 119–125.
    1. Lonze BE, Singer AL, Montgomery RA. Eculizumab and renal transplantation in a patient with CAPS. N Engl J Med 2010; 362: 1744–1745.
    1. Hadaya K, Ferrari-Lacraz S, Fumeaux D, et al.. Eculizumab in acute recurrence of thrombotic microangiopathy after renal transplantation: Eculizumab, TMA and kidney transplantation. Am J Transplant 2011; 11: 2523–2527.
    1. Shapira I, Andrade D, Allen SL, Salmon JE. Brief report: Induction of sustained remission in recurrent catastrophic antiphospholipid syndrome via inhibition of terminal complement with eculizumab. Arthritis Rheum 2012; 64: 2719–2723.
    1. Canaud G, Kamar N, Anglicheau D, et al.. Eculizumab improves posttransplant thrombotic microangiopathy due to antiphospholipid syndrome recurrence but fails to prevent chronic vascular changes: Eculizumab and antiphospholipid syndrome recurrence. Am J Transplant 2013; 13: 2179–2185.
    1. Bakhtar O, Thajudeen B, Braunhut BL, et al.. A case of thrombotic microangiopathy associated with antiphospholipid antibody syndrome successfully treated with eculizumab. Transplantation 2014; 98: e17–e18.
    1. Kronbichler A, Frank R, Kirschfink M, et al.. Efficacy of eculizumab in a patient with Immunoadsorption-Dependent catastrophic antiphospholipid syndrome: a case report. Medicine (Baltimore) ). 2014; 93: e143.
    1. Zikos TA, Sokolove J, Ahuja N, Berube C. Eculizumab induces sustained remission in a patient with refractory primary catastrophic antiphospholipid syndrome. J Clin Rheumatol 2015; 21: 311–313.
    1. Strakhan M, Hurtado-Sbordoni M, Galeas N, Bakirhan K, Alexis K, Elrafei T. 36-year-old female with catastrophic antiphospholipid syndrome treated with eculizumab: a case report and review of literature. Case Rep Hematol 2014; 2014: 704371.
    1. Chaturvedi S, Braunstein EM, Yuan X, et al.. Complement activity and complement regulatory gene mutations are associated with thrombosis in APS and CAPS. Blood 2020; 135: 239–251.
    1. Morales JM, Martinez-Flores JA, Serrano M, et al.. Association of early kidney allograft failure with preformed IgA antibodies to β2-glycoprotein I. J Am Soc Nephrol 2015; 26: 735–745.
    1. Gustavsen A, Skattum L, Bergseth G, et al.. Effect on mother and child of eculizumab given before caesarean section in a patient with severe antiphospholipid syndrome: a case report. Medicine (Baltimore) ). 2017; 96: e6338.
    1. Kim MY, Guerra MM, Kaplowitz E, et al.. Complement activation predicts adverse pregnancy outcome in patients with systemic lupus erythematosus and/or antiphospholipid antibodies. Ann Rheum Dis 2018; 77: 549–555.
    1. Kelly RJ, Höchsmann B, Szer J, et al.. Eculizumab in pregnant patients with paroxysmal nocturnal hemoglobinuria. N Engl J Med 2015; 373: 1032–1039.
    1. Stefanovic V. The extended use of eculizumab in pregnancy and complement activation–associated diseases affecting maternal, fetal and neonatal Kidneys-The future is now? JCM 2019; 8: 407.
    1. Giannakopoulos B, Passam F, Rahgozar S, Krilis SA. Current concepts on the pathogenesis of the antiphospholipid syndrome. Blood 2007; 109: 422–430.
    1. Ostertag MV, Liu X, Henderson V, Pierangeli SS. A peptide that mimics the vth region of beta-2-glycoprotein I reverses antiphospholipid-mediated thrombosis in mice. Lupus 2006; 15: 358–365.
    1. de la Torre YM, Pregnolato F, D’Amelio F, et al.. Anti-phospholipid induced murine fetal loss: novel protective effect of a peptide targeting the β2 glycoprotein I phospholipid-binding site. Implications for human fetal loss. J Autoimmun 2012. May; 38: J209–15.
    1. Kolyada A, Lee CJ, De Biasio A, Beglova N. A novel dimeric inhibitor targeting Beta2GPI in Beta2GPI/antibody complexes implicated in antiphospholipid syndrome. Uversky VN, ed. PLoS ONE 2010; 5: e15345.
    1. Kolyada A, Porter A, Beglova N. Inhibition of thrombotic properties of persistent autoimmune anti-β2GPI antibodies in the mouse model of antiphospholipid syndrome. Blood 2014; 123: 1090–1097.
    1. Kolyada A, Ke Q, Karageorgos I, et al.. Soluble analog of ApoER2 targeting beta2-glycoprotein I in immune complexes counteracts hypertension in lupus-prone mice with spontaneous antiphospholipid syndrome. J Thromb Haemost 2016; 14: 1298–1307.
    1. Ioannou Y, Pericleous C, Giles I, Latchman DS, Isenberg DA, Rahman A. Binding of antiphospholipid antibodies to discontinuous epitopes on domain I of human β2-glycoprotein I: Mutation studies including residues R39 to R43. Arthritis Rheum 2007; 56: 280–290.
    1. Ioannou Y, Romay-Penabad Z, Pericleous C, et al.. In vivo inhibition of antiphospholipid antibody-induced pathogenicity utilizing the antigenic target peptide domain I of beta2-glycoprotein I: proof of concept. J Thromb Haemost 2009; 7: 833–842.
    1. McDonnell TCR, Willis R, Pericleous C, et al.. PEGylated domain I of beta-2-Glycoprotein I inhibits the binding, coagulopathic, and thrombogenic properties of IgG from patients with the antiphospholipid syndrome. Front Immunol 2018; 9: 2413.
    1. Pérez-Sánchez C, Aguirre MÁ, Ruiz-Limón P, et al.. Ubiquinol effects on antiphospholipid syndrome prothrombotic profile: a randomized, Placebo-Controlled trial. Arterioscler Thromb Vasc Biol 2017; 37: 1923–1932.
    1. Garrido-Maraver J, Cordero MD, Oropesa-Avila M, et al.. Clinical applications of coenzyme Q10. Front Biosci (Landmark Ed) 2014; 19: 619–633. Published 2014 Jan 1, doi:10.2741/4231.
    1. Perez-Sanchez C, Ruiz-Limon P, Aguirre MA, et al.. Mitochondrial dysfunction in antiphospholipid syndrome: implications in the pathogenesis of the disease and effects of coenzyme Q10 treatment. Blood 2012; 119: 5859–5870.
    1. Yalavarthi S, Gould TJ, Rao AN, et al.. Release of neutrophil extracellular traps by neutrophils stimulated with antiphospholipid antibodies: a newly identified mechanism of thrombosis in the antiphospholipid syndrome: Release of NETs by neutrophils stimulated with APL. Arthritis Rheumatol 2015; 67: 2990–3003.
    1. Knight JS, Meng H, Coit P, et al.. Activated signature of antiphospholipid syndrome neutrophils reveals potential therapeutic target. JCI Insight 2017; 2: e93897.
    1. Sule G, Kelley WJ, Gockman K, et al.. Increased adhesive potential of antiphospholipid syndrome neutrophils mediated by β2 integrin mac-1. Arthritis Rheumatol 2020; 72: 114–124.
    1. Eby JC, Gray MC, Hewlett EL. Cyclic AMP-Mediated suppression of neutrophil extracellular trap formation and apoptosis by the Bordetella pertussis adenylate cyclase toxin. Blanke SR, ed. Infect Immun 2014; 82: 5256–5269.
    1. Shishikura K, Horiuchi T, Sakata N, et al.. Prostaglandin E 2 inhibits neutrophil extracellular trap formation through production of cyclic AMP: PGE 2 inhibits neutrophil NET formation. Br J Pharmacol 2016; 173: 319–331.
    1. Ali RA, Gandhi AA, Meng H, et al.. Adenosine receptor agonism protects against NETosis and thrombosis in antiphospholipid syndrome. Nat Commun 2019; 10: 1916.
    1. Burcoglu-O’Ral A, Erkan D, Asherson R. Treatment of catastrophic antiphospholipid syndrome with defibrotide, a proposed vascular endothelial cell modulator. J Rheumatol 2002; 29: 2006–2011.
    1. Zhou H, Wolberg AS, Roubey RAS. Characterization of monocyte tissue factor activity induced by IgG antiphospholipid antibodies and inhibition by dilazep. Blood 2004; 104: 2353–2358.
    1. Samudra AN, Dwyer KM, Selan C, et al.. CD39 and CD73 activity are protective in a mouse model of antiphospholipid antibody-induced miscarriages. J Autoimmun 2018; 88: 131–138.
    1. Yadav V, Chi L, Zhao R, et al.. ENTPD-1 disrupts inflammasome IL-1β–driven venous thrombosis. J Clin Invest 2019; 129: 2872–2877.
    1. Knight JS, Mazza LF, Yalavarthi S, et al.. Ectonucleotidase-mediated suppression of lupus autoimmunity and vascular dysfunction. Front Immunol 2018; 9: 1322.
    1. Hisada R, Kato M, Sugawara E, et al.. Circulating plasmablasts contribute to antiphospholipid antibody production, associated with type I interferon upregulation. J Thromb Haemost 2019; 17: 1134–1143.
    1. Grenn RC, Yalavarthi S, Gandhi AA, et al.. Endothelial progenitor dysfunction associates with a type I interferon signature in primary antiphospholipid syndrome. Ann Rheum Dis 2017; 76: 450–457.
    1. Xourgia E, Tektonidou MG. Type I interferon gene expression in antiphospholipid syndrome: Pathogenetic, clinical and therapeutic implications. J Autoimmun 2019; 104: 102311.
    1. Flessa CM, Vlachiotis S, Nezos A, Andreakos E, Mavragani CP, Tektonidou MG. Independent association of low IFNλ1 gene expression and type I IFN score/IFNλ1 ratio with obstetric manifestations and triple antiphospholipid antibody positivity in primary antiphospholipid syndrome. Clin Immunol 2019; 209: 108265.
    1. Newland AC, Sánchez‐González B, Rejtő L, et al.. Phase 2 study of efgartigimod, a novel FcRn antagonist, in adult patients with primary immune thrombocytopenia. Am J Hematol 2020; 95: 178–187.

Source: PubMed

3
订阅