Rheumatoid arthritis: What is refractory disease and how to manage... ⁎ Joaquim Polido-Pereira, Elsa Vieira-Sousa, João Eurico Fonseca Review

Autoimmunity Reviews 10 (2011) 707–713
Contents lists available at ScienceDirect
Autoimmunity Reviews
j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / a u t r ev
Review
Rheumatoid arthritis: What is refractory disease and how to manage it?
Joaquim Polido-Pereira, Elsa Vieira-Sousa, João Eurico Fonseca ⁎
Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon, Portugal
a r t i c l e
i n f o
Available online 5 May 2011
Keywords:
Refractory disease
Remission
Treatment strategies
Rheumatoid arthritis
a b s t r a c t
Despite the enthusiastic progresses in the field of rheumatoid arthritis pharmacotherapy the presence of
prognostic factors associated with an unfavorable outcome and the inappropriate and/or delayed initiation of
DMARDs can diminish the likelihood of achieving remission and increase the probability of refractoriness to
treatment.
During the last decade we have experience exciting developments regarding the approval of new treatment
options but few patients are reaching sustained remission and refractory patients continue to be a problem.
Thus, it is critical to understand how clinicians can decrease the risk of refractoriness by close monitoring
disease activity, using well defined and accepted composite measures, and by early and optimized use of
DMARDs, including biologics.
The goal of this review paper is to offer an evidence based roadmap to prevent and to deal with refractory RA.
Crown Copyright © 2011 Published by Elsevier B.V. All rights reserved.
Contents
1.
Introduction . . . . . . . . . . . . . . . . . . . . . . . .
2.
The ever changing concept of early disease . . . . . . . . . .
3.
New rheumatoid arthritis classification criteria . . . . . . . .
4.
What is refractory rheumatoid arthritis? . . . . . . . . . . .
5.
Response criteria . . . . . . . . . . . . . . . . . . . . . .
6.
Treating a moving target . . . . . . . . . . . . . . . . . . .
7.
Mitigating refractoriness: early and optimal use of DMARDs . .
8.
Refractoriness to synthetic DMARDs. What to do next? . . . .
9.
Refractoriness to TNF inhibitors. What to do next? . . . . . .
10.
Could biologics used as a first line therapy reduce refractoriness
11.
Refractoriness to approved therapies. What to do next? . . . .
12.
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . .
Take-home messages . . . . . . . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
.
.
.
.
.
.
.
.
.
in
.
.
.
.
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
rheumatoid
. . . . . .
. . . . . .
. . . . . .
. . . . . .
1. Introduction
Rheumatoid arthritis (RA) is still an incurable disease, but if
patients are early diagnosed and adequately treated an increasing
number of affected individuals are able to achieve a state where only
⁎ Corresponding author at: Rheumatology Research Unit, Instituto de Medicina
Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
Tel.: + 351 969049532; fax: + 351 217999412.
E-mail address: [email protected] (J.E. Fonseca).
. . . . .
. . . . .
. . . . .
. . . . .
. . . . .
. . . . .
. . . . .
. . . . .
. . . . .
arthritis?
. . . . .
. . . . .
. . . . .
. . . . .
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
707
708
708
708
708
709
709
709
710
711
711
711
711
712
712
mild or no symptoms persist, which, depending on the defined criteria
might be classified as remission [1]. However, the presence of some
disease characteristics associated with a worse prognosis and/or
inappropriate and/or delayed treatment of this disease, leads to
refractoriness to treatment and to progressive joint destruction,
functional impairment and increased mortality. To decrease the risk of
refractoriness it is crucial to early diagnose RA patients, monitor
tightly with appropriate tools disease activity, identify those who
have the worse prognosis and select the adequate treatment strategy.
The goal of this review paper is to offer an evidence based roadmap to
prevent and to deal with refractory RA.
1568-9972/$ – see front matter. Crown Copyright © 2011 Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.autrev.2011.04.023
708
J. Polido-Pereira et al. / Autoimmunity Reviews 10 (2011) 707–713
2. The ever changing concept of early disease
Back in the 1980s, the concept of early RA included disease duration
of up to 5 years and the management strategy was based on the
pyramid concept. Only patients with severe disease and joint
destruction received effective disease modifying antirheumatic drugs
(DMARDs) and this was due to the fact that RA was mostly viewed as a
mild chronic disease, with joint damage and disability occurring very
slowly and late in the disease course. With the increasing evidence of
the efficacy of early RA treatment and the advent of the “window of
opportunity” concept, this paradigm changed, and in the 1990s the
early RA definition evolved into maximum disease duration of 12 to
24 months [2]. In fact, if left improperly treated, most of these patients
develop significant joint damage during this period of time [3]. More
recently, this critical early period has been progressively reduced to
few weeks, as there is increasing evidence that very early patients
(within the first 6–12 weeks) may have a distinct immunopathogenic
process and that they will benefit from earlier intervention [4–8].
3. New rheumatoid arthritis classification criteria
According to the 1987 American College of Rheumatology (ACR)
RA classification criteria, revised by Arnett et al., there has to be at
least 4 out of 7 criteria to establish the diagnosis of RA [9]. Two of
these seven criteria are related with damage and, in early disease,
rheumatoid factor (RF) is frequently negative. Thus, it is not surprising
that these criteria do not perform well in early disease. A systematic
review of the literature showed that sensitivity and specificity of the
1987 ACR criteria in early RA were 77% (68% to 84%) and 77% (68% to
84%) respectively [10,11]. Taking into account the relevance of early
diagnosis, new classification criteria for RA had to be developed. This
was clearly an unmet medical need as many arthritis patients that
were not fulfilling the 1987 criteria were in fact early RA cases and this
misclassification could be hindering effective therapy and was surely
affecting the way epidemiological studies and clinical trials were
being designed and interpreted [12,13]. A major collaborative effort
between the ACR and the European League Against Rheumatism
(EULAR) was settled and this seminal paper was recently published,
emphasizing a major issue in early arthritis management which is to
probabilistically decide whether or not we are facing a persistent
form of arthritis that will likely benefit from immediate methotrexate
(MTX) treatment. These new diagnostic criteria are a score based
algorithm that take into account joint involvement (2–10 large joints
score 1; 1–3 small joints score 2; 4–10 small joint score 3; more than
10 joints, including at least 1 small joint, score 5), serology (RF or anticitrullinated peptide antibodies (ACPA) in low titer score 2; RF or
ACPA in high titer score 3), acute phase reactants (abnormal
erythrocyte sedimentation rate (ESR) or C-reactive protein (CRP)
score 1) and symptom duration (more than 6 weeks score 1). The
scoring system allows a relatively objective way of classifying a
patient as having RA (≥6 points), with potential relevant impact on
clinical practice and research [14].
4. What is refractory rheumatoid arthritis?
Refractory RA is an elusive and moving concept. The definition of
refractory disease is dependent on the ability to delineate predetermined quantitative targets in well defined variables. In the case of RA it
is relatively easy to identify the clinical and laboratorial manifestations
of inflammation (disease activity) as our primary target. Closely monitoring of disease activity and changing treatment according to
predefined targets leads to better disease control, less damage and
disability, as is demonstrated in both the TICORA and BeSt trials [15,16].
Assessment of disease activity is critical to set predetermined targets.
Evaluation of RA clinical activity by an experienced clinician is quite
intuitive. However, in order to adequately monitor patients, quantita-
tive joint evaluation is fundamental. Performing 66 swelling and 68
tenderness joint counts can be difficult to implement in routine clinical
practice. The 28 joint count performs well in most patients and clearly it
works adequately at a group level, even though, by excluding feet, it
might be inappropriate to some individual patients [17]. A good
practical compromise can be to persistently verify the 28 joints and
carefully register other positive joint findings not included in this count.
On top of this, markers of inflammatory activity and the symptoms
reported by the patients are also relevant aspects for the global
assessment of disease activity. In fact, this has been tried to be captured
by composite measures of RA disease activity. Two composite measures
are extensively studied and validated: the EULAR Disease Activity Score
(DAS) and the ACR Core Data Set. The use of composite measures usually
partially overcomes the problem of the heterogeneity of RA clinical and
laboratorial manifestations. In fact, there are patients that have normal
ESR and still maintain painful and swollen joints and develop erosions;
others have essentially high ESR and/or CRP and scarce clinical
manifestations but have a radiologic progression that is equally severe.
Composite measures have been shown to have a closer relationship
with radiographic progression than single measures. Besides, composite
measures diminish the sample size needed in clinical trials to detect
significant differences between groups [18].
The ACR Core Data Set includes three visual analogue scales (VAS)
for the physician and patient global assessment of disease activity and
for patient perception of pain, two joint counts (number of tender and
swollen joint counts), one laboratory measure (CRP or ESR), a measure
of function (usually the Health Assessment Questionnaire, HAQ), as
well as a yearly measure of radiological damage [19]. Nevertheless, the
ACR Core Data Set fails to numerically quantify disease activity. The
DAS score was developed to meet this requirement. It is based on a
mathematical formula that computes four variables: the Ritchie
articular index, the 44 swollen joint count, ESR and VAS for patient
global assessment of wellbeing, which can be used interchangeably
with VAS for disease activity. In the absence of the VAS a three item
DAS (DAS3 variables) can be calculated [18]. Later on a DAS28 was
developed using the 28 joint count for both tenderness and swelling,
simplifying the joint count [17]. Levels of DAS28 are somewhat higher
than DAS values and this is reflected in the way results have to be
clinically interpreted, as is the case of the definition of low disease
activity and remission. Only the DAS28 CRP has been compared with
the DAS28 using ESR. Some studies show equivalence between these
two scores whereas others do not [20–22]. Other simplified scores
were developed, such as the simplified disease activity index (SDAI)
and the clinical disease activity index (CDAI). Despite being less used,
the SDAI offers a very simplified approach (defined as the simple sum
of the tender joint count (using 28 joints), swollen joint count (using
28 joints), patient global assessment (0–10 scale), physician global
assessment (0–10 scale), and C-reactive protein level (mg/dl)) and
was included in the new remission criteria recently published [23,24].
5. Response criteria
Both EULAR and ACR developed response criteria. The ACR response
criteria are based on the previously reviewed core set. ACR 20, 50 or 70
stands for the percentage of improvement in this core set. A response (at
least 20, 50 or 70% improvement) must be observed on both joint
counts, and at least in three out of the remaining five variables. The
EULAR response criteria are based on the DAS or DAS28 and take into
account both the variation and the absolute value. This response can be
classified as “no response”, “moderate response” or “good response”
[19,25]. A DAS28 “no response” corresponds to an improvement of ≤0.6
or, for those with an endpoint DAS28 N5.1, an improvement of ≤1.2. A
“moderate response” is defined as an improvement of N0.6 and ≤1.2
with an endpoint DAS28 ≤5.1 or an improvement of N1.2 with an
endpoint DAS28 of N3.2. A “good response” is reserved for patients
attaining a DAS28 of ≤3.2 and an improvement of N1.2. Additionally,
J. Polido-Pereira et al. / Autoimmunity Reviews 10 (2011) 707–713
DAS28 can classify patients in disease activity classes: remission (≤2.6),
low activity (N2.6 and ≤3.2), moderate activity (N3.2 and b5.1) and
high activity (≥5.1).
709
To define the concept of refractory a target has to be determined.
More than 60 experts from various regions of the world developed
recommendations for achieving optimal therapeutic outcomes in RA
in the Treat to Target Initiative. These recommendations assumed that
the main goal to achieve in RA is remission, being a low disease
activity state an alternative aim for those who failed to reach that
major goal. Regular follow up, every 1–3 months, for disease activity
above the target, and 3–6 months, if patients have already reached the
target, was defined as the best clinical practice (Fig. 1) [26]. According
to this initiative, refractory disease could be defined as the failure to
achieve the predetermined target. The Treat to Target recommendations did not purpose a specific composite measure of disease activity.
Applying the most widely used composite measure (DAS28) in this
setting would mean that the ideal target to reach is a DAS28 ≤2.6
and, for those who cannot achieve it, the goal would be to keep a
DAS28 ≤3.2. Remission could be delineated with more stringed
criteria, based on the recently published definition of remission in RA
proposed by ACR and EULAR: ≤ 1 tender joint, ≤1 swollen joint, CRP
≤1 mg/dl and patient global assessment ≤1 (0 to 10). Alternatively a
SDAI ≤3.3 can also classify a remission state [24].
Currently, there are no biological markers to define refractory
disease, since the concept itself is yet to be clearly defined in clinical
terms. Nevertheless, recent EULAR recommendations on the management of RA state the importance of prognostic markers in treatment decisions. Several factors seem to independently predict worse
outcome: autoantibodies (high levels of rheumatoid factor (RF)
and/or anti-citrullinated peptides antibodies (ACPA)), high disease
activity as measured by composite indices and early occurrence of
erosions. All these factors were included in the creation of a prediction
model. Still, this does not define per se refractory disease, but only
an increased risk of developing refractory RA [27,28]. In addition,
other factors can play a role in the refractoriness of RA in individual
patients, such as genetic variability affecting DMARDs metabolism
and influencing the relevance of pathogenic pathways not targeted by
the available DMARDs.
Delaying the start of treatment can lead to worse outcomes and might
reduce the likelihood of inducing remission or of attaining low disease
activity and increases the risk of refractoriness [29,30]. Indeed, the
first 3–6 months of disease in early RA patients are crucial regarding
joint damage, since around 50% of patients develop erosions during
the first year of disease course and 90% after the second year after
diagnosis [31,32]. Tight monitoring of disease activity, minimum
every 3 months, assessed by validated instruments and followed
by frequent treatment adjustments are also determinant factors to
achieve better outcomes as recently reviewed [33]. Strategies based in
treatment to target and intensive monitoring regimes such as the ones
used in the TICORA, Fransen et al., and the CAMERA studies were
clearly better than treatment decisions according to clinical judgment
and routine care follow-up [15,34,35].
Due to its good safety profile and efficacy both as monotherapy as
well as potentiating the effect of other DMARDs, MTX is nowadays
considered the preferred synthetic drug to be initiated at diagnosis of
RA patients. MTX should be started at a weekly dose of 10 to 15 mg
and increased by adding 5 mg every four weeks up to a dose of 25 mg
(rarely 30 mg), according to tolerability and efficacy. Age, comorbidities and concomitant medications can influence the rate of dose
increment and limit the maximum used dose. In refractory patients in
whom malabsorption is suspected or a dose limit has been determined by gastrointestinal intolerance, changing to parental route
should be tried [36].
Other synthetic DMARDs such as leflunomide and sulphasalazine
are generally placed as alternatives to MTX monotherapy in case of
intolerance or contraindications, while cyclosporine and gold have
limited use mainly due to adverse effects. Antimalarial drugs even if
able to lead to some improvement of disease manifestations are weak
inhibitors of joint damage and should be used as monotherapy only in
exceptionally mild cases, associated with intolerance to other
conventional DMARDs [28].
Glucocorticoids have an unquestionable role as an adjuvant therapy to DMARDs, used ideally for short periods of time to reduce
inflammation, as a bridging therapy at diagnosis or in refractory
patients between switches, particularly when the next treatment
option has a delayed onset of action. Additionally glucocorticoids have
showed to slow radiographic progression and are therefore qualified
as DMARDs [37]. For highly refractory patients continuous low dose
glucocorticoids, in association with other DMARDs, can be considered.
7. Mitigating refractoriness: early and optimal use of DMARDs
8. Refractoriness to synthetic DMARDs. What to do next?
Treating patients early and with the best recognized regimes of
synthetic DMARDs are important factors to mitigate refractoriness.
Synthetic DMARDs should be started in all patients upon diagnosis to
reduce disease activity, control joint damage and prevent disability.
Although combining synthetic DMARDs in patients who do not
respond to monotherapy has additional benefits demonstrated in
some clinical trials, in daily clinical practice the effective response rate
is limited by adverse effects and low compliance. Clearly the SWEFOT
trial has helped us to understand that in early arthritis patients, the
combination of MTX and infliximab is superior to the addition of SSZ
and HCQ [38]. For this reason it is recommended by EULAR task force,
that if predictors of poor outcome are present (RF, ACPA, high disease
activity, early erosions), and patients are refractory to the first synthetic DMARD they should be started on a biologic DMARD [28].
Otherwise, in the absence of severity prognostic factors switching to
another synthetic DMARD (or even to a combination of synthetic
DMARDs) might be considered.
At the moment there are nine biologic therapies approved for RA
treatment, targeting specific components of the immune system pathways, which were developed in a bench to bedside approach. All TNF
inhibitors (infliximab, adalimumab, etanercept, golimumab and
certolizumab) as well as abatacept and tocilizumab are approved by
Food and Drugs Administration (FDA) and European Medicine Agency
(EMEA) for use in DMARDs refractory patients (including MTX).
Rituximab at the moment is only approved after TNF inhibitors (TNFi)
failure, although efficacy similar to abatacept and tocilizumab has been
6. Treating a moving target
Fig. 1. Optimal management of RA [26].
710
J. Polido-Pereira et al. / Autoimmunity Reviews 10 (2011) 707–713
shown in DMARDs failures. Anakinra, also approved for RA treatment,
is less efficacious than the other biologics and its use is therefore
reserved for selected cases, particularly in the setting of autoinflammatory diseases [39].
The fact that TNFi were the first biologics to be approved and have
been used in clinical practice for the last 10 year has allowed the
gathering of solid data on efficacy and safety. This has contributed to
the positioning of TNFi as the first biologics to be started after
DMARDs failure [28]. However it is expected that, as biomarkers of
response to therapy become more specific and reliable, namely at the
individual level, biologic DMARDs will be selected through a more
personalized approach.
Cytokine blocking agents targeting TNF such as infliximab,
etanercept, adalimumab and more recently certolizumab and golimumab have clearly revolutionized the way moderate to severe RA is
treated. Different TNFi exhibit similar efficacy, with an ACR20, ACR50
and ACR70 response attained, respectively, by around 60%, 40% and
20% of the exposed patients [40]. It is well recognized from large
clinical trials that TNFi optimal effect is obtained if they are used in
combination with MTX. It is also possible that leflunomide can
similarly work synergistically but further studies are required [41,42].
Concomitant use of MTX reduces human anti-chimeric antibodies
(HACA) and human anti-human antibodies (HAHA) production,
minimizing immunogenicity and acute infusion reactions. Moreover,
secondary failure to anti-TNF monoclonal antibodies, which is known
to be at least in part dependent on the production of HACAs or HAHAs,
can be diminished by adding MTX [43]. When initial suboptimal
response or a secondary failure occurs during the use of infliximab
increasing the dose (up to 10 mg/kg, but more commonly up to
5 mg/kg) and/or shortening the administrations interval (up to every
4 weeks, but more commonly up to 6 weeks) can help to overcome
lack of efficacy in some cases and by this way, reduce refractoriness to
infliximab [44].
Datasets from registries (e.g., British Society for Rheumatology
Registry, Swedish Registry, German Registry) have highlighted a small
but increased risk of serious infections in comparison with synthetic
DMARDs which seem to be higher in the first 6 months of therapy.
Furthermore patients are more prone to intracellular bacterial
infections such as those caused by Mycobacterium tuberculosis and
this risk is higher in antibody treated patients than in etanercept
exposed individuals [45]. The implementation of preventing strategies has reduced the incidence of latent tuberculosis [46]. Other
preventive strategies like influenza and pneumococcal vaccination,
careful review of dental hygiene, early diagnosis and treatment of
respiratory, urinary tract and skin infections can also contribute to a
safer use of TNFi. All these prophylactic measures can lead to a higher
TNFi retention rate and thus to a decrease in treatment refractoriness.
Recently, two new TNFi, golimumab and certolizumab, have
became available [47,48]. Both were developed based on strategies
aiming at reducing immunogenicity. Golimumab was created upon
human TNF immunization of transgenic mice expressing human
immunoglobulins, being so a fully human monoclonal antibody.
Certolizumab is a humanized antibody without the Fc fragment,
which was substituted by a pegylated tail. Golimumab in association
with MTX is efficacious in MTX inadequate responders, TNFi failures
and MTX naïve patients. In phase III studies golimumab 100 mg was
not superior to 50 mg subcutaneous, neither golimumab monotherapy 100 mg was superior to MTX alone, leading to a final approved
regime of 50 mg subcutaneous once a month [49–52]. Certolizumab
has showed efficacy in MTX (RAPID1 and RAPID2) and in DMARDs
(FAST-4WARD) refractory patients [53–55]. Certolizumab has a quick
onset of action and should be administrated according to an induction
regime of 400 mg at 0, 2 and 4 weeks and 200 mg every two weeks
thereafter.
The addition to RA therapeutic armamentarium of two new
potentially less immunogenic TNFi holds the promise of improving
the chances of response at the individual level, contributing to minimize refractory cases in clinical practice.
9. Refractoriness to TNF inhibitors. What to do next?
The percentage of patients for whom the first TNFi is inefficient
(primary failure) has been estimated to be around 1/3 of the patients
[56]. Furthermore, a significant percentage of patients will also lose
efficacy latter on (secondary failure). For both these subgroups of
patients different treatment options are now available, including
switching to an alternative TNFi or changing to an agent with a different
mechanism of action.
Switching to another TNFi is supported by arguments related to
pharmacology, disease mechanisms and immunogenicity. First of all,
TNFi differ in their half-lives and affinities and this might be translated
into a different duration of TNF neutralization, leading to different
response patterns in individual patients. In addition, in spite of similar
efficacy in RA and Spondyloarthritis the differences in the mechanism
of action between monoclonal antibodies and the TNF receptor fusion
protein is reflected, for instance, in a low effect of etanercept in Crohn's
Disease and in other granulomatosis diseases, and this might be relevant in some nonresponding patients, given the individual heterogeneity of RA physiopathology. Finally, the appearance of neutralizing
antibodies against monoclonal antibodies in nonresponding patients
might be an extra argument for switching.
Therefore lost of efficacy to a TNFi can potentially be recaptured by
switching to another TNFi. The response to a second TNFi has been
reported to be slightly lower in comparison to TNFi naïve patients but
it is clearly reduced with further switches [57]. Additionally, the reason
for switch might also influence the outcome, with discontinuation due
to adverse events and due to secondary failure being predictors of
better response to a second TNFi than when the switch is made after a
primary failure [57,58]. These data suggest that switching to a second
TNFi can be considered, with the possible exception of primary
failures. However, switching to another biologic with a different
mechanism of action is clearly advisable if a second response failure
occurs.
Our armamentarium and experience with the use of other biologics
has markedly increased in recent years. Specifically, rituximab,
tocilizumab and abatacept have proven their efficacy in anti-TNF
refractory patients in large randomized clinical trials (RCT) [59–62].
The rational for the use of rituximab in the treatment of RA is based
in the ability to reduce autoreactive antibodies synthesis, such as RF
and ACPA, and impair antigen presentation to T cells, as well as
cytokine production. Patients with inadequate response to TNFi can
benefit from initiating B cell depleting therapies namely rituximab.
Finckh et al. in a longitudinal cohort study have shown that the
improvement of DAS28 score was better in patients that received
rituximab after the failure of one TNFi than those who were switched
to another TNFi [63]. When the reason for switching was considered
in a later study it was demonstrated that the magnitude of DAS28
improvement between groups was higher in those who switched due
to inefficacy of the TNFi comparing to those who changed due to other
reasons [63,64]. Well known predictors of response to rituximab (RF
and/or ACPA+) have to be taken into account in the selection of
patients, even if a few patients who are not positive for RF or ACPA
might also respond favorably [65]. Furthermore, experience with the
use of rituximab has showed that retreatment might potentiate
efficacy in responders and secondary lost of response is generally rare
[66,67]. However, for primary failures to RTX, switching to a biologic
DMARD targeting other immunologic pathways should be preferred.
At the moment, retreatment using a “treat to target” strategy instead
of an on-demand regime seems to be more effective but longer
follow-ups are needed to clarify this issue and the possible requirements for a fixed schedule are being evaluated [68].
J. Polido-Pereira et al. / Autoimmunity Reviews 10 (2011) 707–713
Encouraging data from five phase III RCT lead to the approval of
tocilizumab both in the US (TNFi failures) and in Europe (DMARDs and
TNFi failures). Blocking the effect of the pleiotropic IL6 cytokine in RA
patients is translated into similar efficacy to TNFi, regarding disease
activity and radiographic progression and clear superiority to MTX [69].
The possibility of using tocilizumab as monotherapy in patients that are
refractory or intolerant to MTX is a distinctive feature of this drug.
Abatacept is a soluble recombinant fully human protein comprises the
extracellular domain of CTLA4 and the Fc portion of an IgG1 molecule that
has been modified to prevent complement activation. The principle of its
use in RA is dependent on the induction of T cell anergy by blocking the
second signal between CD28 and CD80/86 necessary for T cell activation.
Based in data from in vivo experiments it is assumed that abatacept
develops its action by decreasing T cell activation, migration and secretion
of cytokines [70]. Particularly, abatacept is able to reduce progression to
RA in a subgroup of patients with undifferentiated/early rheumatoid
arthritis, highlighting its efficacy in early treatment [71].
Despite the increased number of biologics available, until now
there is not enough evidence that would help clinicians to position
non-TNFi biologics according to disease phenotypes or to helpful
specific biomarkers. Therefore, clinicians are still basing their decisions
on personal experience and on the safety profile of each biologic drug.
10. Could biologics used as a first line therapy reduce
refractoriness in rheumatoid arthritis?
The question if using biologic DMARDs as first line therapy early in the
disease course could reduce refractoriness is still not clearly answered.
Early intervention with biologic agents seems to offer a significant
advantage in the magnitude of their benefits. TNFi including infliximab
(ASPIRE), etanercept (COMET), adalimumab (PREMIER), and golimumab
(Go-Before) have proved that when used in early disease in MTX naïve
patients they were superior to MTX in monotherapy, particularly in their
capacity to arrest radiographic progression [49,72–74]. Similar results
would be expected if certolizumab had been tested in this same
population. Furthermore, recent analysis of the 4 year follow up of the
BeSt study revealed that first line use of infliximab led to a more sustained
remission as compared to patients that received this drug in latter stages
of the treatment strategy [75]. Similarly, the SWEFOT study highlighted
benefits from the combination of infliximab plus MTX in comparison with
triple synthetic DMARDs therapy [38]. Recent data from a prospective
open study suggests that although a faster reduction in DAS28 in patients
starting MTX and adalimumab as first line treatment is observed at
12 weeks, the clinical and radiographic outcomes at 52 weeks did not
differ from those who were changed to the same regime after 3 months, if
they were refractory to MTX [76]. Therefore this data needs further
analysis.
All current biologics (TNFi, tocilizumab, abatacept and rituximab)
have increased benefits when used combined with MTX in DMARDs
naïve patients [77–79].
Despite the ongoing discussion, at the moment there in not
enough evidence to support generalized use of biologics in association
with MTX as fist line therapy. By doing so, we would be over treating
at least 15% to 40% of patients who would respond to MTX
monotherapy [80]. This approach can be however considered in
selected cases, when clear unfavorable prognostic factors are present
and disease activity is very high.
11. Refractoriness to approved therapies. What to do next?
Despite the efficacy and increased number of biologic therapies
available it has been reported that the ACR70 response rate to a second
biologic is of 5–15% and the DAS28 remission is reached by 9–15% of this
patients, leaving the proposed treatment targets clearly unmet [81].
Many exciting new biological agents are in development for the
treatment of RA. Inclusion of selected patients that became refractory
711
to all approved biologics in ongoing clinical trials according to clinical
judgment can bring potential benefits for disease control. For example
new cytokine antagonists, namely blocking IL6, IL17 and GM-CSF are
being studied [82–88]. Other anti-CD20 antibodies including the fully
human ofatumumab and the humanized ocrelizumab and veltuzumab
are in phase II/III trials [89–91]. Neutralization of the B cell survival
and activation cytokines BLyS (or BAFF) and APRIL is an approach that
is in development, despite the disappointing results obtained with
atacicept (which blocks both BLyS and APRIL and could therefore
reversibly suppress B cell function across maturation stages). Signaling pathways including NF-kB and p38 MAPK pathways are being
targeted by small molecules, as is also the case of JAK and Syk kinases
[92,93]. Possible complementary approaches arresting the effects of
inflammation in bone metabolism such as inhibiting osteoclast
differentiation are particularly attractive [94].
The treatment potential of using stem cell transplantation (SCT) for
severe refractory RA patients at risk of high mortality and previously to
irreversible damage should not be forgotten. SCT is relatively well
tolerated by most of the patients, with transplant related mortality after
100-days of 1% and a general survival of 94%. It is associated with
improvement of disease activity and function and inhibition of joint
destruction. Peripheral blood is the most commonly used source of stem
cells and autologous transplant preferred. High doses of cyclophosphamide led to superior disease control than lower doses [95]. Relapses are
however rather frequent with a progression-free rate of 23% at 3-year
and 18% at 5 years and DMARDs such as MTX, cyclosporine and
rituximab are required to maintain disease activity controlled [96]. Since
the advent and broad use of biologic therapies SCT has indeed become
less commonly used for RA treatment as compared to other rheumatic
diseases, such as systemic sclerosis, reflecting the increase of treatment
options, but it can still be considered as a salvage treatment.
Furthermore, mesenchymal stromal cells (MSC) have showed in
vitro and in vivo anti-proliferative and anti-inflammatory properties
that support the rational for their use in inflammatory arthritis [95].
MSC effects on collagen induced arthritis models have been
contradictory, but their potential immunoregulatory effects might
be translated into a valuable therapeutic approach [97].
12. Conclusions
Despite the enthusiastic progresses in the field of RA pharmacotherapy, few patients are reaching sustained remission and refractory
patients continue to be a problem. However, we have the tools to
perform at a higher level. We have now well defined and accepted
composite measures for monitoring RA disease activity, consensual
treatment targets, a managing strategy that has been progressively
updated and a vast option of highly effective drugs. We should now
put the emphasis on the human factor and assume that managing RA
in the 21st century is a complex and highly differentiated task that
should be taken care by specialized and externally certified centers,
through a benchmark approach, where the best balance between
efficacy, safety and costs will be required.
Take-home messages
• Refractoriness can be mitigated by early diagnose of RA patients,
tight monitoring using appropriate tools to measure disease activity,
identification of patients with worse prognosis and starting early
and adequate treatment strategy aiming remission at each individual patient.
• In spite of the increased number of biologics available, until know
there is not enough evidence that would help clinicians to position
biologics according to disease phenotypes or to helpful specific
biomarkers, therefore, clinicians are still basing their decisions on
personal experience and on the safety profile of each biologic drug.
712
J. Polido-Pereira et al. / Autoimmunity Reviews 10 (2011) 707–713
References
[1] Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet 2010;376:1094–108.
[2] Cush JJ. Early rheumatoid arthritis — is there a window of opportunity? J Rheumatol
Suppl 2007;80:1–7.
[3] Emery P. The Roche Rheumatology Prize Lecture. The optimal management of early
rheumatoid disease: the key to preventing disability. Br J Rheumatol 1994;33:765–8.
[4] Raza K, Falciani F, Curnow SJ, Ross EJ, Lee CY, Akbar AN, et al. Early rheumatoid
arthritis is characterized by a distinct and transient synovial fluid cytokine profile
of T cell and stromal cell origin. Arthritis Res Ther 2005;7:R784–95.
[5] Moura RA, Weinmann P, Pereira PA, Caetano-Lopes J, Canhao H, Sousa E, et al.
Alterations on peripheral blood B-cell subpopulations in very early arthritis
patients. Rheumatology (Oxford) 2010;49:1082–92.
[6] Weinmann P, Moura RA, Caetano-Lopes JR, Pereira PA, Canhao H, Queiroz MV,
et al. Delayed neutrophil apoptosis in very early rheumatoid arthritis patients is
abrogated by methotrexate therapy. Clin Exp Rheumatol 2007;25:885–7.
[7] Cascao R, Moura RA, Perpetuo I, Canhao H, Vieira-Sousa E, Mourao AF, et al.
Identification of a cytokine network sustaining neutrophil and Th17 activation in
untreated early rheumatoid arthritis. Arthritis Res Ther 2010;12:R196.
[8] Moura RA, Cascao R, Perpetuo I, Canhao H, Vieira-Sousa E, Mourao AF, et al.
Cytokine pattern in very early rheumatoid arthritis favours B-cell activation and
survival. Rheumatology (Oxford) 2011;50:278–82.
[9] Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The
American Rheumatism Association 1987 revised criteria for the classification of
rheumatoid arthritis. Arthritis Rheum 1988;31:315–24.
[10] Banal F, Dougados M, Combescure C, Gossec L. Sensitivity and specificity of the
American College of Rheumatology 1987 criteria for the diagnosis of rheumatoid
arthritis according to disease duration: a systematic literature review and metaanalysis. Ann Rheum Dis 2009;68:1184–91.
[11] Banal FGL, Dougados M. Ability of American College of Rheumatology 1987
Criteria to predict Rheumatoid Arthritis: a systematic literature review. Ann
Rheum Dis Abstr July 2007;66(supplement II):320.
[12] Wiles N, Symmons DP, Harrison B, Barrett E, Barrett JH, Scott DG, et al. Estimating
the incidence of rheumatoid arthritis: trying to hit a moving target? Arthritis
Rheum 1999;42:1339–46.
[13] van Dongen H, van Aken J, Lard LR, Visser K, Ronday HK, Hulsmans HM, et al. Efficacy
of methotrexate treatment in patients with probable rheumatoid arthritis: a doubleblind, randomized, placebo-controlled trial. Arthritis Rheum 2007;56:1424–32.
[14] Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham III CO, et al. 2010
Rheumatoid arthritis classification criteria: an American College of Rheumatology/
European League Against Rheumatism collaborative initiative. Arthritis Rheum
2010;62:2569–81.
[15] Grigor C, Capell H, Stirling A, McMahon AD, Lock P, Vallance R, et al. Effect of a
treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a
single-blind randomised controlled trial. Lancet 2004;364:263–9.
[16] Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, van Zeben D, Kerstens PJ,
Hazes JM, et al. Clinical and radiographic outcomes of four different treatment
strategies in patients with early rheumatoid arthritis (the BeSt study): a
randomized, controlled trial. Arthritis Rheum 2005;52:3381–90.
[17] Prevoo ML, van't Hof MA, Kuper HH, van Leeuwen MA, van de Putte LB, van Riel
PL. Modified disease activity scores that include twenty-eight-joint counts.
Development and validation in a prospective longitudinal study of patients with
rheumatoid arthritis. Arthritis Rheum 1995;38:44–8.
[18] van der Heijde DM, van't Hof M, van Riel PL, van de Putte LB. Validity of single
variables and indices to measure disease activity in rheumatoid arthritis. J Rheumatol
1993;20:538–41.
[19] Felson DT, Anderson JJ, Boers M, Bombardier C, Furst D, Goldsmith C, et al.
American College of Rheumatology. Preliminary definition of improvement in
rheumatoid arthritis. Arthritis Rheum 1995;38:727–35.
[20] Wells G, Becker JC, Teng J, Dougados M, Schiff M, Smolen J, et al. Validation of the
28-joint Disease Activity Score (DAS28) and European League Against Rheumatism response criteria based on C-reactive protein against disease progression in
patients with rheumatoid arthritis, and comparison with the DAS28 based on
erythrocyte sedimentation rate. Ann Rheum Dis 2009;68:954–60.
[21] Silva I, Mateus M, Branco JC. Assessment of erythrocyte sedimentation rate (ESR)
and C-reactive protein (CRP) on Rheumatoid Arthritis activity prediction. Acta
Reumatol Port 2010;35:456–62.
[22] Hensor EM, Emery P, Bingham SJ, Conaghan PG. Discrepancies in categorizing
rheumatoid arthritis patients by DAS-28(ESR) and DAS-28(CRP): can they be
reduced? Rheumatology (Oxford) 2010;49:1521–9.
[23] Aletaha D, Ward MM, Machold KP, Nell VP, Stamm T, Smolen JS. Remission and
active disease in rheumatoid arthritis: defining criteria for disease activity states.
Arthritis Rheum 2005;52:2625–36.
[24] Felson DT, Smolen JS, Wells G, Zhang B, van Tuyl LH, Funovits J, et al. American
College of Rheumatology/European League Against Rheumatism provisional
definition of remission in rheumatoid arthritis for clinical trials. Arthritis Rheum
2011;63:573–86.
[25] van Gestel AM, Prevoo ML, van't Hof MA, van Rijswijk MH, van de Putte LB, van
Riel PL. Development and validation of the European League Against Rheumatism
response criteria for rheumatoid arthritis. Comparison with the preliminary
American College of Rheumatology and the World Health Organization/International
League Against Rheumatism Criteria. Arthritis Rheum 1996;39:34–40.
[26] Smolen JS, Aletaha D, Bijlsma JW, Breedveld FC, Boumpas D, Burmester G, et al.
Treating rheumatoid arthritis to target: recommendations of an international task
force. Ann Rheum Dis 2010;69:631–7.
[27] Vastesaeger N, Xu S, Aletaha D, St Clair EW, Smolen JS. A pilot risk model for the
prediction of rapid radiographic progression in rheumatoid arthritis. Rheumatology (Oxford) 2009;48:1114–21.
[28] Smolen JS, Landewe R, Breedveld FC, Dougados M, Emery P, Gaujoux-Viala C, et al.
EULAR recommendations for the management of rheumatoid arthritis with
synthetic and biological disease-modifying antirheumatic drugs. Ann Rheum Dis
2010;69:964–75.
[29] Lard LR, Visser H, Speyer I, vander Horst-Bruinsma IE, Zwinderman AH, Breedveld
FC, et al. Early versus delayed treatment in patients with recent-onset rheumatoid
arthritis: comparison of two cohorts who received different treatment strategies.
Am J Med 2001;111:446–51.
[30] Nell VP, Machold KP, Eberl G, Stamm TA, Uffmann M, Smolen JS. Benefit of very
early referral and very early therapy with disease-modifying anti-rheumatic drugs
in patients with early rheumatoid arthritis. Rheumatology (Oxford) 2004;43:
906–14.
[31] Lindqvist E, Jonsson K, Saxne T, Eberhardt K. Course of radiographic damage over
10 years in a cohort with early rheumatoid arthritis. Ann Rheum Dis 2003;62:
611–6.
[32] van der Heijde DM. Joint erosions and patients with early rheumatoid arthritis. Br J
Rheumatol 1995;34(Suppl 2):74–8.
[33] Knevel R, Schoels M, Huizinga TW, Aletaha D, Burmester GR, Combe B, et al.
Current evidence for a strategic approach to the management of rheumatoid
arthritis with disease-modifying antirheumatic drugs: a systematic literature
review informing the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2010;69:987–94.
[34] Fransen J, Moens HB, Speyer I, van Riel PL. Effectiveness of systematic monitoring
of rheumatoid arthritis disease activity in daily practice: a multicentre, cluster
randomised controlled trial. Ann Rheum Dis 2005;64:1294–8.
[35] Verstappen SM, Jacobs JW, van der Veen MJ, Heurkens AH, Schenk Y, ter Borg EJ,
et al. Intensive treatment with methotrexate in early rheumatoid arthritis: aiming
for remission. Computer Assisted Management in Early Rheumatoid Arthritis
(CAMERA, an open-label strategy trial). Ann Rheum Dis 2007;66:1443–9.
[36] Visser K, Katchamart W, Loza E, Martinez-Lopez JA, Salliot C, Trudeau J, et al.
Multinational evidence-based recommendations for the use of methotrexate in
rheumatic disorders with a focus on rheumatoid arthritis: integrating systematic
literature research and expert opinion of a broad international panel of
rheumatologists in the 3E Initiative. Ann Rheum Dis 2009;68:1086–93.
[37] Hoes JN, Jacobs JW, Buttgereit F, Bijlsma JW. Current view of glucocorticoid cotherapy with DMARDs in rheumatoid arthritis. Nat Rev Rheumatol 2010;6:
693–702.
[38] van Vollenhoven RF, Ernestam S, Geborek P, Petersson IF, Coster L, Waltbrand E,
et al. Addition of infliximab compared with addition of sulfasalazine and
hydroxychloroquine to methotrexate in patients with early rheumatoid arthritis
(Swefot trial): 1-year results of a randomised trial. Lancet 2009;374:459–66.
[39] Mitroulis I, Skendros P, Ritis K. Targeting IL-1beta in disease; the expanding role of
NLRP3 inflammasome. Eur J Intern Med 2010;21:157–63.
[40] Salliot C, Finckh A, Katchamart W, Lu Y, Sun Y, Bombardier C, et al. Indirect
comparisons of the efficacy of biological antirheumatic agents in rheumatoid
arthritis in patients with an inadequate response to conventional diseasemodifying antirheumatic drugs or to an anti-tumour necrosis factor agent: a
meta-analysis. Ann Rheum Dis 2011;70:266–71.
[41] De Stefano R, Frati E, Nargi F, Baldi C, Menza L, Hammoud M, et al. Comparison of
combination therapies in the treatment of rheumatoid arthritis: leflunomide-antiTNF-alpha versus methotrexate-anti-TNF-alpha. Clin Rheumatol 2010;29:517–24.
[42] Finckh A, Dehler S, Gabay C. The effectiveness of leflunomide as a co-therapy of
tumour necrosis factor inhibitors in rheumatoid arthritis: a population-based
study. Ann Rheum Dis 2009;68:33–9.
[43] Feldmann M, Maini RN. Anti-TNF therapy, from rationale to standard of care: what
lessons has it taught us? J Immunol 2010;185:791–4.
[44] Perdriger A. Infliximab in the treatment of rheumatoid arthritis. Biologics 2009;3:
183–91.
[45] Martin-Mola E, Balsa A. Infectious complications of biologic agents. Rheum Dis
Clin North Am 2009;35:183–99.
[46] Carmona L, Gomez-Reino JJ, Rodriguez-Valverde V, Montero D, Pascual-Gomez E,
Mola EM, et al. Effectiveness of recommendations to prevent reactivation of latent
tuberculosis infection in patients treated with tumor necrosis factor antagonists.
Arthritis Rheum 2005;52:1766–72.
[47] Zhou H, Jang H, Fleischmann RM, Bouman-Thio E, Xu Z, Marini JC, et al.
Pharmacokinetics and safety of golimumab, a fully human anti-TNF-alpha
monoclonal antibody, in subjects with rheumatoid arthritis. J Clin Pharmacol
2007;47:383–96.
[48] Fleischmann R. The clinical efficacy and safety of certolizumab pegol in
rheumatoid arthritis. Expert Opin Biol Ther 2010;10:773–86.
[49] Emery P, Fleischmann RM, Moreland LW, Hsia EC, Strusberg I, Durez P, et al.
Golimumab, a human anti-tumor necrosis factor alpha monoclonal antibody,
injected subcutaneously every four weeks in methotrexate-naive patients with
active rheumatoid arthritis: twenty-four-week results of a phase III, multicenter,
randomized, double-blind, placebo-controlled study of golimumab before
methotrexate as first-line therapy for early-onset rheumatoid arthritis. Arthritis
Rheum 2009;60:2272–83.
[50] Keystone E, Genovese MC, Klareskog L, Hsia EC, Hall S, Miranda PC, et al.
Golimumab in patients with active rheumatoid arthritis despite methotrexate
therapy: 52-week results of the GO-FORWARD study. Ann Rheum Dis 2010;69:
1129–35.
[51] Kremer J, Ritchlin C, Mendelsohn A, Baker D, Kim L, Xu Z, et al. Golimumab, a new
human anti-tumor necrosis factor alpha antibody, administered intravenously in
J. Polido-Pereira et al. / Autoimmunity Reviews 10 (2011) 707–713
[52]
[53]
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
[68]
[69]
[70]
[71]
[72]
patients with active rheumatoid arthritis: forty-eight-week efficacy and safety
results of a phase III randomized, double-blind, placebo-controlled study. Arthritis
Rheum 2010;62:917–28.
Smolen JS, Kay J, Doyle MK, Landewe R, Matteson EL, Wollenhaupt J, et al. Golimumab
in patients with active rheumatoid arthritis after treatment with tumour necrosis
factor alpha inhibitors (GO-AFTER study): a multicentre, randomised, double-blind,
placebo-controlled, phase III trial. Lancet 2009;374:210–21.
Fleischmann R, Vencovsky J, van Vollenhoven RF, Borenstein D, Box J, Coteur G,
et al. Efficacy and safety of certolizumab pegol monotherapy every 4 weeks in
patients with rheumatoid arthritis failing previous disease-modifying antirheumatic therapy: the FAST4WARD study. Ann Rheum Dis 2009;68:805–11.
Keystone E, Heijde D, Mason Jr D, Landewe R, Vollenhoven RV, Combe B, et al.
Certolizumab pegol plus methotrexate is significantly more effective than placebo
plus methotrexate in active rheumatoid arthritis: findings of a fifty-two-week,
phase III, multicenter, randomized, double-blind, placebo-controlled, parallelgroup study. Arthritis Rheum 2008;58:3319–29.
Smolen J, Landewe RB, Mease P, Brzezicki J, Mason D, Luijtens K, et al. Efficacy and
safety of certolizumab pegol plus methotrexate in active rheumatoid arthritis: the
RAPID 2 study. A randomised controlled trial. Ann Rheum Dis 2009;68:797–804.
Singh JA, Christensen R, Wells GA, Suarez-Almazor ME, Buchbinder R, Lopez-Olivo
MA, et al. Biologics for rheumatoid arthritis: an overview of Cochrane reviews.
Cochrane Database Syst Rev 2009 CD007848.
Karlsson JA, Kristensen LE, Kapetanovic MC, Gulfe A, Saxne T, Geborek P.
Treatment response to a second or third TNF-inhibitor in RA: results from the
South Swedish Arthritis Treatment Group Register. Rheumatology (Oxford)
2008;47:507–13.
Remy A, Avouac J, Gossec L, Combe B. Clinical relevance of switching to a second
tumour necrosis factor-alpha inhibitor after discontinuation of a first tumour
necrosis factor-alpha inhibitor in rheumatoid arthritis: a systematic literature
review and meta-analysis. Clin Exp Rheumatol 2011;29:96–103.
Cohen SB, Emery P, Greenwald MW, Dougados M, Furie RA, Genovese MC, et al.
Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor
therapy: results of a multicenter, randomized, double-blind, placebo-controlled,
phase III trial evaluating primary efficacy and safety at twenty-four weeks.
Arthritis Rheum 2006;54:2793–806.
Weinblatt M, Combe B, Covucci A, Aranda R, Becker JC, Keystone E. Safety of the
selective costimulation modulator abatacept in rheumatoid arthritis patients
receiving background biologic and nonbiologic disease-modifying antirheumatic
drugs: a one-year randomized, placebo-controlled study. Arthritis Rheum
2006;54:2807–16.
Genovese MC, Schiff M, Luggen M, Becker JC, Aranda R, Teng J, et al. Efficacy and
safety of the selective co-stimulation modulator abatacept following 2 years of
treatment in patients with rheumatoid arthritis and an inadequate response to
anti-tumour necrosis factor therapy. Ann Rheum Dis 2008;67:547–54.
Emery P, Keystone E, Tony HP, Cantagrel A, van Vollenhoven R, Sanchez A, et al. IL6 receptor inhibition with tocilizumab improves treatment outcomes in patients
with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals:
results from a 24-week multicentre randomised placebo-controlled trial. Ann
Rheum Dis 2008;67:1516–23.
Finckh A, Ciurea A, Brulhart L, Kyburz D, Moller B, Dehler S, et al. B cell depletion
may be more effective than switching to an alternative anti-tumor necrosis factor
agent in rheumatoid arthritis patients with inadequate response to anti-tumor
necrosis factor agents. Arthritis Rheum 2007;56:1417–23.
Finckh A, Ciurea A, Brulhart L, Moller B, Walker UA, Courvoisier D, et al. Which
subgroup of patients with rheumatoid arthritis benefits from switching to
rituximab versus alternative anti-tumour necrosis factor (TNF) agents after
previous failure of an anti-TNF agent? Ann Rheum Dis 2010;69:387–93.
Jacobi AM, Dorner T. Current aspects of anti-CD20 therapy in rheumatoid arthritis.
Curr Opin Pharmacol 2010;10:316–21.
Emery P, Deodhar A, Rigby WF, Isaacs JD, Combe B, Racewicz AJ, et al. Efficacy and
safety of different doses and retreatment of rituximab: a randomised, placebocontrolled trial in patients who are biological naive with active rheumatoid
arthritis and an inadequate response to methotrexate (Study Evaluating
Rituximab's Efficacy in MTX iNadequate rEsponders (SERENE)). Ann Rheum Dis
2010;69:1629–35.
Rubbert-Roth A, Tak PP, Zerbini C, Tremblay JL, Carreno L, Armstrong G, et al.
Efficacy and safety of various repeat treatment dosing regimens of rituximab in
patients with active rheumatoid arthritis: results of a Phase III randomized study
(MIRROR). Rheumatology (Oxford) 2010.
http://clinicaltrials.gov/ct2/show/NCT01309282?term=portsmab&rank=1.
Kremer JM, Blanco R, Brzosko M, Burgos-Vargas R, Halland AM, Vernon E, et al.
Tocilizumab inhibits structural joint damage in rheumatoid arthritis patients with
inadequate responses to methotrexate: results from the double-blind treatment
phase of a randomized placebo-controlled trial of tocilizumab safety and prevention
of structural joint damage at one year. Arthritis Rheum 2011;63:609–21.
Platt AM, Gibson VB, Patakas A, Benson RA, Nadler SG, Brewer JM, et al. Abatacept
limits breach of self-tolerance in a murine model of arthritis via effects on the
generation of T follicular helper cells. J Immunol 2010.
Emery P, Durez P, Dougados M, Legerton CW, Becker JC, Vratsanos G, et al. Impact
of T-cell costimulation modulation in patients with undifferentiated inflammatory
arthritis or very early rheumatoid arthritis: a clinical and imaging study of
abatacept (the ADJUST trial). Ann Rheum Dis 2010;69:510–6.
Emery P, Breedveld FC, Hall S, Durez P, Chang DJ, Robertson D, et al. Comparison of
methotrexate monotherapy with a combination of methotrexate and etanercept
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]
[81]
[82]
[83]
[84]
[85]
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94]
[95]
[96]
[97]
713
in active, early, moderate to severe rheumatoid arthritis (COMET): a randomised,
double-blind, parallel treatment trial. Lancet 2008;372:375–82.
Breedveld FC, Weisman MH, Kavanaugh AF, Cohen SB, Pavelka K, van Vollenhoven
R, et al. The PREMIER study: a multicenter, randomized, double-blind clinical trial
of combination therapy with adalimumab plus methotrexate versus methotrexate
alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis
who had not had previous methotrexate treatment. Arthritis Rheum 2006;54:
26–37.
Smolen JS, Van Der Heijde DM, St Clair EW, Emery P, Bathon JM, Keystone E, et al.
Predictors of joint damage in patients with early rheumatoid arthritis treated with
high-dose methotrexate with or without concomitant infliximab: results from the
ASPIRE trial. Arthritis Rheum 2006;54:702–10.
van der Kooij SM, le Cessie S, Goekoop-Ruiterman YP, de Vries-Bouwstra JK, van
Zeben D, Kerstens PJ, et al. Clinical and radiological efficacy of initial vs delayed
treatment with infliximab plus methotrexate in patients with early rheumatoid
arthritis. Ann Rheum Dis 2009;68:1153–8.
Soubrier M, Puechal X, Sibilia J, Mariette X, Meyer O, Combe B, et al. Evaluation of
two strategies (initial methotrexate monotherapy vs its combination with
adalimumab) in management of early active rheumatoid arthritis: data from the
GUEPARD trial. Rheumatology (Oxford) 2009;48:1429–34.
Jones G, Sebba A, Gu J, Lowenstein MB, Calvo A, Gomez-Reino JJ, et al. Comparison
of tocilizumab monotherapy versus methotrexate monotherapy in patients with
moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis
2010;69:88–96.
Westhovens R, Robles M, Ximenes AC, Nayiager S, Wollenhaupt J, Durez P, et al.
Clinical efficacy and safety of abatacept in methotrexate-naive patients with early
rheumatoid arthritis and poor prognostic factors. Ann Rheum Dis 2009;68:
1870–7.
Tak PP, Rigby WF, Rubbert-Roth A, Peterfy CG, van Vollenhoven RF, Stohl W, et al.
Inhibition of joint damage and improved clinical outcomes with rituximab plus
methotrexate in early active rheumatoid arthritis: the IMAGE trial. Ann Rheum Dis
2011;70:39–46.
van Vollenhoven RF. Treatment of rheumatoid arthritis: state of the art 2009. Nat
Rev Rheumatol 2009;5:531–41.
Alivernini S, Laria A, Gremese E, Zoli A, Ferraccioli G. ACR70-disease activity score
remission achievement from switches between all the available biological agents
in rheumatoid arthritis: a systematic review of the literature. Arthritis Res Ther
2009;11:R163.
http://clinicaltrials.gov/ct2/show/NCT00966875?term=IL+17&rank=11.
Genovese MC, Van den Bosch F, Roberson SA, Bojin S, Biagini IM, Ryan P, et al.
LY2439821, a humanized anti-interleukin-17 monoclonal antibody, in the
treatment of patients with rheumatoid arthritis: a phase I randomized, doubleblind, placebo-controlled, proof-of-concept study. Arthritis Rheum 2010;62:
929–39.
http://clinicaltrials.gov/ct2/show/NCT00718718?term=IL6+antibody+
arthritis&rank=3.
Cornish AL, Campbell IK, McKenzie BS, Chatfield S, Wicks IP. G-CSF and GM-CSF as
therapeutic targets in rheumatoid arthritis. Nat Rev Rheumatol 2009;5:554–9.
http://clinicaltrials.gov/ct2/show/NCT01023256?term=GM+CSF+arthritis&
rank=1.
http://clinicaltrials.gov/ct2/show/NCT00995449?term=GM+CSF+arthritis&
rank=2.
http://clinicaltrials.gov/ct2/show/NCT01050998?term=GM+CSF+arthritis&
rank=3.
Kausar F, Mustafa K, Sweis G, Sawaqed R, Alawneh K, Salloum R, et al.
Ocrelizumab: a step forward in the evolution of B-cell therapy. Expert Opin Biol
Ther 2009;9:889–95.
Milani C, Castillo J. Veltuzumab, an anti-CD20 mAb for the treatment of nonHodgkin's lymphoma, chronic lymphocytic leukemia and immune thrombocytopenic purpura. Curr Opin Mol Ther 2009;11:200–7.
Ostergaard M, Baslund B, Rigby W, Rojkovich B, Jorgensen C, Dawes PT, et al.
Ofatumumab, a human anti-CD20 monoclonal antibody, for treatment of
rheumatoid arthritis with an inadequate response to one or more diseasemodifying antirheumatic drugs: results of a randomized, double-blind, placebocontrolled, phase I/II study. Arthritis Rheum 2010;62:2227–38.
Riese RJ, Krishnaswami S, Kremer J. Inhibition of JAK kinases in patients with
rheumatoid arthritis: scientific rationale and clinical outcomes. Best Pract Res Clin
Rheumatol 2010;24:513–26.
Boers M. Syk kinase inhibitors for rheumatoid arthritis: trials and tribulations.
Arthritis Rheum 2011;63:329–30.
Deodhar A, Dore RK, Mandel D, Schechtman J, Shergy W, Trapp R, et al.
Denosumab-mediated increase in hand bone mineral density associated with
decreased progression of bone erosion in rheumatoid arthritis patients. Arthritis
Care Res (Hoboken) 2010;62:569–74.
Tyndall A, van Laar JM. Stem cells in the treatment of inflammatory arthritis. Best
Pract Res Clin Rheumatol 2010;24:565–74.
Farge D, Labopin M, Tyndall A, Fassas A, Mancardi GL, Van Laar J, et al. Autologous
hematopoietic stem cell transplantation for autoimmune diseases: an observational study on 12 years' experience from the European Group for Blood and
Marrow Transplantation Working Party on Autoimmune Diseases. Haematologica
2010;95:284–92.
Bouffi C, Djouad F, Mathieu M, Noel D, Jorgensen C. Multipotent mesenchymal
stromal cells and rheumatoid arthritis: risk or benefit? Rheumatology (Oxford)
2009;48:1185–9.