You are on page 1of 53

ASSESSMENT OF RHEUMATOID ARTHRITIS ACTIVITY IN CLINICAL TRIALS

AND CLINICAL PRACTICE


Authors
Josef S Smolen, MD
Daniel Aletaha, MD
Section Editor
Ravinder N Maini, BA, MB BChir, FRCP, FMedSci, FRS
Deputy Editor
Paul L Romain, MD
All topics are updated as new evidence becomes available and our peer review process is
complete.
Literature review current through: Jun 2016. | This topic last updated: Feb 28, 2016.
INTRODUCTION Approaches to the management of rheumatoid arthritis (RA) have evolved
as an increasing number of effective disease-modifying antirheumatic drugs (DMARDs) have
become available. The early introduction of DMARDs has become standard of care, and
depends upon early diagnosis [1]. Such early diagnosis is facilitated by the 2010 American
College of Rheumatology (ACR)/EuropeanLeague Against Rheumatism (EULAR) rheumatoid
arthritis classification criteria [2].

The goals of DMARD use are not only to ameliorate the symptoms and signs of active RA, but
also to prevent structural joint damage and avoid functional impairment or enable its
restoration. The development of new antirheumatic therapies, including biologic and
synthetic agents targeted against specific components of the immunoinflammatory system,
has required the availability of instruments that permit the assessment of disease activity and
the response to therapy. Regardless of whether patients are evaluated in the context of a
clinical trial or longitudinal clinical practice, the successful application of DMARD therapy
requires that the goals of therapy be specified in advance and that the specific choice of
DMARDs be revisited on a regular basis [3,4].

Clinical indicators employed in the assessment of RA activity are discussed here. The roles of
both individual variables (eg, swollen joint counts and acute phase reactant measurements)
and composite indices for disease activity assessment are considered, as are definitions of
remission and criteria for clinically significant responses. Although much of the discussion
centers on clinical trial outcome measures, we also provide recommendations for clinical
practice.

A number of other topic reviews are related to the discussion of RA disease activity and
complement the information provided here. As examples:
The clinical features of RA, including the importance of distinguishing disease activity from
structural joint damage are discussed elsewhere. (See "Clinical manifestations of rheumatoid
arthritis".)

Functional capacity and functional disability indices, which relate to a large extent to the
degree of RA disease activity, are reviewed separately. (See "Disease outcome and functional
capacity in rheumatoid arthritis".)

Descriptions of a variety of biologic markers of disease activity, including those with


established clinical utility and those that are of investigational interest are presented
elsewhere. (See "Biologic markers in the diagnosis and assessment of rheumatoid arthritis"
and "Investigational biologic markers in the diagnosis and assessment of rheumatoid
arthritis".)

The management of RA, including pharmacologic and nonpharmacologic as well as surgical


interventions is surveyed separately. (See "General principles of management of rheumatoid
arthritis in adults" and "Nonpharmacologic therapies and preventive measures for patients
with rheumatoid arthritis".)

GENERAL PRINCIPLES The conceptual framework for the assessment of disease activity in
rheumatoid arthritis (RA) is defined by several principles. (See "General principles of
management of rheumatoid arthritis in adults".)

Active RA leads to severe joint destruction, functional disability, and impaired health status
[5-12], particularly if sustained at high levels for prolonged periods of time. Thus, reduction of
clinical disease activity through prudent DMARD use is an essential therapeutic aim.

Functional impairment may relate to both active RA, manifested by symptoms such as pain,
swelling, and stiffness of the joints, and to structural joint damage occurring as the
consequence of previously active disease [7,9,13-15].

With few exceptions [16], clinically active RA and the processes leading to joint destruction
are linked, such that damage usually progresses in the presence of active disease [16-23].

Monitoring disease activity at regular, short-term intervals (not to exceed three months
when disease is active) and appropriate modifications of disease-modifying antirheumatic
drug (DMARD) therapy to establish and maintain control of disease result in improved
radiographic and functional outcomes in patients with RA [24-26]. To this end, the European
League Against Rheumatism (EULAR) recommendations on the management of RA provide a
treatment strategy, including recommendations for an approach to the institution of various
therapeutic agents [3].

The response to disease-modifying therapy in RA can be assessed by use of one of several


response criteria that allow for an assessment of the degree of disease activity that is present,
the extent of improvement that has occurred, and whether a state of remission has been
achieved. (See 'Response criteria' below and 'Remission' below.)

Therapeutic goals are the achievement of remission (ie, the virtual absence of disease
activity) or a state of low disease activity as the second-best option. These goals are consistent
with the recommendations of an international task force [4]. In the majority of RA patients,
the achievement of sustained disease remission is associated with the cessation of joint
damage [8,18,27-29]. However, in the majority of patients with longstanding disease,
remission may not be an achievable goal, and therefore low disease activity is an acceptable
alternative.

While some progression of damage and residual impairment of physical function may occur in
association with low disease activity [30,31], a state of moderate disease activity, as defined
by composite measures, is generally not acceptable, as it leads to much worse outcomes by
comparison with low disease activity. Nevertheless, patient-specific factors and other risks
need to be taken into account when adapting therapy to attain a good outcome. (See
'Composite indices for disease activity assessment' below.)

The following sections describe individual elements that are assessed in order to determine
RA activity, the concepts of composite indices and response criteria, and the definitions of low
disease activity and remission.

INDIVIDUAL VARIABLES OF DISEASE ACTIVITY Common indicators of disease activity in


rheumatoid arthritis (RA) include the following measurements (reviewed in [32]):

Swollen and tender joint counts

Pain

Patient and evaluator global assessments of disease activity

Erythrocyte sedimentation rate and C-reactive protein (ESR, CRP)

Duration of morning stiffness


Fatigue

Additional measures, which partly reflect disease activity and partly reflect disease outcome,
include:

Measures of function (eg, the Health Assessment Questionnaire)

Health status measures (eg, the Short Form 36)

Core sets of disease activity variables Beginning in the 1990s, several groups defined core
sets of disease activity variables. The American College of Rheumatology (ACR), European
League Against Rheumatism (EULAR), and a group of investigators representing both the
World Health Organization (WHO) and International League of Associations of Rheumatology
(ILAR) all published similar core sets of activity variables [33-35]. The methods used to identify
these variables were driven by clinical data and met several criteria for validity [36-38].

All three core sets include swollen and tender joint counts, patient assessment of pain, patient
global assessment of disease activity, and a measure of the acute phase response. As an
example, the ACR core data set includes a total of seven measures: three performed by the
evaluator (swollen joint count, tender joint count, and global assessment of disease activity);
three collected by patient self-report (functional status, pain, and global assessment of
disease activity); and a laboratory measure (either an ESR or CRP). The ACR
and WHO/ILAR core sets both include evaluator global assessments of disease activity and
physical function. In contrast, in the EULAR core set, physical function is regarded as an
outcome variable rather than a process variable.

Swollen and tender joint counts Joints are typically assessed according to two
characteristics: 1) soft tissue swelling and effusion (the swollen joint count); and 2) pain on
pressure or motion (the tender joint count). In general, neither the weighting of joints by their
size (ie, the cartilage surface area) nor grading them by degree of swelling or tenderness
confers validity and reliability [39,40].

The 28 joint count has become a standard for use in both clinical practice and clinical trials
[40-43]. The 28 joint count excludes assessments of the foot and ankle joints, because the
interpretation of swelling and tenderness in these joints is confounded frequently by disorders
other than RA [40,42,43]. Although the 28 joint count has been criticized for leaving out
assessment of the feet, it has been thoroughly validated and employed reliably in clinical trials
and other analyses [44-46]. It is the basis of todays most frequently employed composite
measures of disease activity. Importantly, employing a 28-joint count for determining levels of
disease activity and response does not obviate the responsibility to also assess ankles and feet
in clinical practice.

Pain Pain is usually measured by visual analog scales [43-45], which most often use
horizontal 100 mm lines. Patients indicate their degree of pain (typically over the preceding
week) by placing a mark between "no pain" (left end, 0 mm) and excruciating pain (right end,
100 mm). Alternatives to visual analog scales include numerical rating scales (ranging from 0
to 10 in steps of 1 or 0.5) and categorical scales (eg, 5-point Likert scales), both of which are
also reliable and sensitive to change [47-49].

Patient and evaluator global assessments Global disease activity rated by the patient, the
evaluator, or both (and termed, respectively, the PGA or EGA, as appropriate) is assessed in a
similar manner using visual analog scales, numerical rating scales, or Likert scales. Measuring
the PGA acknowledges the importance of patient-reported outcomes. Whereas the EGA
typically integrates information from both subjective and objective variables, the PGA is
considered a subjective measure, which is strongly weighted for pain [50]. Because patients
are more likely than medically-trained evaluators to construe functional disability as a
manifestation of active disease, and because medically-trained evaluators have greater
context for comparison with other RA patients, PGAs are usually scored at higher levels than
are EGAs.

The typical question asked of the patient for a PGA is: "Considering all the ways your arthritis
has affected you, how do you feel your arthritis is today?" This assessment is presumed to also
take into account aspects of disease activity that might be less well appreciated by the
clinician (eg, fatigue or sleep disturbances).

Acute phase reactants Acute phase reactant levels, particularly the ESR and CRP, constitute
the most objective disease activity measures. Acute phase reactant levels correlate well with
both clinical disease activity measurements and radiographic progression of joint damage
[6,10,51-54]. The ESR and CRP are the two biomarkers used most widely to assess disease
activity. These tests are widely available, relatively inexpensive, and reflective of the cascade
of inflammatory events associated with active RA; the CRP in particular is well standardized.
Measurements of individual components of this response, eg, levels of the interleukin (IL)-1
beta, IL-6, or tumor necrosis factor (TNF)-alpha, are not more useful in assessing RA activity
and are prone to greater variation from laboratory to laboratory. Moreover, among these
cytokines, only IL-6 can be consistently and reliably measured in patient sera.

Other variables Other variables associated with disease activity include the duration of
morning stiffness, degree of fatigue, and the reduction in functional capacity. Morning
stiffness is not contained among the core set variables because of its higher variability and
lower sensitivity to change compared with other measures. A variety of patient-reported
instruments are available to measure levels of fatigue, including the vitality/fatigue scales that
constitute part of the short form (SF)-36. A specific measure for fatigue, the Functional
Assessment of Chronic Illness Therapy (FACIT) fatigue measure, has been developed. More
commonly, however, fatigue is assessed simply through the use of a visual analog scale.

COMPOSITE INDICES FOR DISEASE ACTIVITY ASSESSMENT The individual variables outlined
above reflect major characteristics of the disease within the population of rheumatoid
arthritis (RA) patients as a whole. Given the heterogeneity of RA, however, the predominance
of these indicators may be highly variable across individual patients, and may even vary with
time within individual patients. As a result, the evaluation of a single core variable (eg, the
erythrocyte sedimentation rate) in patients with RA would not reflect accurately the full
spectrum of the disease. In addition, the evaluation of all variables within core sets often leads
to heterogeneous responses and substantial methodological problems [36-38,55]. Thus,
"composite" indices combining several core set variables have been developed. Several
composite indices are in common use (table 1).

Formulas for calculating the indices vary in complexity, some are challenging to compute
[10,41,56-59]. The Disease Activity Score (DAS), its derivative (the DAS28), the Simplified
Disease Activity Index (SDAI), and the Clinical Disease Activity Index (CDAI) are described in
greater detail below. Interestingly, baseline disease activity by these composite scores, and
especially disease activity after three months of therapy with DMARDs is highly predictive of
disease activity at later points in time (one year) [60] allowing to adopt treatment strategies
that allow rapid, dynamic changes of therapeutic agents in patients who continue with high
(or sometimes moderate) disease activity by three to six months from start of therapy.

Disease activity score (DAS) The basis of the DAS was the clinician's decision to raise or
lower disease-modifying antirheumatic drug (DMARD) doses based on a largely qualitative
assessment of disease activity, and reflected clinical practice in about 1990 [56]. Several
features make the DAS challenging to use in clinical trial and practice settings. First, the DAS
employs the Ritchie Articular Index [57], a measure with major shortcomings in terms of
feasibility and reliability, to evaluate joint tenderness. Second, the DAS employs an extensive
44 joint count to record the number of swollen joints. Finally, once the data required to
complete the DAS are accumulated, the formula for calculating the score is quite complex,
using different weights for each of the variables as well as square roots or logarithmic
transformations in the case of some (table 1). In summary, the original DAS is complicated and
not very user friendly, and therefore not ideally suited to wide use.

DAS28 score The development of the DAS was important because it provided a global
summative and continuous score for disease activity assessment [56]. A modification of the
DAS, the DAS28, eliminated the grading of joints and reduced the number of joints evaluated
to 28 [41,59]; it has been widely used in clinical trials and in practice. A calculator for the
DAS28 is available (calculator 1).

Ranges of DAS28 scores that correspond to high, moderate, and low disease activity have
been proposed (table 1 and figure 1). High disease activity relates to DAS28 >5.1, moderate to
DAS28 of >3.2 to 5.1, low disease activity is regarded in the range of 2.6 to 3.2. A cut-off point
for remission has also been proposed (DAS28 <2.6). However, studies find that nearly 15
percent of patients with DAS28 scores of 2.6 (the cut-point for remission) continue to have at
least two swollen joints; some may have more than 10 swollen joints yet still be categorized as
in "remission" using this composite index [11,61-63] (see 'Remission' below). Other composite
measures also permit a number of inflamed joints among patients who fulfill criteria for
remission.

The greater relative weight the DAS28 gives to acute phase measures can result in a
disproportionately lower estimate of disease activity compared with other measures, such as
the SDAI or CDAI (see 'Simplified disease activity index (SDAI)' below and 'Clinical disease
activity index (CDAI)' below) when used to assess disease activity in patients receiving agents
such as tocilizumab that have more pronounced effects on acute phase reactants relative to
their overall clinical benefit [64]. As revealed by the formula of the DAS28, it also gives half as
much weight to a swollen joint as it does to a tender joint, although both have a similar level
of severity of inflammation.

Simplified disease activity index (SDAI) The SDAI, which employs five of the core set
variables (table 1) is calculated using a linear sum of unweighted, untransformed variables
[59].

The SDAI is defined as the simple sum of the following:

Tender joint count (using 28 joints)

Swollen joint count (using 28 joints)

Patient global assessment (0 to 10 scale)

Physician global assessment (0 to 10 scale)

C-reactive protein level (mg/dL)


The SDAI has been validated for use in both clinical trials and clinical practice [11,65,66].
Moreover, it has been shown to have the highest sensitivity and specificity for predicting
clinicians' decisions to change DMARD therapy in 2005 when compared with other composite
scores [66,67] and to correlate best with sonographic outcomes. A calculator for the SDAI is
available, although the SDAI (and the CDAI (see 'Clinical disease activity index (CDAI)' below))
can potentially be performed without the use of a calculator in daily practice (calculator 2).

Cutpoints have been established for the various activity states (table 1 and figure 1). A
cutpoint of 15 for the SDAI had the best combination of sensitivity and specificity (90 percent
and 86 percent, respectively) when compared with the treating clinicians' decisions to change
DMARDs because of active disease [66]. The cutpoint for remission, an SDAI of 3.3, does not
allow the presence of more than two joints that are swollen or tender; because of its
stringency, it also constitutes the index-based American College of Rheumatology (ACR)-
European League Against Rheumatism (EULAR) remission definition [68,69]. The established
cutpoints of 11 and 26 separate low disease activity (11) from moderate (26) and high (>26)
disease activity [11,70].

Clinical disease activity index (CDAI) A further simplification of the SDAI, the CDAI does not
require the measurement of an acute phase reactant, but otherwise uses the same measures
as the SDAI (table 1 and figure 1). (See 'Simplified disease activity index (SDAI)' above.)

The CDAI correlates well with other disease activity scores and response criteria, as well as
with progression of joint damage and functional impairment [10,59,70]. The cutpoint for
remission is 2.8 and that for low disease activity is 10. The advantage of the CDAI is that it
facilitates immediate treatment decisions based entirely on clinical criteria and includes
assessment of the joints, the principle target organ" in RA. This attribute is useful in clinical
trials and practice, since it circumvents the potential problem of lab to lab variation in the
measurement of acute phase reactants. A calculator for the CDAI is available (calculator 3).

Since the CDAI does not incorporate measures of acute phase reactants, it presumably
constitutes the most valid measure across all agents, including drugs that interfere directly
with the acute phase reactants. However, the contribution of CRP to the SDAI is only minimal
[10] and, therefore, these two scores give very similar results irrespective of the type of drug
used.

PHYSICAL FUNCTION ASSESSMENTS AS MEASURES OF DISEASE ACTIVITY Because active


rheumatoid arthritis (RA) exerts a substantial effect on physical function, instruments
designed to measure physical function are also useful indicators of disease activity. Such
instruments, developed for generic use, are employed commonly in clinical trials, less often in
clinical practice. The two physical function assessment tools used most frequently are the
Health Assessment Questionnaire (HAQ) and the Medical Outcomes Study Short Form-36 (SF-
36) [71-74].

Health Assessment Questionnaire (HAQ) The complete HAQ is a comprehensive instrument


designed to assess patient disability, discomfort, medication side effects, costs, and mortality.
Of these components, only the HAQ Disability Index (HAQ-DI) is used frequently in clinical
trials and clinical practice. The HAQ-DI, a component of the American College of
Rheumatology (ACR) core data set for the evaluation of RA disease activity and outcome,
evaluates patients' ability to perform activities of daily living through their answers to 20
questions designed to assess upper or lower extremity use. These questions are organized
into eight categories: dressing, rising, eating, walking, hygiene, reach, grip, and usual activities.
Each question is answered on a four-level scale of impairment ranging from 0 to 3: 0 = no
difficulty; 1 = some difficulty; 2 = much difficulty; and 3 = inability to do.

The final HAQ index, which ranges from 0 to 3, is the mean of scores from all eight categories.
HAQ scores <0.3 are considered normal, however, the mean HAQ of the population rises with
age [75]. Higher HAQ scores indicate increasing disability. The minimal clinically important
difference in serial HAQ scores has been suggested to be 0.22 on the group level. Several
modifications of the HAQ have been employed in clinical trials and practice [73].

Although the primary influence on the HAQ is disease activity [9,72], the score reflects both
joint damage and disease activity. Therefore, results of the HAQ require careful interpretation
when used for disease activity assessment, as the irreversible, damage-related component of
disability as assessed by HAQ increases with the degree of joint damage and with disease
duration [14,15]. Thus, it has been proposed that the activity-related HAQ (ACT-HAQ) should
be distinguished from its damage-related component (DAM-HAQ).

The short form-36 The short form (SF)-36 is a patient-reported instrument designed to
assess overall health status; it is not disease-specific [74]. The SF-36, usually utilized to
measure patients' quality of life, has been validated in numerous diseases, including RA. The
instrument consists of 36 questions organized into 8 domains: physical function, physical role,
general health, bodily pain, mental health, social function, vitality/fatigue, and emotional role.
Data from these eight domains can be summarized into two categories: a physical component
score and a mental component score. SF-36 results are particularly useful for comparing
quality of life across cohorts of patients with different diseases, but also correlate well with
other measures of RA activity. Increasing SF-36 scores are indicative of improving health
status.

PATIENT-REPORTED INSTRUMENTS Several self-reported instruments are available for the


assessment of RA activity. These include the Rheumatoid Arthritis Disease Activity Index
(RADAI), the RapidAssessment of Disease Activity in Rheumatology (RADAR), and a
modification of the Routine Assessment of Patient Index Data (RAPID3) [76-78].

RADAI and RADAR The RADAI encompasses five items, including patient-assessed joint
counts [76]. A major detraction of the RADAI, similar to that of the Disease Activity Score
(DAS), is the need for a calculator. In contrast, the RADAR is a brief, two-page questionnaire
that includes six items related to arthritis symptoms, physical function, work impact,
psychological status, social health, and satisfaction with health status [77]. Although the use of
patient-reported indices of disease activity such as the RADAI or RADAR is appealing in
rheumatoid arthritis (RA), a condition associated with large personal dimensions, these
instruments are rarely used in clinical trials and almost never in clinical practice. The patient-
reported instruments used most often in clinical trials are global assessments of disease
activity (eg, visual analog scales), Health Assessment Questionnaire (HAQ) scores, and the
short form (SF)-36.

RAPID3 score The RAPID3, as an expansion of the RAPID, constitutes an index that is based
upon patient self-reported outcomes only and is simple to administer. It includes the HAQ,
physical function, pain, and patient global estimate, all normalized to 0 to 10, counted
together, and divided by 3 to yield a score on a scale of 0 to 10 [78,79]. It does not require a
formal joint count, although swollen joint counts correlate with progression of joint
damage [5,12]. It does provide similar quantitative information to the DAS28 or the
Clinical Disease Activity Index (CDAI) regarding disease activity [78]. However, in established
disease where functional impairment, to a large extent, is irreversible due to the accrued joint
damage, the RAPID3 likely does not correlate well with the actual clinical improvement.

COMPOSITE BIOMARKER SCORING A multi-biomarker disease activity (MBDA) assay and


score has been developed which is suggested to reflect disease activity using a blood test
based on a variety of biomarkers [80]. It is not clear when and how to use this blood test,
since while it reflects clinical disease activity, it may be misleading in patients with infections
or other comorbidities, and further study needs to be done in this regard. Indeed, since it
shows good correlation with clinical activity, clinical assessment remains the gold standard
and such tests, if sufficiently validated in various populations, may be useful for research
studies but do not replace clinical assessment. (See "Biologic markers in the diagnosis and
assessment of rheumatoid arthritis", section on 'Multi-protein biomarker algorithms'.)

RESPONSE CRITERIA Response criteria, defined for both moderate and major changes in
disease activity, have been developed for all of the major rheumatoid arthritis (RA) activity
assessment indices. Such criteria can be applied to large numbers of patients in the context of
clinical trials, and also in gauging the treatment responses of individual patients over time.
Response criteria may involve either the comparison of a patient's activity score to a baseline
value for that same patient, or the achievement of a certain disease activity state (eg, either
remission or low disease activity).

ACR response criteria An early attempt to define minimal response requirements was
provided by the Paulus criteria [81]. These criteria formed the basis for the American College
of Rheumatology (ACR) response criteria [82], which (in their initial iteration) outlined the
variables of a 20 percent improvement. The ACR20 response, a standard measure for many
clinical trials performed since 1995, is defined as improvement of at least 20 percent in the
number of both swollen and tender joints, as well as at least 20 percent improvement in three
or more of the five remaining core set variables (table 2).

The categorical response criteria embodied in the ACR20, ACR50, and ACR70 measure the
frequency of benefit (ie, the proportion of patients receiving a certain treatment that achieve
a defined response). In contrast, measuring the mean or median ACR-N (N for numeric)
improvement facilitates an estimation of the cumulative magnitude of benefit that may be
anticipated for a typical patient relative to the baseline disease activity. Both types of
measures are useful for certain situations, and in many cases the information they provide are
complementary. The hybrid-ACR response requires further validation. These measures are
described below.

ACR20 response The ACR20 response discriminates accurately between the effects of active
medications and those of placebo. However, because of improvements in RA treatment, the
ACR20 is no longer considered an optimal measure of clinically meaningful change: patients
who achieve only ACR20 responses may still have substantial disease burdens from active RA.
Moreover, the ACR20 has other potential weaknesses in some applications: 1) it does not
measure directly any responses >20 percent in magnitude over baseline; 2) it characterizes
the percentage of patients who meet this cutoff, rather than the response of the average
patient; and 3) it measures the change in patients' disease activity compared with baseline,
but does not quantify disease activity at the end of the period of interest.

ACR50 and ACR70 responses More ambitious criteria for improvements in disease activity
have been proposed; namely, the ACR50 and ACR70 responses [83], corresponding to 50 and
70 percent improvements, respectively. In contrast to ACR20 responses, patients notice
dramatic differences following the achievement of ACR50 and ACR70 responses. In clinical
trials of biologic agents, disease-modifying antirheumatic drugs (DMARDs), or various
combinations, the percentage of patients achieving ACR20, 50, and 70 responses with the
study agent (or combination of agents) have been on the order of 40 to 80 percent, 25 to 60
percent, and 10 to 40 percent, respectively. An ACR70 response corresponds well with
achievement of a low disease activity state [84].
ACR-N response However, as categorical variables, the ACR50 and ACR70 suffer from some
of the same drawbacks as the ACR20. Thus, some investigators have attempted to express
changes in the ACR core set variables as a continuous measure. One such effort, the ACR-N,
judges changes in the following three variables: swollen joint count, tender joint count, and
the median of the 5 remaining core set variables, using the 0 to 100 percent improvement
that is the smallest among these three measures [59]. Thus, the ACR-N quantifies the patient
response in terms of a single number, providing a continuous variable rather than a
categorical one.

Hybrid-ACR response Another attempt to expand the ACR response criteria is the Hybrid-
ACR measure. To calculate the Hybrid-ACR, first the average improvement in core set measure
is determined as percent improvement (with maximizing and thus censoring worsening at 100
percent). Then, (i) the mean of the percent changes across all core set measures, and (ii) the
traditional ACR response (ie, ACR20, ACR50, or ACR70 response or non-response) is
determined. The Hybrid-ACR is the first of these (ie, the mean percent change across all core
set variables), unless it falls within the respective range of the traditional ACR response. Under
these circumstances, the response is categorized as values of 19.99, 49.99, or 69.99 as long as
mean core set changes lie above the traditional ACR categories that were achieved; if,
however, the mean core set data lie below these traditional ACR categories, they are set to
20, 50, or 70. Thus, calculation of the Hybrid-ACR measure requires a variety of assessments
to be accounted for, including the traditional ACR response evaluation. It requires additional
validation.

EULAR response criteria The European League Against Rheumatism (EULAR) response
criteria are based upon the Disease Activity Score (DAS)28. These criteria categorize
improvement into either good or moderate responses (table 1) [85,86].

EULAR good response For a good response, the decline in score must exceed 1.2 and result
in the achievement of low disease activity (ie, DAS28 <3.2).

EULAR moderate response A moderate response, on the other hand, may be achieved by a
decline in the DAS28 by >1.2 (without reaching low disease activity); or by a decline of 0.6 to
1.2, plus reaching at least moderate disease activity (ie, DAS28 <5.1).

Comparisons of the ACR and EULAR Response Criteria indicate that moderate EULAR
responses are achieved more often than ACR20 responses in most studies. Good EULAR
responses are observed more frequently than are ACR70 responses. (See 'ACR response
criteria' above.)
SDAI and CDAI response criteria Response cutoffs have been defined for the Simplified
Disease Activity Index (SDAI) and the Clinical Disease Activity Index (CDAI) that correspond
with the traditional ACR responses; these definitions of minor, moderate, and major response
have been defined as relative improvements of SDAI or CDAI of 50, 70, or 85 percent,
respectively [87]. (See 'Simplified disease activity index (SDAI)' above and 'Clinical disease
activity index (CDAI)' above.)

REMISSION Provisional definitions of remission were developed jointly by the American


College of Rheumatology (ACR) and the European League Against Rheumatism (EULAR) for use
in clinical trials and in clinical practice [68,69]. These definitions were considered provisional
at the time of their publication, but now have undergone validation in several additional
studies, also in comparisons with other criteria [63,88-90], and have also been adopted by the
US Food and Drug Administration (FDA) draft guidance to industry for developing drugs
products for treatment of rheumatoid arthritis (RA) and European Medicines Agency (EMA)
Guideline on clinical investigation of medicinal products other than nonsteroidal
antiinflammatory drugs (NSAIDs) for treatment of RA.

For clinical trials, the ACR/EULAR remission criteria recommend use of either of the following
definitions as the primary and the other as a secondary outcome measure:

Simplified Disease Activity Index (SDAI) 3.3 (calculator 2) (see 'Simplified disease activity
index (SDAI)' above)

OR

All of the following:

Swollen and tender joint counts (using 28 joint count) each 1

Patient global assessment (on a 0 to 10 scale) 1

C-reactive protein (CRP) (expressed in mg/dL) 1

For clinical practice, either of the clinical trial criteria, modified by exclusion of the CRP, may
be used:

Clinical Disease Activity Index (CDAI) 2.8 (calculator 3) (see 'Clinical disease activity index
(CDAI)' above)

OR
All of the following:

Swollen and tender joint counts (using 28 joint count) each 1

Patient global assessment (on a 0 to 10 scale) 1

In concept, the state of remission constitutes a clinical condition in which no active disease is
present. As noted, definitions of disease remission technically permit within their parameters
some degree of active disease (table 1) [11,66,68,69,91,92]. The identification of remission is
hampered practically by the presence of irreversible joint damage, which may lead to
abnormalities confused with residual disease activity. In a similar manner, comorbidities such
as fibromyalgia or osteoarthritis may confound the designation of remission.

Because progressive joint destruction may occur and function may decline, albeit at
comparatively slow rates, when disease activity persists even at a low level, stringent criteria
are important as a means of distinguishing remission from low disease activity [8,11,25,93]. In
this regard, as briefly noted above, remission criteria by SDAI and CDAI, as well as Boolean-
based criteria, are not only more stringent but also more reliable than Disease Activity Score
(DAS)28 remission criteria and constitute the index-based ACR/EULAR provisional definitions
of remission [68,69].

The above new remission definition has meanwhile been widely validated to convey better
outcomes than other measures [63,88-90].

SUMMARY

The three major points supporting the use of disease activity measures in clinical practice to
try to achieve a goal of remission are the following: 1) Active rheumatoid arthritis (RA) leads
to severe joint destruction, functional disability, and impaired health status; 2) monitoring
disease activity at regular, short-term intervals and appropriate modifications of disease-
modifying antirheumatic drug (DMARD) therapy leads to significant functional and radiologic
improvements; and 3) joint destruction progresses only at a slow rate and much less than in
moderate or high disease activity states; therefore, low disease activity is an alternative
treatment target, especially in established disease.

Several indicators of disease activity are typically assessed in clinical trials of therapeutic
agents in patients with RA. Among them, the most often measured are: swollen and tender
joint counts, pain, patient and evaluator global assessments, acute phase reactants
(erythrocyte sedimentation rate [ESR] and C-reactive protein [CRP]), duration of morning
stiffness, fatigue, measures of function (eg, the Health Assessment Questionnaire), and of
health status (eg, the Short Form 36). (See 'Individual variables of disease activity' above.)

Various sets of individual measures have been characterized that are useful for assessing the
efficacy of drug treatment of RA. These core sets include those developed by the American
College of Rheumatology (ACR), European League Against Rheumatism (EULAR), and World
Health Organization (WHO)/International League of Associations of Rheumatology (ILAR). (See
'Core sets of disease activity variables' above.)

Composite indices of disease activity (Simplified Disease Activity Index [SDAI], and Clinical
Disease Activity Index [CDAI]) represent indices that provide a timely aid to clinical decision
making. The Disease Activity Score using 28 joint counts (DAS28) is a good score for higher
disease activity, but not sufficiently stringent nor reproducible across different agents when
aiming at a good outcome such as remission. The SDAI and CDAI are also easier to calculate
than the DAS or DAS28 and define remission more stringently. Measures that do not include
joint counts may not reflect disease activity sufficiently in patients with longstanding disease
and may not be as well correlated with joint damage as the other indices. (See 'Composite
indices for disease activity assessment' above.)

Responses of groups of patients to treatment in clinical trials may be based on a categorical


criterion (eg, an ACR50 response, a EULAR good response, or a CDAI/SDAI major response) or
the mean change of a numeric index of disease activity (eg, DAS, SDAI, CDAI, ACR-N [N for
numeric], or Hybrid ACR). DAS, SDAI, and CDAI provide measures of actual disease activity,
while the ACR-N and Hybrid ACR represent improvements from a series of baseline values.
(See 'Response criteria' above.)

With some preparation and minor modifications of the usual work flow in clinical settings, all
of the variables comprising core data sets for RA activity assessment can be collected in a
standard fashion: 1) Tender and swollen joint counts can be obtained by any health
professional with appropriate training; 2) visual analogue scales for patient and evaluator
global assessments of disease activity require less than one minute to complete (figure 2); 3)
routine laboratory testing of the ESR, CRP, or both is inexpensive and widely available; 4)
HAQ-DI assessments may be completed by patients in the waiting room.

We suggest use of the CDAI to help guide treatment decisions by clinicians caring for
patients with RA. The CDAI is calculated as a simple numerical sum of swollen and tender
joints (using the 28 joint count), the patient global assessment, and the evaluator global
assessments. It can be used for clinical decision-making purposes even in the absence of
information about acute phase reactants. It is easy to obtain and calculate, despite the
inclusion of formal joint counts. The ESR or CRP, if available, may be useful in validating the
clinician's impressions based on the CDAI or other composite index of choice. (See 'Clinical
disease activity index (CDAI)' above.)

Definitions of remission that may be used in clinical trials and clinical practice, respectively,
have been developed by a joint committee of the ACR and the EULAR and meanwhile widely
validated. (See 'Remission' above.)

Optimal management of patients with RA and some degree of active disease requires serial
assessments. We suggest intervals not greater than every three months, in line with the
recommendations on treating RA to targeting remission or low disease activity.

Use of UpToDate is subject to the Subscription and License Agreement.


REFERENCES

Combe B, Landewe R, Lukas C, et al. EULAR recommendations for the management of early
1 arthritis: report of a task force of the European Standing Committee for International Clinical
Studies Including Therapeutics (ESCISIT). Ann Rheum Dis 2007; 66:34.
Aletaha D, Neogi T, Silman AJ, et al. 2010 rheumatoid arthritis classification criteria: an
2 American College of Rheumatology/European League Against Rheumatism collaborative
initiative. Ann Rheum Dis 2010; 69:1580.
Smolen JS, Landew R, Breedveld FC, et al. EULAR recommendations for the management
3 of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs:
2013 update. Ann Rheum Dis 2014; 73:492.
Smolen JS, Breedveld FC, Burmester GR, et al. Treating rheumatoid arthritis to target: 2014
4
update of the recommendations of an international task force. Ann Rheum Dis 2016; 75:3.
van Leeuwen MA, van der Heijde DM, van Rijswijk MH, et al. Interrelationship of outcome
measures and process variables in early rheumatoid arthritis. A comparison of radiologic
5
damage, physical disability, joint counts, and acute phase reactants. J Rheumatol 1994;
21:425.
van Leeuwen MA, van Rijswijk MH, Sluiter WJ, et al. Individual relationship between
6 progression of radiological damage and the acute phase response in early rheumatoid
arthritis. Towards development of a decision support system. J Rheumatol 1997; 24:20.
Welsing PM, van Gestel AM, Swinkels HL, et al. The relationship between disease activity,
7 joint destruction, and functional capacity over the course of rheumatoid arthritis. Arthritis
Rheum 2001; 44:2009.
Aletaha D, Machold KP, Nell VP, Smolen JS. The perception of rheumatoid arthritis core set
8
measures by rheumatologists. Results of a survey. Rheumatology (Oxford) 2006; 45:1133.
Drossaers-Bakker KW, de Buck M, van Zeben D, et al. Long-term course and outcome of
9 functional capacity in rheumatoid arthritis: the effect of disease activity and radiologic
damage over time. Arthritis Rheum 1999; 42:1854.
Aletaha D, Nell VP, Stamm T, et al. Acute phase reactants add little to composite disease
10 activity indices for rheumatoid arthritis: validation of a clinical activity score. Arthritis Res
Ther 2005; 7:R796.
Aletaha D, Ward MM, Machold KP, et al. Remission and active disease in rheumatoid
11
arthritis: defining criteria for disease activity states. Arthritis Rheum 2005; 52:2625.
Smolen JS, Van Der Heijde DM, St Clair EW, et al. Predictors of joint damage in patients with
12 early rheumatoid arthritis treated with high-dose methotrexate with or without concomitant
infliximab: results from the ASPIRE trial. Arthritis Rheum 2006; 54:702.
Smolen JS, Aletaha D. Patients with rheumatoid arthritis in clinical care. Ann Rheum Dis
13
2004; 63:221.
Aletaha D, Smolen J, Ward MM. Measuring function in rheumatoid arthritis: Identifying
14
reversible and irreversible components. Arthritis Rheum 2006; 54:2784.
Smolen JS, Aletaha D, Grisar JC, et al. Estimation of a numerical value for joint damage-
15 related physical disability in rheumatoid arthritis clinical trials. Ann Rheum Dis 2010;
69:1058.
Smolen JS, Han C, Bala M, et al. Evidence of radiographic benefit of treatment with
infliximab plus methotrexate in rheumatoid arthritis patients who had no clinical
16
improvement: a detailed subanalysis of data from the anti-tumor necrosis factor trial in
rheumatoid arthritis with concomitant therapy study. Arthritis Rheum 2005; 52:1020.
Devlin RD, Reddy SV, Savino R, et al. IL-6 mediates the effects of IL-1 or TNF, but not
17 PTHrP or 1,25(OH)2D3, on osteoclast-like cell formation in normal human bone marrow
cultures. J Bone Miner Res 1998; 13:393.
Kobayashi K, Takahashi N, Jimi E, et al. Tumor necrosis factor alpha stimulates osteoclast
18 differentiation by a mechanism independent of the ODF/RANKL-RANK interaction. J Exp Med
2000; 191:275.
Kudo O, Sabokbar A, Pocock A, et al. Interleukin-6 and interleukin-11 support human
19
osteoclast formation by a RANKL-independent mechanism. Bone 2003; 32:1.
Lam J, Takeshita S, Barker JE, et al. TNF-alpha induces osteoclastogenesis by direct
20 stimulation of macrophages exposed to permissive levels of RANK ligand. J Clin Invest 2000;
106:1481.
Ragab AA, Nalepka JL, Bi Y, Greenfield EM. Cytokines synergistically induce osteoclast
21 differentiation: support by immortalized or normal calvarial cells. Am J Physiol Cell Physiol
2002; 283:C679.
Redlich K, Hayer S, Ricci R, et al. Osteoclasts are essential for TNF-alpha-mediated joint
22
destruction. J Clin Invest 2002; 110:1419.
23 Teitelbaum SL. Bone resorption by osteoclasts. Science 2000; 289:1504.
Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, et al. Clinical and radiographic
24 outcomes of four different treatment strategies in patients with early rheumatoid arthritis
(the BeSt study): a randomized, controlled trial. Arthritis Rheum 2005; 52:3381.
Grigor C, Capell H, Stirling A, et al. Effect of a treatment strategy of tight control for
25 rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet
2004; 364:263.
Smolen JS, Sokka T, Pincus T, Breedveld FC. A proposed treatment algorithm for rheumatoid
26 arthritis: aggressive therapy, methotrexate, and quantitative measures. Clin Exp Rheumatol
2003; 21:S209.
Molenaar ET, Voskuyl AE, Dijkmans BA. Functional disability in relation to radiological
27 damage and disease activity in patients with rheumatoid arthritis in remission. J Rheumatol
2002; 29:267.
Aletaha D, Funovits J, Breedveld FC, et al. Rheumatoid arthritis joint progression in
28 sustained remission is determined by disease activity levels preceding the period of
radiographic assessment. Arthritis Rheum 2009; 60:1242.
Kavanaugh A, Fleischmann RM, Emery P, et al. Clinical, functional and radiographic
consequences of achieving stable low disease activity and remission with adalimumab plus
29
methotrexate or methotrexate alone in early rheumatoid arthritis: 26-week results from the
randomised, controlled OPTIMA study. Ann Rheum Dis 2013; 72:64.
Radner H, Smolen JS, Aletaha D. Remission in rheumatoid arthritis: benefit over low disease
30
activity in patient-reported outcomes and costs. Arthritis Res Ther 2014; 16:R56.
Smolen JS, Han C, van der Heijde DM, et al. Radiographic changes in rheumatoid arthritis
patients attaining different disease activity states with methotrexate monotherapy and
31
infliximab plus methotrexate: the impacts of remission and tumour necrosis factor blockade.
Ann Rheum Dis 2009; 68:823.
Aletaha D, Smolen JS. The definition and measurement of disease modification in
32
inflammatory rheumatic diseases. Rheum Dis Clin North Am 2006; 32:9.
Scott DL, Panayi GS, van Riel PL, et al. Disease activity in rheumatoid arthritis: preliminary
33 report of the Consensus Study Group of the European Workshop for Rheumatology
Research. Clin Exp Rheumatol 1992; 10:521.
Felson DT, Anderson JJ, Boers M, et al. The American College of Rheumatology preliminary
34 core set of disease activity measures for rheumatoid arthritis clinical trials. The Committee
on Outcome Measures in Rheumatoid Arthritis Clinical Trials. Arthritis Rheum 1993; 36:729.
Boers M, Tugwell P, Felson DT, et al. World Health Organization and International League of
35 Associations for Rheumatology core endpoints for symptom modifying antirheumatic drugs
in rheumatoid arthritis clinical trials. J Rheumatol Suppl 1994; 41:86.
Tugwell P, Bombardier C. A methodologic framework for developing and selecting endpoints
36
in clinical trials. J Rheumatol 1982; 9:758.
van der Heijde DM, van't Hof MA, van Riel PL, et al. Validity of single variables and
37 composite indices for measuring disease activity in rheumatoid arthritis. Ann Rheum Dis
1992; 51:177.
Goldsmith CH, Boers M, Bombardier C, Tugwell P. Criteria for clinically important changes in
38 outcomes: development, scoring and evaluation of rheumatoid arthritis patient and trial
profiles. OMERACT Committee. J Rheumatol 1993; 20:561.
Hart LE, Tugwell P, Buchanan WW, et al. Grading of tenderness as a source of interrater
39
error in the Ritchie articular index. J Rheumatol 1985; 12:716.
Prevoo ML, van Riel PL, van 't Hof MA, et al. Validity and reliability of joint indices. A
40 longitudinal study in patients with recent onset rheumatoid arthritis. Br J Rheumatol 1993;
32:589.
Prevoo ML, van 't Hof MA, Kuper HH, et al. Modified disease activity scores that include
41 twenty-eight-joint counts. Development and validation in a prospective longitudinal study of
patients with rheumatoid arthritis. Arthritis Rheum 1995; 38:44.
Fuchs HA, Pincus T. Reduced joint counts in controlled clinical trials in rheumatoid arthritis.
42
Arthritis Rheum 1994; 37:470.
Smolen JS, Breedveld FC, Eberl G, et al. Validity and reliability of the twenty-eight-joint
43
count for the assessment of rheumatoid arthritis activity. Arthritis Rheum 1995; 38:38.
Emery P, Breedveld FC, Lemmel EM, et al. A comparison of the efficacy and safety of
44 leflunomide and methotrexate for the treatment of rheumatoid arthritis. Rheumatology
(Oxford) 2000; 39:655.
Smolen JS, Kalden JR, Scott DL, et al. Efficacy and safety of leflunomide compared with
45 placebo and sulphasalazine in active rheumatoid arthritis: a double-blind, randomised,
multicentre trial. European Leflunomide Study Group. Lancet 1999; 353:259.
van Tuyl LH, Britsemmer K, Wells GA, et al. Remission in early rheumatoid arthritis defined
46 by 28 joint counts: limited consequences of residual disease activity in the forefeet on
outcome. Ann Rheum Dis 2012; 71:33.
Carlsson AM. Assessment of chronic pain. I. Aspects of the reliability and validity of the
47
visual analogue scale. Pain 1983; 16:87.
Scott J, Huskisson EC. Vertical or horizontal visual analogue scales. Ann Rheum Dis 1979;
48
38:560.
Williamson A, Hoggart B. Pain: a review of three commonly used pain rating scales. J Clin
49
Nurs 2005; 14:798.
Studenic P, Smolen JS, Aletaha D. Near misses of ACR/EULAR criteria for remission: effects
50 of patient global assessment in Boolean and index-based definitions. Ann Rheum Dis 2012;
71:1702.
Dawes PT, Fowler PD, Clarke S, et al. Rheumatoid arthritis: treatment which controls the C-
51 reactive protein and erythrocyte sedimentation rate reduces radiological progression. Br J
Rheumatol 1986; 25:44.
Mallya RK, de Beer FC, Berry H, et al. Correlation of clinical parameters of disease activity in
52 rheumatoid arthritis with serum concentration of C-reactive protein and erythrocyte
sedimentation rate. J Rheumatol 1982; 9:224.
Plant MJ, Williams AL, O'Sullivan MM, et al. Relationship between time-integrated C-reactive
53 protein levels and radiologic progression in patients with rheumatoid arthritis. Arthritis
Rheum 2000; 43:1473.
Crowson CS, Rahman MU, Matteson EL. Which measure of inflammation to use? A
54 comparison of erythrocyte sedimentation rate and C-reactive protein measurements from
randomized clinical trials of golimumab in rheumatoid arthritis. J Rheumatol 2009; 36:1606.
Schulz KF, Grimes DA. Multiplicity in randomised trials I: endpoints and treatments. Lancet
55
2005; 365:1591.
van der Heijde DM, van 't Hof M, van Riel PL, van de Putte LB. Development of a disease
56 activity score based on judgment in clinical practice by rheumatologists. J Rheumatol 1993;
20:579.
Ritchie DM, Boyle JA, McInnes JM, et al. Clinical studies with an articular index for the
57
assessment of joint tenderness in patients with rheumatoid arthritis. Q J Med 1968; 37:393.
Smolen JS, Breedveld FC, Schiff MH, et al. A simplified disease activity index for rheumatoid
58
arthritis for use in clinical practice. Rheumatology (Oxford) 2003; 42:244.
Siegel JN, Zhen BG. Use of the American College of Rheumatology N (ACR-N) index of
59
improvement in rheumatoid arthritis: argument in favor. Arthritis Rheum 2005; 52:1637.
Aletaha D, Funovits J, Keystone EC, Smolen JS. Disease activity early in the course of
60 treatment predicts response to therapy after one year in rheumatoid arthritis patients.
Arthritis Rheum 2007; 56:3226.
van der Heijde D, Klareskog L, Boers M, et al. Comparison of different definitions to classify
61
remission and sustained remission: 1 year TEMPO results. Ann Rheum Dis 2005; 64:1582.
Mkinen H, Kautiainen H, Hannonen P, Sokka T. Is DAS28 an appropriate tool to assess
62
remission in rheumatoid arthritis? Ann Rheum Dis 2005; 64:1410.
Balsa A, de Miguel E, Castillo C, et al. Superiority of SDAI over DAS-28 in assessment of
63 remission in rheumatoid arthritis patients using power Doppler ultrasonography as a gold
standard. Rheumatology (Oxford) 2010; 49:683.
Smolen JS, Aletaha D. Interleukin-6 receptor inhibition with tocilizumab and attainment of
64 disease remission in rheumatoid arthritis: the role of acute-phase reactants. Arthritis Rheum
2011; 63:43.
Wong AL, Harker JO, Park GS, Paulus HE. Longitudinal measurement of RA disease activity
65
in a clinical practice setting: Usefulness of the SDAI. Arthritis Rheum 2004; 50:S386.
Soubrier M, Zerkak D, Gossec L, et al. Which variables best predict change in rheumatoid
66
arthritis therapy in daily clinical practice? J Rheumatol 2006; 33:1243.
American College of Rheumatology Committee to Reevaluate Improvement Criteria. A
67 proposed revision to the ACR20: the hybrid measure of American College of Rheumatology
response. Arthritis Rheum 2007; 57:193.
Felson DT, Smolen JS, Wells G, et al. American College of Rheumatology/European League
68 Against Rheumatism provisional definition of remission in rheumatoid arthritis for clinical
trials. Arthritis Rheum 2011; 63:573.
Felson DT, Smolen JS, Wells G, et al. American College of Rheumatology/European League
69 against Rheumatism provisional definition of remission in rheumatoid arthritis for clinical
trials. Ann Rheum Dis 2011; 70:404.
Aletaha D, Smolen J. The Simplified Disease Activity Index (SDAI) and the Clinical Disease
70 Activity Index (CDAI): a review of their usefulness and validity in rheumatoid arthritis. Clin
Exp Rheumatol 2005; 23:S100.
Fries JF, Spitz P, Kraines RG, Holman HR. Measurement of patient outcome in arthritis.
71
Arthritis Rheum 1980; 23:137.
Scott DL, Pugner K, Kaarela K, et al. The links between joint damage and disability in
72
rheumatoid arthritis. Rheumatology (Oxford) 2000; 39:122.
Pincus T, Swearingen C, Wolfe F. Toward a multidimensional Health Assessment
Questionnaire (MDHAQ): assessment of advanced activities of daily living and psychological
73
status in the patient-friendly health assessment questionnaire format. Arthritis Rheum 1999;
42:2220.
Ware JE Jr, Sherbourne CD. The MOS 36-item short-form health survey (SF-36). I.
74
Conceptual framework and item selection. Med Care 1992; 30:473.
Vita AJ, Terry RB, Hubert HB, Fries JF. Aging, health risks, and cumulative disability. N Engl
75
J Med 1998; 338:1035.
Stucki G, Liang MH, Stucki S, et al. A self-administered rheumatoid arthritis disease activity
76 index (RADAI) for epidemiologic research. Psychometric properties and correlation with
parameters of disease activity. Arthritis Rheum 1995; 38:795.
Mason JH, Anderson JJ, Meenan RF, et al. The rapid assessment of disease activity in
77 rheumatology (radar) questionnaire. Validity and sensitivity to change of a patient self-
report measure of joint count and clinical status. Arthritis Rheum 1992; 35:156.
Pincus T, Swearingen CJ, Bergman MJ, et al. RAPID3 (Routine Assessment of Patient Index
78 Data) on an MDHAQ (Multidimensional Health Assessment Questionnaire): agreement with
DAS28 (Disease Activity Score) and CDAI (Clinical Disease Activity Index) activity
categories, scored in five versus more than ninety seconds. Arthritis Care Res (Hoboken)
2010; 62:181.
Pincus T, Amara I, Segurado OG, et al. Relative efficiencies of physician/assessor global
estimates and patient questionnaire measures are similar to or greater than joint counts to
79
distinguish adalimumab from control treatments in rheumatoid arthritis clinical trials. J
Rheumatol 2008; 35:201.
Centola M, Cavet G, Shen Y, et al. Development of a multi-biomarker disease activity test
80
for rheumatoid arthritis. PLoS One 2013; 8:e60635.
Paulus HE, Egger MJ, Ward JR, Williams HJ. Analysis of improvement in individual
rheumatoid arthritis patients treated with disease-modifying antirheumatic drugs, based on
81
the findings in patients treated with placebo. The Cooperative Systematic Studies of
Rheumatic Diseases Group. Arthritis Rheum 1990; 33:477.
Felson DT, Anderson JJ, Boers M, et al. American College of Rheumatology. Preliminary
82
definition of improvement in rheumatoid arthritis. Arthritis Rheum 1995; 38:727.
Felson DT, Anderson JJ, Lange ML, et al. Should improvement in rheumatoid arthritis clinical
83 trials be defined as fifty percent or seventy percent improvement in core set measures,
rather than twenty percent? Arthritis Rheum 1998; 41:1564.
Aletaha D, Funovits J, Smolen JS. The importance of reporting disease activity states in
84
rheumatoid arthritis clinical trials. Arthritis Rheum 2008; 58:2622.
van Gestel AM, Prevoo ML, van 't Hof MA, et al. Development and validation of the European
League Against Rheumatism response criteria for rheumatoid arthritis. Comparison with the
85 preliminary American College of Rheumatology and the World Health
Organization/International League Against Rheumatism Criteria. Arthritis Rheum 1996;
39:34.
van Gestel AM, Haagsma CJ, van Riel PL. Validation of rheumatoid arthritis improvement
86
criteria that include simplified joint counts. Arthritis Rheum 1998; 41:1845.
Aletaha D, Martinez-Avila J, Kvien TK, Smolen JS. Definition of treatment response in
87 rheumatoid arthritis based on the simplified and the clinical disease activity index. Ann
Rheum Dis 2012; 71:1190.
Thiele K, Huscher D, Bischoff S, et al. Performance of the 2011 ACR/EULAR preliminary
88 remission criteria compared with DAS28 remission in unselected patients with rheumatoid
arthritis. Ann Rheum Dis 2013; 72:1194.
Zhang B, Combe B, Rincheval N, Felson DT. Validation of ACR/EULAR definition of remission
89 in rheumatoid arthritis from RA practice: the ESPOIR cohort. Arthritis Res Ther 2012;
14:R156.
Sakellariou G, Scir CA, Verstappen SM, et al. In patients with early rheumatoid arthritis,
the new ACR/EULAR definition of remission identifies patients with persistent absence of
90
functional disability and suppression of ultrasonographic synovitis. Ann Rheum Dis 2013;
72:245.
Prevoo ML, van Gestel AM, van T Hof MA, et al. Remission in a prospective study of patients
91 with rheumatoid arthritis. American Rheumatism Association preliminary remission criteria in
relation to the disease activity score. Br J Rheumatol 1996; 35:1101.
Pinals RS, Masi AT, Larsen RA. Preliminary criteria for clinical remission in rheumatoid
92
arthritis. Arthritis Rheum 1981; 24:1308.
Pincus T, Stein CM, Wolfe F. "No evidence of disease" in rheumatoid arthritis using
93 methotrexate in combination with other drugs: a contemporary goal for rheumatology care?
Clin Exp Rheumatol 1997; 15:591.
Topic 7494 Version 11.0
All rights reserved.
2016 UpToDate, Inc.

GENERAL PRINCIPLES OF MANAGEMENT OF RHEUMATOID


ARTHRITIS IN ADULTS
Authors
Larry W Moreland, MD
Amy Cannella, MD, MS, RhMSUS
Section Editor
James R O'Dell, MD
Deputy Editor
Paul L Romain, MD
All topics are updated as new evidence becomes available and our peer review process is
complete.
Literature review current through: Jun 2016. | This topic last updated: Jun 10, 2016.
INTRODUCTION The treatment of rheumatoid arthritis (RA) is directed toward the control
of synovitis and the prevention of joint injury. The choice of therapies depends upon several
factors, including the severity of disease activity when therapy is initiated and the response of
the patient to prior therapeutic interventions.

Common principles that guide management strategies and the choice of agents have been
derived from an increased understanding of the disease and evidence from clinical trials and
other studies. These strategies include approaches directed at achieving remission or low
disease activity by more rapid and sustained control of inflammation by the institution of
disease-modifying antirheumatic drug (DMARD) therapy early in the disease course.

The general principles and treatment strategies that should be applied to the management of
RA are reviewed here. The initial therapy of RA and the treatment of patients resistant to
initial DMARDs are discussed in greater detail elsewhere. (See "Nonpharmacologic therapies
and preventive measures for patients with rheumatoid arthritis" and "Alternatives to
methotrexate for the initial treatment of rheumatoid arthritis in adults" and "Initial treatment
of rheumatoid arthritis in adults" and "Treatment of rheumatoid arthritis in adults resistant to
initial nonbiologic DMARD therapy" and "Treatment of rheumatoid arthritis in adults resistant
to initial biologic DMARD therapy".)

GENERAL PRINCIPLES Our overall approach to the treatment of patients with rheumatoid
arthritis (RA) depends upon the timely and judicious use of several types of therapeutic
interventions. The appropriate use of these therapies is based upon an understanding of a
group of general principles that have been widely accepted by major working groups and by
professional organizations of rheumatologists [1-5]. (See 'Recommendations by major groups'
below.)

These principles include:

Early recognition and diagnosis (see 'Early recognition and diagnosis' below)

Care by an expert in the treatment of rheumatic diseases, such as a rheumatologist (see


'Care by a rheumatologist' below)

Early use of disease-modifying antirheumatic drugs (DMARDs) for all patients diagnosed with
RA (see 'Early use of DMARDs' below)

Importance of tight control, utilizing a treat-to-target strategy, with a goal of remission or


low disease activity (see 'Tight control' below)

Use of antiinflammatory agents, including nonsteroidal antiinflammatory drugs (NSAIDs) and


glucocorticoids, only as adjuncts to therapy (see 'Adjunctive role of antiinflammatory agents'
below)

The application of these principles has resulted in significant improvement in the outcomes of
treatment, although the etiology and pathogenesis of RA are complex and are not fully
understood [6-8]. Such improvements may owe even more to the therapeutic strategies that
have been adopted than to the development and use of newer and more potent drugs [9].

RA is characterized by acute and chronic inflammation in the synovium, which is associated


with a proliferative and destructive process in joint tissues [8,10,11]. Many elements of innate
and adaptive immunity are involved in this process, and persistent inflammation may lead to
significant and irreversible joint injury as early as the first two years of disease. Therefore,
measures aimed at identifying early active disease and at markedly reducing inflammation are
essential for modifying disease outcome [7,9,12,13]. (See "Pathogenesis of rheumatoid
arthritis" and 'Tight control' below.)
Early recognition and diagnosis Achieving the benefits of early intervention with DMARDs
depends upon making the diagnosis of RA as early as possible. The recognition of RA early in
the course of inflammatory arthritis, before irreversible injury has occurred, is thus an
important element of effective management. (See 'Early use of DMARDs' below.)

In 2010, international collaborative efforts led to the development of new classification


criteria for RA, which have identified factors that support the early diagnosis of RA among
patients presenting with inflammatory arthritis [14]. These criteria are directed at the
recognition of patients with early arthritis who are most likely to develop progressive and
erosive disease. The diagnosis and differential diagnosis of RA are reviewed in detail
elsewhere. (See "Diagnosis and differential diagnosis of rheumatoid arthritis" and "Clinical
manifestations of rheumatoid arthritis".)

Care by a rheumatologist An expert in the treatment of rheumatic diseases, such as a


rheumatologist, should participate in the care of patients with inflammatory arthritis who are
suspected of having RA and in the ongoing care of patients diagnosed with this condition
[3,15]. The early and ongoing care of patients with RA by a rheumatologist is associated with
better disease outcomes, such as reduced joint injury and less functional disability, compared
with care rendered primarily by other clinicians [16-21].

The initial rheumatology consultation allows the diagnosis to be made or reassessed, the
severity of disease to be estimated, and a plan of care to be developed and initiated. The
frequency of subsequent specialist care depends upon the severity of symptoms and joint
inflammation, the patients response to treatment, the complexity and risks associated with
the therapy, and the preferences of the patient and primary care clinician.

NONPHARMACOLOGIC AND PREVENTIVE THERAPIES A number of nonpharmacologic


measures and other medical interventions are important in the comprehensive management
of rheumatoid arthritis (RA) in all stages of disease, in addition to antirheumatic drug
therapies. Patient education that addresses issues related to the disease and its management
is indicated for all patients with RA. Attention to health promotion and specific strategies
aimed at minimizing the adverse effects of RA and of agents used to treat it are also
appropriate. These therapies are discussed in detail elsewhere. (See "Nonpharmacologic
therapies and preventive measures for patients with rheumatoid arthritis".)

Briefly, these measures include:

Patient education

Psychosocial interventions
Rest, exercise, and physical and occupational therapy

Nutritional and dietary counseling

Interventions to reduce risks of cardiovascular disease, including smoking cessation and lipid
control

Screening for and treatment of osteoporosis

Immunizations to decrease risk of infectious complications of immunosuppressive therapies

PHARMACOLOGIC THERAPY

Pretreatment evaluation Prior to starting, resuming, or significantly increasing therapy with


nonbiologic or biologic disease-modifying antirheumatic drugs (DMARDs), we do the following
baseline studies [22]:

General testing for all patients We obtain a baseline complete blood count, serum
creatinine, aminotransferases, erythrocyte sedimentation rate (ESR), and C-reactive protein
(CRP) in all patients.

Hepatitis B and C In all patients without a known history of hepatitis, we screen for
hepatitis B and C before initiating therapy with conventional DMARDs, including methotrexate
(MTX) and leflunomide (LEF); biologic DMARDs; and tofacitinib. Patients should be screened
for hepatitis B with testing for hepatitis B virus (HBV) surface antigen and HBV core antibody
prior to initiating these drugs and prior to treatment with prednisone at doses of 20 mg daily.
Some experts limit screening for hepatitis C to patients at increased risk of hepatitis, such as
those who have a history of intravenous drug abuse, have had multiple sex partners in the
previous six months, or who are healthcare workers. [5,22]. Some patients may require
antiviral treatment prior to initiating DMARD or immunosuppressive therapy, depending upon
their level of risk for HBV reactivation. (See "Hepatitis B virus reactivation associated with
immunosuppressive therapy".)

Ophthalmologic screening for hydroxychloroquine use A complete baseline ophthalmologic


examination should be performed within the first year of treatment, including examination of
the retina through a dilated pupil and automated visual field testing. (See "Antimalarial drugs
in the treatment of rheumatic disease", section on 'Monitoring for toxicity'.)

Testing for latent tuberculosis We screen for latent tuberculosis (TB), consistent with the
US Centers for Disease Control (CDC) and 2015 American College of Rheumatology (ACR)
guidelines, with skin testing or an interferon-gamma release assay prior to all biologic
DMARDs and prior to use of the Janus kinase inhibitor, tofacitinib; such screening is based
upon evidence that these medications, including the tumor necrosis factor (TNF) inhibitors,
other biologics, and tofacitinib, may increase the risk of mycobacterial infection [5,23,24].

We also screen for latent TB in patients about to receive MTX who live, travel or work in areas
with likely TB exposure, although some clinicians prefer to screen all patients starting DMARD
therapy. We obtain a chest radiograph in patients with a history of other risk factors for latent
TB, even if screening tests are negative, given the risks of false-negative testing. Additionally,
skin testing should be repeated in patients with new TB exposures. Glucocorticoids and other
factors may cause false-negative results. Screening for latent TB is discussed in detail
separately. (See "Tumor necrosis factor-alpha inhibitors and mycobacterial infections" and
"Diagnosis of latent tuberculosis infection (tuberculosis screening) in HIV-uninfected adults".)

Interferon-gamma release assays can be used in place of tuberculin skin testing in the United
States, according to CDC recommendations, and may be preferred in patients who have
previously received vaccination with Bacillus Calmette-Guerin (BCG). However, in some
countries, tuberculin skin testing is preferred. (See "Interferon-gamma release assays for
diagnosis of latent tuberculosis infection".)

Choice of therapy Choices between treatment options are based upon multiple factors,
including:

Level of disease activity (eg, mild versus moderate to severe)

Presence of comorbid conditions

Stage of therapy (eg, initial versus subsequent therapy in patients resistant to a given
intervention)

Regulatory restrictions (eg, governmental or health insurance company coverage limitations)

Patient preferences (eg, route and frequency of drug administration, monitoring


requirements, personal cost)

Presence of adverse prognostic signs

A combination of the following types of therapies may be used:


Rapidly acting antiinflammatory medications, including nonsteroidal antiinflammatory drugs
(NSAIDs) and systemic and intraarticular glucocorticoids. (See 'Adjunctive role of
antiinflammatory agents' below.)

DMARDs, including nonbiologic (traditional small molecule or synthetic) and biologic


DMARDs, and an orally administered small molecule kinase inhibitor, which all have the
potential to reduce or prevent joint damage and to preserve joint integrity and function. (See
"Overview of immunosuppressive and conventional (non-biologic) disease-modifying drugs in
the rheumatic diseases".)

The nonbiologic (or conventional synthetic) DMARDs most frequently used include
hydroxychloroquine (HCQ), sulfasalazine (SSZ), MTX, and LEF. (See "Overview of
immunosuppressive and conventional (non-biologic) disease-modifying drugs in the rheumatic
diseases", section on 'Disease-modifying antirheumatic drugs' and "Antimalarial drugs in the
treatment of rheumatic disease" and "Sulfasalazine in the treatment of rheumatoid arthritis"
and "Use of methotrexate in the treatment of rheumatoid arthritis" and "Leflunomide in the
treatment of rheumatoid arthritis".)

Biologic DMARDs, produced by recombinant DNA technology, generally target cytokines or


their receptors or are directed against other cell surface molecules. These include anticytokine
therapies, such as the TNF-alpha inhibitors, etanercept, infliximab, adalimumab, golimumab,
and certolizumab pegol; the interleukin (IL)-1 receptor antagonist anakinra; and the IL-6
receptor antagonist tocilizumab. They also include other biologic response modifiers such as
the T-cell costimulation blocker abatacept (CTLA4-Ig) and the anti-CD20 B-cell depleting
monoclonal antibody rituximab. (See "Overview of biologic agents in the rheumatic diseases"
and "T-cell targeted therapies for rheumatoid arthritis", section on 'Abatacept' and "Rituximab
and other B cell targeted therapies for rheumatoid arthritis", section on 'Rituximab' and
"Randomized clinical trials in rheumatoid arthritis of biologic agents that inhibit IL-1, IL-6, and
RANKL", section on 'Tocilizumab'.)

Targeted synthetic DMARDs, including several kinase inhibitors, are in development for use in
rheumatoid arthritis (RA); one of these, tofacitinib, is available for such clinical use in the
United States and several other countries. Tofacitinib is an orally administered small molecule
DMARD that inhibits cytokine and growth factor signalling through interference with Janus
kinases. (See "Cytokine networks in rheumatic diseases: Implications for therapy", section on
'JAK inhibition'.)

Early use of DMARDs We recommend that all patients diagnosed with RA be started on
DMARD therapy as soon as possible. Our choice of drug therapies in patients with RA and the
evidence supporting these choices is described in detail separately. Briefly, we take the
following approach (see 'Assessment of disease activity' below):

In patients with active RA, we initiate antiinflammatory therapy with either a NSAID or
glucocorticoid, depending upon the degree of disease activity, and generally start DMARD
therapy with MTX. (See "Initial treatment of rheumatoid arthritis in adults".)

Patients unable to take MTX may require an alternative agent, such as HCQ, SSZ, or LEF. (See
"Alternatives to methotrexate for the initial treatment of rheumatoid arthritis in adults".)

In patients resistant to initial DMARD therapy (eg, MTX), we treat with a combination of
DMARDs (eg, MTX plus either SSZ and HCQ or a TNF inhibitor), while also treating the active
inflammation with antiinflammatory drug therapy. (See "Treatment of rheumatoid arthritis in
adults resistant to initial nonbiologic DMARD therapy".)

Much of the joint damage that ultimately results in disability begins early in the course of the
disease [25,26]. As an example, more than 80 percent of patients with RA of less than two
years duration had joint space narrowing on plain radiographs of the hands and wrists, while
two-thirds had erosions [26]. The use of more sensitive imaging techniques, such as magnetic
resonance imaging (MRI) and high-resolution ultrasonography, can identify even earlier
damage than that which is recognized by radiography, although the prognostic implications of
such findings are unknown [27-29].

Better outcomes are achieved by early compared with delayed intervention with DMARDs in
patients with RA [12,30-35]. As an example, one study in Texas found that low socioeconomic
status was associated with a significant delay in starting DMARD therapy; both factors were
independently associated with greater disease activity, joint damage, and physical disability
[35]. Some evidence suggests that the effect upon outcomes is not linear with time, but that
there may be an early window of opportunity for optimal DMARD treatment benefit [36]. As
examples:

The effect of symptom duration prior to DMARD therapy on the likelihood of DMARD-free
sustained remission was examined during five years of follow-up in patients with RA from two
cohorts, the Leiden Early Arthritis Clinic (EAC; n = 738) and tudeet Suivi de POlyarthrites
Indiffrencies Rcentes (ESPOIR; n = 533) [36]. Remission was maintained in 11.5 and 5.4
percent of patients from the two cohorts, respectively. The symptom durations that optimally
discriminated between greater and lesser likelihood of remission were between 11.4 and 19.1
weeks, depending upon the cohort and whether or not patients were anti-citrullinated
peptide antibody (ACPA)-positive, suggesting there is a limited period of several months
following symptom onset during which RA is most susceptible to intervention with DMARDs.
An observational study of 1435 patients involved in 14 trials, primarily of MTX or other
nonbiologic DMARDs, found a progressive decrease over time in the likelihood of a significant
response to DMARD therapy. Response rates were higher in patients with no more than one
year of disease than in those with one to two years of disease, and response rates were
lowest in the group with greater than 10 years of disease (53 versus 43 versus 35 percent)
[30].

ASSESSMENT AND MONITORING Patients should be seen on a regular basis for clinical
evaluation and monitoring of clinical and laboratory assessment of disease activity and for
screening for drug toxicities. The initial evaluation and subsequent monitoring should also
include periodic assessment of disease activity using a quantitative composite measure of
disease activity. (See 'Assessment of disease activity' below.)

Additionally, the ongoing evaluation and monitoring of patients with rheumatoid arthritis (RA)
following the initiation of therapy also involves:

Patient and clinician assessment of symptoms and functional status (see 'Symptoms and
functional status' below)

Evaluation of joint involvement and extraarticular manifestations (see 'Physical examination'


below)

Laboratory markers (see 'Laboratory monitoring of disease activity' below and 'Monitoring
and prevention of drug toxicity' below)

Imaging (see 'Imaging' below)

Assessment of disease activity Disease activity should be evaluated initially and at all
subsequent visits. We recommend that a structured assessment of disease activity using a
composite measure, such as those described here, should be performed initially, and most
patients should be seen at least every three months to monitor the response to therapy using
the same measure. Adjustments to treatment regimens should be made to quickly achieve
and maintain control of disease activity if targeted treatment goals (remission or low disease
activity), rather than an undefined degree of improvement, have not been achieved. (See
"Assessment of rheumatoid arthritis activity in clinical trials and clinical practice", section on
'Composite indices for disease activity assessment' and 'Tight control' below.)

The initial assessment, made once the diagnosis is established, helps to distinguish the smaller
group of patients who present with mild disease from the majority of patients who present
with moderately to severely active disease and who are usually treated initially with
methotrexate (MTX). (See "Alternatives to methotrexate for the initial treatment of
rheumatoid arthritis in adults" and "Initial treatment of rheumatoid arthritis in adults".)

The use of periodic structured assessments of disease activity is complementary to ongoing


regular monitoring of disease manifestations, disease progression, joint injury, disability, and
complications of the disease and to monitoring for adverse effects of medications. (See
'Assessment and monitoring' above.)

Multiple composite measures employing different combinations and weighting of these


variables have been developed for use in clinical research and practice. Among the more than
60 activity measures available for evaluation of patients with RA, the six measures noted
below have been identified by the American College of Rheumatology (ACR) as having the
greatest utility in clinical practice because they accurately reflect disease activity; are sensitive
to change; discriminate well between low, moderate, and high disease activity; have remission
criteria; and are feasible to perform in clinical settings [37]. The choice of measure is based
upon clinician preference; some measures require both patient and clinician input, while
others are based only upon patient-reported data. (See "Assessment of rheumatoid arthritis
activity in clinical trials and clinical practice", section on 'Composite indices for disease activity
assessment'.)

Measures that require both patient and clinician input, as well as calculators for these
measures, include the following:

Disease Activity Score derivative for 28 joints (DAS28) (calculator 1)

Simplified Disease Activity Index (SDAI) (calculator 2)

Clinical Disease Activity Index (CDAI) (calculator 3)

The patient-reported outcome measures include:

Routine Assessment of Patient Index Data 3 (RAPID3) [38]

Patient Activity Scale (PAS) [39]

PAS-II [39]

The RAPID3 correlates well with the results obtained by use of the DAS28 or CDAI [38]. (See
"Assessment of rheumatoid arthritis activity in clinical trials and clinical practice", section on
'RAPID3 score'.)
Symptoms and functional status The clinical assessment of disease activity should include
questions concerning the degree of joint pain, the duration of morning stiffness, and the
severity of fatigue [22,40]. In addition, evidence for and changes in extraarticular
manifestations of RA should be actively sought, including systemic signs such as fever,
anorexia, malaise, weight loss, and symptoms of cardiovascular disease. (See "Clinical
manifestations of rheumatoid arthritis".)

Fever is not a common feature of RA in adults. Infection must be excluded before ascribing
fever to RA.

Patients should be queried regarding their functional capacity, including the performance of
activities of daily living, of vocational activities, and of avocational activities, such as hobbies
or participation in sports. A self-report questionnaire that measures function can also be often
helpful for this purpose. The Stanford Health Assessment Questionnaire (HAQ) is one of the
best known and tested of these questionnaires [41,42]; others include the Functional
Independence Measure [43], the Arthritis Impact Measurement Scale (AIMS), the Short Form
36 (SF36), and the Modified Health Assessment Questionnaire (MHAQ) [44]. (See "Disease
outcome and functional capacity in rheumatoid arthritis", section on 'Functional capacity'.)

We use the evaluation of functional capacity to identify which of a variety of interventions


may be required in addition to pharmacologic therapy, especially in patients with diminished
functional capacity, such as counseling, exercise, and occupational therapy. (See
"Nonpharmacologic therapies and preventive measures for patients with rheumatoid
arthritis".)

Physical examination A physical examination should be performed at regular intervals that


vary with disease activity and severity. As an example, patients with severely active disease
may be seen at four-week intervals, while those with mildly active or well-controlled disease
could be seen every two to four months. At these visits, an examination should be performed
to assess changes in previously affected joints or the appearance of inflammation in
previously uninvolved joints.

A 28-joint examination is appropriate if the hands but not the feet are involved [45]. Examined
joints include the wrists, elbows, shoulders, and knees, as well as the metacarpophalangeal
and proximal interphalangeal joints of the hands. If the feet are involved, the
metatarsophalangeal joints and proximal interphalangeal joints of the feet should also be
assessed. The joints should be evaluated for the presence of swelling, tenderness, loss of
motion, and deformity.
In addition to the articular examination, a periodic general physical examination should be
performed, with particular attention to the skin for rheumatoid nodules or other dermal
manifestations of RA and to the lungs for signs of pleural or interstitial disease, to detect
evidence of systemic or extraarticular involvement. (See "Overview of the systemic and
nonarticular manifestations of rheumatoid arthritis".)

Laboratory monitoring of disease activity The acute phase reactants, such as C-reactive
protein (CRP) or erythrocyte sedimentation rate (ESR), are useful for assessment of disease
activity and are components of several of the formal composite measures used for evaluating
the level of disease activity. In addition to these studies, we obtain other tests primarily for
medication monitoring that may also reflect changes or levels of disease activity. Examples of
the latter include serum hemoglobin, decreases in which may reflect anemia of chronic
inflammation, and serum albumin, which may also be reduced in association with increased
disease activity. Additionally, platelet counts may be mildly elevated (typically up to 400,000
to 450,000/microL) in patients with ongoing inflammation. (See 'Pretreatment evaluation'
above and "Assessment of rheumatoid arthritis activity in clinical trials and clinical practice",
section on 'Acute phase reactants' and "Acute phase reactants" and "Biologic markers in the
diagnosis and assessment of rheumatoid arthritis" and "Anemia of chronic
disease/inflammation" and "Hematologic manifestations of rheumatoid arthritis".)

Monitoring and prevention of drug toxicity Because of the potential risks of serious adverse
effects and the frequency of common side effects of antirheumatic drugs, a careful balance
must be struck between the risks and potential benefits of these agents [46,47]. We generally
follow the recommendations of the ACR for drug monitoring in the treatment of RA (table 1)
[1,5,22,40,48]. Additional precautions, warnings, and the manufacturers recommendations
for clinical and laboratory monitoring are provided in the individual UpToDate drug
information topics for each medication. (See appropriate topic reviews.)

Monitoring for adverse effects, such as osteoporosis, diabetes, and hypertension, should be
performed in patients on glucocorticoids, and appropriate preventive measures should be
undertaken. RA is considered an independent risk factor for osteoporotic fracture, and a
fracture risk assessment should be performed to help guide treatment decisions. (See
"Prevention and treatment of glucocorticoid-induced osteoporosis" and "Osteoporotic
fracture risk assessment", section on 'Assessment of fracture risk' and "Major side effects of
systemic glucocorticoids" and "Antimalarial drugs in the treatment of rheumatic disease",
section on 'Monitoring for toxicity' and "Leflunomide in the treatment of rheumatoid
arthritis", section on 'Liver'.)

Imaging Early in the course of RA, it is appropriate to obtain plain radiographs of the hands
and wrists (one film, posteroanterior [PA] view), as well as at least one anteroposterior (AP)
view of both forefeet to include the metatarsophalangeal joints. These radiographs serve as a
baseline for evaluating change in the joints during treatment. Radiographs should be repeated
every two years in patients in remission or with low disease activity. We view the therapy in
use by the patient as insufficient if radiologic evidence of disease progression, such as
periarticular osteopenia, joint space narrowing, or bone erosions, appears or worsens during
this interval, despite good control of disease activity by other measures. In such patients,
based upon our clinical experience, we may intensify or modify the treatment regimen. The
clinician must be aware that, in hand radiographs of older patients, coexistent osteoarthritis
may account, in part, for joint space narrowing noted near the joints involved with RA [49].

Musculoskeletal ultrasonography (MSUS) and magnetic resonance imaging (MRI) are more
sensitive for the detection of cartilage and bone abnormalities. MSUS is increasingly utilized at
the point of care by rheumatologists for diagnosis and monitoring disease progression and
therapeutic response [27,50]. Likewise, MRI has become increasingly employed to
demonstrate subclinical synovitis, early erosive changes, and therapeutic response [27,51,52].
Some clinicians routinely employ these modalities to follow patients over time. The authors
prefer to use MSUS and MRI to answer specific clinical questions in an individual patient, but
not in the routine management of RA.

TIGHT CONTROL We recommend the use of tight control treatment strategies to quickly
minimize inflammation and disease progression; our therapeutic target is remission or a state
of minimal disease activity, without compromising safety. In patients resistant to initial
disease-modifying antirheumatic drug (DMARD) therapy, we either add additional DMARDs to
the ongoing regimen or switch the patient to a different DMARD, while also treating the active
inflammation with antiinflammatory drug therapy. A description of our treatment approach
and the evidence supporting use of particular medications in patients with active disease
despite initial DMARD therapy are discussed in detail separately. (See 'Drug therapy for flares'
below and "Treatment of rheumatoid arthritis in adults resistant to initial nonbiologic DMARD
therapy".)

In patients with disease exacerbations despite a preceding period of better control of disease
activity (a disease flare), a temporary increase in antiinflammatory therapies, including the
use of glucocorticoids, may be required. (See 'Drug therapy for flares' below.)

The use of strategies for tight control involves frequent periodic reassessment of disease
activity, usually at least every three months; adjustment of DMARD regimens every three to
six months, if needed, as the primary tool to achieve treatment goals; and administration of
antiinflammatory therapies (eg, nonsteroidal antiinflammatory drugs [NSAIDs] and oral and
intraarticular glucocorticoids) as an adjunct to DMARDs to maintain control of disease activity
until DMARD therapies are sufficiently effective to discontinue glucocorticoids or reduce their
use to an acceptably low level. Treatment protocols based upon this general approach are
associated with improved radiographic and functional outcomes compared with less
aggressive approaches [7,53-62].

The periodic reevaluation of disease activity using a quantitative composite measure and the
use of antiinflammatory drugs, including NSAIDs and glucocorticoids, as bridging therapies are
both important elements in the strategy of tight control. Glucocorticoids can rapidly achieve
control of inflammation until DMARDs are sufficiently effective. (See 'Assessment of disease
activity' above and "Assessment of rheumatoid arthritis activity in clinical trials and clinical
practice" and 'Adjunctive role of antiinflammatory agents' below.)

Taken together, studies that have compared tight control with less aggressive approaches
support the following observations:

Most patients should receive a DMARD as soon as possible after diagnosis. Achieving and
maintaining low disease activity using a DMARD or DMARD combinations as quickly as
possible improve long-term outcomes and are cost-effective compared with older, more
gradual approaches to initiating DMARD therapy. (See 'Early use of DMARDs' above.)

Excellent treatment responses can be achieved with a wide variety of nonbiologic and
biologic DMARDs and with regimens that combine either nonbiologic DMARDs alone or
nonbiologic DMARDs with a biologic agent, as described below.

Escalation in the treatment regimen is needed for patients resistant to initial treatment;
both intraarticular glucocorticoids and oral or intramuscular glucocorticoids help minimize
disease activity until DMARDs are sufficient. (See 'Adjunctive role of antiinflammatory agents'
below.)

Regular assessment with composite measures of disease activity is critical to optimize clinical
decision-making. (See 'Assessment of disease activity' above.)

These observations are reflected in the 2008, 2012, and 2015 American College of
Rheumatology (ACR) and 2010 European League Against Rheumatism (EULAR) treatment
recommendations and in the recommendations of an international task force that were
presented in 2010 and updated in 2014 [1-3,7,22,62-65].

The benefits of tight control have been shown in a meta-analysis of six heterogeneous trials
that evaluated tight control strategies in comparison with usual care for rheumatoid arthritis
(RA) [66]. Significantly greater improvement from baseline to one year in the Disease Activity
Score derivative for 28 joints (DAS28) composite measure of disease activity was seen in the
patients randomly allocated to tight control strategies compared with usual care (mean
difference in reduction in DAS28 of 0.59, 95% CI 0.22-0.97). A statistically significantly greater
reduction compared with usual care was observed in the trials in which tight control was
achieved with protocolized treatment adjustments compared with trials without such
protocols (mean difference in DAS28 of 0.91, 95% CI 0.72-1.11, versus 0.25, 95% CI 0.03-0.46).
Four of the six studies analyzed compared functional ability in the two treatment arms using
the Health Assessment Questionnaire (HAQ). Greater improvement in HAQ scores in the tight
control groups that were statistically significant were seen in two of these trials, while
improvements in the HAQ scores did not differ significantly between the treatment arms in
the other two trials. (See "Assessment of rheumatoid arthritis activity in clinical trials and
clinical practice", section on 'Health Assessment Questionnaire (HAQ)'.)

A number of randomized trials and related studies illustrate the range of medications and
approaches that provide evidence favoring a treat-to target approach [53-59,67-69].
Especially notable examples include the Behandel-Strategien voor Reumatoide Artritis (Dutch
for treatment strategies for rheumatoid arthritis or BeSt) trial [53,54]; the Finnish
Rheumatoid Arthritis Combination Therapy (FIN-RACo) trial [56-58,67]; the NEO-RACo trial
[68]; the Treatment of Early Aggressive Rheumatoid Arthritis (TEAR) trial [69]; and the Tight
Control of Rheumatoid Arthritis (TICORA) trial [59]. Taken together, these reports provide
strong support for the view that it is the treat-to-target strategy, more than the specific
agents used, that results in better outcomes for patients with RA [70].

As an example, in the TICORA trial, 111 patients were randomly assigned to intensive or
routine management [59]. Intensively managed patients in this trial had monthly visits, with
calculation of disease activity scores (a validated composite score based upon the erythrocyte
sedimentation rate [ESR], Ritchie articular index, joint swelling count, and patients global
assessment of disease activity defining high, moderate, and low disease activity levels of 3.6,
of 2.4 to <3.6, and of 1.6 to <2.4, respectively); glucocorticoid injections of swollen joints;
and, every three months, adjustment of their treatment regimen by a predefined protocol if
moderate or highly active disease was present. Routinely managed patients were seen every
three months, with no formal measurement of disease activity performed, and glucocorticoid
injections and other treatment adjustments were made at the discretion of the
rheumatologist. After 18 months, the patients receiving intensive management demonstrated
a significantly greater decrease in their disease activity scores compared with the routine
management group (-3.5 versus -1.9), and a higher proportion achieved a good response by
EULAR criteria (82 versus 44 percent). Additionally, physical function assessed by the HAQ was
improved to a statistically and clinically significantly greater degree in the patients receiving
intensive management (change in HAQ of -0.97 versus -0.47).
Adjunctive role of antiinflammatory agents We use antiinflammatory therapies, including
systemic and intraarticular glucocorticoids and NSAIDs, primarily as adjuncts for temporary
control of disease activity in patients in whom treatment is being started with DMARDs, in
patients in whom the DMARD regimen requires modification, or in patients who are
experiencing disease flares. Although NSAIDs act rapidly to control inflammation, they do not
provide adequate benefit on their own for longer-term control of disease or for prevention of
joint injury.

There is strong evidence that glucocorticoids retard radiographic progression in patients with
RA in the short to medium term (ie, up to two years of therapy) [71-73]. However, these
agents should not be used alone for an extended period. We avoid long-term use, if possible,
because chronic use for inflammatory disease is often associated with adverse effects [74].
However, patients with severe RA sometimes require sustained therapy with low doses of
glucocorticoids (less than 10 mg/day); such doses in RA are generally well-tolerated and may
have some benefit in retarding disease progression [73,75] (see "Use of glucocorticoids in the
treatment of rheumatoid arthritis", section on 'Efficacy of chronic use'). More detailed
discussions of NSAID and glucocorticoids in RA are presented elsewhere. (See "Use of
glucocorticoids in the treatment of rheumatoid arthritis" and "Initial treatment of rheumatoid
arthritis in adults", section on 'Symptomatic treatment with antiinflammatory drugs'.)

In patients who receive glucocorticoids, we taper the medication as rapidly as tolerated once
disease control is achieved and can be maintained, with the ideal goal of discontinuing
systemic glucocorticoid therapy.

Intraarticular injections of long-acting glucocorticoids are used to reduce synovitis in particular


joints that are more inflamed than others. Occasional patients benefit from intramuscular
rather than oral administration. (See "Intraarticular and soft tissue injections: What agent(s)
to inject and how frequently?".)

Drug therapy for flares RA has natural exacerbations (also known as flares) and reductions
of continuing disease activity. It is important to distinguish a disease flare, characterized by
symptoms and by physical and laboratory findings of increased inflammatory synovitis, from
noninflammatory causes of local or generalized increased pain. Patients with recurrent flares
may require adjustment in the background DMARD therapy. (See "Clinical manifestations of
rheumatoid arthritis" and 'Assessment of disease activity' above.)

The severity of the flare and background drug therapy influence the choice of therapies. The
following is a brief summary of glucocorticoid therapy, which is discussed in detail separately.
(See "Use of glucocorticoids in the treatment of rheumatoid arthritis".)
With respect to the severity of the flare:

In patients with a single or few affected joints, intraarticular glucocorticoid injections may be
effective and avoid the need for additional or prolonged systemic therapy.

More widespread flares may be treated by initiating glucocorticoid therapy or by increasing


the dose of oral glucocorticoid, with the intention of reducing the dose once the flare is under
control. The magnitude of dose increase varies with the baseline dose and with the severity of
the flare. An alternative to an increase in the oral dose is the administration of a single deep
intramuscular injection of methylprednisolone acetate (120 mg in 3 mL) or triamcinolone
acetonide (60 mg in 1.5 mL).

Pulse intravenous methylprednisolone therapy, usually consisting of three daily infusions of


up to 1000 mg, is generally limited to severe flares, particularly those associated with systemic
manifestations, such as rheumatoid vasculitis.

With respect to background drug therapy, an escalation in dose or a modification in drugs is


warranted if the patient is flaring frequently or severely. The strategy depends upon the
background DMARDs being used. As examples:

Patients on methotrexate (MTX) who will tolerate a slower resolution of their flare may
respond to optimization of the MTX dose by either an increase in the dose of MTX or
switching from oral to subcutaneous therapy [76]. (See "Use of methotrexate in the treatment
of rheumatoid arthritis", section on 'Dosing of MTX'.)

Patients initially controlled with a regimen that includes infliximab may benefit from a
decrease in the interval between infliximab doses or from higher doses [77,78]. However,
increasing the dose from 3 to 5 mg/kg was not beneficial in one well-designed trial [79,80].

Increases in doses of etanercept (greater than 50 mg weekly) or adalimumab (weekly rather


than every two weeks), with or without MTX, do not appear to increase efficacy [81,82].

Patients who require multiple treatment courses with glucocorticoids for recurrent disease
flares and whose medication doses have been increased to the maximally tolerated or
acceptable level should be treated as patients with sustained disease activity. (See "Treatment
of rheumatoid arthritis in adults resistant to initial nonbiologic DMARD therapy" and
"Treatment of rheumatoid arthritis in adults resistant to initial biologic DMARD therapy".)

OTHER CONSIDERATIONS IN RA MANAGEMENT The focus of therapeutic decision-making is


control of disease activity, as described above, but additional factors, such as the degree of
joint injury or disability, may influence the choice of specific therapies in individual patients
[2,8]. Additionally, the efficacy of particular medications may be affected by the presence or
absence of some of these factors, which are associated with an adverse prognosis. (See
'Prognosis' below.)

The relative importance of these factors depends upon the individual treatment choice; these
are discussed in more detail in the appropriate individual treatment topics. We consider the
following factors, depending upon the specific treatment decision:

Disability and function General scales that measure disability may not identify specific
limitations of greater impact on an individual patient. As an example, specific vocational
requirements, family responsibilities, or recreational interests may affect a patients
willingness to accept the risks of a given intervention that would help to achieve a greater
degree of disease control than low disease activity. We therefore incorporate patient-specific
needs in our assessment of the severity of disease-related disability.

Joint injury Good control of disease activity may not result in complete elimination of
progressive joint injury in all patients. In patients with low disease activity but with worsening
findings on imaging studies, either changes in medications or increased dosing may be of
benefit. However, there is insufficient evidence to determine whether treating to targets that
are based upon imaging findings provides additional benefit for long-term outcomes,
compared with targets based upon measures of disease activity alone.

Comorbidities The presence of comorbidities, such as renal or hepatic disease, may affect
medication choices and may influence the degree of risk inherent in attempting to reach a
goal of remission or of low disease activity in a given patient.

Comorbidities A number of medical conditions that often coexist with or result from
rheumatoid arthritis (RA) may influence the choice of medications; our approach is consistent
with expert opinion, including the American College of Rheumatology (ACR) treatment
guidelines [1,5,22,83].

Infection

Serious infection In patients with an active serious infection, conventional and biologic
disease-modifying antirheumatic drugs (DMARDs) and tofacitinib should be temporarily held
until resolution of infection and completion of antimicrobial therapy. In patients with a history
of a serious infection, we recommend conventional DMARDs over biologic agents.
Medications administered more frequently are preferred in patients in whom there is
heightened concern regarding infection or with recurrent infections because of the relative
greater ease of discontinuing the therapy and its immunomodulatory effect if needed.

Hepatitis B In patients with natural immunity to hepatitis B virus (HBV; HBV core antibody
[HBc]-positive, normal liver chemistries, HBV surface antibody [HBs]-positive, and HBV surface
antigen [HBsAg]-negative), treatment for RA should be the same as for HBV-unexposed RA
patients, but viral loads should be monitored every 6 to 12 months to ensure there is no
reactivation. For patients with active untreated hepatitis, referral for antiviral therapy should
be obtained before immunosuppressive therapy, and patients should be treated in
collaboration with their hepatologist. In the absence of additional harms, RA treatment may
proceed for patients with active HBV on concomitant antiretroviral treatment. (See "Diagnosis
of hepatitis B virus infection" and "Overview of the management of hepatitis B and case
examples".)

Hepatitis C Patients with active hepatitis C virus (HCV) and normal liver function should not
be treated differently than RA patients without HCV. If underlying liver disease is present,
non-hepatotoxic DMARDs (sulfasalazine [SSZ] or hydroxychloroquine [HCQ]) are preferred
initially. Patients with HCV infection should be managed in collaboration with their
hepatologist. (See "Overview of the management of chronic hepatitis C virus infection".)

Tuberculosis Prior to initiation of immunomodulatory therapy, patients with risk factors


for tuberculosis (TB) should be screened for latent TB and treated if indicated (see
'Pretreatment evaluation' above). In patients in whom latent TB is diagnosed, at least one
month of treatment should be completed prior to the initiation of immunosuppressive agents.

Malignancy Management of RA in patients with malignancy or a history of malignancy is


based upon findings from observational studies involving relatively small numbers of patients
with typically imprecise results, together with expert opinion [5,84-88].

Non-melanoma skin cancer (basal cell and squamous cell carcinoma) In patients with a
history of non-melanoma skin cancer, we use conventional DMARDs over biologic DMARDs or
tofacitinib. There is no contraindication to escalation of therapy to include biologics, but
routine skin cancer surveillance is indicated. (See "Epidemiology and clinical features of basal
cell carcinoma" and "Clinical features and diagnosis of cutaneous squamous cell carcinoma
(SCC)".)

Melanoma skin cancer In patients with a history of melanoma, we use conventional


DMARDs over biologic DMARDs or tofacitinib. If a biologic agent is needed, the first choice
would be rituximab, and we would avoid use of abatacept. Routine skin cancer surveillance is
indicated. (See "Screening and early detection of melanoma".)
History of lymphoproliferative disorder In patients with a history of a lymphoproliferative
disorder, we prefer conventional DMARDs, and if a biologic agent is needed, the first choice
would be rituximab, given its use in the treatment of lymphoproliferative disorders and a lack
of evidence for increased cancer risk with its use.

Solid organ malignancy In patients with a treated solid organ malignancy within the past
five years, we use conventional DMARDs over biologic DMARDs. If a biologic agent is needed,
the first choice would be rituximab, given the lack of evidence for increased cancer risk with
its use.

In patients who are more than five years out from a treated solid organ malignancy, excluding
melanoma, RA treatment is no different than from those without malignancy.

Pregnancy RA often improves or remits completely during pregnancy. Issues related to the
pregnant woman with RA, including the use of immunosuppressive drugs, are discussed
separately. (See "Rheumatoid arthritis and pregnancy" and "Use of antiinflammatory and
immunosuppressive drugs in rheumatic diseases during pregnancy and lactation".)

Lung disease Comorbid pulmonary disease is common in patients with RA and may also be
a complication of therapy or of the disease [83]. Therapeutic agents with potential for causing
adverse pulmonary effects include methotrexate (MTX), leflunomide (LEF), tumor necrosis
factor (TNF) inhibitors, SSZ, parenteral gold, abatacept, and rituximab. (See "Overview of lung
disease associated with rheumatoid arthritis".)

Cardiovascular disease Comorbid cardiovascular disease can occur in patients with RA and
may also be a complication of therapy [83]. Both glucocorticoids and nonsteroidal
antiinflammatory drugs (NSAIDs) may increase cardiovascular risk. Active RA is associated with
an increased risk of cardiovascular disease, but good control of disease activity has been
associated with reduced cardiovascular complications. (See "Coronary artery disease in
rheumatoid arthritis: Pathogenesis, risk factors, clinical manifestations, and diagnostic
implications" and "Coronary artery disease in rheumatoid arthritis: Implications for prevention
and management" and "Major side effects of systemic glucocorticoids", section on
'Cardiovascular disease' and "Nonselective NSAIDs: Overview of adverse effects", section on
'Cardiovascular effects'.)

TNF inhibitors should be avoided in patients with moderate or severe heart failure (HF), as
they can worsen HF. Such patients should be treated instead with traditional nonbiologic
DMARDs, non-TNF inhibitor biologics, or tofacitinib. (See "Tumor necrosis factor-alpha
inhibitors: An overview of adverse effects", section on 'Heart failure'.)
Neurologic manifestations Neurologic manifestations of RA and the presence of coexistent
neurologic disease are generally uncommon, other than the occurrence of impingement
neuropathies such as carpal tunnel syndrome. However, TNF inhibitors should be avoided in
those with a history of or an ongoing demyelinating disorder because of case reports of such
disorders in patients being treated for RA and because of increased risk of disease worsening
in trials of TNF blockade in patients with multiple sclerosis (MS). Some RA experts are also
cautious about using TNF-alpha inhibitors in patients with family histories of MS [83]. (See
"Neurologic manifestations of rheumatoid arthritis" and "Tumor necrosis factor-alpha
inhibitors: An overview of adverse effects", section on 'Demyelinating disease'.)

Diabetes The risk of diabetes mellitus is not increased in patients with RA. However, in
patients with both diabetes and RA, glucocorticoids should be used with particular caution
because they may worsen control of the diabetes [83]. By contrast, patients being treated
with HCQ or with TNF inhibitors for RA have a lower risk of diabetes [89], and SSZ may have
glucose-lowering effects [90]. (See "Major side effects of systemic glucocorticoids", section on
'Glucose metabolism' and "Antimalarial drugs in the treatment of rheumatic disease", section
on 'Reduction of diabetes risk'.)

Renal disease RA infrequently affects the kidney, but, if renal disease coexists, it increases
mortality risk [83]. In addition to NSAIDs, the use of some medications occasionally or only
historically used in the treatment of patients with RA may adversely affect renal function,
including gold, penicillamine, and cyclosporine. Some nonbiologic DMARDs, particularly MTX
and cyclosporine, should be avoided or used with particular caution in patients with
significantly decreased renal function. (See "NSAIDs: Acute kidney injury (acute renal failure)"
and "Causes and diagnosis of membranous nephropathy", section on 'Drugs, particularly
penicillamine and gold' and "Cyclosporine and tacrolimus nephrotoxicity".)

Extraarticular disease RA has many extraarticular manifestations. The treatment of these


specific features, such as vasculitis, interstitial lung disease, and others, is reviewed in detail
elsewhere. (See "Overview of the systemic and nonarticular manifestations of rheumatoid
arthritis" and "Treatment of rheumatoid vasculitis".)

Use of analgesics Drugs that primarily or only provide analgesia, including topical
medications (eg, capsaicin) and oral agents, such as acetaminophen (paracetamol), tramadol,
and more potent opioids (eg, oxycodone, hydrocodone), have a limited role in most patients
with active disease but may be helpful in patients with end-stage disease and, occasionally, in
patients with more severe involvement or during disease flares for added temporary benefit.
These medications should not be used as the sole or primary therapy in patients with active
inflammatory disease. An additional concern regarding opioid analgesics is an increased risk of
hospitalization for serious infection, which has been associated with their use by patients with
RA and may be due to impairment of certain immune functions by these agents [91].

Apparent need for additional analgesic medications when inflammatory disease is well-
controlled (other than acetaminophen or occasional NSAIDs) should prompt a search for
alternative comorbid diagnoses, such as fibromyalgia, to explain the patients symptoms. (See
"Clinical manifestations and diagnosis of fibromyalgia in adults".)

THERAPY OF END-STAGE DISEASE Despite therapeutic intervention, some patients progress


to disabling, destructive joint disease. Symptoms in such patients may be present in the
absence of active inflammatory joint disease and may be due to the secondary degenerative
changes alone. The accurate evaluation of such patients is essential, since deterioration
associated with mechanical problems of the muscle or joint is treated much differently from
ongoing inflammation or systemic manifestations of rheumatoid arthritis (RA). Disease
exacerbations and their systemic effects are usually easily recognized by the presence of many
inflamed joints, fever, anemia, an elevated erythrocyte sedimentation rate (ESR), or an
elevated serum C-reactive protein (CRP) concentration.

The goals of therapy in the patient with end-stage disease are:

Pain relief

Protection of remaining articular structures

Maintenance of function

Relief from fatigue and weakness

In the absence of inflammation, which requires antiinflammatory medications and disease-


modifying antirheumatic drugs (DMARDs), treatments other than medications may be
particularly important in management. These include nonpharmacologic interventions, such
as physical and occupational therapy and the use of adaptive devices, and surgery. (See
"Evaluation and medical management of end-stage rheumatoid arthritis".)

The indications for surgical intervention in patients with RA include intractable pain or severe
functional disability due to joint destruction, as well as impending tendon rupture. The timing
of surgery is often critical. If one waits too long, there may be so much muscle atrophy from
disuse that postoperative rehabilitation is unsuccessful. On the other hand, a decision about
joint replacement should take into account the average life of the artificial joint. (See "Total
joint replacement for severe rheumatoid arthritis".)
RECOMMENDATIONS BY MAJOR GROUPS Recommendations for the management of
patients with rheumatoid arthritis (RA) have been developed by major professional
organizations, including the American College of Rheumatology (ACR) and the European
League Against Rheumatism (EULAR) [1,3,4,22,65]. In addition, an international task force has
developed recommendations for treating RA to targeted goals [2,7,62,64]. Our approach to
treatment is generally consistent with these recommendations. The 2015 ACR
recommendations and supporting documents can be accessed online.

The EULAR recommendations for the management of RA represent a European consensus on


the management of RA with disease-modifying antirheumatic drugs (DMARDs) and
glucocorticoids and on the strategies to reach optimal outcomes, based upon evidence and
expert opinion [3,4]. The evidence used was detailed in a series of systematic literature
reviews, and the recommendations were updated in 2013 [4,6,92-95]. The 2013
recommendations include three overarching principles for the care of patients with RA and a
set of 14 recommendations covering major issues in disease management. The 2013 EULAR
recommendations can be accessed online.

PROGNOSIS Rheumatoid arthritis (RA) was associated with a high degree of economic loss,
morbidity, and early mortality prior to the widespread use of methotrexate (MTX) that began
in the 1980s, more aggressive treatment of early disease, and the availability of targeted
biologic agents since the later part of the 1990s. As an example, almost 20 percent of patients
in one center were severely disabled after 20 years of follow-up (from 1967 to 1987); an
additional one-third had died [96]. Patients with RA that required hospital care had at least a
twofold increased mortality when compared with those without disease [97], and more
severe RA was associated with higher mortality rates due to higher rates of myocardial
infarction, infection, and certain malignancies, comparable to three-vessel coronary artery
disease or to stage IV Hodgkin lymphoma [98].

Clinical outcomes have improved significantly with changes in drug therapy and in the
approach to treatment. These improvements have decreased the need for joint surgery and
reduced disability in patients with RA. As an example, in a study involving data from 57
hospitals in Ireland, the number of total hip and knee joint arthroplasties in patients with RA
decreased from 1995 to 2010, despite substantial increases in these procedures among the
general population [99].

An analysis of 1007 patients with RA diagnosed between 1993 and 2011 in the Leiden Early
Arthritis Clinic found that more intensive treatment strategies adopted over the years of study
increased the chances for DMARD-free sustained remissions [100]. Moreover, those patients
achieving DMARD-free remission had normal functional status when compared with the
general population. The average level of disability in RA was found in a longitudinal study of
over 3000 patients to have declined by about 40 percent from 1977 to 1998, even prior to the
introduction of the biologic DMARDs [101]. Similarly, patients with RA seen in a single
university clinic from 1984 through 1986 had significantly more disability and greater
radiographic evidence of joint injury compared with a cohort from the same clinic seen from
1999 through 2001 (modified Health Assessment Questionnaire [HAQ] disability score on a 0
to 3 scale of 1 versus 0.4, respectively, and Larsen radiographic score on a 0 to 100 scale of 20
versus 3, respectively) [13]. Patients able to achieve remission with combination therapy had
significantly less work disability over five years of follow-up compared with patients with
incomplete responses to treatment [102]. (See "Disease outcome and functional capacity in
rheumatoid arthritis".)

A number of factors have been associated with poorer outcomes in patients with RA. The
following four markers of adverse prognosis can be used to identify patients who may require
more aggressive pharmacotherapy, especially in early stages of disease [22]:

Functional limitation

Extraarticular disease

Rheumatoid factor positivity or presence of anticyclic citrullinated peptide (CCP) antibodies

Bony erosions documented radiographically

Other factors associated with a worse prognosis include concurrent medical disorders,
cigarette smoking, lack of formal education, and lower socioeconomic status [103]. Older age,
female sex, and the presence of the shared epitope have also been associated with a poorer
prognosis [22]. Some studies have derived models to estimate prognosis, such as persistent
erosive disease [104]. However, these models have not been validated in other cohorts. The
individual factors associated with a poor prognosis are discussed in detail separately. (See
"Epidemiology of, risk factors for, and possible causes of rheumatoid arthritis" and "Disease
outcome and functional capacity in rheumatoid arthritis" and "Biologic markers in the
diagnosis and assessment of rheumatoid arthritis" and "Overview of the systemic and
nonarticular manifestations of rheumatoid arthritis" and "HLA and other susceptibility genes
in rheumatoid arthritis".)

INFORMATION FOR PATIENTS UpToDate offers two types of patient education materials,
The Basics and Beyond the Basics. The Basics patient education pieces are written in plain
language, at the 5thto 6th grade reading level, and they answer the four or five key questions a
patient might have about a given condition. These articles are best for patients who want a
general overview and who prefer short, easy-to-read materials. Beyond the Basics patient
education pieces are longer, more sophisticated, and more detailed. These articles are written
at the 10th to 12th grade reading level and are best for patients who want in-depth information
and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to
print or e-mail these topics to your patients. (You can also locate patient education articles on
a variety of subjects by searching on patient info and the keyword(s) of interest.)

Beyond the Basics topics (see "Patient information: Rheumatoid arthritis symptoms and
diagnosis (Beyond the Basics)" and "Patient information: Rheumatoid arthritis treatment
(Beyond the Basics)" and "Patient information: Disease-modifying antirheumatic drugs
(DMARDs) (Beyond the Basics)" and "Patient information: Complementary and alternative
therapies for rheumatoid arthritis (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

In patients with rheumatoid arthritis (RA), affected areas may be irreversibly damaged or
destroyed if inflammation persists. Thus, prompt diagnosis, early recognition of active disease,
and measures to quickly achieve and maintain control of inflammation and the underlying
disease process, with the goal of remission or low disease activity, are central to modifying
disease outcome. The application of these principles in the management of patients with RA,
together with the development and use of newer and more potent drugs, has resulted in
significant improvement in the outcomes of treatment. (See 'General principles' above and
'Early recognition and diagnosis' above.)

An expert in the care of rheumatic disease, such as a rheumatologist, should participate in


the care of patients suspected of having RA and in the ongoing care of patients diagnosed with
this condition. The treatment of patients with RA by a rheumatologist is associated with better
disease outcomes compared with care rendered primarily by other clinicians. (See 'Care by a
rheumatologist' above.)

Nonpharmacologic measures, such as patient education, psychosocial interventions, and


physical and occupational therapy, should be used in addition to drug therapy. Other medical
interventions that are important in the comprehensive management of RA in all stages of
disease include cardiovascular risk reduction and immunizations to decrease the risk of
complications of drug therapies. (See 'Nonpharmacologic and preventive therapies' above.)

We recommend that all patients diagnosed with RA be started on disease-modifying


antirheumatic drug (DMARD) therapy as soon as possible following diagnosis, rather than
using antiinflammatory drugs alone, such as nonsteroidal antiinflammatory drugs (NSAIDs)
and glucocorticoids (Grade 1B). Better outcomes are achieved by early compared with
delayed intervention with DMARDs. (See 'Early use of DMARDs' above.)

We recommend the use of tight control treatment strategies to guide adjustments in the
treatment regimen, rather than less aggressive approaches (Grade 1B). Tight control involves
reassessment of disease activity on a regularly planned basis with the use of quantitative
composite measures and adjustment of treatment regimens to quickly achieve and maintain
control of disease activity if targeted treatment goals (remission or low disease activity),
rather than an undefined degree of improvement, have not been achieved. Such tight control
treatment strategies are associated with improved radiographic and functional outcomes
compared with less aggressive approaches. (See 'Tight control' above and 'Assessment of
disease activity' above.)

Laboratory testing prior to therapy should include a complete blood count, erythrocyte
sedimentation rate (ESR), C-reactive protein (CRP), aminotransferases, blood urea nitrogen,
and creatinine. Patients receiving hydroxychloroquine (HCQ) should have a baseline
ophthalmologic examination, and most patients who will receive a biologic agent should be
tested for latent tuberculosis (TB) infection. Screening for hepatitis B and C should be
performed in all patients. Some patients may require antiviral treatment prior to initiating
DMARD or immunosuppressive therapy, depending upon their level of risk for hepatitis B virus
(HBV) reactivation. (See 'Pretreatment evaluation' above.)

We use antiinflammatory drugs, including NSAIDs and glucocorticoids, as bridging therapies


to rapidly achieve control of inflammation until DMARDs are sufficiently effective. Some
patients may benefit from longer-term therapy with low doses of glucocorticoids. (See
'Adjunctive role of antiinflammatory agents' above.)

RA has natural exacerbations (also known as flares) and reductions of continuing disease
activity. The severity of the flare and background drug therapy influence the choice of
therapies. Patients who require multiple treatment courses with glucocorticoids for recurrent
disease flares and whose medication doses have been increased to the maximally tolerated or
acceptable level should be treated as patients with sustained disease activity. Such patients
require modifications of their baseline drug therapies. (See 'Drug therapy for flares' above.)

The monitoring that we perform on a regular basis includes testing that is specific to
evaluation of the safety of the drugs being used; periodic assessments of disease activity with
composite measures; monitoring for extraarticular manifestations of RA, other disease
complications, and joint injury; and functional assessment. (See 'Assessment and monitoring'
above.)
Other factors in RA management that may influence the target or choice of therapy include
the disabilities or functional limitations important to a given patient, progressive joint injury,
comorbidities, and the presence of adverse prognostic factors. (See 'Other considerations in
RA management' above and 'Prognosis' above.)

ACKNOWLEDGMENT The editorial staff at UpToDate would like to acknowledge Peter


Schur, MD, who contributed to an earlier version of this topic review.

Use of UpToDate is subject to the Subscription and License Agreement.


REFERENCES

Singh JA, Furst DE, Bharat A, et al. 2012 update of the 2008 American College of
Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and
1
biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken)
2012; 64:625.
Smolen JS, Aletaha D, Bijlsma JW, et al. Treating rheumatoid arthritis to target:
2
recommendations of an international task force. Ann Rheum Dis 2010; 69:631.
Smolen JS, Landew R, Breedveld FC, et al. EULAR recommendations for the management
3 of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs.
Ann Rheum Dis 2010; 69:964.
Smolen JS, Landew R, Breedveld FC, et al. EULAR recommendations for the management
4 of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic
drugs: 2013 update. Ann Rheum Dis 2014; 73:492.
Singh JA, Saag KG, Bridges SL Jr, et al. 2015 American College of Rheumatology Guideline
5
for the Treatment of Rheumatoid Arthritis. Arthritis Care Res (Hoboken) 2016; 68:1.
Knevel R, Schoels M, Huizinga TW, et al. Current evidence for a strategic approach to the
management of rheumatoid arthritis with disease-modifying antirheumatic drugs: a
6
systematic literature review informing the EULAR recommendations for the management of
rheumatoid arthritis. Ann Rheum Dis 2010; 69:987.
Schoels M, Knevel R, Aletaha D, et al. Evidence for treating rheumatoid arthritis to target:
7
results of a systematic literature search. Ann Rheum Dis 2010; 69:638.
McInnes IB, O'Dell JR. State-of-the-art: rheumatoid arthritis. Ann Rheum Dis 2010;
8
69:1898.
Sokka T, Pincus T. Rheumatoid arthritis: strategy more important than agent. Lancet 2009;
9
374:430.
10 Klareskog L, Catrina AI, Paget S. Rheumatoid arthritis. Lancet 2009; 373:659.
11 Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet 2010; 376:1094.
Lard LR, Visser H, Speyer I, et al. Early versus delayed treatment in patients with recent-
12 onset rheumatoid arthritis: comparison of two cohorts who received different treatment
strategies. Am J Med 2001; 111:446.
Pincus T, Sokka T, Kautiainen H. Patients seen for standard rheumatoid arthritis care have
13 significantly better articular, radiographic, laboratory, and functional status in 2000 than in
1985. Arthritis Rheum 2005; 52:1009.
Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis classification criteria: an
14 American College of Rheumatology/European League Against Rheumatism collaborative
initiative. Arthritis Rheum 2010; 62:2569.
15 Huizinga TW, Pincus T. In the clinic. Rheumatoid arthritis. Ann Intern Med 2010; 153:ITC1.
Ward MM, Leigh JP, Fries JF. Progression of functional disability in patients with rheumatoid
16 arthritis. Associations with rheumatology subspecialty care. Arch Intern Med 1993;
153:2229.
Solomon DH, Bates DW, Panush RS, Katz JN. Costs, outcomes, and patient satisfaction by
17 provider type for patients with rheumatic and musculoskeletal conditions: a critical review
of the literature and proposed methodologic standards. Ann Intern Med 1997; 127:52.
Criswell LA, Such CL, Yelin EH. Differences in the use of second-line agents and prednisone
18 for treatment of rheumatoid arthritis by rheumatologists and non-rheumatologists. J
Rheumatol 1997; 24:2283.
Rat AC, Henegariu V, Boissier MC. Do primary care physicians have a place in the
19
management of rheumatoid arthritis? Joint Bone Spine 2004; 71:190.
Lacaille D, Anis AH, Guh DP, Esdaile JM. Gaps in care for rheumatoid arthritis: a population
20
study. Arthritis Rheum 2005; 53:241.
van der Linden MP, le Cessie S, Raza K, et al. Long-term impact of delay in assessment of
21
patients with early arthritis. Arthritis Rheum 2010; 62:3537.
Saag KG, Teng GG, Patkar NM, et al. American College of Rheumatology 2008
22 recommendations for the use of nonbiologic and biologic disease-modifying antirheumatic
drugs in rheumatoid arthritis. Arthritis Rheum 2008; 59:762.
Fleischmann R, Kremer J, Cush J, et al. Placebo-controlled trial of tofacitinib monotherapy
23
in rheumatoid arthritis. N Engl J Med 2012; 367:495.
van Vollenhoven RF, Fleischmann R, Cohen S, et al. Tofacitinib or adalimumab versus
24
placebo in rheumatoid arthritis. N Engl J Med 2012; 367:508.
van der Heijde DM, van Leeuwen MA, van Riel PL, et al. Biannual radiographic assessments
25 of hands and feet in a three-year prospective followup of patients with early rheumatoid
arthritis. Arthritis Rheum 1992; 35:26.
Fuchs HA, Kaye JJ, Callahan LF, et al. Evidence of significant radiographic damage in
26
rheumatoid arthritis within the first 2 years of disease. J Rheumatol 1989; 16:585.
Wakefield RJ, Gibbon WW, Conaghan PG, et al. The value of sonography in the detection of
27 bone erosions in patients with rheumatoid arthritis: a comparison with conventional
radiography. Arthritis Rheum 2000; 43:2762.
Weidekamm C, Kller M, Weber M, Kainberger F. Diagnostic value of high-resolution B-
28 mode and doppler sonography for imaging of hand and finger joints in rheumatoid arthritis.
Arthritis Rheum 2003; 48:325.
29 McQueen FM. The use of MRI in early RA. Rheumatology (Oxford) 2008; 47:1597.
Anderson JJ, Wells G, Verhoeven AC, Felson DT. Factors predicting response to treatment
30
in rheumatoid arthritis: the importance of disease duration. Arthritis Rheum 2000; 43:22.
van der Heide A, Jacobs JW, Bijlsma JW, et al. The effectiveness of early treatment with
31 "second-line" antirheumatic drugs. A randomized, controlled trial. Ann Intern Med 1996;
124:699.
Nell VP, Machold KP, Eberl G, et al. Benefit of very early referral and very early therapy
32 with disease-modifying anti-rheumatic drugs in patients with early rheumatoid arthritis.
Rheumatology (Oxford) 2004; 43:906.
Sokka T, Mkinen H. Drug management of early rheumatoid arthritis - 2008. Best Pract
33
Res Clin Rheumatol 2009; 23:93.
Emery P, Kvien TK, Combe B, et al. Combination etanercept and methotrexate provides
better disease control in very early (<=4 months) versus early rheumatoid arthritis (>4
34
months and <2 years): post hoc analyses from the COMET study. Ann Rheum Dis 2012;
71:989.
Molina E, Del Rincon I, Restrepo JF, et al. Association of socioeconomic status with
35 treatment delays, disease activity, joint damage, and disability in rheumatoid arthritis.
Arthritis Care Res (Hoboken) 2015; 67:940.
van Nies JA, Tsonaka R, Gaujoux-Viala C, et al. Evaluating relationships between symptom
duration and persistence of rheumatoid arthritis: does a window of opportunity exist?
36
Results on the Leiden early arthritis clinic and ESPOIR cohorts. Ann Rheum Dis 2015;
74:806.
Anderson J, Caplan L, Yazdany J, et al. Rheumatoid arthritis disease activity measures:
37 American College of Rheumatology recommendations for use in clinical practice. Arthritis
Care Res (Hoboken) 2012; 64:640.
Pincus T, Swearingen CJ, Bergman MJ, et al. RAPID3 (Routine Assessment of Patient Index
Data) on an MDHAQ (Multidimensional Health Assessment Questionnaire): agreement with
38 DAS28 (Disease Activity Score) and CDAI (Clinical Disease Activity Index) activity
categories, scored in five versus more than ninety seconds. Arthritis Care Res (Hoboken)
2010; 62:181.
Wolfe F, Michaud K, Pincus T. A composite disease activity scale for clinical practice,
39 observational studies, and clinical trials: the patient activity scale (PAS/PAS-II). J
Rheumatol 2005; 32:2410.
American College of Rheumatology Subcommittee on Rheumatoid Arthritis Guidelines.
40 Guidelines for the management of rheumatoid arthritis: 2002 Update. Arthritis Rheum
2002; 46:328.
Fries JF, Spitz PW, Young DY. The dimensions of health outcomes: the health assessment
41
questionnaire, disability and pain scales. J Rheumatol 1982; 9:789.
Fries JF, Spitz P, Kraines RG, Holman HR. Measurement of patient outcome in arthritis.
42
Arthritis Rheum 1980; 23:137.
McDowell I, Newell C. health: A guide to rating scales and questionnaires, 2nd, Oxford
43
University Press, New York 1996. p.106.
Wolfe F. Which HAQ is best? A comparison of the HAQ, MHAQ and RA-HAQ, a difficult 8
44 item HAQ (DHAQ), and a rescored 20 item HAQ (HAQ20): analyses in 2,491 rheumatoid
arthritis patients following leflunomide initiation. J Rheumatol 2001; 28:982.
Smolen JS, Breedveld FC, Schiff MH, et al. A simplified disease activity index for
45
rheumatoid arthritis for use in clinical practice. Rheumatology (Oxford) 2003; 42:244.
O'Dell JR. Combination DMARD therapy for rheumatoid arthritis: a step closer to the goal.
46
Ann Rheum Dis 1996; 55:781.
Felson DT, Anderson JJ, Meenan RF. Use of short-term efficacy/toxicity tradeoffs to select
47 second-line drugs in rheumatoid arthritis. A metaanalysis of published clinical trials.
Arthritis Rheum 1992; 35:1117.
Guidelines for monitoring drug therapy in rheumatoid arthritis. American College of
48
Rheumatology Ad Hoc Committee on Clinical Guidelines. Arthritis Rheum 1996; 39:723.
Khanna D, Ranganath VK, Fitzgerald J, et al. Increased radiographic damage scores at the
onset of seropositive rheumatoid arthritis in older patients are associated with
49
osteoarthritis of the hands, but not with more rapid progression of damage. Arthritis
Rheum 2005; 52:2284.
Ruta S, Reginato AM, Pineda C, et al. General applications of ultrasound in rheumatology:
50
why we need it in our daily practice. J Clin Rheumatol 2015; 21:133.
McGonagle D, Conaghan PG, O'Connor P, et al. The relationship between synovitis and
51 bone changes in early untreated rheumatoid arthritis: a controlled magnetic resonance
imaging study. Arthritis Rheum 1999; 42:1706.
stergaard M, Mller-Bisgaard S. Optimal use of MRI in clinical trials, clinical care and
52
clinical registries of patients with rheumatoid arthritis. Clin Exp Rheumatol 2014; 32:S.
Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, et al. Clinical and radiographic
53 outcomes of four different treatment strategies in patients with early rheumatoid arthritis
(the BeSt study): a randomized, controlled trial. Arthritis Rheum 2005; 52:3381.
Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, et al. Comparison of treatment
54
strategies in early rheumatoid arthritis: a randomized trial. Ann Intern Med 2007; 146:406.
de Vries-Bouwstra JK, Goekoop-Ruiterman YP, Verpoort KN, et al. Progression of joint
damage in early rheumatoid arthritis: association with HLA-DRB1, rheumatoid factor, and
55
anti-citrullinated protein antibodies in relation to different treatment strategies. Arthritis
Rheum 2008; 58:1293.
Mttnen T, Hannonen P, Leirisalo-Repo M, et al. Comparison of combination therapy with
56 single-drug therapy in early rheumatoid arthritis: a randomised trial. FIN-RACo trial group.
Lancet 1999; 353:1568.
Puolakka K, Kautiainen H, Mttnen T, et al. Impact of initial aggressive drug treatment
with a combination of disease-modifying antirheumatic drugs on the development of work
57
disability in early rheumatoid arthritis: a five-year randomized followup trial. Arthritis
Rheum 2004; 50:55.
Rantalaiho V, Korpela M, Hannonen P, et al. The good initial response to therapy with a
combination of traditional disease-modifying antirheumatic drugs is sustained over time:
58
the eleven-year results of the Finnish rheumatoid arthritis combination therapy trial.
Arthritis Rheum 2009; 60:1222.
Grigor C, Capell H, Stirling A, et al. Effect of a treatment strategy of tight control for
59 rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet
2004; 364:263.
van der Kooij SM, Goekoop-Ruiterman YP, de Vries-Bouwstra JK, et al. Drug-free
60 remission, functioning and radiographic damage after 4 years of response-driven treatment
in patients with recent-onset rheumatoid arthritis. Ann Rheum Dis 2009; 68:914.
Finckh A, Bansback N, Marra CA, et al. Treatment of very early rheumatoid arthritis with
61 symptomatic therapy, disease-modifying antirheumatic drugs, or biologic agents: a cost-
effectiveness analysis. Ann Intern Med 2009; 151:612.
Stoffer MA, Schoels MM, Smolen JS, et al. Evidence for treating rheumatoid arthritis to
62
target: results of a systematic literature search update. Ann Rheum Dis 2016; 75:16.
Bathon JM, Cohen SB. The 2008 American College of Rheumatology recommendations for
63 the use of nonbiologic and biologic disease-modifying antirheumatic drugs in rheumatoid
arthritis: where the rubber meets the road. Arthritis Rheum 2008; 59:757.
Smolen JS, Breedveld FC, Burmester GR, et al. Treating rheumatoid arthritis to target:
64 2014 update of the recommendations of an international task force. Ann Rheum Dis 2016;
75:3.
Singh JA, Saag KG, Bridges SL Jr, et al. 2015 American College of Rheumatology Guideline
65
for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol 2016; 68:1.
Schipper LG, van Hulst LT, Grol R, et al. Meta-analysis of tight control strategies in
66 rheumatoid arthritis: protocolized treatment has additional value with respect to the clinical
outcome. Rheumatology (Oxford) 2010; 49:2154.
Rantalaiho V, Korpela M, Laasonen L, et al. Early combination disease-modifying
antirheumatic drug therapy and tight disease control improve long-term radiologic outcome
67
in patients with early rheumatoid arthritis: the 11-year results of the Finnish Rheumatoid
Arthritis Combination Therapy trial. Arthritis Res Ther 2010; 12:R122.
Leirisalo-Repo M, Kautiainen H, Laasonen L, et al. Infliximab for 6 months added on
combination therapy in early rheumatoid arthritis: 2-year results from an investigator-
68
initiated, randomised, double-blind, placebo-controlled study (the NEO-RACo Study). Ann
Rheum Dis 2013; 72:851.
Moreland LW, O'Dell JR, Paulus HE, et al. A randomized comparative effectiveness study of
oral triple therapy versus etanercept plus methotrexate in early aggressive rheumatoid
69
arthritis: the treatment of Early Aggressive Rheumatoid Arthritis Trial. Arthritis Rheum
2012; 64:2824.
Pincus T, Castrejn I. Evidence that the strategy is more important than the agent to treat
rheumatoid arthritis. Data from clinical trials of combinations of non-biologic DMARDs, with
70
protocol-driven intensification of therapy for tight control or treat-to-target. Bull Hosp Jt
Dis (2013) 2013; 71 Suppl 1:S33.
Kirwan JR. The effect of glucocorticoids on joint destruction in rheumatoid arthritis. The
71 Arthritis and Rheumatism Council Low-Dose Glucocorticoid Study Group. N Engl J Med
1995; 333:142.
Kirwan JR, Bijlsma JW, Boers M, Shea BJ. Effects of glucocorticoids on radiological
72
progression in rheumatoid arthritis. Cochrane Database Syst Rev 2007; :CD006356.
Bakker MF, Jacobs JW, Welsing PM, et al. Low-dose prednisone inclusion in a
73 methotrexate-based, tight control strategy for early rheumatoid arthritis: a randomized
trial. Ann Intern Med 2012; 156:329.
Hoes JN, Jacobs JW, Verstappen SM, et al. Adverse events of low- to medium-dose oral
74
glucocorticoids in inflammatory diseases: a meta-analysis. Ann Rheum Dis 2009; 68:1833.
Da Silva JA, Jacobs JW, Kirwan JR, et al. Safety of low dose glucocorticoid treatment in
75 rheumatoid arthritis: published evidence and prospective trial data. Ann Rheum Dis 2006;
65:285.
Visser K, Katchamart W, Loza E, et al. Multinational evidence-based recommendations for
the use of methotrexate in rheumatic disorders with a focus on rheumatoid arthritis:
76
integrating systematic literature research and expert opinion of a broad international panel
of rheumatologists in the 3E Initiative. Ann Rheum Dis 2009; 68:1086.
Ariza-Ariza R, Navarro-Sarabia F, Hernndez-Cruz B, et al. Dose escalation of the anti-TNF-
77 alpha agents in patients with rheumatoid arthritis. A systematic review. Rheumatology
(Oxford) 2007; 46:529.
Rahman MU, Strusberg I, Geusens P, et al. Double-blinded infliximab dose escalation in
78
patients with rheumatoid arthritis. Ann Rheum Dis 2007; 66:1233.
Pavelka K, Jarosov K, Such D, et al. Increasing the infliximab dose in rheumatoid
79 arthritis patients: a randomised, double blind study failed to confirm its efficacy. Ann
Rheum Dis 2009; 68:1285.
van Vollenhoven RF. How to dose infliximab in rheumatoid arthritis: new data on a serious
80
issue. Ann Rheum Dis 2009; 68:1237.
Johnsen AK, Schiff MH, Mease PJ, et al. Comparison of 2 doses of etanercept (50 vs 100
81 mg) in active rheumatoid arthritis: a randomized double blind study. J Rheumatol 2006;
33:659.
Weinblatt ME, Schiff MH, Ruderman EM, et al. Efficacy and safety of etanercept 50 mg
twice a week in patients with rheumatoid arthritis who had a suboptimal response to
82
etanercept 50 mg once a week: results of a multicenter, randomized, double-blind, active
drug-controlled study. Arthritis Rheum 2008; 58:1921.
Makol A, Wright K, Matteson EL. Safe use of antirheumatic agents in patients with
83
comorbidities. Rheum Dis Clin North Am 2012; 38:771.
Raaschou P, Simard JF, Holmqvist M, et al. Rheumatoid arthritis, anti-tumour necrosis
84 factor therapy, and risk of malignant melanoma: nationwide population based prospective
cohort study from Sweden. BMJ 2013; 346:f1939.
Dixon WG, Watson KD, Lunt M, et al. Influence of anti-tumor necrosis factor therapy on
cancer incidence in patients with rheumatoid arthritis who have had a prior malignancy:
85
results from the British Society for Rheumatology Biologics Register. Arthritis Care Res
(Hoboken) 2010; 62:755.
Chakravarty EF, Michaud K, Wolfe F. Skin cancer, rheumatoid arthritis, and tumor necrosis
86
factor inhibitors. J Rheumatol 2005; 32:2130.
Kameda T, Dobashi H, Miyatake N, et al. Association of higher methotrexate dose with
87 lymphoproliferative disease onset in rheumatoid arthritis patients. Arthritis Care Res
(Hoboken) 2014; 66:1302.
Raaschou P, Frisell T, Askling J, ARTIS Study Group. TNF inhibitor therapy and risk of
88 breast cancer recurrence in patients with rheumatoid arthritis: a nationwide cohort study.
Ann Rheum Dis 2015; 74:2137.
Solomon DH, Massarotti E, Garg R, et al. Association between disease-modifying
89 antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis.
JAMA 2011; 305:2525.
Haas RM, Li P, Chu JW. Glucose-lowering effects of sulfasalazine in type 2 diabetes.
90
Diabetes Care 2005; 28:2238.
Wiese AD, Griffin MR, Stein CM, et al. Opioid Analgesics and the Risk of Serious Infections
91 Among Patients With Rheumatoid Arthritis: A Self-Controlled Case Series Study. Arthritis
Rheumatol 2016; 68:323.
Gaujoux-Viala C, Nam J, Ramiro S, et al. Efficacy of conventional synthetic disease-
modifying antirheumatic drugs, glucocorticoids and tofacitinib: a systematic literature
92
review informing the 2013 update of the EULAR recommendations for management of
rheumatoid arthritis. Ann Rheum Dis 2014; 73:510.
Nam JL, Ramiro S, Gaujoux-Viala C, et al. Efficacy of biological disease-modifying
antirheumatic drugs: a systematic literature review informing the 2013 update of the
93
EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis
2014; 73:516.
Schoels M, Wong J, Scott DL, et al. Economic aspects of treatment options in rheumatoid
94 arthritis: a systematic literature review informing the EULAR recommendations for the
management of rheumatoid arthritis. Ann Rheum Dis 2010; 69:995.
Ramiro S, Gaujoux-Viala C, Nam JL, et al. Safety of synthetic and biological DMARDs: a
95 systematic literature review informing the 2013 update of the EULAR recommendations for
management of rheumatoid arthritis. Ann Rheum Dis 2014; 73:529.
Scott DL, Symmons DP, Coulton BL, Popert AJ. Long-term outcome of treating rheumatoid
96
arthritis: results after 20 years. Lancet 1987; 1:1108.
Wolfe F, Mitchell DM, Sibley JT, et al. The mortality of rheumatoid arthritis. Arthritis Rheum
97
1994; 37:481.
Pincus T, Callahan LF. What is the natural history of rheumatoid arthritis? Rheum Dis Clin
98
North Am 1993; 19:123.
Harty L, O'Toole G, FitzGerald O. Profound reduction in hospital admissions and
99 musculoskeletal surgical procedures for rheumatoid arthritis with concurrent changes in
clinical practice (1995-2010). Rheumatology (Oxford) 2015; 54:666.
Ajeganova S, van Steenbergen HW, van Nies JA, et al. Disease-modifying antirheumatic
100 drug-free sustained remission in rheumatoid arthritis: an increasingly achievable outcome
with subsidence of disease symptoms. Ann Rheum Dis 2016; 75:867.
Krishnan E, Fries JF. Reduction in long-term functional disability in rheumatoid arthritis
101
from 1977 to 1998:a longitudinal study of 3035 patients. Am J Med 2003; 115:371.
Puolakka K, Kautiainen H, Mttnen T, et al. Early suppression of disease activity is
102 essential for maintenance of work capacity in patients with recent-onset rheumatoid
arthritis: five-year experience from the FIN-RACo trial. Arthritis Rheum 2005; 52:36.
Symmons DP. Environmental factors and the outcome of rheumatoid arthritis. Best Pract
103
Res Clin Rheumatol 2003; 17:717.
Visser H, le Cessie S, Vos K, et al. How to diagnose rheumatoid arthritis early: a prediction
104
model for persistent (erosive) arthritis. Arthritis Rheum 2002; 46:357.
Topic 7516 Version 30.0
All rights reserved.
2016 UpToDate, Inc.

You might also like