Sei sulla pagina 1di 9

RHEUMATOLOGY Rheumatology 2012;51:vi28–vi36

doi:10.1093/rheumatology/kes278

Evolution of treatment for rheumatoid arthritis


Katherine S. Upchurch1 and Jonathan Kay1

Abstract
Treatment for RA has changed profoundly over the past 25 years, evolving from a strategy of providing
symptomatic relief, to implementation of therapeutic regimens that impact disease activity and ultimately
have been shown to slow or arrest structural joint damage. Drug therapy for RA has evolved from sali-
cylates, to NSAIDs, CSs, DMARDs, MTX, and finally to biologic response modifiers. MTX has become the
initial drug of choice in most patients with RA, and some do well on MTX monotherapy without the
addition of other agents. Combination regimens including MTX and other conventional DMARDs may
be an effective early approach to treatment of RA. The biologic response modifiers (biologics) became
available in the late 1990s, based on our understanding of the molecular mediators of synovial inflamma-
tion in RA. The first biologics inhibited TNF-a, a cytokine active in host defences against some infections
and malignancies, but which also promotes inflammation and bone erosion. Inhibitors of TNF-a are mostly
given with MTX, although some can be given as monotherapy. Studies consistently show that combination
MTX + TNF-a inhibitor therapy leads to better outcomes than with either agent alone. Tight control stra-
tegies, employing objective measures, also lead to improved outcomes. When patients fail treatment with
one or more TNF-a inhibitor + MTX, a number of other possible alternatives may be tried, including treat-
ment with biologics having other mechanisms, such as antibodies to certain ILs, other cytokines and
inflammatory mediators. Current therapy for RA is such that progression from symptom onset to signifi-
cant disability is now no longer inevitable, and RA patients can anticipate comfortable and productive lives
on medical therapy.
Key words: rheumatoid arthritis, salicylates, NSAIDs, DMARDs, methotrexate, biologic response modifiers,
interleukins, cytokines.

Introduction could impact disease activity in a meaningful way.


These therapies subsequently expanded to include other
Both the objectives and the results of treatment for RA agents (including parenteral gold salts, SSZ, chloroquine,
have changed profoundly over the past 25 years, dictated HCQ, D-Pen, ciclosporin and AZA), and these drugs were
largely by an enhanced understanding of the pathogen- optimistically termed DMARDs. The approach recom-
esis of this disease. During the first half of the 20th cen- mended to treat patients with newly diagnosed RA was
tury, RA treatment regimens included drugs that could pyramidal: initially with analgesics, then with NSAIDs and
provide only symptomatic benefit (salicylates, from ultimately with DMARDs, an approach that led to improve-
which were derived NSAIDs), analgesics and physical ment in some—but not all—RA patients, with reduced
measures such as bed rest, splinting and physical ther- symptoms and in some cases decreased disease activity.
apy. Following the early reports of the beneficial effects of Large early clinical trials confirming DMARD reduction in
gold salts in many patients with RA [1] and the subsequent the progression of structural joint damage, however, were
discovery of the efficacy of CSs in RA [2], rheumatologists initially lacking.
for the first time had access to drugs that they hoped During the 1980s, in addition to new drugs to treat RA,
new measurements were developed to assess the out-
1
Division of Rheumatology, Department of Medicine, University of
comes of therapeutic intervention. The application of
Massachusetts Medical School, Worcester, MA, USA. these initial outcome measures, such as the Sharp radio-
Submitted 10 January 2012; revised version accepted graphic scoring system [3], to assess response to early
14 September 2012. DMARDs prescribed according to the traditional treat-
Correspondence to: Katherine S. Upchurch, Division of Rheumatology, ment pyramid led to the recognition that many of these
Department of Medicine, University of Massachusetts Medical School,
55 Lake Avenue North, Worcester, MA 01605, USA. drugs did not, in fact, modify the course of the disease.
E-mail: katherine.upchurch@umassmed.edu In addition, it was observed that (i) the early DMARDs

! The Author 2012. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For Permissions, please email: journals.permissions@oup.com
Evolution of treatment for RA

were often poorly tolerated for longer than 2 years, erosions). Combination DMARDs (two or more DMARDs,
(ii) structural damage often became evident on radio- most of which are MTX based) were recommended for all
graphs within the first 2 years after the onset of RA [4] others with early disease, underscoring this approach in
and (iii) low-dose weekly oral MTX, a drug widely used patients before the initiation of biologic response modifiers
to treat psoriasis, not only was effective in treating PsA [30].
but also was both safe and well tolerated when given to
patients with RA [5, 6]. Taken together, these observations
led to US Food and Drug Administration (FDA) approval of
Current treatment practice: biologic
MTX for the treatment of RA in 1988 and to inversion of response modifiers
the traditional pyramidal approach to the medical man- In the late 1990s, a sea change took place in the treatment
agement of RA [5–8]. of RA, with the introduction of biologic targeted therapies,
By the 1990s, MTX had become the initial drug of choice also called biologic response modifiers. The development
of most rheumatologists to treat patients with RA in the of these therapeutic proteins was the culmination of re-
USA, although early efficacy studies suggested that, at search over the previous two decades that had elucidated
least within the first year of use, other drugs, including key molecular mediators of the inflammatory process that
SSZ, were equally efficacious [9]. Efforts were devoted to drives RA and results in structural damage to joints. These
study approaches by which to improve the tolerability of agents include mAbs and genetically engineered proteins
higher MTX doses (such as through s.c. administration), directed against cytokines or cell-surface molecules. The
thereby increasing its efficacy, and patients with RA were earliest agents inhibited the biological activity of TNF-a, a
often able to tolerate MTX for sustained periods of time, cytokine known not only to contribute to host defence
which was not usually the case with earlier DMARDs against infection and certain malignancies, but also to be
[10–14]. In spite of this, some RA patients were unable to key in perpetuating the inflammatory response in RA, which
tolerate the dose required to achieve maximal benefit or leads to synovial proliferation and bony destruction [31, 32].
remained MTX inadequate responders. Additionally, it To date, five drugs that inhibit TNF-a biological activity have
was contraindicated in others, such as young women con- been approved in the USA for clinical use in the treatment
templating pregnancy or those who used alcohol regularly. of patients with RA, each of which has been shown in rigor-
Furthermore MTX alone did not typically result in drug-free ous testing to improve outcomes, while reducing disease
remission; sustained MTX treatment was required to main- activity and structural joint damage. These include, in order
tain a clinical response and many patients had persistent of FDA approval, etanercept, infliximab, adalimumab, cer-
disease activity even while on this drug [15]. A number of tolizumab pegol and golimumab. Although subtly different
early studies supported the possibility that combination (route of administration, dose interval, chemical structure,
therapy with DMARDs ± CSs might prove more useful for example), these drugs are similar in efficacy and side-
than monotherapy with MTX alone [16, 17], and this is a effect profile, though the experience with certolizumab
line of investigation that continues nearly 20 years later. To pegol and golimumab is more recent and therefore not as
review the body of literature devoted to the different non- extensively studied as that of earlier agents. Treatment with
biologic treatment options for both early and established TNF inhibitors often dramatically improves RA disease ac-
(active) RA is beyond the scope of this introduction. tivity and, in patients who respond, may slow or arrest dis-
Emerging from these studies, however, has been the gen- ease progression as assessed by clinical, radiographic and
eral conclusion that combinations of traditional DMARDs patient-reported outcome measures. TNF inhibitors are
are both safe and effective in many patients with RA and generally used in combination with MTX. Although most
thus constitute a reasonable approach to early treatment of TNF inhibitors are effective as monotherapy, studies of
disease [18–27]. Although monotherapy with MTX is effect- TNF inhibitors used in combination with MTX have demon-
ive in many patients with newly diagnosed active RA strated consistently that both the clinical and structural out-
[26–28], based on health economic modelling comes of combination therapy are superior to those
(cost-effectiveness), the National Institute for Health and achieved with either agent alone. As a class, TNF inhibitors
Clinical Excellence (NICE) in the UK recommends a com- are generally well tolerated; however, adverse effects such
bination of DMARDs plus short-term glucocorticoids as as decreased resistance to both routine and opportunistic
first-line therapy as soon as possible after onset of symp- infections can be devastating and must be aggressively
toms, but ideally within 3 months of the onset of persistent sought and treated [33].
symptoms of RA in appropriate patients who do not have a Following the introduction of the first TNF inhibitors,
contraindication to this approach [29]. More recently, in the biologic response modifiers targeting other components
2012 update of previously published guidelines, the ACR, of the immune response involved in the pathogenesis of
with a target of low disease activity or remission, recom- RA have been approved for clinical use. Anakinra, a re-
mends DMARD monotherapy (agent not specified) in pa- combinant human soluble IL-1 receptor antagonist, can
tients with disease duration of <6 months with low disease be administered subcutaneously daily to treat patients
activity (activity defined by accepted DAS ranges) and for with RA. Abatacept, a recombinant fusion protein that
moderate or high disease activity without poor prognostic combines the T-cell co-stimulation inhibitory molecule
features (presence of one or more of functional limitation, CTLA-4 with the Fc region of human IgG, may be given
extra-articular disease, positive RF or ACPAs, and bony subcutaneously weekly or intravenously monthly.

www.rheumatology.oxfordjournals.org vi29
Katherine S. Upchurch and Jonathan Kay

Rituximab, a chimeric mAb directed against the surface optimal place of agents in the treatment algorithm. When
molecule CD20 on B cells, is administered in two i.v. should the addition of a biologic be considered after initi-
doses given 2 weeks apart, usually every 6 months. ation of MTX therapy? Is 3 months an adequate trial of MTX
Most recently, tocilizumab, a humanized mAb directed monotherapy? Should combination agents be tried before
against the IL-6 receptor, has been approved for monthly turning to biologics and are there patients for whom bio-
i.v. administration to patients with RA. Specifics regarding logic therapy should commence early in the course of their
the biologic response modifiers approved for the treat- disease to improve outcomes? The 2012 update of the
ment of RA are summarized in Table 1 [34–63]. 2008 ACR recommendations for the use of disease-
modifying antirheumatic drugs and biologic agents in the
treatment of rheumatoid arthritis recommends targeting
Strategies in patients who have low disease activity or remission, that patients with estab-
responded inadequately to one or lished RA should receive 3 months of DMARD therapy
more TNF inhibitors before adding or switching between DMARDs or switching
from DMARDs to biologic agents [30].
Several possible strategies may be tried when a patient
Another issue is that of agent selection. Individualized
has not responded to treatment with MTX and a TNF in-
therapy using pharmacogenetics or certain patient char-
hibitor. Expert opinion does not point to one strategy as
acteristics as a predictor of response to guide therapy
being superior to the others; all treatment plans are
selection is under investigation. For example, patients
guided by observation of the individual patient’s
who are positive for RF or ACPA are more likely to re-
response, combined with clinical judgement. Of the pos-
spond to rituximab than patients who lack these antibo-
sible approaches, two deserve careful consideration:
dies. The identification of additional biomarkers that might
(i) sequential TNF inhibitor use (i.e. trying a second TNF
guide medication choices and the likelihood of response
inhibitor if the first one has failed); and (ii) switching to a
to therapy will be useful in this regard in the future. Until
biologic DMARD with a different mechanism of action.
then, the choice of therapy should be based on the effi-
Both of these strategies are supported by efficacy and
cacy and safety profile of each agent, patient preference
safety data from randomized, placebo-controlled clinical
concerning dosing and route of administration and, of
trials (Table 1) [34–63].
course, third-party reimbursement. Clinicians are con-
The results of switching from one TNF inhibitor to a
strained in their decision-making capacity by the limited
second TNF inhibitor were assessed in an analysis of
amount of data on the comparative benefits and risks of
data from the British Society for Rheumatology Biologics the various agents currently available. Fortunately a
Registry, using rates of discontinuation due either to lack number of trials are currently under way to address
of efficacy or to adverse events (AEs) [64]. Of 6739 pa- these important questions and may bring answers.
tients, 35% discontinued their first TNF inhibitor. Of these,
856 switched to a second TNF inhibitor. At the end of 30
months of follow-up, 73% of patients who had switched
Future therapies for RA
to a second TNF inhibitor continued on treatment. In addition to those targets for which drug therapies have
Patients who discontinued the initial TNF inhibitor be- been approved, there are other potential targets for drugs
cause of lack of efficacy were more likely to have discon- to treat RA, including ILs such as IL-17 and receptor tyro-
tinued the second TNF inhibitor for the same reason; sine kinases. Of these, a target that has yielded promising
similarly, patients who discontinued the first TNF inhibitor results is JAK3, one of the Janus kinases (JAKs), which
because of AEs were more likely to have discontinued the mediates signal transduction of cell surface receptors for
second TNF inhibitor because of another AE. cytokines involved in the pathogenesis of inflammatory
An alternative to switching between TNF inhibitors is to diseases such as RA. An oral inhibitor of this enzyme,
initiate treatment with another biologic response modifier tofacitinib (formerly CP-690,550) has demonstrated effi-
that has a different mechanism of action. Retrospective cacy in several trials conducted in patients with RA:
analysis of 116 patients from the Swiss Clinical Quality those who had failed at least one earlier DMARD [66],
Management Program for Rheumatoid Arthritis cohort those with disease activity despite MTX therapy [67, 68]
who had an inadequate response to at least one TNF in- and those previously exposed to TNF inhibitors [69].
hibitor revealed greater improvement in DAS28 at 3, 6 and Tofacitinib has been studied both as monotherapy [66,
9 months among the 50 patients who had subsequently 70] and in combination with MTX [67–69]. Fostamatinib,
received two infusions of rituximab 1000 mg with con- an oral inhibitor of spleen tyrosine kinase (Syk), which is
comitant i.v. glucocorticoids, 14 days apart, as compared an intracellular non-receptor tyrosine kinase, has demon-
with the 66 patients who had switched to treatment with a strated efficacy superior to that of placebo when given in
second or third TNF inhibitor [65]. combination with MTX to patients inadequately respon-
sive to MTX [71]. However, fostamatinib was not superior
Current issues in the management of RA to placebo when given in addition to stable DMARD ther-
apy to patients inadequately responsive to a TNF inhibitor
With the rapidly increasing number of biologic options [66]. A potentially important characteristic of these tyro-
available to treat patients with RA, a number of important sine kinase inhibitors is that each can be taken orally
questions have arisen. The first question is that of the rather than by injection or as an infusion, which may

vi30 www.rheumatology.oxfordjournals.org
TABLE 1 Summary of biologic response modifiers approved for the treatment of RA [33–63]

Agent Mechanism of action Administration and dosage Indications Studies for RA indication

Etanercept Cloned fusion protein of human s.c. injection; 50 mg weekly RA (with MTX or alone), ETN monotherapy [34]
TNF-a receptor 2 with Fc region JIA, PsA ETN + MTX in MTX-refractory RA [35]
of human IgG1; inhibits TNF-a ETN vs MTX in patients with ERA [36]
binding to its receptors TEMPO (ETN + MTX vs ETN or MTX alone) [37]
COMET (ETN + MTX vs MTX alone) [38]
Infliximab Chimeric mAb to TNF-a; inhibits i.v. infusion; for patients with RA, RA (with MTX only), ATTRACT (IFX + MTX in MTX-refractory RA) [39]
TNF-a binding to its receptors dosing is 3 mg/kg at weeks 0, 2 Crohn’s disease, UC, ASPIRE (IFX + MTX in MTX-naı̈ve patients) [40]

www.rheumatology.oxfordjournals.org
and 6, then every 8 weeks; AS, PsA, PsO START (IFX + MTX in MTX-refractory patients with
some patients may benefit from co-morbidities) [41]
doses up to 10 mg/kg or given
as frequently as every 4 weeks
Adalimumab Humanized mAb to TNF-a; s.c. injection; 40 mg every other RA (with MTX or alone), ARMADA (ADA + MTX in patients with
inhibits TNF-a binding to its week; when used alone, some JIA, PsA, AS, Crohn’s DMARD-refractory RA) [42]
receptors patients may benefit from disease, PsO DE011 (ADA alone in patients with
dosing at 40 mg weekly DMARD-refractory RA) [43]
DE019 (ADA + MTX in patients with MTX-refractory
RA) [44]
STAR (ADA + DMARD in inadequate responders to
standard treatment) [45]
PREMIER (ADA alone or + MTX in MTX-naı̈ve
patients with ERA <3 years’ disease duration)
[46]
Golimumab Human mAb to TNF-a; inhibits s.c. injection; 50 mg monthly RA (with MTX); PsA, AS Phase 2 (GLM+ MTX in MTX-refractory RA) [47]
TNF-a binding to its receptors GO-AFTER (GLM ± DMARD in patients treated with
one or more TNF inhibitor) [48]
GO-FORWARD (GLM alone or + MTX in patients
with MTX-refractory RA) [49]
GO-BEFORE 0GLM + MTX in MTX-naı̈ve patients
with early-onset RA) [50]
Certolizumab Pegylated Fab0 fragment of a s.c. injection; 400 mg initially, RA, Crohn’s disease RAPID 1 (CZP + MTX in patients with
pegol humanized mAb to TNF-a; then 200 mg every other week; MTX-refractory RA) [51]
inhibits TNF-a binding to its 400 mg every 4 weeks may be RAPID 2 (CZP + MTX in patients with
receptors considered for maintenance MTX-refractory RA) [52]
dosing FAST4WARD (CZP alone in patients previously
treated with 51 DMARD) [53]
Abatacept Cloned fusion protein of i.v. infusion; in adult patients with RA, JIA ATTAIN (ABA alone in patients with an inadequate
extracellular domain of human RA: <60 kg, 500 mg; 60–100 kg, response to one or more TNF inhibitor) [54, 55]
CTLA-4 with the hinge, CH2 750 mg; >100 kg, 1000 mg, AIM (ABA alone in patients with
and CH3 domains of human following initial infusion at MTX-refractory RA) [56, 57]
IgG1; inhibits T-cell weeks 2 and 4, thereafter every ABA s.c. vs i.v. in patients with MTX-refractory RA
co-stimulation by 4 weeks OR after a single i.v. [58]
antigen-presenting cells infusion as a loading dose, s.c.
injection, 125 mg, should be
given within a day, followed by
125 mg weekly for maintenance

vi31
Evolution of treatment for RA

dosing
(continued)
vi32
TABLE 1 Continued

Agent Mechanism of action Administration and dosage Indications Studies for RA indication

Rituximab Chimeric mouse/human mAb to Two 1000 mg i.v. infusions 2 RA with MTX in adult DANCER (RTX + MTX in patients with RA and in-
CD20 antigens on B weeks apart every 24 weeks; patients with RA and adequate response to DMARDs including MTX)
lymphocytes based on clinical evaluation not inadequate response [59]
more frequently than every 16 to ne or more TNF REFLEX (RTX + MTX in patients with RA and inad-
Katherine S. Upchurch and Jonathan Kay

weeks inhibitor, NHL, CLL, equate response to one ore more TNF inhibitor
granulomatosis with [60]
polyangiitis
Tocilizumab Recombinant humanized mAb to i.v. infusion; in patients with RA, RA with inadequate OPTION (TCZ + MTX in patients with active RA) [61]
IL-6R, blocking signalling and 8 mg/kg every 4 weeks response to existing RADIATE (TCZ + MTX in patients with RA refractory
activation of B cells therapies, polyarticu- to One or more TNF inhibitor) [62]
lar JIA, systemic JIA, ROSE (TCZ + background DMARDs in patients with
symptomatic inadequate response to DMARDs [63]
treatment of
Castleman disease

ABA: abatacept; ADA: adalimumab; AIM: Abatacept in Inadequate Responders to Methotrexate; ARMADA: Anti-TNF Research Study Program of the Monoclonal Antibody D2E7 in
patients with Rheumatoid Arthritis; ASPIRE: Active Controlled Study of Patients Receiving Infliximab for Treatment of Rheumatoid Arthritis of Early Onset; ATTAIN: Abatacept Trial in
Treatment of Anti-TNF Inadequate Responders; ATTRACT: Anti-TNF Trial in Rheumatoid Arthritis With Concomitant Therapy; COMET: Combination of Methotrexate and Etanercept in
Active Early Rheumatoid Arthritis; CLL: chronic lymphocytic leukaemia; CZP: certolizumab pegol; DANCER: Dose-Ranging Assessment: International Clinical Evaluation of Rituximab in
Rheumatoid Arthritis; ETN: etanercept; ERA: early RA; FAST4WARD: Efficacy and Safety of Certolizumab Pegol—4 Weekly Dosage in Rheumatoid Arthritis; GLM: golimumab;
GO-AFTER: Golimumab in Patients With Active Rheumatoid Arthritis After Treatment With Tumour Necrosis Factor Alpha Inhibitors; GO-BEFORE: Golimumab Before Employing
Methotrexate as the First-line Option in the Treatment of Rheumatoid Arthritis of Early Onset; GO-FORWARD: Golimumab for Subjects With Active RA Despite MTX; NHL: non-Hodgkin
lymphoma; IFX: infliximab; OPTION: Tocilizumab Pivotal Trial in Methotrexate Inadequate Responders; PsO: plaque psoriasis; RADIATE: Rheumatoid Arthritis Study in Anti-TNF
Failures; RAPID: Rheumatoid Arthritis Prevention of Structural Damage; REFLEX: Randomized Evaluation of Long-term Efficacy of Rituximab in RA; ROSE: Rapid Onset and Systemic
Efficacy; RTX: rituximab; STAR: Safety Trial of Adalimumab in Rheumatoid Arthritis; START: Safety Trial for Rheumatoid Arthritis With Remicade Therapy; TCZ: tocilizumab; TEMPO:
Trial of Etanercept and Methotrexate With Radiographic Patient Outcomes; UC: ulcerative colitis.

www.rheumatology.oxfordjournals.org
Evolution of treatment for RA

impact patient acceptance of these drugs. Additional Nordisk Inc., Pfizer Inc. and UCB SA and has received re-
agents targeting signal transduction kinases including search support paid to the University of Massachusetts
other JAKs, cytokines other than TNF-a, IL-1 and IL-6, Medical School from Abbott Laboratories, Ardea Inc., F.
and various inflammatory mediators are in development Hoffmann-La Roche Ltd, Fidia Farmaceutici s.p.a. and
and may augment the therapeutic armamentarium avail- sanofi-aventis US LLC. K.S.U. has been a consultant for
able to manage patients with RA. These agents may bring Human Genome Sciences.
additional options to patients who fail to respond to cur-
rently available biologic response modifiers.
References
Conclusion 1 Forestier J. The treatment of rheumatoid arthritis with gold
salts. Lancet 1932;219:441–4.
The advances in RA treatment over the past 25 years have 2 Hench PS, Kendall EC et al. The effect of a hormone of the
been profound. Previously the progression of RA from adrenal cortex (17-hydroxy-11-dehydrocorticosterone;
symptom onset to significant disability was often inevit- compound E) and of pituitary adrenocorticotropic hor-
able and, in some cases, rapid. Now, with the availability mone on rheumatoid arthritis. Proc Staff Meet Mayo Clin
of medications that can slow or halt disease progression 1949;24:181–97.
and prevent irreversible joint damage, joint replacement 3 Sharp JT, Bluhm GB, Brook A et al. Reproducibility of
surgery is not always the ultimate outcome and patients multiple-observer scoring of radiologic abnormalities in
with RA may live comfortable and productive lives on the hands and wrists of patients with rheumatoid arthritis.
medical therapy. However, the biologic response modi- Arthritis Rheum 1985;28:16–24.
fiers are expensive and may be beyond the financial 4 Wolfe F, Sharp JT. Radiographic outcome of recent-onset
means of some patients who are in need of these effective rheumatoid arthritis: a 19-year study of radiographic pro-
treatments. The development of biosimilars over the next gression. Arthritis Rheum 1998;41:1571–82.
several years may help to provide more affordable ver- 5 Weinblatt ME, Coblyn JS, Fox DA et al. Efficacy of
sions of these successful therapies [72]. low-dose methotrexate in rheumatoid arthritis. N Engl J
Currently available drug therapy for RA has made Med 1985;312:818–22.
remission a feasible treatment goal. Patients with RA 6 Kremer JM, Lee JK. The safety and efficacy of the use of
should be diagnosed early in their disease course and be methotrexate in long-term therapy for rheumatoid arthritis.
assessed regularly using objective quantitative measures Arthritis Rheum 1986;29:822–31.
of disease activity. The recent revision of the definition of 7 Wilske KR, Healey LA. Remodeling the pyramid—a con-
remission in RA by a joint ACR/European League Against cept whose time has come. J Rheumatol 1989;16:565–7.
Rheumatism (EULAR) committee exemplifies the improved
8 Wilske KR. Inverting the therapeutic pyramid: observa-
outcomes that are achievable with the advances in RA
tions and recommendations on new directions in
treatment made over the past quarter century [73]. The rheumatoid arthritis therapy based on the author’s
life-limiting and, in some cases, life-shortening conse- experience. Semin Arthritis Rheum 1993;23(2 Suppl. 1):
quences of RA are no longer predestined. Thus patients 11–8.
with RA can now expect to experience a quality of life that
9 Felson DT, Anderson JJ, Meenan RF. The comparative
previously was unavailable to patients during the 20th efficacy and toxicity of second-line drugs in rheumatoid
century. arthritis. Results of two metaanalyses. Arthritis Rheum
1990;33:1449–61.
Rheumatology key messages 10 Pincus T, Marcum SB, Callahan LF. Longterm drug ther-
apy for rheumatoid arthritis in seven rheumatology private
. DMARDs modify disease activity, but not structural practices: II. Second line drugs and prednisone. J
outcomes, in many RA patients. Rheumatol 1992;19:1885–94.
. TNF inhibitors, alone or with MTX, may improve
11 Kremer JM, Phelps CT. Long-term prospective study of
outcomes in most RA patients.
the use of methotrexate in the treatment of rheumatoid
. Other biologic agents should be tried in RA patients
arthritis. Update after a mean of 90 months. Arthritis
with inadequate response to TNF inhibitors.
Rheum 1992;35:138–45.
12 Weinblatt ME, Maier AL, Fraser PA, Coblyn JS. Longterm
Supplement: This paper forms part of the supplement prospective study of methotrexate in rheumatoid arthritis:
‘Reducing the toll of autoimmune disease: a focus on conclusion after 132 months of therapy. J Rheumatol
rheumatoid arthritis’. This supplement was supported by 1998;25:238–42.
unrestricted education grants from Centocor and 13 Wluka A, Buchbinder R, Mylvaganam A et al. Longterm
Genentech. methotrexate use in rheumatoid arthritis: 12 year followup
of 460 patients treated in community practice. J
Disclosure statement: J.K. has been a consultant for Rheumatol 2000;27:1864–71.
Amgen, Bristol-Myers Squibb Company, Celgene Cor- 14 Hoekstra M, van de Laar MA, Bernelot Moens HJ,
poration, Crescendo BioScience Inc., F. Hoffmann-La Kruijsen MW, Haagsma CJ. Longterm observational study
Roche Ltd, Genentech Inc., Horizon Pharma, Janssen of methotrexate use in a Dutch cohort of 1022 patients
Pharmaceuticals Inc., Molecular Partners AG, Novo with rheumatoid arthritis. J Rheumatol 2003;30:2325–9.

www.rheumatology.oxfordjournals.org vi33
Katherine S. Upchurch and Jonathan Kay

15 Kremer JM. The changing face of therapy for rheumatoid 29 The National Collaborating Centre for Chronic Conditions.
arthritis. Rheum Dis Clin North Am 1995;21:845–52. Rheumatoid arthritis: national clinical guidelines for man-
16 O’Dell JR, Haire CE, Erikson N et al. Treatment of agement and treatment in adults. London: Royal College
rheumatoid arthritis with methotrexate alone, sulfasalazine of Physicians, 2009. http://www.nice.org.uk/nicemedia/
and hydroxychloroquine, or a combination of all three live/12131/43326/43326.pdf (20 March 2012, date last
medications. N Engl J Med 1996;334:1287–91. accessed).

17 Weinblatt ME, Kremer JM, Coblyn JS et al. 30 Singh JA, Furst DE, Bharat A et al. 2012 Update of the
Pharmacokinetics, safety, and efficacy of combination 2008 American College of Rheumatology recommenda-
treatment with methotrexate and leflunomide in patients tions for the use of disease-modifying antirheumatic drugs
with active rheumatoid arthritis. Arthritis Rheum 1999;42: and biologic agents in the treatment of rheumatoid arth-
1322–8. ritis. Arthritis Care Res 2012;64:625–39.

18 Korpela M, Laasonen L, Hannonen P et al. Retardation of 31 Criscione LG, St Clair EW. Tumor necrosis factor-alpha
joint damage in patients with early rheumatoid arthritis by antagonists for the treatment of rheumatic diseases. Curr
initial aggressive treatment with disease-modifying anti- Opin Rheumatol 2002;14:204–11.
rheumatic drugs: five-year experience from the FIN-RACo 32 Furst DE, Keystone EC, Braun J et al. Updated consensus
study. Arthritis Rheum 2004;50:2072–81. statement on biological agents for the treatment of
19 Mäkinen H, Kautiainen H, Hannonen P et al. Sustained rheumatic diseases, 2010. Ann Rheum Dis 2011;
remission and reduced radiographic progression 70(Suppl. 1):i2–36.
with combination disease modifying antirheumatic 33 Ruderman EM. Overview of safety of non-biologics and
drugs in early rheumatoid arthritis. J Rheumatol 2007;34: biologic DMARDs. Rheumatology 2012;51(Suppl. 6):
316–21. vi35–41.
20 Möttönen T, Hannonen P, Leirisalo-Repo M et al. 34 Moreland LW, Schiff MH, Baumgartner SW et al.
Comparison of combination therapy with single-drug Etanercept therapy in rheumatoid arthritis. A randomized,
therapy in early rheumatoid arthritis: a randomised trial. controlled trial. Ann Intern Med 1999;130:478–86.
Lancet 1999;353:1568–73. 35 Weinblatt ME, Kremer JM, Bankhurst AD et al. A trial of
21 Puolakka K. Impact of initial aggressive drug treatment etanercept, a recombinant tumor necrosis factor receptor:
with a combination of disease-modifying antirheumatic Fc fusion protein, in patients with rheumatoid arthritis
drugs on the development of work disability in early receiving methotrexate. N Engl J Med 1999;340:253–9.
rheumatoid arthritis: a five-year randomized followup trial. 36 Bathon JM, Martin RW, Fleischmann RM et al. A com-
Arthritis Rheum 2004;50:55–62. parison of etanercept and methotrexate in patients with
22 van Jaarsveld CH, Jacobs JW, van der Veen MJ et al. early rheumatoid arthritis. N Engl J Med 2000;343:
Aggressive treatment in early rheumatoid arthritis: a ran- 1586–93.
domised controlled trial. Ann Rheum Dis 2000;59:468–77. 37 Klareskog L, van der Heijde D, de Jager JP et al.
23 Boers M. Randomised comparison of combined Therapeutic effect of the combination of etanercept and
step-down prednisolone, methotrexate and sulphasala- methotrexate compared with each treatment alone in pa-
zine with sulphasalazine alone in early rheumatoid arthritis. tients with rheumatoid arthritis: double-blind randomised
Lancet 1997;350:309–18. controlled trial. Lancet 2004;363:675–81.
24 Landewé RB, Boers M, Verhoeven AC et al. COBRA 38 Emery P, Breedveld FC, Hall S et al. Comparison of
combination therapy in patients with early rheumatoid methotrexate monotherapy with a combination of metho-
arthritis: long-term structural benefits of a brief interven- trexate and etanercept in active, early, moderate to severe
tion. Arthritis Rheum 2002;46:347–56. rheumatoid arthritis (COMET): a randomised,
25 Capell HA, Madhok R, Porter DR et al. Combination ther- double-blind, parallel treatment trial. Lancet 2008;372:
apy with sulfasalazine and methotrexate is more effective 375–82.
than either drug alone in patients with rheumatoid arthritis 39 Lipsky PE, van der Heijde DM, St Clair EW et al. Infliximab
with a suboptimal response to sulfasalazine: results from and methotrexate in the treatment of rheumatoid arthritis.
the double-blind placebo-controlled MASCOT study. Ann Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis
Rheum Dis 2007;66:235–41. with Concomitant Therapy Study Group. N Engl J Med
26 Allaart CF, Goekoop-Ruiterman YP, de Vries-Bouwstra JK 2000;343:1594–602.
et al. Aiming at low disease activity in rheumatoid arthritis 40 St Clair EW, van der Heijde DM, Smolen JS et al.
with initial combination therapy or initial monotherapy Combination of infliximab and methotrexate therapy for
strategies: the BeSt study. Clin Exp Rheumatol 2006;24(6 early rheumatoid arthritis: a randomized, controlled trial.
Suppl. 43):1–77. Arthritis Rheum 2004;50:3432–43.
27 Goekoop-Ruiterman YP, de Vries BJK, Allaart CF et al. 41 Westhovens R, Houssiau F, Joly J et al. A phase I study
Comparison of treatment strategies in early rheumatoid assessing the safety, clinical response, and pharmaco-
arthritis: a randomized trial. Ann Intern Med 2007;146: kinetics of an experimental infliximab formulation
406–15. for subcutaneous or intramuscular administration in
28 Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF patients with rheumatoid arthritis. J Rheumatol 2006;33:
et al. Clinical and radiographic outcomes of four different 847–53.
treatment strategies in patients with early rheumatoid 42 Weinblatt ME, Keystone EC, Furst DE et al. Adalimumab, a
arthritis (the BeSt study): a randomized, controlled trial. fully human anti-tumor necrosis factor alpha monoclonal
Arthritis Rheum 2005;52:3381–90. antibody, for the treatment of rheumatoid arthritis in

vi34 www.rheumatology.oxfordjournals.org
Evolution of treatment for RA

patients taking concomitant methotrexate: the ARMADA rheumatoid arthritis: the RAPID 2 study. A randomised
trial. Arthritis Rheum 2003;48:35–45. controlled trial. Ann Rheum Dis 2009;68:797–804.
43 van de Putte LB, Atkins C, Malaise M et al. Efficacy and 53 Fleischmann R, Vencovsky J, van Vollenhoven RF et al.
safety of adalimumab as monotherapy in patients with Efficacy and safety of certolizumab pegol monotherapy
rheumatoid arthritis for whom previous disease modifying every 4 weeks in patients with rheumatoid arthritis failing
antirheumatic drug treatment has failed. Ann Rheum Dis previous disease-modifying antirheumatic therapy: the
2004;63:508–16. FAST4WARD study. Ann Rheum Dis 2009;68:805–11.
44 Keystone EC, Kavanaugh AF, Sharp JT et al. 54 Genovese MC, Becker JC, Schiff M et al. Abatacept for
Radiographic, clinical, and functional outcomes of treat- rheumatoid arthritis refractory to tumor necrosis factor
ment with adalimumab (a human anti-tumor necrosis alpha inhibition. N Engl J Med 2005;353:1114–23.
factor monoclonal antibody) in patients with active 55 Genovese MC, Schiff M, Luggen M et al. Efficacy and
rheumatoid arthritis receiving concomitant methotrexate safety of the selective co-stimulation modulator abatacept
therapy: a randomized, placebo-controlled, 52-week trial.
following 2 years of treatment in patients with rheumatoid
Arthritis Rheum 2004;50:1400–11.
arthritis and an inadequate response to anti-tumour ne-
45 Furst DE, Schiff MH, Fleischmann RM et al. Adalimumab, crosis factor therapy. Ann Rheum Dis 2008;67:547–54.
a fully human anti tumor necrosis factor-alpha monoclonal
56 Genant HK, Peterfy CG, Westhovens R et al. Abatacept
antibody, and concomitant standard antirheumatic ther-
inhibits progression of structural damage in rheumatoid
apy for the treatment of rheumatoid arthritis: results of
arthritis: results from the long-term extension of the AIM
STAR (Safety Trial of Adalimumab in Rheumatoid Arthritis).
trial. Ann Rheum Dis 2008;67:1084–9.
J Rheumatol 2003;30:2563–71.
57 Kremer JM, Russell AS, Emery P et al. Long-term safety,
46 Breedveld FC, Weisman MH, Kavanaugh AF et al. The
efficacy and inhibition of radiographic progression with
PREMIER study: a multicenter, randomized, double-blind
abatacept treatment in patients with rheumatoid arthritis
clinical trial of combination therapy with adalimumab plus
and an inadequate response to methotrexate: 3-year re-
methotrexate versus methotrexate alone or adalimumab
sults from the AIM trial. Ann Rheum Dis 2011;70:1826–30.
alone in patients with early, aggressive rheumatoid arth-
ritis who had not had previous methotrexate treatment. 58 Genovese MC, Covarrubias A, Leon G et al.
Arthritis Rheum 2006;54:26–37. Subcutaneous abatacept versus intravenous abatacept: a
phase IIIb noninferiority study in patients with an inad-
47 Kay J, Matteson EL, Dasgupta B et al. Golimumab in pa-
equate response to methotrexate. Arthritis Rheum 2011;
tients with active rheumatoid arthritis despite treatment
63:2854–64.
with methotrexate: a randomized, double-blind,
placebo-controlled, dose-ranging study. Arthritis Rheum 59 Emery P, Fleischmann R, Filipowicz-Sosnowska A et al.
2008;58:964–75. The efficacy and safety of rituximab in patients with active
rheumatoid arthritis despite methotrexate treatment: re-
48 Smolen JS, Kay J, Doyle MK et al. Golimumab in
sults of a phase IIB randomized, double-blind,
patients with active rheumatoid arthritis after treatment
placebo-controlled, dose-ranging trial. Arthritis Rheum
with tumour necrosis factor alpha inhibitors (GO-AFTER
2006;54:1390–400.
study): a multicentre, randomised, double-blind,
placebo-controlled, phase III trial. Lancet 2009;374: 60 Cohen SB, Emery P, Greenwald MW et al. Rituximab for
210–21. rheumatoid arthritis refractory to anti-tumor necrosis
factor therapy: results of a multicenter, randomized,
49 Keystone EC, Genovese MC, Klareskog L et al.
double-blind, placebo-controlled, phase III trial evaluating
Golimumab, a human antibody to tumour necrosis factor
primary efficacy and safety at twenty-four weeks. Arthritis
{alpha} given by monthly subcutaneous injections, in
Rheum 2006;54:2793–806.
active rheumatoid arthritis despite methotrexate therapy:
the GO-FORWARD Study. Ann Rheum Dis 2009;68: 61 Smolen JS, Beaulieu A, Rubbert-Roth A et al. Effect of
789–96. interleukin-6 receptor inhibition with tocilizumab in pa-
tients with rheumatoid arthritis (OPTION study): a
50 Emery P, Fleischmann RM, Moreland LW et al.
double-blind, placebo-controlled, randomised trial. Lancet
Golimumab, a human anti-tumor necrosis factor alpha
2008;371:987–97.
monoclonal antibody, injected subcutaneously every four
weeks in methotrexate-naive patients with active 62 Emery P, Keystone E, Tony HP et al. IL-6 receptor inhib-
rheumatoid arthritis: twenty-four-week results of a phase ition with tocilizumab improves treatment outcomes in
III, multicenter, randomized, double-blind, patients with rheumatoid arthritis refractory to anti-tumour
placebo-controlled study of golimumab before metho- necrosis factor biologicals: results from a 24-week multi-
trexate as first-line therapy for early-onset rheumatoid centre randomised placebo-controlled trial. Ann Rheum
arthritis. Arthritis Rheum 2009;60:2272–83. Dis 2008;67:1516–23.
51 Keystone E, van der Heijde D, Mason D Jr et al. 63 Yazici Y, Curtis JR, Ince A et al. Efficacy of tocilizumab in
Certolizumab pegol plus methotrexate is significantly patients with moderate to severe active rheumatoid arth-
more effective than placebo plus methotrexate in active ritis and a previous inadequate response to
rheumatoid arthritis: findings of a fifty-two-week, phase III, disease-modifying antirheumatic drugs: the ROSE study.
multicenter, randomized, double-blind, placebo- Ann Rheum Dis 2012;71:198–205.
controlled, parallel-group study. Arthritis Rheum 2008;58: 64 Hyrich KL, Lunt M, Watson KD, Symmons DP, Silman AJ.
3319–29. Outcomes after switching from one anti-tumor necrosis
52 Smolen J, Landewé RB, Mease P et al. Efficacy and safety factor alpha agent to a second anti-tumor necrosis factor
of certolizumab pegol plus methotrexate in active alpha agent in patients with rheumatoid arthritis: results

www.rheumatology.oxfordjournals.org vi35
Katherine S. Upchurch and Jonathan Kay

from a large UK national cohort study. Arthritis Rheum 69 Burmester GR, Blanco R, Charles-Schoeman C et al.
2007;56:13–20. Tofacitinib (CP-690,550), an oral Janus kinase inhibitor,
65 Finckh A, Ciurea A, Brulhart L et al. B cell depletion may be in combination with methotrexate, in patients with
more effective than switching to an alternative anti-tumor active rheumatoid arthritis with an inadequate response
necrosis factor agent in rheumatoid arthritis patients with to tumor necrosis factor-inhibitors: a 6-month phase
inadequate response to anti-tumor necrosis factor agents. 3 study [abstract]. Arthritis Rheum 2011;63(10 Suppl.):
Arthritis Rheum 2007;56:1417–23. S279.
66 Fleischmann R, Cutolo M, Genovese MC et al. Phase IIb 70 Fleischmann R, Kremer J, Cush J et al. Placebo-controlled
dose-ranging study of the oral JAK inhibitor tofacitinib
trial of tofacitinib monotherapy in rheumatoid arthritis. N
(CP-690,550) or adalimumab monotherapy versus pla-
Engl J Med 2012;367:495–507.
cebo in patients with active rheumatoid arthritis with an
inadequate response to DMARDs. Arthritis Rheum 2012; 71 Weinblatt ME, Kavanaugh A, Genovese MC et al. An oral
64:617–29. spleen tyrosine kinase (Syk) inhibitor for rheumatoid arth-
ritis. N Engl J Med 2010;363:1303–12.
67 Kremer JM, Cohen S, Wilkinson BE et al. A Phase IIb
dose-ranging study of the oral JAK inhibitor tofacitinib 72 Scheinberg MA, Kay J. The advent of biosimilar therapies
(CP-690,550) versus placebo in combination with back- in rheumatology—‘O Brave New World’. Nature Rev
ground methotrexate in patients with active rheumatoid Rheumatol 2012;8:430–436.
arthritis and inadequate response to methotrexate alone. 73 Felson DT, Smolen JS, Wells G et al. American
Arthritis Rheum 2012;64:970–81. College of Rheumatology/European League Against
68 van Vollenhoven RF, Fleischmann R, Cohen S et al. Rheumatism provisional definition of remission in
Tofacitinib or adalimumab versus placebo in rheumatoid rheumatoid arthritis for clinical trials. Arthritis Rheum 2011;
arthritis. N Engl J Med 2012;367:508–19. 63:573–86.

vi36 www.rheumatology.oxfordjournals.org

Potrebbero piacerti anche