quarta-feira, 13 de julho de 2016

Preclinical Rheumatoid
Arthritis: Identification,
Evaluation, and Future
Directions for
Investigation
Kevin D. Deane, MDa,*, Jill M. Norris, MPH, PhDb,
V. Michael Holers, MDa
‘‘When the rheumatic poison is in the system, any disturbing circumstance, even
of temporary duration, such as over fatigue, anxiety, grief or anger, by rendering
the system more susceptible of its influence, may prove the accidental or exciting
cause of the disease’’
Henry William Fuller in ‘‘On Rheumatism, Rheumatic Gout, and Sciatica’’
Publishers: Samuel S & William Wood, New York, 1854.
The discovery of genetic and environmental factors associated with rheumatoid
arthritis (RA), and elevations in autoantibodies and inflammatory markers prior to
the onset of symptomatic disease, coupled with similar findings in other autoimmune
diseases including type 1 diabetes mellitus (T1DM), has led to the creation of a shared
model of autoimmune disease development. In this model, the development of RA
follows a natural history divided into phases wherein genetic and environmental interactions initially lead to a period of asymptomatic autoimmunity, evidenced by the
presence of RA-related autoantibodies, that later evolves into clinically apparent
disease. It is the initial phases of risk and asymptomatic autoimmunity that encompass
‘‘preclinical’’ RA.
To understand the genetic and environmental influences that are important to the
evolution of RA, as well as develop predictive models and preventive strategies for
a Division of Rheumatology, University of Colorado School of Medicine, 1775 Aurora Court,
Mail Stop B-115, Aurora, CO 80045, USA
b Department of Epidemiology, Colorado School of Public Health, University of Colorado,
13001 East 17th Place, Aurora, CO 80045, USA
* Corresponding author.
E-mail address: Kevin.Deane@UCDenver.edu
KEYWORDS
Rheumatoid arthritis Preclinical period
Anticyclic citrullinated peptide antibodies
Rheum Dis Clin N Am 36 (2010) 213–241
doi:10.1016/j.rdc.2010.02.001 rheumatic.theclinics.com
0889-857X/10/$ – see front matter ª 2010 Elsevier Inc. All rights reserved.
future symptomatic disease, this preclinical period must be investigated. Herein are
discussed the following issues related to preclinical RA: (1) what is known about the
preclinical development of autoimmunity and inflammation in RA as well as other autoimmune diseases that may follow a similar model of development as RA, (2) how RA
development can be modeled based on studies in preclinical RA and other autoimmune diseases, (3) practical issues related to the challenge of defining for research
studies ‘‘preclinical’’ RA, as compared with clinically apparent disease, and (4) what
aspects of RA evolution including genetic and environmental influences, and predictive and preventive models, could be addressed in studies of the preclinical period.
Finally, potential methodologies and areas of focus going forward for research into
preclinical RA are discussed.
PART 1. AUTOANTIBODIES AND INFLAMMATION IN PRECLINICAL RHEUMATOID
ARTHRITIS AS WELL AS OTHER AUTOIMMUNE DISEASES
Studies of Preclinical Rheumatoid Arthritis
Multiple studies have shown that RA-related autoantibodies are present years before
the diagnosis of RA (Table 1).1–11 del Puente and colleagues,1 who investigated RA in
the Pima Indians in the Southwestern United States, showed that rheumatoid factor
was present before the onset of clinically apparent RA. Aho and colleagues,2,12–14
who investigated preclinical RA in Finland using a biobank of stored prediagnosis
samples, and Jonsson and colleagues,15 who used Icelandic biobank samples, also
demonstrated that rheumatoid factor (RF) (by various methodologies) was present
prior to the onset of clinically apparent RA. Also, in a prospective study of initially
healthy family members of patients with RA, Silman and colleagues11 showed that
the presence of RF preceded the onset of clinically apparent RA.
Later studies also showed preclinical RA positivity for RF, as well as positivity for
the highly RA-specific antibodies to citrullinated protein antigens (ACPAs). Again
using a Finnish biobank, Aho and colleagues3,4 demonstrated preclinical RA positivity
of antikeratin (AKA) and antiperinuclear factor (APF) antibodies, and antifillagrin antibodies (AFA)6—autoantibody targets that, based on later findings, represented citrullinated antigens.16 Using stored blood samples available through the Medical
Biobank of Northern Sweden, Rantapaa-Dahlqvist and colleagues7 evaluated for
elevations of RF isotypes (immunoglobulins [Ig] M, G and A) and the anticyclic citrullinated peptide (anti-CCP) antibody in 98 preclinical samples from 83 RA patients,
collected a median of 2.5 years prior to symptomatic disease onset. In this study,
approximately 34% of patients were positive for anti-CCP within 1.5 years prior to
diagnosis of RA, and the prevalence of RF isotype positivity ranged from about
17% to 34% during this same period. In addition, in comparison to controls, a combination of both anti-CCP and any RF isotype was highly specific (99%) for the future
development of classifiable RA. This report was followed by a similarly designed
retrospective cohort study by Nielen and colleagues8 that evaluated preclinical RA
RF and anti-CCP positivity using pre-RA diagnosis samples stored in a Dutch blood
donor biobank. In this study, 79 RA patients with stored prediagnosis samples were
identified, with a median of 13 preclinical samples per case. Of these 79 cases,
approximately 28% and 41% had pre-RA diagnosis elevation of RF (IgM isotype) or
anti-CCP, respectively. RF was positive a median of 2.0 years prior to RA diagnosis
(range 0.3–10.3 years), and anti-CCP was positive a median of 4.5 years prior to
RA diagnosis (range 0.1–13.8 years).
Additional studies using stored biobank samples have also demonstrated preclinical
RA positivity for autoantibodies. Using stored pre-RA samples from 83 United States
214 Deane et al
military subjects with RA, Majka and colleagues9 demonstrated pre-RA diagnosis positivity for RF and anti-CCP. In addition, they demonstrated that individuals that were
older at the time of diagnosis of RA had longer duration of preclinical positivity of autoantibodies, a finding that was supported by work by Bos and colleagues.17 Chibnik and
colleagues18 utilized the Nurses’ Health Study (NHS) biobank to demonstrate anti-CCP
positivity prior to RA diagnosis, and showed that a lower cut-off value for anti-CCP than
the kit suggested was also highly specific for future RA.
Multiple studies have also evaluated elevations of inflammatory markers prior to
diagnosis of RA, with varying results (see Table 1).19–24 In their Finnish biobank study,
Aho and colleagues25 found no significant elevation of C-reactive protein (CRP) in preRA diagnosis samples, although the time of sample collection prediagnosis was not
reported. Using the same Medical Biobank of Northern Sweden as in the study of
pre-RA RF and anti-CCP positivity, Rantapaa-Dahlqvist and colleagues21 showed
a significant elevation of monocyte chemoattractant protein-1 (MCP-1) in 92 pre-RA
cases versus controls, but not secretory phospholipase A2 (sPLA2), high-sensitivity
CRP (hsCRP) or interleukin (IL)-6. Nielen and colleagues19 demonstrated, in the Dutch
pre-RA sample cohort described above, that CRP was elevated in the pre-RA period,
most commonly about 2 years before diagnosis, regardless of prediagnosis autoantibody positivity, although these investigators were unable in this study to demonstrate
the chronologic sequence of appearance of CRP versus autoantibodies. Nielen and
colleagues20 later additionally demonstrated pre-RA elevation of sPLA2 in this cohort,
but were unable to determine whether CRP or sPLA2 preceded autoantibodies in the
preclinical period, and they concluded that the temporal development of autoantibodies and these 2 markers were probably similar. Jorgensen and colleagues24
utilized samples from a Norwegian biobank to examine in 49 cases with RA prediagnosis elevations of autoantibodies (RF and anti-CCP) and multiple cytokines. These
investigators found that RF and anti-CCP were elevated in RA cases prediagnosis;
however, they could not demonstrate any cytokine elevations prior to 5 years before
disease onset, and only tumor necrosis factor a (TNFa) was statistically significantly
elevated in cases (vs controls) during the 5-year period just before diagnosis of
RA.24 Using a single pre-RA diagnosis blood sample from 90 incident RA cases
(case status confirmed by chart review) identified in the Women’s Health Study
(WHS), Shadick and colleagues23 were unable to demonstrate significant elevations
in CRP levels in cases versus controls (with case samples available a mean of 6.6
years prior to diagnosis), and they were unable to use a single CRP level to predict
future RA. However, in a later study using 170 RA cases from a combination of the
NHS and WHS cohorts, the same group22 was able to demonstrate statistically significant elevations of soluble tumor necrosis factor receptor II (sTNFRII) prior to RA diagnosis, but not IL-6 or hsCRP. Most recently, Rantaapa-Dahlqvist and colleagues used
an expanded sample set from their Swedish Biobank, to demonstrate elevations of
multiple cytokines and chemokines prior to the diagnosis of RA, with these elevations
likely indicating pre-clinical RA activity of Th1, Th2, and T regulatory processes.26
There are caveats when interpreting the data regarding pre-clinical elevations of
biomarkers. Certainly the prospective studies by del Puente and colleagues1 and Silman and colleagues11 suggest that autoantibodies are truly elevated before the onset
of clinically apparent disease. However, in the studies of preclinical autoantibody and
inflammatory marker elevations using stored samples from biobanks, there were not
detailed joint-directed questionnaires or examinations performed at baseline or over
time to ensure that no clinically apparent arthritis was present at the time of presumed
preclinical RA blood collections. As such, it may be that the duration of pre-RA diagnosis autoantibody positivity is overestimated in the studies utilizing stored biobank
Preclinical Rheumatoid Arthritis 215
Table 1
Summary of selected studies of preclinical rheumatoid arthritis (RA)
Study Study Design
Biomarkers
Assessed Findings
Implications for Prediction
of Future RA
del Puente et al,
19881
Prospective; Native Americans,
Southwest United States
RF RF precedes diagnosis of RA Increased incidence of RA in RF1
individuals. Rates 2.4–48.3 per
1000 person-years, with highest
rates in those with highest RF
titers at baseline
Silman et al,
199211
Prospective; British; FDRs from
families with R2 RA cases
RF RF precedes diagnosis of RA Average incidence of RA 8 per
1000 person-years in FDRs;
highest rate in FDRs with RF1:
34.8 per 1000 person-years
Jonsson et al,
199215
Retrospective; Icelandic;
biobank
RF (isotypes) RF isotype elevations precede
diagnosis of RA
Not analyzed
Aho et al,
1985–19912,13,14
Retrospective; Finnish;
biobank
RF (isotypes) RF precedes diagnosis of RA Not analyzed
Aho et al,
1993, 20004,6
Retrospective; Finnish;
biobank
AKA, AFA, APF (later studies
showed target antigens
likely citrullinated)
AKA, AFA, APF precede
diagnosis of RA
RA-related antibodies may be
present in subjects who did not
develop RA in follow-up period
Rantapaa-Dahlqvist
et al, 20037
Retrospective; Swedish;
biobank; N 5 83 RA cases
RF, anti-CCP RF, anti-CCP precede diagnosis
of RA; 34% anti-CCP1 %1.5
years prior to RA diagnosis;
RF-isotypes1 in ~17%–34%
of cases pre-RA diagnosis
PPV 22%for future RA if RF-IgA
and anti-CCP positive (estimated
population prevalence of RA of
1%)
Nielen et al, 20048 Retrospective; Dutch;
biobank; N 5 79 RA cases
RF-IgM, anti-CCP RF-IgM, anti-CCP precede RA
diagnosis; 49% positive for
RF-IgM and/or anti-CCP pre-RA.
RF-IgM1 or anti-CCP1 median
of 2.0 or 4.8 years prior to
diagnosis of RA, respectively
PPV up to 100% for RA diagnosis
within 5 years based on 5-year
incidence rates of 0.001 (general
population) or 3.9% (estimated
from high-risk multicase RA
families)
216 Deane et al
Majka et al, 20089 Retrospective; United States
Military; biobank; N 5 83
RA cases
RF, anti-CCP Pre-RA diagnosis: RF1 57%
of cases, median 6.0 years;
anti-CCP1 61% of cases,
median 5.4 years
Increased age-at-diagnosis of RA
associated with longer duration
of preclinical autoantibody
positivity (replicated by Bos
et al17)
Rantapaa-Dahlqvist
et al, 200721
Retrospective; Swedish;
biobank
MCP-1, IL-6, CRP, sPLA2 After adjustment, only MCP-1
elevated prior to RA diagnosis
Not analyzed
Nielen et al,
2004, 200619,20
Retrospective; Dutch;
biobank
RF, anti-CCP, CRP, sPLA2 CRP and sPLA2 elevations precede
RA diagnosis; CRP ~2 years prior;
timing similar to RF/anti-CCP
Not analyzed
Jorgensen et al,
200824
Retrospective; Norwegian;
biobank; N 5 49 RA cases
RF-IgM, anti-CCP,
multiple cytokines
RF-IgM and/or anti-CCP precede
RA diagnosis by as much as 20
years; anti-TNF elevated <5 years
prior to diagnosis of RA. EBV
and Parvovirus serologies not
different between cases and
controls
Suggests that cytokine elevation
may indicate symptomatic
disease within 5 years
Berglin et al,
2004115
Retrospective; Swedish;
biobank; N 5 59 RA cases
RF, anti-CCP, HLA-DRB1
alleles *0404, *0401
Anti-CCP 1 DRB1*0404 or 0401
high-risk for future RA (OR ~67)
Johansson et al,
2006116
Retrospective; Swedish;
biobank; N 5 92 RA cases
RF, anti-CCP; PTPN22
(1858T)
Anti-CCP 1 PTPN22: 100% specific
for future RA, and high-risk
(OR >132)
Abbreviations: AFA, antifilaggrin antibodies; AKA, antikeratin antibodies; anti-CCP, anticyclic citrullinated peptide antibodies; APF, antiperinuclear factor antibodies; CRP, C-reactive protein; FDR, first-degree relative of proband with RA; HLA, human leukocyte antigen; IL-6, interleukin-6; MCP-1, monocyte chemoattractant protein-1; OR, odds ratio; PPV, positive predictive value; PTPN22, protein phosphatase 22; RF, rheumatoid factor; sPLA2, secretory phospholipase A2.
Preclinical Rheumatoid Arthritis 217
samples, as subjects may have had mild or fluctuating inflammatory symptoms long
before a confirmed diagnosis of RA. Also of importance, these pre-RA studies show
that not all RA patients have detectable pre-RA diagnosis autoantibody positivity.
Methodologic issues may in part explain these findings. For example, patients may
not have a stored blood sample available for analysis from the correct time period
to demonstrate their prediagnosis autoantibody positivity. Also, current assays for
autoantibodies may not detect the earliest preclinical autoantibody specificities;
perhaps an as-of-yet unknown citrullinated antigen is the earliest autoantibody target
in preclinical RA? In addition, not all patients with RA may develop detectable circulating autoantibodies in the preclinical period. For example, some patients may
develop circulating autoantibodies after clinically apparent disease develops. Of
note, in the Nielen study, while only about 28% of patients with RA were RF-positive
prediagnosis, about 67% of these same patients were positive for RF by 6 years post
diagnosis, suggesting that circulating RF develops in some cases after symptomatic
onset of RA.8 All of these issues will need to be considered in studies of preclinical
RA going forward.
The Contribution of Genetic and Environmental Factors Associated
with Rheumatoid Arthritis to Understanding Preclinical Development
Although the exact etiology of RA is unknown, there are multiple genetic and environmental factors that have been associated with disease. Estimates of the genetic
contribution to RA have ranged between 30% and 60%.27,28 Of the known genetic
factors associated with RA, a DRB1 allele containing the ‘‘shared epitope’’ is the
most important genetic factor associated with RA.29,30 Of the HLA alleles, HLADRB1*0401 and HLA-DRB1*0404 (within the HLA-DR4 group) are the most strongly
associated with RA, with an approximate relative risk for RA of almost 4 in Caucasians.31 In addition, several genes outside the HLA-DRB1 gene have recently been
associated with RA. These genes include PTPN22, TRAF1/C5, CTLA4, and STAT4,
and the list is rapidly growing.32,33
Several potential environmental factors that could play important roles in modifying
either susceptibility to RA or disease severity have been identified. High levels of
coffee consumption,34 in particular decaffeinated coffee,35 as well as a positive
smoking history,31,36 exposure to air pollution,37 and environmental exposure to
silica-containing dust38,39 are also associated with increased risk for RA. In addition,
infections have been associated with RA including pathogens such as the EpsteinBarr virus (EBV), Mycoplasma or Proteus species.27,40–43 Finally, several factors
have been identified as possibly protective against development of RA including
a history of successful pregnancy,44,45 oral contraceptive pill use,46–48 and higher
vitamin D intake.49
Several studies have examined the association of environmental factors with
asymptomatic RA-related autoantibody positivity. These studies suggest that lack
of oral contraceptive use and a history of heavy smoking are associated with an
increased prevalence of RF,50–52 whereas 25,hydroxyvitamin D levels are not associated with RF positivity in individuals without RA.53 Active pulmonary infection with
tuberculosis has also been associated with RF and anti-CCP positivity in individuals
without clinically apparent synovitis.54,55 These studies suggest that environmental
factors affect RA-related autoimmunity, although prospective evaluation of future
risk for RA in these types of autoantibody positive subjects is lacking.
Of recent interest regarding the pathogenesis of RA are interactions between
genetic and environmental factors that may lead to RA-related autoimmunity and
disease. For example, several studies have reported that smoking in individuals with
218 Deane et al
specific HLA alleles is associated with ACPA-positive RA, suggesting that a geneenvironment interaction leads to RA-related autoimmunity.56–59
However, while genetic and environmental factors have been associated with RA,
the exact role that these factors play in the development of RA-related autoimmunity
is not yet clear. Also, it is unclear whether these factors may lead to initial RA-related
immune dysregulation or transition from asymptomatic autoimmunity to clinically
apparent RA. It is important that, as discussed later, prospective study of the early
phases of RA development should help to clarify these issues.
Preclinical Studies in Other Autoimmune Diseases
In addition to the information about preclinical RA available from RA-specific studies,
much about the evolution of autoimmunity can be learned by examining the preclinical
natural history of other autoimmune diseases including T1DM and systemic lupus
erythematosus (SLE). In particular, prospective studies of T1DM have led to the development of a model of disease evolution that may be of importance to RA.
T1DM results from autoimmune-mediated destruction of the pancreatic b cells, and
it affects approximately 1 in 300 children. Numerous studies of T1DM have established
that there are specific high-risk HLA genotypes that predispose to disease.60,61 Moreover, the autoimmune attack on the pancreatic b cells can be detected years before
clinical onset of T1DM via the presence of any of the T1DM-related autoantibodies
in the blood including antibodies to the following antigens: islet cell (ICA), insulin
(IAA), protein tyrosine phosphatase (IA2), and glutamic acid decarboxylase
(GAD65).62,63
Prospective studies of T1DM have established that T1DM exhibits several phases
during its development.63 The first phase is defined as the presence of genetic risk
factors that, either alone or in combination, may predispose to the loss of self tolerance. The second phase is reached when transformation from the risk state to a state
of immunologic autoreactivity occurs; this second phase of ‘‘asymptomatic autoimmunity’’ is measurable by testing for T1DM-related biomarkers, but this phase is not
yet associated with the presence of clinically apparent hyperglycemia. This transition
from genetic risk to asymptomatic autoimmunity occurs either because of the introduction of an environmental factor, or perhaps because of the stochastic nature of
the immune response. In T1DM, this preclinical autoimmune phase is marked by initial
immune reactivity to only a small number of autoantigens. The third phase of T1DM is
the development of clinically apparent hyperglycemia; this final phase is characterized
immunologically by autoreactivity to numerous antigens and extensive immune-mediated tissue destruction of islet cells caused by many proinflammatory pathways.
The presence of these 3 phases of diseases in T1DM is well established, as is the
value of prospectively studying children with highly predictive autoantibodies in the
preclinical phases of disease for epidemiologic and genetic associations.60,64–70 Of
importance, due to the strong association of T1DM with b-cell (or islet) autoimmunity
(IA), defined as the presence of autoantibodies specific to b-cell autoantigens, IA has
become an alternative end point to clinically apparent hyperglycemia in T1DM
research. Advantages of this approach include the opportunity to study the pathologic
process underlying T1DM in a preclinical state, and to verify that a candidate risk
factor is not only related to diabetes but also to the alteration of immune function
preceding clinical onset of disease. Studies using as end points both autoimmunity
and clinically apparent diabetes have allowed for differentiation of the risk factors
that initiate humoral autoimmunity from those that promote progression from subclinical autoimmunity to diabetes.60,70 Of note, prospective studies using asymptomatic
IA as an outcome measure have identified important findings regarding dietary and
Preclinical Rheumatoid Arthritis 219
other factors in T1DM, including the lack of association of islet-cell autoimmunity with
childhood vaccines,68 and the important relationship of timing of cereal exposure
during the first year of life to the later development of IA.66 This latter finding has
been of importance in terms of potentially reducing risk for T1DM based on dietary
considerations.
The presence of autoantibodies can be also used in the prediction of future T1DM. In
first-degree relatives (FDRs) of diabetic individuals, the presence of a single autoantibody on one occasion has been found to have a sensitivity and specificity of 95%
and 99%, respectively, and a positive predictive value (PPV) of 46% for type 1 diabetes
with 5 years of follow-up.71 Of note, the number of unique T1DM-related autoantibodies
that are positive also predicts risk for future T1DM. For example, a person who tests
positive for multiple autoantibodies is at a much higher risk of developing T1DM than
a person with a single detectable autoantibody.72 However, T1DM-related autoantibodies may be transiently positive, and there are not yet enough longitudinal data to
determine if all people that develop autoantibodies will eventually develop
T1DM.63,72,73 Although not 100% predictive of future T1DM development, IA still serves
as a very useful intermediate end point when studying the autoimmune disease process
and potential risk factors for T1DM. In terms of RA, given the high specificity of certain
autoantibodies (notably ACPAs) for disease, using autoantibody positivity as a surrogate end point for autoimmunity in preclinical RA is likely to be of similar great value.
SLE is a multisystem autoimmune disease of unknown etiology. More than 98% of
SLE patients are positive for antinuclear antibodies (ANA) and many SLE patients have
additional reactivity to specific nuclear antigens including double-stranded DNA
(dsDNA), Smith, ribonuclear proteins, and Ro and La.74–77 Some of these autoantibodies are associated with specific clinical manifestations of disease. For example,
autoantibodies to dsDNA are associated with nephritis, and levels of anti-dsDNA antibodies may fluctuate in conjunction with disease activity.78,79 In addition, anti-Ro antibodies have also been shown to be associated with specific manifestations of SLE
including skin disease and neutropenia.75–77 Although these autoantibody systems
have been extensively studied for more than 50 years, the initiating events for the
development of these autoantibodies and the actual roles these antibody specificities
play in clinical SLE are not known. However, these SLE-related autoantibodies are
present in the serum of lupus patients many years prior to the first clinical evidence
of disease.80,81 Furthermore, the spectrum/number of autoantibody specificities
increases up to the time of diagnosis.82 The discovery of preclinical autoantibody positivity and evolution of antibody specificity has led to important investigations into
potential etiologic agents such as EBV infection in SLE, investigations that could not
be performed without preclinical studies.83
Animal Models and Preclinical Rheumatoid Arthritis
While animal models of arthritis are not an exact match to human disease, they have
provided important insights into the preclinical period of arthritis development. In
particular, investigations into the relationships between the major histocompatibility
complex (MHC) class II dependence of disease, a requirement in some instances
for the specific exposure to citrullinated autoantigen, and the timing between the
evolution of ACPA and clinically apparent arthritis have been helpful in understanding
important factors in preclinical arthritis. An early example was provided by evidence
that citrullinated proteins are found in the synovium after immunization with bovine
collagen type II in DBA/1j mice, a strain that is commonly used to study the type II
collagen-induced arthritis (CIA) model of RA.84 However, in this study no ACPAs
were detected. In a second study of the same model, ACPAs were found to develop
220 Deane et al
before clinically detectable joint inflammation.85 In this latter study, 2 methods were
used to show that the ACPAs are pathogenic: first, treatment of mice with a ‘‘tolerogenic’’ form of a citrullinated peptide decreased the arthritis disease severity by
approximately 60% and decreased the levels of ACPAs and epitope spreading to
other citrullinated peptides; and second, a mouse monoclonal antibody that recognized citrullinated peptides, and could be used as a representative example of an
ACPA, was found to greatly amplify joint inflammation. This process occurred only
following the administration of low doses of antibodies specific for type II collagen,
which induced the generation of citrullinated target antigens, presumably through
the actions of macrophages and neutrophils containing peptidyl arginine deiminase
(PAD) enzymes that participated in the inflammatory response. This second experiment suggested that, in the absence of citrullinated target antigens, there is no target
injury; however, when these antigens are present, such as during treatment with
monoclonal antibodies specific for type II collagen, ACPAs can greatly amplify inflammation and damage. Recently, additional monoclonal antibodies reactive with citrullinated type II collagen, with or without cross-reactivity to other epitopes, were also
found to amplify the development of arthritis in mice in vivo.86 In addition, another
study in rats has shown an increased incidence and severity of arthritis when collagen
type II is citrullinated before immunization.87
Other animal studies have focused on the relationship between cellular and humoral
autoimmunity to citrullinated antigens and the shared epitope and class II molecules.
For example, immunization with citrullinated vimentin peptide leads to CD41 T-cell
activation and proliferation in transgenic mice expressing human HLADRB1*0401.88 In addition, immunization of human HLA-DRB1*0401-transgenic
C57BL/6 mice with citrullinated human fibrinogen, but not native human fibrinogen
or either form of mouse fibrinogen, resulted in the development of arthritis in a substantial proportion of animals.89 In this setting, a relatively low level of inflammation was
present, but this was accompanied by specific antibody responses to citrullinated
human fibrinogen peptide, as well as T-cell responses to citrullinated protein.
ACPA-positive arthritis was also found to develop in mice that are immunized with
low doses of collagen type II and that have dysregulated MHC expression in the joints
owing to transgenic expression of class II transactivator.90 Finally, a particularly
intriguing study has recently shown that immunization of a subset of strains of mice
with human fibrinogen alone resulted in destructive arthritis as well as T- and B-cell
reactivity to fibrinogen.91 In this study, epitope spreading to citrullinated fibrinogen
as well as many synovial citrullinated and noncitrullinated autoantigens developed.
Remarkably, arthritis could be transferred to naı¨ve mice with either serum or fibrinogen-reactive T cells from immunized mice.
PART 2. THE PHASES OF RHEUMATOID ARTHRITIS DEVELOPMENT: FROM GENETIC
RISK TO CLINICALLY APPARENT DISEASE
Based on the above discussion, including the presence of preclinical RA-related autoantibodies and inflammatory markers, the genetic and environmental factors associated with RA, the models of autoimmune disease development established in
prospective studies of T1DM, and the animal models of disease, it is likely that RA
develops 3 phases, outlined as follows and in Fig. 1A. The initial phase (Phase 1) is
characterized by genetic risk for RA, during which no biomarkers of active autoimmunity and inflammation or symptoms are present. This Phase 1 is followed by environmental and additional genetic influences that lead to a Phase 2 of disease
development—asymptomatic autoimmunity—characterized by the presence of
Preclinical Rheumatoid Arthritis 221
Fig. 1. A model of rheumatoid arthritis (RA) development. (A) Based on RA studies as well as prospective studies in other autoimmune disease (type 1
diabetes mellitus), RA may evolve through 3 phases of disease: Phase 1 5 genetic risk, Phase 2 5 asymptomatic autoimmunity (identified by presence of
autoantibodies) and Phase 3 5 clinically apparent disease. Transition between phases may be caused by interactions between genetic and environmental factors, and/or changes in immune reactivity. (B) RA-related factors that can be measured during the preclinical phases of disease development.
(Adapted from Kolfenbach J, Deane KD, Derber LA, et al. A prospective approach to investigating the natural history of preclinical rheumatoid arthritis
(RA) using first-degree relatives of probands with RA. Arthritis Rheum 2009;61(12):1735–4; with permission.)
222 Deane et al
Fig. 1. (continued)
Preclinical Rheumatoid Arthritis 223
RA-related autoantibodies and other immunologic factors such as T- and B-cell autoreactivity, and perhaps elevated inflammatory markers. Phase 2 may be of variable
length, perhaps influenced by genetic, environmental, or endogenous factors such
as age or gender. As Phases 1 and 2 are asymptomatic, they may collectively be
termed ‘‘preclinical’’ RA. Finally, there is a transition from asymptomatic autoimmunity
to Phase 3, or clinically apparent disease. During this final phase, patients will have
symptoms and signs of active RA, with numerous autoimmune and inflammatory
biomarkers present as well as clear evidence of end-organ damage.
Of note, although in comparison with T1DM the natural history of RA is much less
well understood, there are several studies that have demonstrated results very consistent with this model of disease development for RA. These studies include several
associating the presence of high-risk HLA-DR alleles containing the shared epitope
with RA29,30 and, as described above, the finding of increased levels of autoantibodies
in individuals who later develop RA. In addition, studies in patients with established RA
demonstrating altered synovial pathology in clinically unaffected joints suggest that
there may be a period of time that presymptomatic joint inflammation is present during
RA development.92
PART 3. DEFINING PRECLINICAL RHEUMATOID ARTHRITIS AND TRANSITION
INTO CLINICALLY APPARENT DISEASE
A key aspect of this 3-phase model of RA development is that there is a transition
period from the ‘‘preclinical’’ state (or Phases 1 and 2), where specific disease
markers may be present but there are no symptoms or signs of active inflammatory
disease, to a ‘‘clinical’’ period of RA, when symptoms and signs of active inflammatory
disease are present. There may be agreement that RA-related autoantibody positivity,
in the absence of joint symptoms or other organ injury, in subjects that eventually
develop fully classifiable RA, represents preclinical RA. However, defining this transition from preclinical to clinical disease is more difficult: at what point does clinically
apparent RA begin?
To date, given that the main anatomic site affected by RA is the synovial joint, the
criteria for determining the presence of ‘‘clinically apparent’’ RA have largely focused
on the joints. In the American College of Rheumatology (ACR) 1987 Revised Classification Criteria for RA, 4 of 7 criteria need to be fulfilled for RA to be defined.93
However, it is important to remember that these criteria were developed to help standardize research studies in RA, that all of the disease manifestations that result in
criteria fulfillment take time to develop, and that a patient meeting these criteria
may have truly transitioned from preclinical to clinical RA years prior to fulfilling the
ACR criteria for RA.94,95
In an attempt to investigate early joint disease that may not meet the 1987
Revised ACR Criteria, there are several investigative groups that have sought to
classify inflammatory arthritis (IA) or undifferentiated arthritis (UA) as a distinct clinical entity. Such classification allows for the establishment of prospective clinical
studies to determine the long-term outcomes of patients who present with such
findings, as well as to identify factors that may predict evolution to classifiable
and factors that may indicate that early aggressive therapy to prevent long-term
disability is warranted. Of note, Verweij and colleagues have shown that, in patients
with arthralgia and ACPA positivity but who do not meet classification criteria for RA,
upregulation of certain genes in peripherally-circulating blood cells predicts risk for
future development of fully-classificiable RA (by ACR criteria) within a defined time
period.96
224 Deane et al
The most commonly utilized criteria for IA/UA are the Norfolk criteria and the Leiden
criteria.97–101 The Norfolk criteria define IA as 2 or more swollen joints of duration
greater than 4 weeks.97 In the Leiden criteria, IA/UA is defined as 1 or more joint
with inflammatory findings confirmed by a rheumatologist, with the joint findings not
otherwise classifiable (ie, gout or pseudogout).98 In addition, together the ACR and
the European League Against Rheumatism (EuLAR) are currently developing revised
RA criteria, in part to address the need to link IA to the same long-term prognostic
considerations and need for treatment, as are typical for RA as defined by the earlier
ACR criteria.102
The Norfolk and Leiden criteria for UA/IA, and the new ACR/EuLAR criteria for RA,
may be more useful than the current ACR RA criteria in identifying patients earlier in the
transition from preclinical to clinical RA, and it will be important for studies in preclinical RA going forward to have clearly defined criteria for transition from preclinical
disease in order to understand the evolution of RA, define when in the spectrum of
clinical presentations therapeutic intervention may be necessary, and define a clinical
outcome that is a suitable end point for prevention trials.
PART 4. WHAT CAN WE LEARN BY INVESTIGATING PRECLINICAL RHEUMATOID
ARTHRITIS?
Genetic and Environmental Interactions in Preclinical Rheumatoid Arthritis
As discussed earlier, the genetic and environmental influences that lead to the preclinical phase of disease development are likely key features in the development of RArelated autoimmunity. However, given the observed length of time between the
appearance of autoimmunity and diagnosis of RA, standard case-control study
approaches using subjects with established RA may not optimally allow for the identification of environmental risk factors for RA or at what point the risk factors influence
the development of autoimmunity and disease. For example, as mentioned earlier,
there is now considerable data that supports that gene-environmental interactions
between smoking and specific HLA alleles are associated with ACPA positivity in
established RA.36,56–59,103,104 However, investigating the relationship between these
factors in the preclinical period would help determine whether HLA alleles, smoking,
and ACPA positivity are associated in the absence of synovitis—a finding that would
support a role for smoking as an initial and perhaps causative step in RA-related
autoimmunity.
Inherent in the ‘Phases’ model of RA development discussed above is that there is
a physical location where genetic and environmental factors interact to lead to the
initial RA-related immune dysregulation and break in tolerance. For example, given
the strong association of smoking with RA, and in particular ACPA-positive RA, there
is a theory that initial RA-related immune dysregulation occurs at the mucosal surfaces
of the mouth (periodontal surfaces) or lung, with later spread of RA-related autoimmunity and inflammation to the joints resulting in clinically-apparent synovial
disease.37,39,57,58,105 Real-time prospective study of subjects with RA-related autoimmunity, evidenced by autoantibodies (either prevalent or incident) but no synovitis,
would allow for detailed exploration of mucosal surface immune function. Supporting
this approach, in preliminary work the authors have identified evidence of inflammatory lung injury, including airway and alveolar disease, using high-resolution computed
tomography of the lungs of individuals with RA-related autoantibody positivity but
without joint symptoms or findings, suggesting that RA-related inflammation may
indeed occur initially in the lung.106
Preclinical Rheumatoid Arthritis 225
Evaluation of Autoantibody and Inflammatory Marker Evolution in the Preclinical
Period of Rheumatoid Arthritis Development
There is a growing body of work suggesting that factors involving RA-related autoantibodies including isotype switching, antigen specificity, posttranslational modification and effector function (such as autoantibody glycosylation) may be related to
disease severity and perhaps evolution from UA to RA.107–112 For example, Verpoort
and colleagues111 reported that patients who evolved from UA to RA had a broader
spectrum of ACPA isotypes compared with those with persistent UA at follow-up. In
addition, a study by Ioan-Facsinay and colleagues113 comparing autoantibodies
in North American natives with RA and their healthy FDRs showed ACPA reactivity
in both groups (19% in healthy FDRs vs about 91% in RA cases); however, the fine
specificities of ACPAs differed between RA cases and healthy controls, with reactivity
to citrullinated fibrinogen and vimentin only seen in RA cases. Also in this study, ACPA
isotypes were more limited in healthy FDRs compared with RA cases. As a whole,
these findings suggest that ACPA isotype evolution and specificity may be important
for disease development. As such, investigating the evolution of the isotype changes
and specificity during the preclinical period when the possible transformations from
nonpathogenic to pathogenic states may occur, may lead to key insights into RA
development, and perhaps identification of the ‘‘original sin’’ in RA; that is, the initial
autoantigen that leads to RA-related autoimmunity. In regard to this latter point, based
on the work of several groups, ACPA reactivity to citrullinated a-enolase CEP-1
epitope may indicate that this citrullinated antigen most strongly links exposure to
cigarette smoke and HLA risk alleles in RA.59 If autoimmunity to this citrullinated
antigen could be demonstrated in the preclinical period prior to other autoimmune
responses to citrullinated antigens, the argument that this antigen is the initial one in
RA would be compelling. In addition, prospective evaluation of cytokine/chemokine
type (ie, IL-17, TNFa, or paradigm-specific small molecule profiles) may allow for identification of key inflammatory processes in RA development that may help to identify
the inciting factors for this disease as well as allow us to specifically target aspects of
the immune system for prevention.
Predictive Models for Future Rheumatoid Arthritis
Several prospective and retrospective studies suggest that autoantibodies or combinations of genetic factors and autoantibodies indicate high risk for future RA. In
prospective evaluation (including joint examinations and radiographs of hands and
feet approximately every 2 years) of approximately 2700 Pima and/or Papago Native
Americans in Arizona, USA (a population with an overall prevalence rate of RA of
approximately 5%), followed up to 19 years, del Puente and colleagues1 demonstrated increased risk for incident RA in RF-positive (by sheep cell agglutination) individuals, with the highest risk of 48.3 cases per 1000 person-years seen in those with
a titer of greater than 1:256 at baseline testing. Also in prospective evaluation of 370
FDRs from multicase RA families followed with periodic joint symptom assessment for
a mean of 5 years, Silman and colleagues11 found the overall rate for incident RA to be
approximately 8 per 1000 person-years, with a rate of incident RA in subjects that
were positive for RF at baseline of 34.8 cases per 1000 person-years (calculated
from 4 incident cases of RA out of 24 individuals).
Retrospective biobank studies have also estimated risk for future RA, although
these results are more limited due to lack of prospectively collected data. In their
Swedish biobank study, Rantapaa-Dahlqvist and colleagues7 estimated PPVs for
future RA of 4% if RF-IgM positive alone, 16% if anti-CCP positive alone, and 22%
226 Deane et al
if anti-CCP and RF-IgA positive (these PPV calculations assumed a prevalence of RA
in the general population of 1%). Nielen and colleagues8 estimated a PPV for onset of
classifiable RA within 5 years of 5.9% for subjects that were anti-CCP positive alone,
assuming an incidence rate of RA over 5 years in the general population of 0.001%.
However, this PPV increased to 69.4% if the incidence rate of RA in the population
over 5 years used for calculation was 3.9%. Of note, this higher rate of incident RA
(3.9% vs 0.001%) used by Nielen and colleagues in their PPV calculations was estimated from Silman and colleagues11 1992 study of incident RA in individuals from
multicase RA families, in which a strong family history of RA (R2 members with RA)
may serve as a possible surrogate for additional genetic and/or environmental risk
for RA. Also, in the Nielen study,8 if both RF and anti-CCP were positive, the PPV
for RA diagnosis within 5 years was 100%, regardless of which background incidence
of RA was used for comparison.
Studies that have evaluated risk for future RA have shown that certain genetic
factors, notably HLA alleles containing the ‘‘shared epitope,’’ may increase risk for
future RA.114 However, the ability of genetic factors alone to predict future RA may
be limited, in part due to the high prevalence of these factors in the population—in
North American controls, at least one HLA allele containing the ‘‘shared epitope’’ is
present in about 40% of the population, and PTPN22 risk alleles are present in about
16% of the population.57 As such, prediction of risk for future RA will likely best be
done using combinations of genetic, environmental, and autoantibody factors.
Several studies have already examined the relationship between genetic factors and
autoantibodies in prediction of future RA. Berglin and colleagues,115 using the
Northern Sweden Health and Disease biobank, performed a nested case-control
study to show a strong association between pre-RA diagnosis anti-CCP positivity
and HLA-DRB1*0404 or *0401 alleles and risk for future RA (odds ratio 66.8, 95%
confidence interval [CI] 8.3–539.4). In addition, using data from the same Swedish biobank and similar study design, Johansson and colleagues116 showed a strong association of anti-CCP positivity and 2 copies of the 1858T variant allele of PTPN22 with
future RA (odds ratio 132.0, 95% CI ~18–2721). Studies are also underway to use gene
and environment interactions for prediction of RA, as well as prediction of preclinical
RA-related autoantibody positivity as a surrogate outcome for evolving RA (Elizabeth
Karlson, personal communication, 2009).
There may be other factors that act separately or in concert with genetic and environmental factors to influence transition from asymptomatic to symptomatic RArelated autoimmunity. Majka and colleagues9 and Bos and colleagues17 reported in
their retrospective cohort analyses that subjects with older age at time of diagnosis
of RA had longer duration of preclinical RA-related autoantibody positivity. These findings may be explained by differences in genetic or environmental exposures that vary
by age, or by age-related immune senescence. In addition, these findings suggest that
age or other endogenous factors such as gender may have relevance in predicting
future onset of RA in at-risk populations.
Also, prospective studies of cohorts of patients initially with early symptomatic IA/
UA have allowed for investigation of the effects of genetic and other factors in the
persistence and evolution of symptomatic arthritis. Several Dutch studies have shown
that factors such as age, sex, number of swollen joints, and autoantibody and CRP
status may predict persistent IA at 1 year.98,100,117 In addition, in separate studies,
Feitsma and colleagues118 and van der Helm-van Mil and colleagues119 showed
that the presence of PTPN22 or HLA risk alleles may not significantly increase risk
of transition from UA to RA, but that these genetic risk factors do influence autoantibody presence (HLA) or level (PTPN22). Finally, several preclinical studies including
Preclinical Rheumatoid Arthritis 227
those using the Swedish, Dutch, and NHS biobanks noted an increase in autoantibody
titer and/or inflammatory marker levels in the time period immediately preceding diagnosis.7,8,18,19,21,22 Regarding this latter point, prospective study of biomarker changes
may also be of key interest in understanding the kinetics of preclinical RA and in
predictive models for symptomatic disease.
Although these studies are supportive of the ability of predictive models to determine
risk for future RA, it should be noted that estimates of risk for future RA using stored
samples from biobanks are limited because such studies were not truly prospective.
Of importance is that participants were not subjected to detailed baseline or followup joint examinations, and pre-RA diagnosis samples were assessed retrospectively
in comparison with selected controls after RA had been diagnosed. For these reasons,
the relationship between symptom onset and biomarker positivity is inexact.
Preclinical Prevention of Rheumatoid Arthritis
Several clinical trials have demonstrated that treatment with aggressive therapy in
early RA (that meets 1987 ACR RA criteria) leads to improved clinical outcomes,
and perhaps increased rates of drug-free remission.120,121 As such, currently a major
goal for RA treatment is early identification and treatment of disease.122 In addition,
several investigative groups have evaluated the efficacy of disease-modifying therapy
in individuals who have UA/IA not yet meeting established ACR criteria for RA. For
example, in the Probable Rheumatoid Arthritis Methotrexate versus Placebo Treatment (PROMPT) trial, in patients with UA of duration 2 years or less, use of methotrexate delayed progression to RA as defined by the 1987 ACR criteria, with the
most benefit seen in anti-CCP positive subjects.99 Another small study investigating
abatacept in UA showed a trend toward decreased progression to classifiable
RA.123 Also, several studies utilizing immunomodulation to prevent progression of
T1DM have been performed, and although T1DM has not yet been prevented, studies
have shown that intervention may lead to decreased insulin usage, and decreased risk
for death from ketoacidosis early in disease.124 Finally, in SLE, a retrospective study of
military patients with SLE by James and colleagues125 found that treatment with
hydroxychloroquine in early disease led to decreased progression of SLE-specific
autoantibodies as well as clinical findings. Overall, the results from these studies
suggest that intervention in an even earlier period of RA-related autoimmunity (ie, in
the preclinical period) may lead to prevention of disease.
Unfortunately, as of yet there are no published results of preclinical RA prevention
trials. However, interesting data obtained from database studies suggests that certain
agents such as HMG-CoA reductase inhibitors (statins) that may have some benefit in
treatment of active RA may modify future RA risk.126–128 Jick and colleagues129 have
shown, using a national database in the United Kingdom (N 5 313 RA cases and 1252
controls), a decreased risk for incident RA in patients using statins for hyperlipidemia
(odds ratio 0.59, 95% CI 037–0.96).
Ultimately, these findings and further research may lead to preclinical pharmacologic immunomodulation to prevent disease. Alternatively, perhaps nonpharmacologic tolerization (based on murine models of disease) or risk factor modification in
the preclinical period may be used to abrogate clinically apparent disease; for
example, if a person at high-risk for future RA stopped smoking, this may decrease
the risk for developing clinically apparent arthritis. However, while prevention of RA
is of key importance, implementation of prevention trials will only be possible after
researchers have first developed models in prospective studies to identify accurately
those at high risk for future RA in a definable and relatively short time period necessary
to conduct such studies.
228 Deane et al
PART 5. FUTURE RESEARCH IN PRECLINICAL RHEUMATOID ARTHRITIS
There are multiple approaches to investigate preclinical RA, and these are outlined in
Table 2. The investigation of preclinical RA would ideally take an approach similar to
the Framingham study, which followed a large population over time to investigate
causes of cardiovascular disease using serial measures of biomarkers and clinical
outcomes.130 However, due to the low prevalence of RA, large-scale population
studies at single centers may be impractical due to the cost of screening and relatively
few outcomes of disease. As such, preclinical RA may best be investigated by using
research models similar to those used in studies in T1DM, that is, using a multicenter
approach and prospectively following subjects who are likely to be at higher risk for
RA-related autoimmunity as well as development of clinically apparent disease. One
example of a population at higher risk for RA is FDRs of RA probands, as these
FDRs may have 5- to 7-fold increased risk for incident RA.11 In a multicenter study
in the United States, the authors are currently following prospectively a large cohort
of FDRs of probands with RA (goal N 5 2100).131 To date, a substantial proportion
(~14%, N~1200) of these FDRs exhibit RA-related autoantibody positivity.131 In addition, in Canada, El-Gabalawy and colleagues113,132,133 are currently prospectively
following a cohort of North American Natives who are FDRs of probands with RA,
and have demonstrated intriguing findings regarding differences in ACPA isotypes
and specificities between RA cases and asymptomatic FDRs. These cohorts will be
valuable in investigating the effects of genetic and environmental factors on RArelated autoimmunity, as well as in investigating the evolution of RA-related autoimmunity and inflammation over time. While identification of asymptomatic individuals with
RA-related autoantibody positivity is of interest, these prospective cohort analyses will
also allow for the identification of factors that impact the transition from Phase 1
(genetic risk) to Phase 2 (asymptomatic autoimmunity)—a transition during which
the key early interactions leading to RA-related autoimmunity occur. Examples of
RA-related measures that may be assessed prospectively in preclinical study are
provided in Fig. 1B.
Also, based on the prior success of biobank studies of preclinical RA, additional
samples from yet untapped existing biobanks may be evaluated for RA-related
biomarkers. However, as these retrospective cohorts may lack detailed joint evaluations, the ability to investigate the relationship between immune parameters and
symptomatic disease may be limited.
As an alternative to the prospective or biobank studies discussed earlier, a large
general population could be screened for RA-related genetic traits or RA-related autoantibody positivity to identify those at high risk for disease. From this initial screening,
a smaller group of individuals with high-risk genetic and/or autoantibody markers may
be followed over time to evaluate the evolution of RA-related autoimmunity. For
example, in a health-fair screen of approximately 5000 individuals, the authors have
identified approximately 40 anti-CCP positive subjects without synovitis on examination, and these subjects are now being evaluated prospectively for evolution of
RA-related autoimmunity.134 Such population screening is complicated by cost issues
and the potential for subjects participating in such screening to have existing symptomatic joint disease, thereby missing the ‘‘preclinical’’ window of disease. However,
with appropriate methodologies, population screening may be a reasonable approach
to identify individuals with asymptomatic RA-related autoimmunity.
Of note, given the growing availability of array technologies, it may be possible to
screen large populations at risk of developing a wide variety of autoimmune diseases.
By testing for multiple autoimmune diseases, this approach might also allow for
Preclinical Rheumatoid Arthritis 229
Table 2
Potential methodologies for investigating preclinical rheumatoid arthritis
Study Design Examples Strengths Weaknesses
Retrospective case-control studies
using stored prediagnosis samples
(biobanks):
Finnish, Swedish, and Dutch biobank
studies6–8
United States Military/Department
of Defense Serum Repository9
Nurses’ Health Study18,22
Women’s Health Study22
Biobanks already established and
preclinical samples may be obtained
from cases with known RA
Decreased ability to determine future
risk for disease; unable to ascertain
exact relationship between timing
of appearance of RA-related
biomarkers and symptoms; limited
samples available
Prospective studies:
Populations at high risk for RA
(such as first-degree relatives
of probands with RA, twin studies,
or populations such as Native
Americans with high prevalence
of disease)
Ongoing:
Holers et al131: Studies of the
Etiologies of Rheumatoid Arthritis
(SERA): prospective evaluation of
first-degree relatives of probands
with RA
El-Gabalawy et al113,132,133:
North American Native people in
Central Canada (Cree and Ojibway)
prospectively followed for
evolution of RA-related
autoimmunity. Background
prevalence of RA in population
~2%
Prior:
Native American (Pima and Papago)
studies1
Real-time evaluation of evolution of
RA-related autoimmunity to
determine timing of appearance of
biomarkers in relationship to
environmental exposures and
development of symptomatic
disease; possibility for detailed
assessment of mucosal biology in
preclinical RA
Costly in terms of money and time;
relatively low prevalence of
incident RA and prevalence of
RA-related autoantibodies limits
statistical power
230 Deane et al
Other prospective studies:
Large-scale population screening to
identify individuals with high-risk
features for RA including genetic
factors and RA-related
autoantibody positivity
Health-fair screening in Colorado
identifying anti-CCP positive
asymptomatic individuals who are
recruited for follow-up134
Potentially identifies subjects at risk
for ‘‘sporadic RA’’(as are most cases
in population); may help to
establish prevalence of RA-related
autoantibodies in general
populations
Expensive, and relatively low
prevalence of RA-related
autoantibodies requires large-scale
screening to identify at-risk
individuals
Coupling of prospective RA studies
with other research projects (such
as cardiovascular disease studies)
Multiple studies are likely available Allows for coupling of research
projects perhaps resulting in
decreased costs
Costly in terms of money and effort to
establish screening
Other approaches:
Animal models of disease
Multiple Allows for detailed evaluation of
antigen interactions; may provide
for testing of methodologies for
tolerization
Animal models of arthritis have
substantial differences from human
disease
Population screening for multiple
autoimmune diseases
Multiple Multiple autoimmune diseases may be
studied; array technology for
multiple autoantibody testing
Expensive; optimal diagnostic
accuracy and reproducibility
of testing would need to be
established
Preclinical Rheumatoid Arthritis 231
a more cost-effective way to identify individuals at risk for RA-related autoimmunity as
well as those at risk for other autoimmune diseases that also exhibit preclinical
disease-related autoantibodies, including T1DM and SLE already mentioned, as
well as autoimmune thyroid disease (Graves disease and Hashimoto thyroiditis),
primary biliary cirrhosis, and celiac disease.135,136 With regard to such a strategy, it
is important to understand that in aggregate these diseases affect up to 7% of the
population.135,136 Also, there are data to support the possibility that risk for cardiovascular disease (CVD), the leading cause of death in RA patients, may be increased even
in the preclinical period of RA.137 As such, preclinical evaluation of RA may not only
identify RA-specific factors but also lead to a better understanding of CVD risk in
RA, and perhaps CVD prevention.
In addition to the design and implementation of prospective studies to investigate
the preclinical period of RA development, several other issues regarding research
methodologies in preclinical RA need to be addressed. First, what constitutes positivity for RA-related autoantibodies? At present there are multiple methodologies for
testing RFs and ACPAs, with each kit having different cut-offs for positivity resulting
in a variety of sensitivities and specificities. As such, standardized methodologies
for testing RFs and ACPAs as well as for determining what result is considered positive
will need to be established in order to homogenize testing results and development of
predictive models for future RA. International studies in T1DM have already adopted
standardized autoantibody testing, and these tests are performed in a limited number
of research laboratories to ensure accuracy and reproducibility of testing. Second, as
discussed earlier, we will need to define what constitutes ‘‘preclinical’’ RA as well as
what constitutes transition from asymptomatic RA-related autoimmunity to clinically
apparent disease. Likely this will need standardized symptom and joint count assessments, as well as potentially standardized techniques for imaging of the synovium
using magnetic resonance imaging and/or ultrasound, and standardized synovial
biopsy techniques and tissue processing. Also, it may be useful in preclinical studies
to establish biomarker end points that could be employed in prevention trials. Such
profiles, which may include autoantibodies and inflammatory markers, may be used
as surrogate outcomes for increased risk for future RA. For example, if an asymptomatic individual in a prevention trial has RA-related autoantibodies suggesting high risk
for future RA as well as an elevated CRP suggesting active inflammation, a decrease of
CRP in this individual due to a pharmacologic intervention (in absence of joint symptoms) may be considered a clinically appropriate success in prevention of disease.
This approach is aimed at allowing for a more timely assessment of intervention
success, as joint symptoms may take years to develop. Of note, in terms of symptoms
or examination findings, swollen joints may not be the first indication of transition from
preclinical RA to clinical RA, and perhaps patient-reported measures such as stiffness
or pain in absence of detectable swelling are the first indications of symptomatic RA.
Detailed studies of the preclinical period, closely examining joint symptoms, and
perhaps investigating asymptomatic organ injury using synovial biopsy to discover
asymptomatic synovitis, or joint or lung imaging to detect asymptomatic RA-related
injury, may be necessary to understand truly the evolution from preclinical to clinical
RA. Third, what phenotype of RA will be best to investigate? Will it be best to focus
on seropositive disease, as it will be easier to identify preclinical seropositive RA
due to the presence of specific autoantibodies? Given that seronegative RA may be
distinct from seropositive RA in terms of genetic and environmental risk factors, it
may be worthwhile to focus efforts going forward on seropositive disease.138 Fourth,
to understand the predictive power of RA-related biomarkers for future disease,
relevant control populations for prospective studies of preclinical RA need to be
232 Deane et al
established. This factor is of particular importance when considering that several
preclinical studies in RA have already shown that not all subjects with RA-related autoantibody positivity, including ACPAs, may develop symptomatic RA.4,15 Finally, standardized approaches for assessing epidemiologic exposures that might impact RA
development will need to be established.
In addition to studies in humans, animal models of autoimmune disease may yield
important insights into preclinical autoimmunity. In particular, animal models may be
utilized to understand initial citrullinated protein-specific autoimmunity, or the role of
mucosal surfaces in the generation of autoimmunity. Also, animal models may help
to identify methods to tolerize individuals to citrullinated or other antigens important
to RA in the preclinical period of RA development, a time period in which nonpharmaceutical, immunomodulatory strategies may be more effective prior to the extensive
epitope spreading that has occurred in RA by the time clinical symptoms and signs
of disease appear. Animal studies may also allow the determination of whether novel
treatments focused on the citrullination process itself may be beneficial.
SUMMARY
Based on multiple studies in RA as well as other autoimmune diseases, RA likely
develops in phases exhibited by genetic risk, asymptomatic autoimmunity and, finally,
clinically apparent disease, with transformations between these phases occurring due
to combinations of genetic, environmental, and immunologic factors. Investigating the
preclinical phases of RA development will provide key insights into the factors that
lead to disease, as well as allow for development of predictive models for disease,
and ultimately prevention strategies for RA. Going forward, studies of preclinical RA
may use bio-repositories, but the most effective approach will likely be the use of
prospective cohort studies, with subjects chosen from populations at greater risk
for RA such as FDRs or twin studies, or those identified with high-risk markers for
RA, either genetic or autoantibody, through community screening. In addition to
design and implementation of such prospective studies, the rheumatology community
will need to define with greater clarity what constitutes the transition from preclinical to
clinical RA, as well as standardize autoantibody and inflammatory marker testing.
Although these latter tasks are daunting, investigation of preclinical RA likely gives
us the greatest hope for curing or preventing this disease.
REFERENCES
1. del Puente A, Knowler WC, Pettitt DJ, et al. The incidence of rheumatoid arthritis
is predicted by rheumatoid factor titer in a longitudinal population study. Arthritis
Rheum 1988;31(10):1239–44.
2. Aho K, Heliovaara M, Maatela J, et al. Rheumatoid factors antedating clinical
rheumatoid arthritis. J Rheumatol 1991;18(9):1282–4.
3. Kurki P, Aho K, Palosuo T, et al. Immunopathology of rheumatoid arthritis. Antikeratin antibodies precede the clinical disease. Arthritis Rheum 1992;35(8):
914–7.
4. Aho K, von Essen R, Kurki P, et al. Antikeratin antibody and antiperinuclear
factor as markers for subclinical rheumatoid disease process. J Rheumatol
1993;20(8):1278–81.
5. Aho K, Heliovaara M, Knekt P, et al. Serum immunoglobulins and the risk of rheumatoid arthritis. Ann Rheum Dis 1997;56(6):351–6.
Preclinical Rheumatoid Arthritis 233
6. Aho K, Palosuo T, Heliovaara M, et al. Antifilaggrin antibodies within ‘‘normal’’
range predict rheumatoid arthritis in a linear fashion. J Rheumatol 2000;
27(12):2743–6.
7. Rantapaa-Dahlqvist S, de Jong BA, Berglin E, et al. Antibodies against cyclic
citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritis. Arthritis Rheum 2003;48(10):2741–9.
8. Nielen MM, van Schaardenburg D, Reesink HW, et al. Specific autoantibodies
precede the symptoms of rheumatoid arthritis: a study of serial measurements
in blood donors. Arthritis Rheum 2004;50(2):380–6.
9. Majka DS, Deane KD, Parrish LA, et al. Duration of preclinical rheumatoid
arthritis-related autoantibody positivity increases in subjects with older age at
time of disease diagnosis. Ann Rheum Dis 2008;67(6):801–7.
10. MacGregor AJ, Silman AJ. Rheumatoid factors as predictors of rheumatoid
arthritis. J Rheumatol 1991;18(9):1280–1.
11. Silman AJ, Hennessy E, Ollier B. Incidence of rheumatoid arthritis in a genetically
predisposed population. Br J Rheumatol 1992;31(6):365–8.
12. Aho K, Palosuo T, Heliovaara M. Predictive significance of rheumatoid factor.
J Rheumatol 1995;22(11):2186–7.
13. Aho K, Palosuo T, Raunio V, et al. The timing of rheumatoid factor seroconversions. Arthritis Rheum 1987;30(6):719–20.
14. Aho K, Palosuo T, Raunio V, et al. When does rheumatoid disease start? Arthritis
Rheum 1985;28(5):485–9.
15. Jonsson T, Thorsteinsson J, Kolbeinsson A, et al. Population study of the importance of rheumatoid factor isotypes in adults. Ann Rheum Dis 1992;51(7):
863–8.
16. van Venrooij WJ, Pruijn GJ. Citrullination: a small change for a protein with great
consequences for rheumatoid arthritis. Arthritis Res 2000;2(4):249–51.
17. Bos WH, Nielen MM, Dijkmans BA, et al. Duration of pre-rheumatoid arthritis
anti-cyclic citrullinated peptide positivity is positively associated with age at
seroconversion. Ann Rheum Dis 2008;67(11):1642.
18. Chibnik LB, Mandl LA, Costenbader KH, et al. Comparison of threshold cutpoints and continuous measures of anti-cyclic citrullinated peptide antibodies
in predicting future rheumatoid arthritis. J Rheumatol 2009;36(4):706–11.
19. Nielen MM, van Schaardenburg D, Reesink HW, et al. Increased levels of
C-reactive protein in serum from blood donors before the onset of rheumatoid
arthritis. Arthritis Rheum 2004;50(8):2423–7.
20. Nielen MM, van Schaardenburg D, Reesink HW, et al. Simultaneous development of acute phase response and autoantibodies in preclinical rheumatoid
arthritis. Ann Rheum Dis 2006;65(4):535–7.
21. Rantapaa-Dahlqvist S, Boman K, Tarkowski A, et al. Up regulation of monocyte
chemoattractant protein-1 expression in anti-citrulline antibody and immunoglobulin M rheumatoid factor positive subjects precedes onset of inflammatory
response and development of overt rheumatoid arthritis. Ann Rheum Dis 2007;
66(1):121–3.
22. Karlson EW, Chibnik LB, Tworoger SS, et al. Biomarkers of inflammation and
development of rheumatoid arthritis in women from two prospective cohort
studies. Arthritis Rheum 2009;60(3):641–52.
23. Shadick NA, Cook NR, Karlson EW, et al. C-reactive protein in the prediction of
rheumatoid arthritis in women. Arch Intern Med 2006;166(22):2490–4.
24. Jorgensen KT, Wiik A, Pedersen M, et al. Cytokines, autoantibodies and viral
antibodies in premorbid and postdiagnostic sera from patients with rheumatoid
234 Deane et al
arthritis: case-control study nested in a cohort of Norwegian blood donors. Ann
Rheum Dis 2008;67(6):860–6.
25. Aho K, Palosuo T, Knekt P, et al. Serum C-reactive protein does not predict rheumatoid arthritis. J Rheumatol 2000;27(5):1136–8.
26. Kokkonen H, Soderstrom I, Rocklow J, et al. Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis. Arthritis Rheum 2010;62(2):
383–91.
27. Silman AJ. Epidemiology of rheumatoid arthritis. APMIS 1994;102(10):721–8.
28. Alarcon GS. Epidemiology of rheumatoid arthritis. Rheum Dis Clin North Am
1995;21(3):589–604.
29. Reveille JD. The genetic contribution to the pathogenesis of rheumatoid arthritis.
Curr Opin Rheumatol 1998;10(3):187–200.
30. Nepom GT, Nepom BS. Prediction of susceptibility to rheumatoid arthritis by
human leukocyte antigen genotyping. Rheum Dis Clin North Am 1992;18(4):
785–92.
31. Karlson EW, Chang SC, Cui J, et al. Gene-environment interaction between
HLA-DRB1 shared epitope and heavy cigarette smoking in predicting incident
rheumatoid arthritis. Ann Rheum Dis 2010;69(1):54–60.
32. Plenge RM. Rheumatoid arthritis genetics: 2009 update. Curr Rheumatol Rep
2009;11(5):351–6.
33. Barton A, Worthington J. Genetic susceptibility to rheumatoid arthritis: an
emerging picture. Arthritis Rheum 2009;61(10):1441–6.
34. Heliovaara M, Aho K, Knekt P, et al. Coffee consumption, rheumatoid factor, and
the risk of rheumatoid arthritis. Ann Rheum Dis 2000;59(8):631–5.
35. Mikuls TR, Cerhan JR, Criswell LA, et al. Coffee, tea, and caffeine consumption
and risk of rheumatoid arthritis: results from the Iowa Women’s Health Study.
Arthritis Rheum 2002;46(1):83–91.
36. Padyukov L, Silva C, Stolt P, et al. A gene-environment interaction between
smoking and shared epitope genes in HLA-DR provides a high risk of seropositive rheumatoid arthritis. Arthritis Rheum 2004;50(10):3085–92.
37. Hart JE, Laden F, Puett RC, et al. Exposure to traffic pollution and increased risk
of rheumatoid arthritis. Environ Health Perspect 2009;117(7):1065–9.
38. Parks CG, Conrad K, Cooper GS. Occupational exposure to crystalline
silica and autoimmune disease. Environ Health Perspect 1999;107(Suppl 5):
793–802.
39. Koeger AC, Lang T, Alcaix D, et al. Silica-associated connective tissue disease.
A study of 24 cases. Medicine (Baltimore) 1995;74(5):221–37.
40. Blaschke S, Schwarz G, Moneke D, et al. Epstein-Barr virus infection in peripheral blood mononuclear cells, synovial fluid cells, and synovial membranes of
patients with rheumatoid arthritis. J Rheumatol 2000;27(4):866–73.
41. Oliver JE, Silman AJ. Risk factors for the development of rheumatoid arthritis.
Scand J Rheumatol 2006;35(3):169–74.
42. Dixon B. Bacteria and arthritis. BMJ 1990;301(6759):1043.
43. Ebringer A. Rheumatoid arthritis and proteus. Clin Med 2005;5(4):420–1.
44. Silman AJ, Hochberg MC. Rheumatoid arthritis. In: Silman AJ, Hochberg MC,
editors. Epidemiology of the rheumatic diseases. New York: Oxford University
Press; 1993. p. 7–68.
45. Spector TD, Silman AJ. Is poor pregnancy outcome a risk factor in rheumatoid
arthritis? Ann Rheum Dis 1990;49(1):12–4.
46. Pladevall-Vila M, Delclos GL, Varas C, et al. Controversy of oral contraceptives
and risk of rheumatoid arthritis: meta-analysis of conflicting studies and review
Preclinical Rheumatoid Arthritis 235
of conflicting meta-analyses with special emphasis on analysis of heterogeneity.
Am J Epidemiol 1996;144(1):1–14.
47. Spector TD, Hochberg MC. The protective effect of the oral contraceptive pill on
rheumatoid arthritis: an overview of the analytic epidemiological studies using
meta-analysis. J Clin Epidemiol 1990;43(11):1221–30.
48. Doran MF, Crowson CS, O’Fallon WM, et al. The effect of oral contraceptives and
estrogen replacement therapy on the risk of rheumatoid arthritis: a population
based study. J Rheumatol 2004;31(2):207–13.
49. Merlino LA, Curtis J, Mikuls TR, et al. Vitamin D intake is inversely associated
with rheumatoid arthritis: results from the Iowa Women’s Health Study. Arthritis
Rheum 2004;50(1):72–7.
50. Bhatia SS, Majka DS, Kittelson JM, et al. Rheumatoid factor seropositivity is
inversely associated with oral contraceptive use in women without rheumatoid
arthritis. Ann Rheum Dis 2007;66(2):267–9.
51. Korpilahde T, Heliovaara M, Knekt P, et al. Smoking history and serum cotinine
and thiocyanate concentrations as determinants of rheumatoid factor in nonrheumatoid subjects. Rheumatology (Oxford) 2004;43(11):1424–8.
52. Tuomi T, Heliovaara M, Palosuo T, et al. Smoking, lung function, and rheumatoid
factors. Ann Rheum Dis 1990;49(10):753–6.
53. Feser M, Derber LA, Deane KD, et al. Plasma 25, OH vitamin D concentrations
are not associated with rheumatoid arthritis (RA)-related autoantibodies in individuals at elevated risk for RA. J Rheumatol 2009;36(5):943–6.
54. Elkayam O, Caspi D, Lidgi M, et al. Auto-antibody profiles in patients with active
pulmonary tuberculosis. Int J Tuberc Lung Dis 2007;11(3):306–10.
55. Elkayam O, Segal R, Lidgi M, et al. Positive anti-cyclic citrullinated proteins and
rheumatoid factor during active lung tuberculosis. Ann Rheum Dis 2006;65(8):
1110–2.
56. Criswell LA, Saag KG, Mikuls TR, et al. Smoking interacts with genetic risk
factors in the development of rheumatoid arthritis among older Caucasian
women. Ann Rheum Dis 2006;65(9):1163–7.
57. 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(5):867–75.
58. Klareskog L, Stolt P, Lundberg K, et al. A new model for an etiology of rheumatoid
arthritis: smoking may trigger HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by citrullination. Arthritis Rheum 2006;54(1):38–46.
59. Mahdi H, Fisher BA, Kallberg H, et al. Specific interaction between genotype,
smoking and autoimmunity to citrullinated alpha-enolase in the etiology of rheumatoid arthritis. Nat Genet 2009;41(12):1319–24.
60. Steck AK, Zhang W, Bugawan TL, et al. Do non-HLA genes influence development of persistent islet autoimmunity and type 1 diabetes in children with highrisk HLA-DR, DQ genotypes? Diabetes 2009;58(4):1028–33.
61. Sheehy MJ. HLA and insulin-dependent diabetes. A protective perspective.
Diabetes 1992;41(2):123–9.
62. Taplin CE, Barker JM. Autoantibodies in type 1 diabetes. Autoimmunity 2008;
41(1):11–8.
63. Achenbach P, Bonifacio E, Koczwara K, et al. Natural history of type 1 diabetes.
Diabetes 2005;54(Suppl 2):S25–31.
64. Norris JM, Beaty B, Klingensmith G, et al. Lack of association between early
exposure to cow’s milk protein and beta-cell autoimmunity. Diabetes Autoimmunity Study in the Young (DAISY). JAMA 1996;276(8):609–14.
236 Deane et al
65. Ziegler AG, Schmid S, Huber D, et al. Early infant feeding and risk of developing
type 1 diabetes-associated autoantibodies. JAMA 2003;290(13):1721–8.
66. Norris JM, Barriga K, Klingensmith G, et al. Timing of initial cereal exposure in
infancy and risk of islet autoimmunity. JAMA 2003;290(13):1713–20.
67. Graves PM, Rotbart HA, Nix WA, et al. Prospective study of enteroviral infections
and development of beta-cell autoimmunity. Diabetes autoimmunity study in the
young (DAISY). Diabetes Res Clin Pract 2003;59(1):51–61.
68. GravesPM,BarrigaKJ,NorrisJM, etal. Lackofassociationbetweenearly childhood
immunizations and beta-cell autoimmunity. Diabetes Care 1999;22(10):1694–7.
69. Norris JM, Yin X, Lamb MM, et al. Omega-3 polyunsaturated fatty acid intake
and islet autoimmunity in children at increased risk for type 1 diabetes. JAMA
2007;298(12):1420–8.
70. Lamb MM, Yin X, Barriga K, et al. Dietary glycemic index, development of islet
autoimmunity, and subsequent progression to type 1 diabetes in young children. J Clin Endocrinol Metab 2008;93(10):3936–42.
71. Verge CF, Gianani R, Kawasaki E, et al. Number of autoantibodies (against
insulin, GAD or ICA512/IA2) rather than particular autoantibody specificities
determines risk of type I diabetes. J Autoimmun 1996;9(3):379–83.
72. Taplin CE, Barker JM. Natural evolution, prediction, and prevention of type 1 diabetes in youth. Endocr Res 2008;33(1–2):17–33.
73. Kishiyama CM, Chase HP, Barker JM. Prevention strategies for type 1 diabetes.
Rev Endocr Metab Disord 2006;7(3):215–24.
74. Maddison PJ, Reichlin M. Quantitation of precipitating antibodies to certain
soluble nuclear antigens in SLE. Arthritis Rheum 1977;20(3):819–24.
75. Lee LA, Gaither KK, Coulter SN, et al. Pattern of cutaneous immunoglobulin G
deposition in subacute cutaneous lupus erythematosus is reproduced by
infusing purified anti-Ro (SSA) autoantibodies into human skin-grafted mice.
J Clin Invest 1989;83(5):1556–62.
76. Miranda-Carus ME, Askanase AD, Clancy RM, et al. Anti-SSA/Ro and anti-SSB/
La autoantibodies bind the surface of apoptotic fetal cardiocytes and promote
secretion of TNF-alpha by macrophages. J Immunol 2000;165(9):5345–51.
77. Kurien BT, Newland J, Paczkowski C, et al. Association of neutropenia in systemic
lupus erythematosus (SLE) with anti-Ro and binding of an immunologically crossreactive neutrophil membrane antigen. Clin Exp Immunol 2000;120(1):209–17.
78. Hahn BH. Antibodies to DNA. N Engl J Med 1998;338(19):1359–68.
79. Davis P, Percy JS, Russell AS. Correlation between levels of DNA antibodies and
clinical disease activity in SLE. Ann Rheum Dis 1977;36(2):157–9.
80. 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(16):1526–33.
81. McClain MT, Arbuckle MR, Heinlen LD, et al. The prevalence, onset, and clinical
significance of antiphospholipid antibodies prior to diagnosis of systemic lupus
erythematosus. Arthritis Rheum 2004;50(4):1226–32.
82. Arbuckle MR, James JA, Dennis GJ, et al. Rapid clinical progression to diagnosis among African-American men with systemic lupus erythematosus. Lupus
2003;12(2):99–106.
83. Gross AJ, Hochberg D, Rand WM, et al. EBV and systemic lupus erythematosus: a new perspective. J Immunol 2005;174(11):6599–607.
84. Vossenaar ER, Nijenhuis S, Helsen MM, et al. Citrullination of synovial
proteins in murine models of rheumatoid arthritis. Arthritis Rheum 2003;
48(9):2489–500.
Preclinical Rheumatoid Arthritis 237
85. Kuhn KA, Kulik L, Tomooka B, et al. Antibodies against citrullinated proteins
enhance tissue injury in experimental autoimmune arthritis. J Clin Invest 2006;
116(4):961–73.
86. Uysal H, Bockermann R, Nandakumar KS, et al. Structure and pathogenicity of
antibodies specific for citrullinated collagen type II in experimental arthritis.
J Exp Med 2009;206(2):449–62.
87. Lundberg K, Nijenhuis S, Vossenaar ER, et al. Citrullinated proteins have
increased immunogenicity and arthritogenicity and their presence in arthritic
joints correlates with disease severity. Arthritis Res Ther 2005;7(3):R458–67.
88. 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(2):538–41.
89. 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(4):
967–79.
90. Kanazawa S, Ota S, Sekine C, et al. Aberrant MHC class II expression in mouse
joints leads to arthritis with extraarticular manifestations similar to rheumatoid
arthritis. Proc Natl Acad Sci U S A 2006;103(39):14465–70.
91. Ho PP, Lee LY, Zhao X, et al. Autoimmunity against fibrinogen mediates inflammatory arthritis in mice. J Immunol 2010;184(1):379–90.
92. Kraan MC, Versendaal H, Jonker M, et al. Asymptomatic synovitis precedes
clinically manifest arthritis. Arthritis Rheum 1998;41(8):1481–8.
93. 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(3):315–24.
94. Aletaha D, Breedveld FC, Smolen JS. The need for new classification criteria for
rheumatoid arthritis. Arthritis Rheum 2005;52(11):3333–6.
95. Silman AJ, Symmons DP. Selection of study population in the development of
rheumatic disease criteria: comment on the article by the American College of
Rheumatology Diagnostic and Therapeutic Criteria Committee. Arthritis Rheum
1995;38(5):722–3.
96. van Baarsen LG, Bos WH, Rustenburg F, et al. Gene expression profiling in autoantibody-positive patients with arthralgia predicts development of arthritis.
Arthritis Rheum 2010;62(3):694–704.
97. Symmons DP, Silman AJ. The Norfolk Arthritis Register (NOAR). Clin Exp Rheumatol 2003;21(5 Suppl 31):S94–9.
98. van der Helm-van Mil AH, le Cessie S, van Dongen H, et al. A prediction rule
for disease outcome in patients with recent-onset undifferentiated arthritis:
how to guide individual treatment decisions. Arthritis Rheum 2007;56(2):
433–40.
99. 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;56(5):1424–32.
100. Verpoort KN, van Dongen H, Allaart CF, et al. Undifferentiated arthritis–disease
course assessed in several inception cohorts. Clin Exp Rheumatol 2004;22(5
Suppl 35):S12–7.
101. van Aken J, van Bilsen JH, Allaart CF, et al. The Leiden early arthritis clinic. Clin
Exp Rheumatol 2003;21(5 Suppl 31):S100–5.
102. Panel ACoRAaELARE, editor. New rheumatoid arthritis criteria. American
College of Rheumatology Annual Meeting. Philadelphia, PA, 2009.
238 Deane et al
103. Pedersen M, Jacobsen S, Garred P, et al. Strong combined gene-environment
effects in anti-cyclic citrullinated peptide-positive rheumatoid arthritis: a nationwide case-control study in Denmark. Arthritis Rheum 2007;56(5):1446–53.
104. Verpoort KN, Papendrecht-van der Voort EA, van der Helm-van Mil AH, et al.
Association of smoking with the constitution of the anti-cyclic citrullinated
peptide response in the absence of HLA-DRB1 shared epitope alleles. Arthritis
Rheum 2007;56(9):2913–8.
105. Smolik I, Robinson D, El-Gabalawy HS, et al. Periodontitis and rheumatoid
arthritis: epidemiologic, clinical, and immunologic associations. Compend Contin Educ Dent 2009;30(4):188–90 [92, 94 passim; quiz: 98, 210].
106. Deane KD, Simonian PL, Lynch DA, et al. Rheumatoid arthritis (RA)-related autoantibodies are associated with pulmonary airway abnormalities in individuals
without RA [abstract 340]. Arthritis Rheum 2008;58(9):S292.
107. Aho K, Tuomi T, Heliovaara M, et al. Rheumatoid factors and rheumatoid
arthritis. Scand J Rheumatol Suppl 1988;74:41–4.
108. Dorner T, Egerer K, Feist E, et al. Rheumatoid factor revisited. Curr Opin Rheumatol 2004;16(3):246–53.
109. van der Helm-van Mil AH, Verpoort KN, Breedveld FC, et al. Antibodies to citrullinated proteins and differences in clinical progression of rheumatoid arthritis.
Arthritis Res Ther 2005;7(5):R949–58.
110. Verpoort KN, Cheung K, Ioan-Facsinay A, et al. Fine specificity of the anti-citrullinated protein antibody response is influenced by the shared epitope alleles.
Arthritis Rheum 2007;56(12):3949–52.
111. Verpoort KN, Jol-van der Zijde CM, Papendrecht-van der Voort EA, et al. Isotype
distribution of anti-cyclic citrullinated peptide antibodies in undifferentiated
arthritis and rheumatoid arthritis reflects an ongoing immune response. Arthritis
Rheum 2006;54(12):3799–808.
112. Malhotra R, Wormald MR, Rudd PM, et al. Glycosylation changes of IgG associated with rheumatoid arthritis can activate complement via the mannosebinding protein. Nat Med 1995;1(3):237–43.
113. Ioan-Facsinay A, Willemze A, Robinson DB, et al. Marked differences in fine
specificity and isotype usage of the anti-citrullinated protein antibody in health
and disease. Arthritis Rheum 2008;58(10):3000–8.
114. 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(11):1205–13.
115. Berglin E, Padyukov L, Sundin U, et al. A combination of autoantibodies to cyclic
citrullinated peptide (CCP) and HLA-DRB1 locus antigens is strongly associated
with future onset of rheumatoid arthritis. Arthritis Res Ther 2004;6(4):R303–8.
116. Johansson M, Arlestig L, Hallmans G, et al. PTPN22 polymorphism and anticyclic citrullinated peptide antibodies in combination strongly predicts future
onset of rheumatoid arthritis and has a specificity of 100% for the disease.
Arthritis Res Ther 2006;8(1):R19.
117. van der Helm-van Mil AH, Detert J, le Cessie S, et al. Validation of a prediction
rule for disease outcome in patients with recent-onset undifferentiated arthritis:
moving toward individualized treatment decision-making. Arthritis Rheum 2008;
58(8):2241–7.
118. Feitsma AL, Toes RE, Begovich AB, et al. Risk of progression from undifferentiated arthritis to rheumatoid arthritis: the effect of the PTPN22 1858T-allele in
anti-citrullinated peptide antibody positive patients. Rheumatology (Oxford)
2007;46(7):1092–5.
Preclinical Rheumatoid Arthritis 239
119. van der Helm-van Mil AH, 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(4):1117–21.
120. Allaart CF, Breedveld FC, Dijkmans BA. Treatment of recent-onset rheumatoid arthritis: lessons from the BeSt study. J Rheumatol Suppl 2007;80:
25–33.
121. Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, et al. Clinical and
radiographic outcomes of four different treatment strategies in patients with
early rheumatoid arthritis (the BeSt study): a randomized, controlled trial.
Arthritis Rheum 2008;58(Suppl 2):S126–35.
122. Cush JJ. Early rheumatoid arthritis—is there a window of opportunity? J Rheumatol Suppl 2007;80:1–7.
123. Emery P, Durez P, Dougados M, et al. Impact of T-cell costimulation modulation
in patients with undifferentiated inflammatory arthritis or very early rheumatoid
arthritis: a clinical and imaging study of abatacept (the ADJUST trial). Ann
Rheum Dis 2010;69(3):510–6.
124. Rewers M, Gottlieb P. Immunotherapy for the prevention and treatment of type 1
diabetes: human trials and a look into the future. Diabetes Care 2009;32(10):
1769–82.
125. James JA, Kim-Howard XR, Bruner BF, et al. Hydroxychloroquine sulfate treatment is associated with later onset of systemic lupus erythematosus. Lupus
2007;16(6):401–9.
126. Endres M. Statins: potential new indications in inflammatory conditions. Atheroscler Suppl 2006;7(1):31–5.
127. McInnes IB, McCarey DW, Sattar N. Do statins offer therapeutic potential in
inflammatory arthritis? Ann Rheum Dis 2004;63(12):1535–7.
128. Klareskog L, Hamsten A. Statins in rheumatoid arthritis—two birds with one
stone? Lancet 2004;363(9426):2011–2.
129. Jick SS, Choi H, Li L, et al. Hyperlipidaemia, statin use and the risk of developing rheumatoid arthritis. Ann Rheum Dis 2009;68(4):546–51.
130. Dawber TR, Meadors GF, Moore FE Jr. Epidemiological approaches to heart
disease: the Framingham Study. Am J Public Health Nations Health 1951;
41(3):279–81.
131. Kolfenbach JR, Deane KD, Derber LA, et al. A prospective approach to investigating the natural history of preclinical rheumatoid arthritis (RA) using firstdegree relatives of probands with RA. Arthritis Rheum 2009;61(12):1735–42.
132. Oen K, Robinson DB, Nickerson P, et al. Familial seropositive rheumatoid
arthritis in North American Native families: effects of shared epitope and cytokine genotypes. J Rheumatol 2005;32(6):983–91.
133. El-Gabalawy HS, Robinson DB, Hart D, et al. Immunogenetic risks of anticyclical citrullinated peptide antibodies in a North American Native population
with rheumatoid arthritis and their first-degree relatives. J Rheumatol 2009;
36(6):1130–5.
134. Deane KD, Striebich CC, Goldstein BL, et al. Identification of undiagnosed
inflammatory arthritis in a community health fair screen. Arthritis Rheum 2009;
61(12):1642–9.
135. Bizzaro N. The predictive significance of autoantibodies in organ-specific autoimmune diseases. Clin Rev Allergy Immunol 2008;34(3):326–31.
136. Rose NR. Predictors of autoimmune disease: autoantibodies and beyond. Autoimmunity 2008;41(6):419–28.
240 Deane et al
137. Maradit-Kremers H, Crowson CS, Nicola PJ, et al. Increased unrecognized
coronary heart disease and sudden deaths in rheumatoid arthritis: a population-based cohort study. Arthritis Rheum 2005;52(2):402–11.
138. Alarcon GS, Koopman WJ, Acton RT, et al. Seronegative rheumatoid arthritis.
A distinct immunogenetic disease? Arthritis Rheum 1982;25(5):502–7.
Preclinical Rheumatoid Arthritis 241

Nenhum comentário:

Postar um comentário