Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

Contents lists available at ScienceDirect

Best Practice & Research Clinical Rheumatology journal homepage: www.elsevierhealth.com/berh

9

Seronegative rheumatoid arthritis: Pathogenetic and therapeutic aspects Arthur G. Pratt*, John D. Isaacs 1 NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK

a b s t r a c t Keywords: Rheumatoid arthritis Seronegative Autoantibody-negative Pathogenesis Treatment

Rheumatoid arthritis (RA) has long been recognised as a highly heterogeneous disease of immune dysregulation. Despite an evergrowing appreciation of the role of circulating autoantibodies in the development of ‘seropositive’ disease, the pathogenesis of seronegative RA remains poorly understood. Accumulating evidence suggests that RA ‘serotypes’, in fact, reflect distinct disease entities that, despite their clinical overlap, diverge in respect of genetic architecture, cellular pathology and even therapeutic responsiveness. Focussing on seronegative RA, this review considers these concepts and their implications for the management of patients with this challenging, though sometimes overlooked, condition. Crown Copyright © 2014 Published by Elsevier Ltd. All rights reserved.

Seronegative rheumatoid arthritis as an evolving entity For classification purposes, seronegative rheumatoid arthritis (RA) has become something of a moving target, reflecting both an evolving understanding of disease-associated autoantibodies [1] and recent modifications in RA classification overall [2]. Such factors inevitably impact upon the clinical spectrum and natural history of the syndrome described. RA ‘seropositivity’ historically referred only to

* Corresponding author. Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Level 4 Cookson Building, The Medical School, Framlington Place, Newcastle upon Tyne, UK. Tel.: þ44 (0)191 222 5337. E-mail addresses: [email protected] (A.G. Pratt), [email protected] (J.D. Isaacs). 1 Tel.: þ44 (0)191 222 5337.

http://dx.doi.org/10.1016/j.berh.2014.10.016 1521-6942/Crown Copyright © 2014 Published by Elsevier Ltd. All rights reserved.

652

A.G. Pratt, J.D. Isaacs / Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

the presence of immunoglobulin M (IgM) rheumatoid factor (RF), but this term must now also take account of circulating anti-citrullinated protein antibodies (ACPAs) [3] whose presence is independently associated with accelerated joint destruction [4]. Consequently, by excluding from the definition of ‘seronegative RA’ the 10% of RF-negative patients now determined ACPA positive, a subtly more benign disease subset may be being described. ACPA status is typically measured using the now widely available anti-cyclic citrullinated peptide 2 (CCP2) assay [5], and efforts to develop comparably diseasespecific but more disease-sensitive ACPA tests may further advance this trend [6]. Moreover, a novel system of RA-associated autoantibodies was recently described, directed against carbamylated proteins (anti-CarP); 16% of ACPA-negative RA patients were found to be anti-CarP positive, having an adverse disease course compared with their ‘double-negative’ counterparts [7]. Although the classification criteria for RA are not yet informed by the new antibody system, they were modified in 2010 to incorporate consideration of the ACPA status alongside RF in an effort to enhance their ability to identify early RA [2]. Interestingly, this replacement of the pre-existing 1987 ACR classification criteria [8] results in an increased prevalence of classifiable RA in the early arthritis clinic [9]. Although no difference in the proportion of patients seropositive for either RF or ACPA has ensued, the modified criteria appear to describe an early RA cohort with a milder disease course than before [9,10] e a phenomenon likely to apply to both seronegative and seropositive subgroups. Such a rapidly changing landscape complicates efforts to define seronegative RA, raising legitimate questions as to whether a stable phenotype for the condition exits [11]. We here argue that it does, starting with a consideration of the clinical spectrum of the condition, and then moving through pathogenetic and therapeutic aspects. Although challenging, we conclude that improving our understanding of seronegative RA is a worthwhile endeavour, with the potential to improve disease outcomes through the development of robust diagnostic tools and more targeted treatment strategies. Prevalence and clinical spectrum Depending on the population studied, the proportion of classifiable RA patients reportedly seronegative for both RF and ACPA ranges widely between 0.10 [12] and 0.48 [13]. A recent meta-analysis provided a pooled ‘double-seronegativity’ rate estimate of 0.22 (95% confidence interval (CI) 0.20e0.24) in this setting [14]. Even allowing for slight diminution of this figure attributable to the availability of anti-CarP testing in the future, an overall population prevalence for seronegative RA that approaches 20 per 10,000 may be extrapolated from epidemiological data [15,16] e comparable, for example, to that of coeliac disease [17] and twice that of psoriatic arthritis [18]. The literature has long recognised particular clinical presentations of RA as being associated with autoantibody seronegativity, particularly in older patients. Indeed, employing a cut-off of 60 years of age, lower rates of both RF and ACPA have been observed in late-onset RA (LORA) compared with youngonset RA [19,20]. A syndrome of explosive-onset, seronegative LORA with polymyalgic features and a soaring acute phase markers is well recognised [21e23], which has been described as having a relatively mild course [21,24]. Distinguishing seronegative LORA from polymyalgia rheumatica (PMR) may therefore be challenging at disease onset, as the latter condition can also present with evidence of peripheral joint synovitis [25], but longitudinal study has identified particular patterns of joint involvement as being discriminatory for LORA in this context [26]. Importantly, >30% of RF-negative LORA patients, and no PMR patients, developed bony erosions at follow-up, justifying their classification as distinct entities and giving the lie to the notion that seronegative RA is a ‘benign’ condition. Patients with remitting symmetrical seronegative synovitis with pitting oedema (RS3PE) may also describe myalgic symptoms alongside characteristic peripheral soft tissue oedema and/or tenosynovitis [27]. Although initially considered a form of RA [27,28], the condition's unique associations with raised circulating serum vascular endothelial growth factor (VEGF), distinct tissue types and, in some cases, particular malignancies have increasingly led experts to classify it separately [29,30]. Of course, many seronegative RA patients do not map to any of the above descriptions and, in practical terms, are essentially indistinguishable from their seropositive counterparts at presentation [31]. Yet diagnosing seronegative RA with confidence remains particularly challenging [32], and the introduction of new classification criteria for RA [2] has had the effect of reducing the prevalence of ACPA seropositivity amongst undifferentiated arthritis (UA) patients from 24% to 10% [10]. The

A.G. Pratt, J.D. Isaacs / Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

653

conclusion that diagnostic delay may increasingly be a feature of seronegative disease is borne out by a review of >130 early RA patients at our own unit, amongst whom a diagnosis was achieved within 6 weeks of referral for proportionately fewer ‘double-seronegative’ patients than for RF and/or ACPA seropositive individuals (51% vs. 73%, p ¼ 0.02, unpublished). It is therefore anticipated that renewed efforts to understand aspects of RA pathogenesis, and the extent to which they are concordant or discordant between serotypes, should yield tools of clinical utility for diagnosis as well as treatment. Aspects of pathogenesis Although genetic variation has long been supposed to account for around 60% of RA risk in Caucasian populations [33], recent work suggests that this figure for heritability, may be considerably lower in seronegative RA [34,35]. It follows that environmental factors should play a more important role in seronegative RA, but much has yet to be understood in this area [36]. Interestingly, whilst smoking is considered a major environmental determinant of seropositive disease, in turn interacting with genetic risk, it plays a lesser, and perhaps negligible, role in the induction of seronegative RA [37,38]. Nonetheless, the likelihood that a first-degree relative of a seronegative patient will share the diagnosis is more than twice the RA population prevalence, implying a substantial contribution of genetic variation to disease risk in this group. The increasingly well-characterised genetic architecture of RA now supports a paradigm in which disease serotypes delineate aetiologies with both distinct and overlapping features. The strongest and most established genetic association with RA is with ‘shared epitope’ (SE) alleles at the highly polymorphic human leucocyte antigen (HLA) DRB1 locus, which encode a conserved amino acid sequence within the peptide-binding groove of major histocompatibility complex (MHC) class II molecules, which are expressed by antigen-presenting cells [39]. The contribution to the RA risk of two additional amino acid positions in the binding groove, encoded at HLA-B and HLA-DPB1 loci, respectively, has since been confirmed [40]. Consistent with the notion that the SE allelic expression provides a mechanism for citrullinated peptides to generate antibody responses [41], it was initially thought that the HLA-DRB1 association represented a primary risk factor for ACPA positivity rather than RA per se [42,43]. A more nuanced reality has recently been confirmed by increasingly wellpowered studies, clearly demonstrating HLA association in both serotypes, albeit with diminished effect sizes in ACPA-negative disease [44e46]. Indeed, recent work suggests that individual HLA alleles confer differing risk according to ACPA status, and that distinct antigens may therefore be involved in the pathogenesis of both conditions [47]. A similar pattern of association with seropositive and seronegative RA is seen for missense mutation at the common autoimmune susceptibility locus protein tyrosine phosphatase, non-receptor type 22 (PTPN22), encoding lymphoid protein tyrosine phosphatase (Lyp) [48] e with a diminished effect size in ACPA-negative individuals [45]. The functional role of this variant in conferring RA risk may vary within and between cellular contexts [49,50], but important regulatory effects on T-cell-receptor-mediated signalling seem beyond doubt [51,52]. A differential role of T cells between RA serotypes is also implied by scrutiny of synovial tissue in established disease. van Oosterhout et al. studied 34 ACPA-positive and 23 ACPA-negative patients (median 9 years duration), and identified significantly more fibrosis and synovial lining thickening, with less lymphocyte infiltration, in the ACPA-negative group [53]. A non-significant trend towards a reduced likelihood of ectopic germinal centre formation in synovial infiltrates of ACPA-negative patients was also noted. Intriguingly, Cantaert et al. by contrast observed comparable numbers of infiltrating synovial T cells between serotypes, but demonstrated a relatively restricted T-cell repertoire in the synovial compartment of ACPA-positive patients compared with ACPA-negative counterparts [54] e consistent with a role for T cells in both disease subsets, but a more classical ‘antigen-driven’ pathology in seropositive disease. Gomez-Puerta et al. found that synovial fluid from ACPA-positive patients contained higher levels of T-cell-derived pro-inflammatory cytokines, including interleukin (IL)-17, than seronegative patients [55]. These data must now be considered alongside recently highlighted cluster of differentiation (CD)4þ T-cell-mediated mechanisms of ACPA-negative disease induction. For example, the most compelling non-HLA genetic association with seronegative RA thus far identified, with an effect size equivalent to that seen in seropositive disease, is with an intronic variant of the ankyrin repeat domain-55 (ANKRD55)

654

A.G. Pratt, J.D. Isaacs / Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

Fig. 1. RA serotypes delineate aetiologies with distinct, but overlapping, features. Select environmental and genetic risks are depicted in the upper part of the figure, with individual factors, respectively, positioned to the left (light grey) or right (dark grey) according to their postulated importance in the development of seronegative versus seropositive disease. In our speculative model, these elements interact to promote qualitatively distinct mechanisms of T-cell dysregulation, which lead to a common pathway of persistent joint inflammation, and define serological status. Pathogenetic mechanisms of seronegative disease may, in fact, apply to both serotypes. See text.

locus on chromosome 5 [44,45,56] e a gene highly expressed in CD4þ T cells but of unknown functional significance [57]. Intriguingly, the locus lies ~185 kilobases upstream of the gp130-encoding IL6ST transcriptional start site, and within a transcription factor-binding domain for that gene [58]. Glycoprotein 130 (gp130) is a ubiquitously expressed co-receptor for numerous cytokines including the pro-inflammatory cytokine IL-6. It is notable that polymorphism within several other components of this cytokine's signalling pathway [59], including the IL-6 receptor (IL-6R), tyrosine kinase-2 (TYK2) and signal transduction and activator of transcription-3 (STAT3) genes, have been associated with RA e in some cases specifically ACPA-negative disease [44,60]. Moreover, we recently described a STAT3 target-enriched gene signature in circulating CD4þ T cells of drug-naïve, early arthritis patients, which predicted progression to RA most effectively in ACPA-negative disease [61]. As this was validated in an independent cohort, we found that the phenomenon most likely reflects increased circulating IL-6 in early ACPA-negative RA, together with a maximal capacity for STAT3 phosphorylation in response to the cytokine amongst CD4þ T cells (manuscript submitted). The extent to which these observations may be mechanistically linked to genetic risk remains the subject of ongoing investigation, but the precedent for the importance of this pathway in the induction of inflammatory arthritis continues to emerge from animal models [62,63]. Accumulating evidence now implicates T-cell-mediated immune dysregulation in RA pathogenesis, irrespective of autoantibody status. It may be that qualitatively distinct cellular mechanisms during disease induction not only define serotype but also drive parallel pathways towards apparently indistinguishable clinical phenotypes. Hence, T cells non-specifically activated by cytokines as a feature

A.G. Pratt, J.D. Isaacs / Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

655

of generalised ‘immune ageing’ [64,65] may be relatively important in sustaining chronic joint inflammation without autoantibody production, and auto-antigen-mediated adaptive immune induction may represent a potent additional disease mechanism under particular environmental and/or genetic conditions [66]. Such a model is highly speculative and has much about it yet to be explained (Fig. 1). It nonetheless illustrates how a fuller appreciation of seronegative RA pathogenesis should inform our understanding of RA as a whole, and could ultimately contribute to more ‘personalised’ strategies for managing the condition in the future. Therapeutic considerations The ability to use biomarkers for defining disease subtypes that reflect targetable pathobiology, and so permit stratification by therapeutic responsiveness, has become a major research goal in rheumatology, as in other fields [67]. Attempts to determine the extent to which RA autoantibody serotype may represent such a biomarker are summarised here, and have generally arisen from observational studies and retrospective analyses of clinical trials. Methotrexate (MTX) has emerged as the widely accepted ‘first-line’ disease-modifying anti-rheumatic drug (DMARD) for the management of newly diagnosed RA, and its use in the management of inflammatory UA has also been advocated [68]. In the PROMPT study, which followed newly presenting UA patients, rates of progression to classifiable RA, radiographic progression and drug-free remission were all comparable in MTX- and placebo-treated arms [69,70]. However, MTX did delay RA progression uniquely in the ACPA-positive UA subgroup, perhaps suggesting its relatively diminished effect on the natural history of ACPA-negative disease. In established RA, neither RF nor ACPA status have in general proved effective in predicting responsiveness to MTX therapy [71e73], despite observations that drug-free remission is less likely, and radiographic progression more likely, in ACPApositive individuals [73,74]. The introduction of combination DMARDs, such as sulphasalazine and hydroxychloroquine, alongside MTX with corticosteroid ‘induction’, has been advocated for the treatment of newly diagnosed RA [68,75]. It seems that the advantages of initial DMARD combination (more rapid reductions in DAS28) should be weighed up alongside potential disadvantages (need for dose adjustments due to side effects) on a case-by-case basis, as a recent randomised controlled trial (RCT) has reported no difference between them in terms of radiological or functional outcome at 12 months providing a responsive, treat-to-target approach is maintained [76]. A reanalysis of the Combination Anti-Rheumatic Drugs in Early RA (CARDERA) trial [77] suggests that the autoantibody status may yet prove a useful biomarker for guiding such decisions, as the use of combination strategies (including the use of oral steroids) significantly reduced radiologic progression over 2 years in ACPApositive, but not ACPA-negative patients [78]. Whilst such data seem to favour a more ‘aggressive’ combination DMARD approach for the treatment of ‘poor-prognosis’ ACPA-positive early RA, radiological progression in the ACPA-negative sub-cohort of this study was not trivial, and the need to identify alternative therapeutic approaches for this disease subtype is therefore emphasised. As regards biologic use, retrospective analyses of registry and cohort data have suggested superior efficacy of anti-tumour necrosis factor (TNF) therapy for the treatment of seronegative RA compared with seropositive disease [79e81]. In such studies, the contribution of autoantibody status to the variability of response nonetheless remains small e perhaps influenced by protracted disease duration at the time of recruitment and/or analysis. Indeed, a prospective study found an association between high baseline ACPA and RF titres with good responses to infliximab, but concluded that the predictive value of autoantibodies in this setting was unlikely to be clinically useful [82]. Although focussed pharmacogenetic analyses have yet to conclusively identify genetic associations with anti-TNF responsiveness [83], an intriguing report that polymorphism at the tumour necrosis factor receptor II (TNFRII) locus predicts therapeutic response preferentially in seronegative RA awaits full publication [84]. Despite a dearth of large prospective studies addressing the issue for anti-TNF, a meta-analysis of phase II or III clinical trials for the B-cell-depleting agent rituximab demonstrated increased efficacy in seropositive patients [85] e including those naïve to MTX and with relatively short disease duration [86]. A similar enhanced efficacy amongst ACPA-positive RA patients has been reported in an extensive registry study of the cytotoxic T-lymphocyte-associated protein 4 (CTLA4)eIg fusion protein abatacept

656

A.G. Pratt, J.D. Isaacs / Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

[87], perhaps reflecting differential effects of co-stimulation blockade on T-cell function according to autoantibody serotype [88]. By contrast, no difference in the efficacy of the IL-6R-targeting monoclonal antibody tocilizumab has to date been demonstrated with respect to autoantibody serotype [89,90].

Conclusion and future directions Seronegative RA is a chronic, painful condition that affects around 100,000 adults in the UK. Inevitable comparisons with its seropositive counterpart may on occasion contribute to the misconception that it is a ‘benign’ condition, but close scrutiny of observational data combined with experience in the clinic confirm that, from the patient's perspective, this is certainly not the case. Whether the activation of distinct pathobiological pathways between the two serotypes underpins subtle differences thus far identified in the efficacy of various treatment approaches in the management of RA is uncertain. Looking ahead, systematic characterisation of the events that drive disease induction, their triggers and the extent to which they differ or overlap according to autoantibody status will together inform our ability to diagnose and treat RA more effectively. These studies will demand an integrated consideration of genetic and epigenetic factors alongside a detailed study of pathological and clinical parameters in large-scale, collaborative projects. Although challenging, the rewards will include more effective tools for diagnosing and treating seronegative RA, and improved outcomes for patients.

Practice points  Seronegative RA represents a significant disease burden, for which prompt therapeutic intervention continues to be hampered by diagnostic challenges.  RA serotypes delineate aetiologies with distinct, yet overlapping, features, orchestrated in part by deregulated CD4þ T-cell function.  Currently, there is insufficient evidence to support a bespoke treatment strategy for the seronegative RA subset, although the value of initial combination DMARD use, and that of Bcell depletion therapy, may be less than in seropositive disease.

Research agenda  Epidemiological studies should focus on delineating the environmental risks for seronegative RA development; large-scale analyses will continue to inform our understanding of interplay with genetic and epigenetic determinants.  An improved understanding of both common and distinct features in the cellular pathology of RA between autoantibody serotypes should inform the development of diagnostic and therapeutic biomarkers in the future, resulting in improved outcomes for patients.

Conflict of interest statement The authors declare no conflict of interest.

Acknowledgement The authors wish to thank Mr. Ben Hargreaves for administrative support during the preparation of the manuscript.

A.G. Pratt, J.D. Isaacs / Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

657

References [1] Trouw LA, Huizinga TW, Toes RE. Autoimmunity in rheumatoid arthritis: different antigens e common principles. Ann Rheum Dis 2013;72(Suppl. 2):ii132e6. [2] Aletaha D, Neogi T, Silman AJ, et al. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League against rheumatism collaborative initiative. Arthritis Rheum 2010;69:1580e8. [3] Schellekens GA, Visser H, de Jong BA, et al. The diagnostic properties of rheumatoid arthritis antibodies recognizing a cyclic citrullinated peptide. Arthritis Rheum 2000;43:155e63. [4] Meyer O, Nicaise-Roland P, Santos MD, et al. Serial determination of cyclic citrullinated peptide autoantibodies predicted five-year radiological outcomes in a prospective cohort of patients with early rheumatoid arthritis. Arthritis Res Ther 2006;8:R40. [5] van Venrooij WJ, Zendman AJW. Anti-CCP2 antibodies: an overview and perspective of the diagnostic abilities of this serological marker for early rheumatoid arthritis. Clin Rev Allergy Immunol 2008;34:36e9. [6] Szekanecz Z, Szabo Z, Zeher M, et al. Superior performance of the CCP3.1 test compared to CCP2 and MCV in the rheumatoid factor-negative RA population. Immunol Res 2013;56:439e43. [7] Shi J, Knevel R, Suwannalai P, et al. Autoantibodies recognizing carbamylated proteins are present in sera of patients with rheumatoid arthritis and predict joint damage. Proc Natl Acad Sci U S A 2011;108:17372e7. [8] Arnett FC, Edworthy SM, Bloch DA, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum 1988;31:315e24. [9] Burgers LE, van Nies JA, Ho LY, et al. Long-term outcome of rheumatoid arthritis defined according to the 2010classification criteria. Ann Rheum Dis 2014;73:428e32. [10] Krabben A, Huizinga TW, van der Helm-van Mil AH, et al. Undifferentiated arthritis characteristics and outcomes when applying the 2010 and 1987 criteria for rheumatoid arthritis. Ann Rheum Dis 2012;71:238e41. *[11] De Rooy DP, Willemze A, Mertens B, et al. Can anti-cyclic citrullinated peptide antibody-negative RA be subdivided into clinical subphenotypes? Arthritis Res Ther 2011;13:R180. [12] Greiner A, Plischke H, Kellner H, et al. Association of anti-cyclic citrullinated peptide antibodies, anti-citrulline antibodies, and IgM and IgA rheumatoid factors with serological parameters of disease activity in rheumatoid arthritis. Ann N Y Acad Sci 2005;1050:295e303. [13] Ates A, Karaaslan Y, Aksaray S. Predictive value of antibodies to cyclic citrullinated peptide in patients with early arthritis. Clin Rheumatol 2007;26:499e504. *[14] Sun J, Zhang Y, Liu L, et al. Diagnostic accuracy of combined tests of anti cyclic citrullinated peptide antibody and rheumatoid factor for rheumatoid arthritis: a meta-analysis. Clin Exp Rheumatol 2014;32:11e21. [15] Symmons D, Turner G, Webb R, et al. The prevalence of rheumatoid arthritis in the United Kingdom: new estimates for a new century. Rheumatology 2002;41:793e800. [16] NAO. Services for people with rheumatoid arthritis. London: National Audit Office Report; 2009. http://www.nao.org.uk/ publications/0809/services_for_people_with_rheum.aspx. [17] McLoughlin R, Sebastian SS, Qasim A, et al. Coeliac disease in Europe. Aliment Pharmacol Ther 2003;18(Suppl. 3):45e8. [18] O'Neill T, Silman AJ. Psoriatic arthritis. Historical background and epidemiology. Baillieres Clin Rheumatol 1994;8: 245e61. [19] Turkcapar N, Demir O, Atli T, et al. Late onset rheumatoid arthritis: clinical and laboratory comparisons with younger onset patients. Arch Gerontol Geriatr 2006;42:225e31. [20] Lopez-Hoyos M, Ruiz de Alegria C, Blanco R, et al. Clinical utility of anti-CCP antibodies in the differential diagnosis of elderly-onset rheumatoid arthritis and polymyalgia rheumatica. Rheumatol Oxf 2004;43:655e7. [21] Deal CL, Meenan RF, Goldenberg DL, et al. The clinical features of elderly-onset rheumatoid arthritis. A comparison with younger-onset disease of similar duration. Arthritis Rheum 1985;28:987e94. [22] Olivieri I, Pipitone NSDA, Padula A, et al. Late-onset rheumatoid arthritis and late-onset spondyloarthritis. Clin Exp Rheumatol 2009;27:S139e45. [23] Pease CT, Bhakta BB, Devlin J, et al. Does the age of onset of rheumatoid arthritis influence phenotype?: a prospective study of outcome and prognostic factors. Rheumatol Oxf 1999;38:228e34. [24] Olivieri I, Palazzi C, Peruz G, et al. Management issues with elderly-onset rheumatoid arthritis: an update. Drugs Aging 2005;22:809e22. [25] Pease CT, Haugeberg G, Morgan AW, et al. Diagnosing late onset rheumatoid arthritis, polymyalgia rheumatica, and temporal arteritis in patients presenting with polymyalgic symptoms. A prospective long term evaluation. J Rheumatol 2005;32:1043e6 [Erratum appears in J Rheumatol. 2005 Sep;32(9):1852]. [26] Pease CT, Haugeberg G, Montague B, et al. Polymyalgia rheumatica can be distinguished from late onset rheumatoid arthritis at baseline: results of a 5-yr prospective study. Rheumatol Oxf 2009;48:123e7. [27] McCarty DJ, O'Duffy JD, Pearson L, et al. Remitting seronegative symmetrical synovitis with pitting edema. RS3PE syndrome. JAMA 1985;254:2763e7. [28] Bhakta BB, Pease CT. Late-onset rheumatoid arthritis: is pitting oedema of the hands at onset a good prognostic indicator? Br J Rheumatol 1997;36:214e9. [29] Schaeverbeke T, Fatout E, Marce S, et al. Remitting seronegative symmetrical synovitis with pitting oedema: disease or syndrome? Ann Rheum Dis 1995;54:681e4. [30] Yao Q, Su X, Altman RD. Is remitting seronegative symmetrical synovitis with pitting edema (RS3PE) a subset of rheumatoid arthritis? Semin Arthritis Rheum 2010;40:89e94. [31] Cader MZ, Filer AD, Buckley CD, et al. The relationship between the presence of anti-cyclic citrullinated peptide antibodies and clinical phenotype in very early rheumatoid arthritis. BMC Musculoskelet Disord 2010;11:187. [32] Pratt AG, Isaacs JD, Wilson G. The clinical utility of a rule for predicting rheumatoid arthritis in patients with early undifferentiated arthritis: comment on the article by van der Helm-van Mil et al.[comment]. Arthritis Rheum 2009;60: 905. author reply 6.

658

A.G. Pratt, J.D. Isaacs / Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

[33] MacGregor AJ, Snieder H, Rigby AS, et al. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum 2000;43:30e7. [34] van der Woude D, Houwing-Duistermaat JJ, Toes RE, et al. Quantitative heritability of anti-citrullinated protein antibodypositive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum 2009;60:916e23. *[35] Frisell T, Holmqvist M, Kallberg H, et al. Familial risks and heritability of rheumatoid arthritis: role of rheumatoid factor/ anti-citrullinated protein antibody status, number and type of affected relatives, sex, and age. Arthritis Rheum 2013;65: 2773e82. [36] Tobon GJ, Youinou P, Saraux A. The environment, geo-epidemiology, and autoimmune disease: rheumatoid arthritis. J Autoimmun 2010;35:10e4. [37] Kallberg H, Padyukov L, Plenge RM, et al. Gene-gene and gene-environment interactions involving HLA-DRB1, PTPN22, and smoking in two subsets of rheumatoid arthritis. Am J Hum Genet 2007;80:867e75. [38] Stolt P, Bengtsson C, Nordmark B, et al. Quantification of the influence of cigarette smoking on rheumatoid arthritis: results from a population based case-control study, using incident cases. Ann Rheum Dis 2003;62:835e41. [39] Gregersen PK, Silver J, Winchester RJ. The shared epitope hypothesis. An approach to understanding the molecular genetics of susceptibility to rheumatoid arthritis. Arthritis Rheum 1987;30:1205e13. [40] Raychaudhuri S, Sandor C, Stahl EA, et al. Five amino acids in three HLA proteins explain most of the association between MHC and seropositive rheumatoid arthritis. Nat Genet 2012;44:291e6. [41] Hill JA, Southwood S, Sette A, et al. Cutting edge: the conversion of arginine to citrulline allows for a high-affinity peptide interaction with the rheumatoid arthritis-associated HLA-DRB1*0401 MHC class II molecule. J Immunol 2003;171: 538e41. [42] van der Helm-van Mil AHM, Verpoort KN, Breedveld FC, et al. The HLA-DRB1 shared epitope alleles are primarily a risk factor for anti-cyclic citrullinated peptide antibodies and are not an independent risk factor for development of rheumatoid arthritis. Arthritis Rheum 2006;54:1117e21. [43] Padyukov L, Seielstad M, Ong RT, et al. A genome-wide association study suggests contrasting associations in ACPApositive versus ACPA-negative rheumatoid arthritis. Ann Rheum Dis 2011;70:259e65. [44] Eyre S, Bowes J, Diogo D, et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat Genet 2012;44:1336e40. *[45] Viatte S, Plant D, Bowes J, et al. Genetic markers of rheumatoid arthritis susceptibility in anti-citrullinated peptide antibody negative patients. Ann Rheum Dis 2012;71:1984e90. [46] Mackie SL, Taylor JC, Martin SG, et al. A spectrum of susceptibility to rheumatoid arthritis within HLA-DRB1: stratification by autoantibody status in a large UK population. Genes Immun 2012;13:120e8. *[47] Han B, Diogo D, Eyre S, et al. Fine mapping seronegative and seropositive rheumatoid arthritis to shared and distinct HLA alleles by adjusting for the effects of heterogeneity. Am J Hum Genet 2014;94:522e32. [48] Begovich AB, Carlton VEH, Honigberg LA, et al. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. Am J Hum Genet 2004;75:330e7. [49] Burn GL, Svensson L, Sanchez-Blanco C, et al. Why is PTPN22 a good candidate susceptibility gene for autoimmune disease? FEBS Lett 2011;585:3689e98. [50] Fousteri G, Liossis SN, Battaglia M. Roles of the protein tyrosine phosphatase PTPN22 in immunity and autoimmunity. Clin Immunol 2013;149:556e65. [51] Vang T, Congia M, Macis MD, et al. Autoimmune-associated lymphoid tyrosine phosphatase is a gain-of-function variant. Nat Genet 2005;37:1317e9. [52] Zhang J, Zahir N, Jiang Q, et al. The autoimmune disease-associated PTPN22 variant promotes calpain-mediated Lyp/Pep degradation associated with lymphocyte and dendritic cell hyperresponsiveness. Nat Genet 2011;43:902e7. [53] van Oosterhout M, Bajema I, Levarht EWN, et al. Differences in synovial tissue infiltrates between anti-cyclic citrullinated peptide-positive rheumatoid arthritis and anti-cyclic citrullinated peptide-negative rheumatoid arthritis. Arthritis Rheum 2008;58:53e60. *[54] Cantaert T, Brouard S, Thurlings RM, et al. Alterations of the synovial T cell repertoire in anti-citrullinated protein antibody-positive rheumatoid arthritis. Arthritis Rheum 2009;60:1944e56. [55] Gomez-Puerta JA, Celis R, Hernandez MV, et al. Differences in synovial fluid cytokine levels but not in synovial tissue cell infiltrate between anti-citrullinated peptide/protein antibody-positive and -negative rheumatoid arthritis patients. Arthritis Res Ther 2013;15:R182. [56] Stahl EA, Raychaudhuri S, Remmers EF, et al. Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci. Nat Genet 2010;42:508e14. [57] Hu X, Kim H, Stahl E, et al. Integrating autoimmune risk loci with gene-expression data identifies specific pathogenic immune cell subsets. Am J Hum Genet 2011;89:496e506 [Erratum appears in Am J Hum Genet. 2011 Nov 11;89(5):682]. [58] Lill CM, Schjeide BM, Graetz C, et al. Genome-wide significant association of ANKRD55 rs6859219 and multiple sclerosis risk. J Med Genet 2013;50:140e3. [59] Rose-John S, Scheller J, Elson G, et al. Interleukin-6 biology is coordinated by membrane-bound and soluble receptors: role in inflammation and cancer. J Leukoc Biol 2006;80:227e36. [60] Seddighzadeh M, Gonzalez A, Ding B, et al. Variants within STAT genes reveal association with anticitrullinated protein antibody-negative rheumatoid arthritis in 2 European populations. J Rheumtol 2014;39:1509e16. *[61] Pratt AG, Swan DC, Richardson S, et al. A CD4 T cell gene signature for early rheumatoid arthritis implicates interleukin 6mediated STAT3 signalling, particularly in anti-citrullinated peptide antibody-negative disease. Ann Rheum Dis 2012;71: 1374e81. [62] Sawa Si, Kamimura D, Jin GH, et al. Autoimmune arthritis associated with mutated interleukin (IL)-6 receptor gp130 is driven by STAT3/IL-7-dependent homeostatic proliferation of CD4þ T cells. J Exp Med 2006;203:1459e70. [63] Murakami M, Okuyama Y, Ogura H, et al. Local microbleeding facilitates IL-6- and IL-17-dependent arthritis in the absence of tissue antigen recognition by activated T cells. J Exp Med 2011;208:103e14 [Erratum appears in J Exp Med. 2012 Nov 19;209(12):2321]. [64] Weyand CM, Yang Z, Goronzy JJ. T-cell aging in rheumatoid arthritis. Curr Opin Rheumatol 2014;26:93e100.

A.G. Pratt, J.D. Isaacs / Best Practice & Research Clinical Rheumatology 28 (2014) 651e659

659

[65] Tchkonia T, Zhu Y, van Deursen J, et al. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest 2013;123:966e72. [66] Hill JA, Bell DA, Brintnell W, et al. Arthritis induced by posttranslationally modified (citrullinated) fibrinogen in DR4-IE transgenic mice. J Exp Med 2008;205:967e79. [67] Isaacs JD, Ferraccioli G. The need for personalised medicine for rheumatoid arthritis. Ann Rheum Dis 2011;70:4e7. [68] Combe B, Landewe R, Lukas C, et al. EULAR recommendations for the management of early arthritis: report of a task force of the European Standing Committee for International Clinical Studies Including Therapeutics (ESCISIT).[see comment]. Ann Rheum Dis 2007;66:34e45. [69] van Dongen H, van Aken J, Lard LR, et al. Efficacy of methotrexate treatment in patients with probable rheumatoid arthritis: a double-blind, randomized, placebo-controlled trial. Arthritis Rheum 2007:1424e32. [70] van Aken J, Heimans L, Gillet-van Dongen H, et al. Five-year outcomes of probable rheumatoid arthritis treated with methotrexate or placebo during the first year (the PROMPT study). Ann Rheum Dis 2014;73:396e400. [71] Hider SL, Silman AJ, Thomson W, et al. Can clinical factors at presentation be used to predict outcome of treatment with methotrexate in patients with early inflammatory polyarthritis? Ann Rheum Dis 2009;68:57e62. [72] Wessels JAM, van der Kooij SM, le Cessie S, et al. A clinical pharmacogenetic model to predict the efficacy of methotrexate monotherapy in recent-onset rheumatoid arthritis. Arthritis Rheum 2007;56:1765e75. [73] van den Broek M, Dirven L, Klarenbeek NB, et al. The association of treatment response and joint damage with ACPAstatus in recent-onset RA: a subanalysis of the 8-year follow-up of the BeSt study. Ann Rheum Dis 2012;71:245e8. [74] Klarenbeek NB, van der Kooij SM, Guler-Yuksel M, et al. Discontinuing treatment in patients with rheumatoid arthritis in sustained clinical remission: exploratory analyses from the BeSt study. Ann Rheum Dis 2011;70:315e9. [75] NICE. Rheumatoid arthritis: the management of rheumatoid arthritis in adults. London: National Institute of Health and Clinical Excellence; 2009. http://www.nice.org.uk/CG79. [76] de Jong PH, Hazes JM, Han HK, et al. Randomised comparison of initial triple DMARD therapy with methotrexate monotherapy in combination with low-dose glucocorticoid bridging therapy; 1-year data of the tREACH trial. Ann Rheum Dis 2014;73:1331e9. [77] Choy EH, Smith CM, Farewell V, et al. Factorial randomised controlled trial of glucocorticoids and combination disease modifying drugs in early rheumatoid arthritis. Ann Rheum Dis 2008;67:656e63. *[78] Seegobin SD, Ma MH, Dahanayake C, et al. ACPA-positive and ACPA-negative rheumatoid arthritis differ in their requirements for combination DMARDs and corticosteroids: secondary analysis of a randomised controlled trial. Arthritis Res Ther 2014;16:R13. [79] Bobbio-Pallavicini F, Caporali R, Alpini C, et al. High IgA rheumatoid factor levels are associated with poor clinical response to tumour necrosis factor alpha inhibitors in rheumatoid arthritis. Ann Rheum Dis 2007;66:302e7. [80] Fisher BA, Plant D, Lundberg K, et al. Heterogeneity of anticitrullinated peptide antibodies and response to anti-tumor necrosis factor agents in rheumatoid arthritis. J Rheumatol 2012;39:929e32. [81] Potter C, Hyrich KL, Tracey A, et al. Association of rheumatoid factor and anti-cyclic citrullinated peptide positivity, but not carriage of shared epitope or PTPN22 susceptibility variants, with anti-tumour necrosis factor response in rheumatoid arthritis. Ann Rheum Dis 2009;68:69e74 [Erratum appears in Ann Rheum Dis. 2011 Aug;70(8):1519]. [82] Klaasen R, Cantaert T, Wijbrandts CA, et al. The value of rheumatoid factor and anti-citrullinated protein antibodies as predictors of response to infliximab in rheumatoid arthritis: an exploratory study. Rheumatology 2011;50:1487e93. [83] Potter C, Gibbons LJ, Bowes JD, et al. Polymorphisms spanning the TNFR2 and TACE genes do not contribute towards variable anti-TNF treatment response. Pharmacogenet Genomics 2010;20:338e41. [84] Pers YM, Cadart D, Rittore C, et al. TNFRII polymorphism is associated with response to TNF blockers in rheumatoid arthritis patients seronegative for ACPA. Jt Bone Spine 2014. http://dx.doi.org/10.1016/j.jbspin.2013.12.005. *[85] Isaacs JD, Cohen SB, Emery P, et al. Effect of baseline rheumatoid factor and anticitrullinated peptide antibody serotype on rituximab clinical response: a meta-analysis. Ann Rheum Dis 2013;72:329e36. [86] Tak PP, Rigby WF, Rubbert-Roth A, et al. Inhibition of joint damage and improved clinical outcomes with rituximab plus methotrexate in early active rheumatoid arthritis: the IMAGE trial. Ann Rheum Dis 2011;70:39e46. *[87] Gottenberg JE, Ravaud P, Cantagrel A, et al. Positivity for anti-cyclic citrullinated peptide is associated with a better response to abatacept: data from the ‘Orencia and Rheumatoid Arthritis’ registry. Ann Rheum Dis 2012;71:1815e9. [88] Pieper J, Herrath J, Raghavan S, et al. CTLA4-Ig (abatacept) therapy modulates T cell effector functions in autoantibodypositive rheumatoid arthritis patients. BMC Immunol 2013;14:34. [89] Wang J, Platt A, Upmanyu R, et al. IL-6 pathway-driven investigation of response to IL-6 receptor inhibition in rheumatoid arthritis. BMJ Open 2013;3:e003199. [90] Pers YM, Fortunet C, Constant E, et al. Predictors of response and remission in a large cohort of rheumatoid arthritis patients treated with tocilizumab in clinical practice. Rheumatol Oxf 2014;53:76e84.

Seronegative rheumatoid arthritis: pathogenetic and therapeutic aspects.

Rheumatoid arthritis (RA) has long been recognised as a highly heterogeneous disease of immune dysregulation. Despite an ever-growing appreciation of ...
554KB Sizes 2 Downloads 12 Views