Sie sind auf Seite 1von 35

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/10735125

Therapeutic strategies for Rheumatoid arthritis

Article in Nature Reviews Drug Discovery · July 2003


DOI: 10.1038/nrd1109 · Source: PubMed

CITATIONS READS

876 6,025

2 authors, including:

Günter Steiner
Medical University of Vienna
433 PUBLICATIONS 14,991 CITATIONS

SEE PROFILE

All content following this page was uploaded by Günter Steiner on 14 August 2014.

The user has requested enhancement of the downloaded file.


Kurs “ Allgemeine und systematische Pharmakologie und Toxikologie”
Wintersemester 2011/12

Seminarthema II: Antiphlogistische und antipyretische Analgetika

Der Inhalt bzw. die Gliederung der Referate ist frühzeitig mit der/dem zuständigen
Dozentin/en abzusprechen. Alle Referate sollten max. 15 Minuten dauern und den Einsatz
von Hilfsmitteln (Folien) umfassen.
Bei Wiederverwendung von Overhead-Folien von Kolleginnen/en vorangegangener
Seminare werden keine Jokerpunkte (siehe Link "Creditsystem") vergeben.

PD Dr. Barbara Möpps N 26-5204  500-65505/65515


Referat I: Prothrombotische, antithrombotische und cardiovaskuläre Effekte von NSAIDs
und Coxiben
Salinas, G., Rangasetty, U. C., Uretsky, B. F., and Birnbaum, Y. (2007): The
cyclooxygenase 2 (COX-2) story; it´s time to explain, not inflame. J. Cardiovasc.
Pharmacol. Ther. 12: 98-111.
Grosser, T, Fries, S., and FitzGerald, G.A. (2006): Biological basis for the
cardiovascular consequences of COX-2 inhibition: therapeutic challenges and
opportunities. J. Clin. Invest. 116: 4-15.
Amer, M., Bead, V.R., Bathon, J., Blumenthal, R.S., and Edwards, D.N. (2010): Use
of nonsteroidal anti-inflammatory drugs in patients with cardiovascular disease.
Cardiol Rev. 18: 204-212.

Ihr Referat sollte folgende Punkte umfassen:


- Strukturelle Unterschiede und Physiologie der COX1- und COX2-Enzyme
- Vorstellung COX-2-selektiver Hemmer, Wirkungen und unerwünschten Wirkungen
- prothrombotische, antithrombotische und kardiovaskuläre Effekte von NSAIDs und Coxiben

Referat II: Unterschiede in der Pharmakotherapie der Rheumatoiden Arthritis und


Osteoarthritis
Smolen, J.S., and Steiner, G. (2003): Therapeutic Strategies for Rheumatoid
Arthritis. Nature Reviews 2: 473-488.
Wieland, H.A., Michaelis, M., Kirschbaum, B.J., and Rudolphi, K.A. (2005):
Osteoarthritis-an untreatable disease? Nat. Rev. Drug Discov. 4: 331-344.
Krüger, K. (2008): Quantensprünge in der Therapie der rheumatoiden Arthritis.
MMW Fortschr. Med. 150: 120-122.
Michael, J. W.-P., Schlüter-Brust, K.U., and Eysel, P. (2010): The epidemiology,
etiology, diagnosis, and treatment of osteoarthritis of the knee. Dtsch. Ärztebl. 107:
152-162.
Ihr Referat sollte folgende Punkte umfassen:
- Gegenüberstellung der Pharmakotherapie der Rheumatoiden Arthritis und Osteoarthritis
- Welche Rolle spielen NSAIDs? Nebenwirkungen von NSAIDs und Paracetamol.

Referat III: Neuropathischer Schmerz: therapeutische Ansätze


Galluzzi, K.E. (2005): Management of neuropathic pain. J. Am. Osteopath. Assoc.
105: S12-S19.
Baron, R. (2006): Diagnostik und Therapie neuropathischer Schmerzen. Dtsch.
Ärztebl. 41: A2720- A2730.
Jensen, T.S., Madsen, C.S., and Finnerup, N.B. (2009): Pharmacology and
treatment of neuropathic pains. Curr. Opin. Neurol. 22: 467-474.

Ihr Referat sollte folgende Punkte umfassen:


- Evidenzbasierte Therapie des neuropathischen Schmerzes
- Welche Rolle spielen NSAIDs?
REVIEWS

THERAPEUTIC STRATEGIES FOR


RHEUMATOID ARTHRITIS
Josef S. Smolen and Günter Steiner
Recent years have seen considerable advances in our understanding of both the clinical and
basic-research aspects of rheumatoid arthritis. Clinical progress has come from a better
recognition of the natural history of the disease, the development and validation of outcome
measures for clinical trials and, consequently, innovative trial designs. In parallel, basic research
has provided clues to the pathogenic events underlying rheumatoid arthritis, and advances in
biotechnology have facilitated the development of new classes of therapeutics. Here, we
summarize the fruits of these advances: innovative approaches to the use of existing, traditional
disease-modifying antirheumatic drugs; novel agents approved very recently; and further
avenues that are presently under investigation or which are of more distant promise.

Rheumatoid arthritis (RA) is a chronic inflammatory anti-inflammatory drugs (NSAIDs) and disease-modi-
and destructive joint disease that affects 0.5–1% of the fying antirheumatic drugs (DMARDs). NSAIDs only
population in the industrialized world and commonly interfere with a small segment of the inflammatory cas-
leads to significant disability and a consequent reduc- cade, namely prostaglandin generation by cyclooxy-
tion in quality of life1,2. It is two to three times more genases (COXs), but do not interfere with the underlying
frequent in women than in men and can start at any immuno-inflammatory events or retard joint destruc-
age, with a peak incidence between the fourth and sixth tion. By contrast, DMARDs ‘modify’ the disease process
decade of life. RA is associated with high costs and, if in all these respects, and, once DMARDs are effective, no
not treated appropriately, with a reduction in life further symptomatic therapies are needed. It is these lat-
expectancy3–5. In addition to the joint swelling and ter forms of therapeutic intervention that will constitute
pain caused by the inflammatory process, the ultimate the focus of this review.
hallmark of RA is joint destruction (FIG. 1).
RA is a polyarthritis, that is, it involves many joints Pathogenesis of RA
(six or more), although in the early stages of the dis- RA is a disease in which the immune and inflamma-
Division of Rheumatology, ease, only one or a few joints might be afflicted. tory systems are intimately linked to the destruction of
Department of Internal
Medicine III, University
Virtually all peripheral joints can be affected by the cartilage and bone. Although the penultimate link of
of Vienna, and Center disease; however, the most commonly involved joints the two systems remains elusive, and the cause of RA
of Molecular Medicine, are those of the hands, feet and knees 6, and distal unknown, many pathways involved in the generation
Austrian Academy of interphalangeal joints are usually spared. In addition, of the disease have been recognized and some of these
Sciences, A-1090, Vienna, RA can affect the spine, and atlanto-axial joint have been unequivocally identified as important by
and 2nd Department
of Medicine, Center for involvement is common in longer-standing disease therapeutic proof-of-principle studies. It has long been
Rheumatic Diseases, and constitutes a directly joint-related cause of mor- speculated that RA could be triggered by infectious
Lainz Hospital, A-1130 tality. Extra-articular involvement is another hallmark agents7–9, but proof of this is still lacking. Interestingly
Vienna, Austria. of RA, and this can range from rheumatoid nodules in this respect, RA patients respond favourably to drugs
Correspondence to J. S. S.
e-mail:
to life-threatening vasculitis. that have antibiotic activity or are derived from anti-
josef.smolen@wienkav.at Drug therapy for RA rests on two principal biotics10–12, although non-antibacterial mechanisms of
doi:10.1038/nrd1109 approaches: symptomatic treatment with non-steroidal action are usually implicated in this effect.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 2 | JUNE 2003 | 4 7 3

© 2003 Nature Publishing Group


REVIEWS

The reason for the joint-specific localization of the


ensuing immuno-inflammatory response is also
unknown. It could result from an arthrotropism of the
trigger, cross-reactivity of the trigger or its products with
structures primarily present in the joint tissues, or acti-
vation of other mechanisms that lead to homing of
involved cells to the joint. The products of such foreign
agent(s) could activate the innate immune system by
binding to Toll-like receptors (TLRs)13 or CD14 (REF. 14),
and subsequently triggering a T-cell response. T cells
undergo polarization into either TH1 or TH2 cells15,
which can be mutually inhibitory. TH2 cells are induced
by interleukin-4 (IL-4), whereas IL-12 is the strongest
TH1-inducing cytokine. TH1 cells mainly secrete inter-
feron-γ (IFN-γ) and tumour-necrosis factor-β (TNF-β);
TH2 cells produce IL-4, IL-5, IL-13 and IL-10. The polar-
ity of TH cells is decisive for the type of B-cell activation.
TH1 cells exert pro-inflammatory activities and promote
certain humoral responses, whereas TH2 cells have anti-
inflammatory potential and promote other types of
humoral responses, including immunoglobulin (Ig) E
production. In this context, it is important to bear in
mind that RA is regarded as an autoimmune disease16. In
particular, there is a strong association between RA and
several types of autoantibodies17; the longest known and
most important is rheumatoid factor (RF), which
is directed against the Fc fragment of IgG. Aside from
RF, responses to other autoantigens occur very com-
monly, both at the B- and T-cell level18,19. Whether such
autoantigens initiate the T-cell activation cascade and
the consequent inflammatory changes, or step in at a
later point in time to bolster and/or perpetuate the
process, is unknown. The potential role of autoimmune
responses in the chronicity and destructiveness of the
disease will be discussed later.
RA has a polygenic basis20, although the genes
involved have not yet been defined. So, it is assumed
Figure 1 | Schematic view of a normal joint and its changes in rheumatoid arthritis. that, in a genetically predisposed host, TH1 cells become
a | The synovial joint is composed of two adjacent bony ends each covered with a layer of cartilage, activated by arthritogenic antigen(s) in conjunction
separated by a joint space and surrounded by the synovial membrane and joint capsule. The with co-stimulatory signals and an appropriate cytokine
synovial membrane is normally <100 µm thick and the synovial lining (facing the cartilage and bone) environment (FIG. 2). The earliest event(s) might involve
consists of a thin (1–3 cells) layer of synoviocytes (type A are macrophage derived, and type B are
fibroblast derived); there is no basement membrane. Only a few, if any, mononuclear cells are
activation of the innate immune response, such as the
interspersed in the sublining connective tissue layer, which has considerable vascularity. The synovial triggering of dendritic cells (DCs) through TLRs (several
membrane covers all intra-articular structures except for cartilage and small areas of exposed bone of which are known to be expressed on synovial cells) by
(‘bare areas’) and inserts near the cartilage–bone junction. The ‘radiographic joint space’ (seen in c, exogenous material or by a combination of such foreign
which shows a radiograph of the second, third and fourth metacarpophalangeal joints in a normal stimuli together with autologous antigens, before or in
hand), in contrast to the usually minute ‘anatomic joint space’, consists of the latter, as well as of the
parallel to T-cell involvement.
two neighbouring, radiotranslucent portions of cartilage covering the non-translucent subchondral
bone. b | Like many other forms of arthritis, rheumatoid arthritis (RA) is initially characterized by an The T cells infiltrating the synovial membrane are
inflammatory response of the synovial membrane (‘synovitis’) that is conveyed by a transendothelial primarily CD4+ memory cells, which produce IL-2 and
influx and/or local activation of a variety of mononuclear cells, such as T cells, B cells, plasma IFN-γ to a similar extent as antigen-triggered T cells18,21
cells, dendritic cells, macrophages, mast cells, as well as by new vessel formation. The lymphoid and so clearly have a TH1 bias22–24. This polarity of the
infiltrate can be diffuse or, commonly, form lymphoid-follicle-like structures. The lining layer T-cell response in RA is further supported by a vast pre-
becomes hyperplastic (it can have a thickness of >20 cells) and the synovial membrane expands
and forms villi. However, in addition, the hallmark of RA is bone destruction (seen in d, which shows
ponderance of TH1 T-cell clones derived from RA
three metacarpophalangeal joints from a hand radiograph of a patient with established RA: the joint patients18,25,26 and by the presence of a milieu favouring
spaces have narrowed or disappeared as a sign of cartilage degradation and destructions of the the generation of myeloid DCs that preferentially activate
adjacent bone, also termed ‘erosions’, have occurred). The destructive portion of the synovial TH1 cells27. These T cells, by cell–cell contact — for
membrane is termed ‘pannus’, and the destructive cellular element is the osteoclast; destruction example, through CD11- and CD69-mediation28,29 —
mostly starts at the cartilage–bone–synovial membrane junction. Bone repair by osteoblasts
and activation by different cytokines, such as IFN-γ,
usually does not occur in active RA. Polymorphonuclear leukocytes are found in high numbers in
the joint fluid, but very rarely are seen in the synovial membrane, suggesting very rapid TNF-α and IL-17, activate monocytes, macrophages and
transgression from blood to the joint space. The neutrophils’ enzymes, together with enzymes synovial fibroblasts30–33 (FIG. 2). These latter cells then over-
secreted by synoviocytes and chondrocytes, lead to cartilage degradation. produce pro-inflammatory cytokines — mainly TNF-α,

474 | JUNE 2003 | VOLUME 2 www.nature.com/reviews/drugdisc

© 2003 Nature Publishing Group


REVIEWS

IL-1 and IL-6 — which seem to constitute the pivotal


event leading to chronic inflammation21,34–40. Moreover,
several lines of evidence indicate that TNF-α might reside
at the apex of this particular pro-inflammatory cytokine
cascade and drive the production of IL-1 and IL-6
(REFS 41,42); however, IL-1 can also induce TNF-α42.
Moreover, aside from those mentioned, many other
cytokines and chemokines are involved in RA pathogene-
sis, including IL-15 and IL-18 (REF. 43) and angiogenic
factors44. These soluble molecules, once engaged to their
receptors, trigger various signal transduction cascades,
such as the MAPK, nuclear factor-κB (NF-κB) or
Jak/STAT pathways45–47, which lead to the activation of
transcription factors and the subsequent induction of
genes whose products mediate inflammation and tissue
degradation.Among these products are various cytokines,
chemokines and tissue-degrading enzymes, such as the
matrix metalloproteinases (MMPs), but also cell-surface
molecules that enhance cell activation and cell–cell inter-
actions, such as co-stimulatory and adhesion molecules.
The latter comprise selectins, integrins and their coun-
terparts, which mediate cell rolling, adhesion and migra-
tion into inflammatory sites via endothelial cells and
other important intercellular interactions48, and so are
pivotal in the generation of the inflammatory response.
Overall, many cell populations are involved in disease
pathogenesis: T cells, B cells, monocyte/macrophages,
mast cells, DCs and fibroblasts are found in highly
increased numbers in the synovial membrane in RA.
Aside from the inflammatory infiltrate in the perivascu-
lar and sublining regions, the lining layer consisting of
type A (macrophage-like) and type B (fibroblast-like)
synoviocytes becomes hyperplastic and, at the cartilage–
bone junction, transforms into an aggressive tissue, the
‘pannus’, which contains osteoclasts49,50 (FIGS 1 and 2). The
growth of the synovial membrane is accompanied by
neovascularization44. Neutrophils, although not demon-
strably present within the synovial membrane, accumu-
late within the synovial fluid and participate in the
degradative processes. Whereas the degradation of
cartilage is mainly mediated by the plethora of metallo-
proteinases present in the joint, bone destruction is
mediated by the generation and activation of osteo-
clasts50–52, an event that apparently does not occur in
non-destructive arthritides.
Figure 2 | Schematic representation of events occurring in rheumatoid arthritis. The T cells
The role of B cells and autoantibodies, and/or
invading the synovial membrane are primarily CD4+ memory cells, which produce interleukin-2 (IL-2) immune complexes, could lie in the propagation and
and interferon-γ (IFN-γ) to a similar extent as antigen-triggered T cells21, and which are either already enhancement of the inflammatory process: it has long
pre-activated or become (further) activated by antigen-presenting cells (APCs) in conjunction with been known that complement is activated in RA syn-
arthritogenic (auto)antigen(s) and appropriate major histocompatibility complex (MHC) class II mole- ovial fluids and complement components are even
cules, co-stimulation (mainly through CD80/81 and CD28) and certain cytokines (IL-1, IL-15, IL-18). locally produced53; moreover, immune complexes are
Through cell–cell contact (for example, through CD11- and CD69-mediation28,29) and through
different cytokines, such as IFN-γ, tumour-necrosis factor (TNF)-α and IL-17, these T cells activate
deposited in cartilage54, and, in experimental arthritis, a
monocytes, macrophages and synovial fibroblasts30–33. The latter then overproduce pro-inflammatory lack of Fcγ-receptors abrogates the induction of disease55.
cytokines, mainly TNF-α, IL-1 and IL-6, which seem to constitute the pivotal event leading to chronic Endogeneous anti-inflammatory agents, such as
inflammation21,34–40. Through complex signal transduction cascades (FIG. 4), these cytokines soluble cytokine receptors and receptor antagonists,
activate a variety of genes characteristic of inflammatory responses, including genes coding for anti-inflammatory cytokines56, or regulatory T cells57,
various cytokines and matrix metalloproteinases (MMPs) involved in tissue degradation. TNF-α and are apparently insufficient to counterbalance the cas-
IL-1 also induce RANK expression on macrophages which, when interfering with RANKL on stromal
cells or T cells, differentiate into osteoclasts that resorb and destroy bone. In addition, chondrocytes
cade of pro-inflammatory events in chronic inflamma-
also become activated, leading to the release of MMPs. Initial events might also involve activation of tion. Targeting these events by mimicking the action of
APCs through Toll-like receptors (TLRs) before T-cell engagement. RANK, receptor activator of such natural inhibitors is one of the modern therapeutic
nuclear factor-κB; RANKL, RANK ligand; RF, rheumatoid factor; TCR, T-cell receptor. approaches, as discussed later.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 2 | JUNE 2003 | 4 7 5

© 2003 Nature Publishing Group


REVIEWS

DMARDs a more detailed referenced version) have limited efficacy,


DMARDs are agents that impede both the inflammatory toxicity problems or both. Efficacies in clinical trials
and destructive processes of RA. Many such drugs only range between ~40% and 70% for ACR20 responses, the
start acting after several weeks (for example, methotrexate usual primary endpoint (BOX 1), compared with placebo
(MTX)) or months (for example, gold salts), but once rates of between 15% and 30%. Drug retention rates in
they show effect, they reduce both pain and swelling, as clinical practice are relatively low, meaning that within
well as the progression of destruction. If efficacious, 1–2 years, the majority of patients have to stop a given
patients often do not need other anti-inflammatory medi- DMARD course, with the exception of MTX, which has
cations, such as NSAIDs or glucocorticoids. Remissions a median retention rate of ~3–4 years65,66. Patients who
occur in only 20–25% of the patients in clinical practice, failed DMARDs previously can respond to subsequent
but continued DMARD therapy is warranted, as stop- DMARD courses, but often to a lesser extent67. Only
ping DMARDs is accompanied by a significant risk of recently has it been appreciated that aggressive DMARD
flares58.Aside from the direct implications for RA therapy, therapy early in the course of the disease is beneficial68,69
these data, complemented by the finding of renewed effi- and that the therapeutic aim of RA therapy must be to
cacy after re-institution of DMARDs59, also support the fully arrest the inflammatory and destructive process
idea that DMARDs confer significant benefit. rather than just alleviating it, as is suggested by even the
We will divide DMARDs into small molecules and present clinical trial endpoints (BOX 1).
biological agents. The more well-established drugs will Combination therapy has also been advocated for
only be reviewed briefly; more emphasis will be placed RA, and most combination therapy strategies include
on recent insights into mode of action, and possible MTX as one of the components. Some combinations
routes to novel therapies. seemed to give better results than the monotherapies70–72,
whereas others did not73–75; however, toxicity was not
Small-molecule DMARDs presently used in RA accelerated in the majority of the more recent trials.
Small-molecule DMARDs have been used since the Careful analyses of the doses employed in comparator
1920s, and even in the days of biological therapies, one groups are necessary to fully appreciate potential differ-
such drug — MTX60,61 — is still the most commonly ences, and it is probable that the improved efficacy in at
used DMARD, and continues to be the gold standard of least one of the studies derived from high-dose glucocor-
DMARD therapy. MTX sets the level of efficacy that new ticoid therapy70,76. However, none of the combinations
compounds have to match, and its efficacy has still not tested so far have led to a clear-cut and reproducible
been unequivocally surmounted in head-to-head trials. advantage in the achievement of higher rates of a
The latest addition to the group of small chemical remission-like state77. Importantly, all the mentioned
DMARDs is leflunomide62, an active metabolite of which trials were head-to-head comparisons of combinations
inhibits dihydro-orotate-dehydrogenase, an enzyme that versus one of the drugs used as a single agent.
is pivotally involved in de novo pyrimidine synthesis. Another type of combination therapy involves the
Inhibition of this enzyme affects various signal-trans- addition of a second agent once an initial agent has
duction mechanisms, the generation of cytokines, and failed or demonstrated insufficient efficacy (step-up).
cell proliferation and migration63,64. However, all This was first introduced into clinical trials for insuffi-
presently used DMARDs (TABLE 1; see ONLINE TABLE 1 for cient responders to MTX by the addition of cyclosporin
A78, and has subsequently been used in trials of anti-
cytokine agents (see below). The most recent addition to
Table 1 | Small-molecule DMARDs in clinical use* the list of compounds suitable for combination use with
Agent First used Comments MTX is leflunomide79. All of these trials allow important
Gold salts 1920s Relatively long period of administration before information on efficacy and toxicity of the combina-
clinical effects are seen; rarely used today tions under study to be retrieved. However, none of
Sulphasalazine 1940s Second most common DMARD used in Europe them provide an answer to the question of whether such
during the 1990s combinations are at all advantageous, as — with one
Antimalarials‡ 1950s Less efficacious than other DMARDs, but also exception — they all involved only an MTX + new drug
less toxic arm, and an MTX + placebo arm, but not a new-drug-
Methotrexate 1950s Gold-standard therapy only arm. The single exception was a relatively small
D-penicillamine 1960s One of the more toxic DMARDs; rarely used today Phase II trial of MTX + infliximab (a monoclonal anti-
Azathioprine, 1960s Some clinical benefit
TNF antibody; see below) in which the addition of
mycophenolic acid infliximab to pre-existing MTX did not appear to be
Cyclosporine A, 1980s Efficacious, but relative toxicity has precluded superior to switching from MTX to infliximab80.
tacrolimus, sirolimus widespread use
Minocycline 1990s Mildly beneficial Biological DMARDs presently used in RA
The pro-inflammatory role of cytokines, and the
Leflunomide 1990s Overall, has similar effects to sulphasalazine
and methotrexate involvement of different cell types and their surface
Glucocorticoids 1950s Rapid anti-inflammatory effects, but also qualities molecules in the pathogenesis of RA, provides the ratio-
of DMARDs; however, have long-term side effects nale for the development of highly specific therapeutics
*See ONLINE TABLE 1 for a more detailed referenced version. ‡ Chloroquine, hydroxychloroquine. to target these molecules. Targeting of pro-inflamma-
DMARD, disease-modifying antirheumatic drug. tory cytokines can be achieved by several strategies.

476 | JUNE 2003 | VOLUME 2 www.nature.com/reviews/drugdisc

© 2003 Nature Publishing Group


REVIEWS

Box 1 | Development of outcome measures for RA clinical trials TNF blockade has been the major breakthrough in the
therapy of RA during the past ten years, both with respect
When compared with other disorders in which a single, or a group, of objectively to transferring basic work to the bedside and regarding its
assessable laboratory or clinical variables can be used as ‘activity’ or ‘outcome’ measures, effects in otherwise often refractory patients. This, how-
rheumatoid arthritis (RA) is difficult to study for a variety of reasons. First, there is a wide ever, should not detract from the fact that more than half
variability between patients. Second, there are dozens of potential outcome measures of the patients in clinical trials do not achieve ACR50
(ONLINE BOX 1), such as tender or swollen joint counts, global disease assessments by
responses, that remissions are rare, and that these drugs
patients or physicians, functional assessments and radiographic changes. And third,
do have side effects61,83–91(ONLINE TABLE 2).
patients with RA are amenable to a tremendous placebo response for most of these
variables (see ONLINE BOX 4 for details).
IL-1 blockade. IL-1 is well established as another key pro-
To improve the reliability of disease assessment, different groups of clinical researchers
inflammatory cytokine involved in RA. Binding of IL-1
have independently determined and validated core sets of clinical variables that were
sensitive to change, had long-term predictive value, had only little, if any, redundancy, and to IL-1 receptor I (IL-1RI) allows the engagement of the
were therefore reliable in determining disease activity and/or outcome in clinical IL-1R accessory protein (IL-1RacP) and subsequent cell
trials253–255. To distinguish therapeutic from placebo responses in clinical trials, the signalling and activation. The natural inhibitor IL-1
American College of Rheumatology improvement criteria rely on relative changes of a receptor antagonist (IL-1ra), which belongs to the IL-1
composite of these core set disease activity variables, whereas the European League Against family, competes with IL-1 for its receptor but does not
Rheumatism response criteria give absolute changes (see ONLINE BOX 1 for details)254. allow engagement of IL-1RacP, thereby blocking activa-
tion of signal transduction mechanisms92. The impor-
tance of the role of IL-1ra in regulating the inflammatory
First, monoclonal antibodies, soluble receptors, binding response is exemplified by the occurrence of a destruc-
proteins or receptor antagonists can bind to pro- tive arthritis in IL-1ra-deficient animals93.
inflammatory molecules or their receptors and interfere On the basis of several randomized double-blind con-
with receptor ligation and its consequences. Second, trolled clinical trials94–96, anakinra (TABLE 2) — a recombi-
anti-inflammatory cytokines, such as IL-4, IL-10 or IL-13, nant form of IL-1ra — became the first, and so far the
can antagonize the production or action of the pro- only, IL-1-blocking drug to be approved for the treatment
inflammatory cytokines. Third, monoclonal antibodies of RA. Published results seem to indicate that the effects
targeted against differentiation- or function-associated of anakinra might be more modest than those seen with
cell-surface antigens can lead to either elimination of other agents; however, head-to-head-trials of different
the targeted cells or interference with the cell’s function. biological compounds have not yet been performed.
Agents that exploit each of these strategies have
entered clinical trials: some have failed, others are being Summary of recently approved DMARDs
further evaluated in controlled trials (discussed below) FIGURE 3 summarizes the principal results obtained with
and a few have been approved. All of the approved ther- adalimumab, etanercept, infliximab and anakinra, and
apies, which are shown in TABLE 2 (see ONLINE TABLE 2 also with the recently introduced small-molecule com-
for a more detailed version), belong to the first group of pound leflunomide. As the patient populations were
the previous paragraph: they prevent pro-inflamma- different in these studies, it is difficult to compare the
tory cytokines — in particular TNF and IL-1 — from results of each trial with those of the others. However, as
interacting with their receptors. most trials involved placebo groups, differences between
placebo and the agent studied can be extrapolated
Anti-TNF therapies. The key role of TNF in the patho- between studies. Only leflunomide was studied against
genesis of RA was elucidated in detail during the late an active comparator and placebo, and among the bio-
1980s and throughout the 1990s by Feldmann, Maini and logicals, only etanercept was studied against an active
others, in both experimental animals and in patients with comparator. Such data for infliximab and adalimumab
RA (reviewed in REF. 81). Key evidence for the importance are expected in the near future. The safety concerns
of TNF is also provided by the observation of the occur- associated with the new biologicals mainly involve
rence of a severe arthritis in TNF-overexpressing trans- application-specific issues, such as infusion- or injec-
genic mice82, which, in common with other mouse tion-site reactions, and their potential to increase the
arthritis models, could be prevented by inhibiting TNF-α. risk of infections, particularly tuberculosis and oppor-
Three drugs that block the activity of TNF have been tunistic infections with the TNF-blockers. Another issue
approved (TABLE 2). Infliximab (which was the first TNF- to be mentioned here is the high financial burden of the
blocker to be clinically tested in RA) and adalimumab are new biological therapies, which is one of the reasons for
antibodies against TNF, and etanercept (which was the the increased interest in finding novel small-molecule
first biological DMARD to be approved for treating RA) DMARDs that can be produced more cheaply.
is a fusion protein of the TNF receptor II. All of these
agents lead to clinical improvements in RA, and patients Candidates for future small-molecule DMARDs
typically achieve ~50–70% ACR20 and 30–50% ACR50 Most new candidate DMARDs are enzyme inhibitors,
responses (FIG. 3). In addition, patients also experience a which target either secreted enzymes involved in tissue
significant improvement in function-related quality of destruction, such as matrix metalloproteinases
life, and, most importantly, radiographic progression is (MMPs), enzymes that liberate active cytokines from
very significantly retarded 61,80,83–89, and might even be their precursor or membrane-associated forms, or
halted in a considerable fraction of the patients. As such, kinases of various signal-transduction cascades.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 2 | JUNE 2003 | 4 7 7

© 2003 Nature Publishing Group


REVIEWS

Table 2 | Approved biological DMARDs and related drugs in development*


Agent Status‡ Properties Comments
Anti-TNF therapies
Infliximab Approved Chimaeric monoclonal antibody First biological DMARD to be clinically
(RA: 1999) to TNF tested; trials showed that TNF blockade is
clinically effective short- and long-term
Etanercept Approved Construct of TNF-RII and the Fc Efficacy in RA comparable to infliximab used
(1998) portion of IgG1 as a monotherapy or combination therapy
Adalimumab Approved Human monoclonal antibody to TNF Efficacy in RA comparable to infliximab used
(2002) as a monotherapy or combination therapy
CDP870 In study PEGylated Fab fragment of CDP571,
a humanized antibody to TNF
PEG-TNF-RI In study PEGylated form of soluble TNF-RI
IL-1-blocking agents
Anakinra Approved IL-1R antagonist Clinically effective as a monotherapy and in
(2001) combination with MTX
IL-1 trap Phase I Construct of two IL-1R chains with
an IgG-Fc domain
*See ONLINE TABLE 2 for a more detailed version, including adverse events. ‡Years shown are those of the first approvals in a major
pharmaceutical market. Anti-TNF therapies are described in REFS 61,80–91; IL-1-blocking agents are described in REFS 92–96. DMARD,
disease-modifying anti-rheumatic drug; IgG1, immunoglobulin G1; IL-1, interleukin-1; IL-1R, interleukin-1 receptor; MTX, methotrexate;
RA, rheumatoid arthritis; TNF, tumour-necrosis factor; TNF-R, tumour-necrosis factor receptor.

Inhibitors of proteases. MMPs seem to be important in led to discontinuation of this programme and, likewise,
RA, as they are present in increased amounts and in to termination of Ro 113-0830 trials in OA.
active forms in the inflamed joint tissue97,98. This group There are potentially several reasons for the failure of
of molecules comprise more than 20 members that MMP inhibition for the treatment of arthritis. First,
include the collagenases (now termed MMP-1, MMP-8 MMPs could be less important in tissue destruction than
and MMP-13), the gelatinases (now named MMP-2, originally assumed. Second, the doses necessary to reach
MMP-9), the stromelysins (MMP-3, MMP-10, MMP-11 sufficient MMP inhibition locally might be higher than
and MMP-7) and the adamalysins99. TNF-α-converting can be achieved given the safety profile of the drugs con-
enzyme (TACE), which releases soluble TNF from its cerned. Third, the MMPs targeted so far might not be the
membrane-bound form, is an adamalysin; indeed, the most important ones in the process of joint destruction,
adamalysins are involved in the shedding of many cell- or it could be that the redundancy of MMP activities
surface molecules. Therefore, MMP-inhibition consti- allows tissue destruction to continue unless all or most of
tutes an interesting and important avenue to explore in them are inhibited sufficiently. So, at present, the only
developing therapies. compound in use that is also an MMP inhibitor is
Synthetic MMP inhibitors were originally developed minocycline; however, this drug seems to have limited
as anticancer agents, and were intended to inhibit the efficacy and probably exerts its effect mostly through
invasive capacity of malignant cells and to interfere with mechanisms other than MMP inhibition.
angiogenesis; however, results from cancer trials have MMPs, however, are already naturally regulated by
mostly been disappointing. MMP inhibitors include col- different molecules, such as the tissue inhibitors of metal-
lagen peptidomimetics and non-peptidomimetics100,101, loproteinases (TIMPs)98,104. In fact, in experimental mod-
bisphosphonates and tetracyclines. The inhibition pro- els of destructive arthritis, overexpression of TIMP-1 can
files of these compounds differ: for example, trocade almost abrogate disease105. The short half-life of TIMPs
(Ro 32-3555) selectively inhibits the collagenases could be overcome by various molecular or biochemical
(MMP-1, MMP-8 and MMP-13) and has low activity techniques that are already used for TNF-blocking
against other MMPs, whereas Ro 113-0830 interferes agents, although, as with the small-molecule MMP
efficiently with MMP-2, MMP-9, MMP-8 and MMP-13, inhibitors, safety issues might be a concern.
but does not inhibit MMP-1 (REF. 102). Yet another type of MMP inhibitor has entered
Several MMP inhibitors are efficacious in experi- clinical trials in RA: drugs that interfere with TACE,
mental models of arthritis and have therefore consti- which cleaves TNF-α from its membrane-associated
tuted interesting compounds for the therapy of RA: form and thereby enables the cytokine to act in a
BAY12-9566 (which inhibits stromelysins and gelati- paracrine, endocrine and autocrine manner106. TACE is
nases), Ro 113-0830, GI 168, MDL 101, BB 1101, ONO overexpressed in RA107 and its inhibition by compounds
4817, and trocade. BAY 12-9566 was developed for such as GW 3333, TMI-1, or Ro 32-7315 has shown effi-
osteoarthritis (OA) and had reached Phase II trials, but cacy in experimental arthritis108–111. However, it has to be
all trials were discontinued because of safety issues aris- considered that inhibition of TACE increases levels of
ing in the cancer trials102. Trocade was evaluated in the membrane-associated TNF-α. The consequence of
Phase III trials in RA, but its lack of clinical efficacy, this is unclear: on the one hand, the presence of mem-
including failure to improve radiographic scores102,103, brane TNF in the absence of secreted TNF might be

478 | JUNE 2003 | VOLUME 2 www.nature.com/reviews/drugdisc

© 2003 Nature Publishing Group


REVIEWS

insufficient to mount a full-blown inflammatory TNF was shown to be sufficient for the induction of
response112; on the other hand, even such ‘suboptimal’ severe destructive arthritis in TNF transgenic mice113.
inflammation might be sufficient to lead to tissue Moreover, an MMP inhibitor that blocked both the pro-
destruction. In fact, overexpression of membrane-bound cessing of TNF receptors as well as TNF was unable to
inhibit further downstream generation of other pro-
A Clinical response to new disease-modifying antirheumatic drugs inflammatory cytokines114. Phase I studies of TACE
80
a b c d e f
inhibitors have been performed and results from Phase
II trials in RA are awaited with great interest.
% of patients achieving an ACR20 response

70 72
71 Like TNF-α, IL-1β (and likewise IL-18) has to be lib-
65 erated from a precursor form to become an active soluble
60
cytokine. This process is mediated by the interleukin-1
50 52 59 converting enzyme (ICE), which is identical to caspase-1
(REF. 115). Pro-IL-1β and pro-IL-18, although biologically
46
40 42 active molecules, reside in the cytosol and, in addition to
ICE, can also be cleaved by other proteases116,117.
30 Inhibiting IL-1β and IL-18 release by inhibiting ICE
27 could be a valuable avenue to investigate for RA therapy,
20 23 24 but it should be borne in mind that other enzymes can
20
17
also cleave the precursors of these cytokines. In addition,
10
ATP can lead to release of mature and precursor forms
0 of IL-1β through the P2X7 receptor118. One of several
bo ab bo ep
t
bo nra bo ide TX pt o ab ICE/caspase-1 inhibitors is pralnacasan119, which
ce ixim ce erc ce aki ce om rc
e eb
a a a a M
lac um reduced inflammation and joint destruction by ~60% in
p l f l p l
an p l n p l
lun ne p m
+) ) in +) et +) X+)
a
+) lef e ta +) d ali
TX TX+ TX X+) TX T TX +) TX )a experimental arthritis120. In a Phase II clinical trial,
( M ( M T ( M (M ( M X (M +
(M (M (M
T TX pralnacasan showed a dose-dependent increase in
(M
response which, however, did not reach statistical signifi-
B Radiographic progression cance compared with placebo121. Nevertheless, as only
8 mild adverse events were observed, further investigation
a e f g h
of higher doses might be warranted and could lead to
Increase in modified Sharp score (12 months)

7 7.2 better results. The P2X7 receptor, which, as mentioned


6
above, is involved in the release of IL-1 and IL-18, is like-
wise an attractive target. Indeed, inflammatory pain
5 resulting from the local inoculation of adjuvant was alle-
viated by injection of oxidized ATP, a selective inhibitor
4 of the P2X7 receptor, into rats’ paws122. In addition,
3.6 arthritis induced by monoclonal antibodies to collagen
3 was alleviated in P2X7-deficient mice123.
2.7
The novel small molecules discussed so far either
2 2.2
1.9 target enzymes secreted by cells into the extracellular
1.6
1
space or cellular enzymes that enable the secretion of
0.6
1.0
0.1 0.9 mature forms of pro-inflammatory cytokines, and so
0.5
0 interfere mainly with late effector phases of the
o b TX pt o b o n) o TX ide immuno-inflammatory events. However, the engage-
eb ma M rc
e eb ma eb ea eb M
lac flixi e lac imu Pl
ac (m Pl
ac o m
ment of cytokines with their receptors leads to a
+)
p
) in Et
an p
+) ada
l ra f lun
in Le
TX X+ TX ) ak plethora of redundant signal-transduction cascades and
(M (MT (M TX+ An
(M the consequent generation of activated transcription
Figure 3 | Data for recently approved antirheumatic agents. A | Clinical results of randomized factors that mediate the whole inflammatory process
controlled clinical trials. Only trials in which the newly approved agents (or placebo) were applied in through respective gene activation. This redundancy is
addition to methotrexate (MTX) to patients with insufficient response to MTX therapy or (in the case not only related to the instigation of several signal-
of etanercept) direct comparison between a new agent and high-dose MTX are shown. The transduction pathways in parallel, but also to the fact
anakinra + MTX, the etanercept + MTX and the leflunomide + MTX trial reflect six-month data,
that several cytokines can trigger the same cascades.
whereas all other trials reflect one-year data. In trials a, b, c, d and f, the differences between new
compound + MTX were statistically significantly different from placebo + MTX; at the 12-month
endpoint, the difference between the etanercept and the MTX group (e) was not significantly Inhibition of signal-transduction cascades. TNF-α and
different. Data from all trials are published as full papers61,79,84,85,87,95. B | Radiographic data of two IL-1 induce a variety of signal-transduction cascades
trials shown in A (a, e), a trial comparing anakinra with placebo96 (g) and a leflunomide 12-month that lead to the activation of a series of transcription
trial comparing leflunomide with intermediate-dose MTX and placebo256 (h); data from the 12- factors (FIG. 4), including the AP-1 complex and NF-κB
month adalimumab trial are published only in abstract form. The anakinra study used the Genant- (reviewed in REFS 45,46,124), and consequently to the
modified Sharp score, all others used the van der Heijde-modified Sharp score. It should be noted
that each trial can only stand on its own and that it is difficult to compare with other trials because
induction of a variety of genes, such as those coding for
of the non-comparative nature of the individual trials, as well as to significant differences in patient various cytokines, other inflammatory mediators
populations investigated. At best, an estimation of similarities or differences between trials is including prostaglandins, and MMPs125,126. The two
potentially possible where placebo arms exist. principal pathways activated by TNF-α and IL-1 are the

NATURE REVIEWS | DRUG DISCOVERY VOLUME 2 | JUNE 2003 | 4 7 9

© 2003 Nature Publishing Group


REVIEWS

MAPK- and the NF-κB pathways (FIG. 4), and molecules option; it will therefore be interesting to learn more about
of these signal-transduction cascades have been detected the effects of LCK inhibitors in experimental arthritis156.
in the RA synovial membrane127,128. As p38-MAPK is B cells are also likely to be important in RA, as evidenced
predominant both in endothelial cells and in the lining by the presence of autoantibodies in the vast majority of
layer of the RA synovial membrane, the inhibition of RA patients and the relation of the severity of disease to
p38-MAPK could be of particular interest. A variety of the presence of such autoantibodies, as discussed above.
p38-MAPK inhibitors have been developed, such as SB In fact, in some experimental models, such as collagen-
203580, SB 220025, RWJ 67657, VX-745, RPR 200765A, induced arthritis and the K/BxN model, autoantibodies,
RPR 203494 and BIRB 796 (REFS 129–140). These agents and hence B cells, are of significant importance157.
usually inhibit two of the four isoforms of p38-MAPK Moreover, mice with B-cell deficiency, such as mice with
(α and β)137, and many have anti-arthritic activity in the xid mutation, are resistant to developing collagen
experimental models129,132,133,138,139. RWJ 67657 also effec- arthritis and less susceptible to Staphylococcus-induced
tively inhibited endotoxin-induced clinical effects and arthritis158. The xid mouse carries a Bruton’s tyrosine
cytokine release in Phase I studies140. Phase II clinical trials kinase (BTK) defect, and in humans BTK mutations are
of p38-MAPK inhibition in RA are in progress. Several of associated with X-linked agammaglobulinaemia159. BTK
these compounds have shown toxicities, many of which is essential for activation of NF-κB through the B-cell
do not seem to be related to the mode of action but to receptor160 and also seems to be involved in the induction
the chemistry of the particular agents; these toxicities of macrophage effector functions161. So, targeting BTK
involve the liver, heart and central nervous system. might allow a combination of a B-cell-selective approach
Another important MAPK is c-JUN N-terminal by inhibiting (auto)antibody-mediated inflammation
kinase (JNK), which has several isoforms that phosphory- and also interfering with inflammation-associated effec-
late specific sites on c-JUN. Specific JNK-inhibitors, such tor functions of macrophages162. Interestingly, BTK can be
as SP 600125, CEP 1347, or CEP 11004 (REFS 141,142), phosphorylated by c-ABL, the tyrosine kinase responsible
might ameliorate experimental arthritis143. The JNK path- for chronic myelogeneous leukaemia163, an observation
way is particularly involved in the regulation of MMPs. that opens new avenues to explore in the use of kinase
Finally, inhibitors of extracellular signal-regulated kinase inhibitors in RA. Moreover, BTK has recently been
(ERK) or the upstream kinase MEK, such as PD 98059, shown to be involved in the LPS-induced generation of
NAMI-A, or U0126 (REFS 144,145), have also been used, but TNF-α164. In addition to BTK, other tyrosine kinases,
did not ameliorate experimental arthritis143. such as LYN and SYK, are involved in signalling through
The second major family of transcription factors acti- B-cell receptors (and also other ones in other cells) and
vated in the course of the inflammatory response is the it will be interesting to learn more about the effects of
NF-κB family. NF-κB exists as homo- or heterodimers of specific inhibitors in arthritis165.
five DNA-binding proteins, RelA/p65, Rel B, c-Rel, p52 B-lymphocyte stimulator (BLys), also termed BAFF
and p50, which are usually bound to the inhibitory IκB (B-cell activating factor of the TNF family) leads to B-cell
proteins. When IκB is phosphorylated by IκB kinases activation and production of immunoglobulins, includ-
(IKK), NF-κB is freed and can translocate to the ing autoantibodies. BLys levels are, in fact, increased in
nucleus45. An adenovirus vector encoding IκBα can several autoimmune disorders, including RA. Targeting
inhibit the in vitro production of pro-inflammatory Blys either with monoclonal antibodies or receptor con-
cytokines and MMPs, and interfere with DC function; structs might be a further interesting targeted approach.
the latter could also be achieved by a small-molecule In summary, a plethora of kinases and transcription
inhibitor146. Various compounds that are already used in factors might constitute promising therapeutic targets
NF-κB PROTEINS the therapy of RA act as NF-κB inhibitors, such as gluco- for combating inflammatory disorders in general and
NF-κB consists of homo- or corticoids, cyclosporin A and leflunomide; in addition, RA in particular. The reason why it is difficult to pin-
heterodimers, and the most flavonoids and proteasome inhibitors, such as gliotoxin, point a single or a few such molecules lies in the com-
important of its components are
PSI or parthenolide, interfere with the degradation of plexity of the pathogenesis of RA and the redundancy of
the p50 and the p65 proteins.
IκB147–149. The potential of selective inhibitors of NF-κB the cellular and molecular pathways underlying the
TRANSCRIPTION FACTOR AP-1 in the treatment of RA is also evidenced by the effects of inflammatory and arthritogenic response. In any case,
The transcription factor AP-1 is NF-κB inhibition in experimental animals: overexpres- the use of kinase inhibitors will have to be watched
composed of homo- or sion of IκBα, or the application of small-molecule agents intensively for their tolerability. In particular, it will
heterodimers of proteins that
primarily belong to the JUN and
that target the NF-κB pathway at various points, inhibits have to be seen whether the inhibition of such essential
FOS (but also other) families. experimental inflammation and colitis147,148,150,151. At pre- intracellular pathways is afflicted with an increased
Whereas JUN proteins can sent, the most promising target among the many mole- propensity towards infections and/or malignancy, but
homodimerize, FOS proteins do cules composing the NF-κB pathway is IKK-β. other safety issues could also arise.
not form stable homodimers but
As T cells have a role in the pathogenesis of RA and
form heterodimers with JUN
proteins. Once phosphorylated many forms of experimental arthritis, inhibiting T-cell Possible targets for new biological DMARDs
and dimerized, the AP-1 functions by interfering with pivotal kinases involved in TNF, other pro-inflammatory cytokines and lymphokines.
proteins gain enhanced DNA- T- and/or B-cell activation is an attractive possibility. In Additional TNF blockers are being evaluated at present,
binding capacity and modify fact, leflunomide has been shown to also inhibit the tyro- such as CDP 870, a pegylated Fab fragment of a human-
transcriptional activity. AP-1
proteins are phosphorylated
sine kinase LCK64, which is an essential protein for T-cell ized IgG monoclonal anti-TNF antibody166, and oner-
(and thereby activated) by the activation. More specific LCK inhibitors have been char- cept, a fusion protein of recombinant human TNF
MAPK pathways. acterized152–155, and targeting LCK might be an attractive receptor I (TNF-RI, also known as p55) with IgG1.

480 | JUNE 2003 | VOLUME 2 www.nature.com/reviews/drugdisc

© 2003 Nature Publishing Group


REVIEWS

Onercept (in contrast to etanercept) seems to have effi-


cacy in Crohn’s disease167 and constitutes another can-
didate TNF blocker for RA. Finally, pegsunercept, a
pegylated soluble TNF-RI, has shown anti-arthritic activ-
ity in experimental arthritis168. Like TNF, IL-1 constitutes
an important target in the pathogenic pathways under-
lying RA and, as mentioned above, anakinra (recombi-
nant IL-1ra) is approved for the treatment of the disease.
Other IL-1 blockers, such as soluble recombinant IL-1
receptors type I and II, are candidate agents169.
IL-6 is a pro-inflammatory cytokine that has been
consistently found in highly increased levels in RA and
linked to the acute-phase response. However, the role of
IL-6 in the pathogenesis of RA is not yet fully known. It
seems to be mainly induced by TNF-α and IL-1, and
TNF blockade leads to a significant decrease in IL-6
(REF. 170). Furthermore, IL-6 deficiency ameliorates some
forms of experimental arthritis171. Anti-IL-6 mono-
clonal antibodies were tested in RA patients in open
trials with minor success more than a decade ago172.
However, a recombinant human anti-IL-6 receptor
monoclonal antibody that inhibits the function of IL-6
has recently undergone Phase II clinical trials. In a small
Phase II trial, this anti-IL-6R antibody induced ACR20
responses in 56% of patients with established RA173.
This molecule is being further evaluated in RA and
might also be effective in multiple myeloma174.
IL-2 and its receptor were also targeted in experimen-
tal models by using monoclonal antibodies to the IL-2R
or recombinant human IL-2 coupled to diphtheria toxin
(DAB 389IL-2, DAB 486-IL-2)175,176. Although somewhat
effective in malignancies177 and psoriasis178, response rates
in RA were unremarkable179. However, the efficacy of
recombinant monoclonal anti-IL-2R antibodies, such as
daclizumab and basiliximab, in the treatment of trans-
plant rejection180, indicates that such intervention might
also be useful in RA, possibly in combination with other
Figure 4 | Simplified overview of pathways involved in TNF-α and IL-1 signalling. Two major agents, and particularly in the early stages of the disease.
pathways, the NF-κB (left) and the MAPK pathway (right) are activated by the pro-inflammatory As some of the functions of IL-2 can be substituted
cytokines TNF-α and IL-1. a | On the left of the figure, in the cytoplasm, NF-κB PROTEINS are usually for by IL-15, this cytokine is a focus of interest. IL-15 is
associated with inhibitors of NF-κB (IκBs). Various stimuli, including ligation of receptors of pro- produced by a variety of cell types, including mono-
inflammatory cytokines, activate the IκB kinase (IKK) complex, which consists of IKK1 (α), IKK2 (β),
and the regulatory subunit NEMO (also known as IKKγ). IKK phosphorylates IκBs, leading to their
cytes, DCs and fibroblasts. Its receptor comprises the
degradation, thereby allowing NF-κB to enter the nucleus and activate genes coding for various IL-2Rβ- and γ-chains and a unique IL-15R α-chain.
molecules, such as cytokines, chemokines, cyclooxygenase-2, anti-apoptotic and stress proteins. IL-15 is expressed in the synovial membrane and is also
NF-κB can be activated by a variety of engaged receptors and various other signalling pathways found in synovial fluids. TNF-α production by
than the one depicted here, which all involve activation of IKKs. b | On the right of the figure, MAPKs macrophages as induced by T-cells is partly dependent
are regulated by several (two or more) upstream phosphorylation cascades initialized after receptor
on the activity of IL-15 (REF. 181). IL-15 blockade by solu-
ligation and recruitment of adaptor proteins. The kinases immediately upstream of the MAPKs
belong to the MAPK or ERK kinase (MKK or MEK) family. The three major MAPK families are the ble mouse IL-15Rα inhibited experimental arthritis in
extracellular-signal-regulated kinases (ERKs), the c-JUN N-terminal kinases (JNKs) and the p38 mice182, and soluble human IL-15Rα has also shown
enzymes. The ERK subgroup of MAPKs is mainly activated by growth factors, whereas pro- efficacy against collagen-induced arthritis in a primate
inflammatory cytokines mostly induce the JNK and p38 MAPK cascades. After phosphorylation of model43. Results of a Phase I trial in RA have recently
its components (primarily members of the Fos and Jun families), the TRANSCRIPTION FACTOR AP-1 been publicized and further data are awaited.
induces activation of genes coding for various cytokines (including TNF-α and IL-1 themselves),
other inflammatory molecules, such as prostaglandins (PGs), and proteases, such as the matrix
Another potent pro-inflammatory cytokine is IL-12,
metalloproteinases (MMPs). In addition, AP-1 regulates cell proliferation, survival and apoptosis. which is the major inducer of TH1 cells. So, interference
As also shown, capture of TNF-α by TNF-blocking agents, or interaction of IL-1ra with the IL-1 with TH1 responses can be achieved by blocking IL-12,
receptor, prevents TNF-α and IL-1, respectively, from ligation to their receptors, induction of signal and anti-IL-12 antibodies suppress experimental
transduction cascades and activation of transcription factors AP-1 and NF-κB. Likewise, the arthritis both as monotherapy and even more effi-
different kinases, as well as the transcription factor activity itself, constitute therapeutic targets. AP-1,
ciently in synergy with anti-TNF therapy183. Clinical
activator protein-1; IL, interleukin; IRAK, IL-1 receptor-associated kinase; MAPK, mitogen-activated
protein kinase; MyD88, myeloid differentiation protein 88; NEMO, NF-κB essential modulator; trials of anti-IL-12 have been initiated in Crohn’s dis-
NF-κB, nuclear factor-κB; TNF, tumour-necrosis factor; TRADD, TNF receptor-associated death ease and RA, but results might not be impressive as
domain protein;TRAF, TNF receptor-associated factor. they have not yet been published.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 2 | JUNE 2003 | 4 8 1

© 2003 Nature Publishing Group


REVIEWS

As discussed previously, IL-18 belongs to the IL-1 Anti-inflammatory cytokines. Several cytokines possess
family and is derived from its precursor by ICE. IL-18 anti-inflammatory activity, particularly in the context of
can be found in RA synovial membrane and joint fluid, TH1-mediated diseases. The TH2 cytokines IL-4 and IL-10
and it induces TNF-α, IFN-γ, IL-1 and other cytokines have already been employed in clinical trials in RA.
through lymphocyte activation, as well as direct action Despite efficacy in experimental models199, a Phase I trial
on macrophages. In addition, IL-18 increases the pro- of recombinant human (rh) IL-4 in RA, although well
inflammatory activity of T-cell–macrophage interactions tolerated, did not reveal significant clinical benefit200.
in synergy with IL-15 and IL-12 (REF. 184), and has an Daily subcutaneous injection of rhIL-10 initially seemed
important effect on the induction of IFN-γ, which is why to be beneficial201 and led to pronounced increases of
it was originally described as IFN-γ-inducing factor185. circulating TNF-R and IL-1ra202. However, thrombo-
IL-18 signals through a receptor comprising an α- and cytopaenia occurred in some patients and efficacy was
β-chain, with IL-18Rβ serving a similar function to the not discerned in subsequent studies. So, it seems that
IL-1RacP. In addition, there exists an IL-18-binding pro- both these anti-inflammatory cytokines did not lead to
tein (IL-18BP) that has no sequence homology with the the expected shift to a TH2 phenotype or that the
IL-18Rα or β-chain and which serves as a decoy receptor. assumption of a beneficial role of changes of the
A deficiency, or neutralization, of IL-18 ameliorates TH1/TH2 balance in RA might be incorrect. The devel-
experimental arthritis186,187. There are four ways to opment of both IL-4 and IL-10 for RA was discontin-
block the actions of IL-18: anti-IL-18 antibodies, anti- ued, but it is still possible that the combination of both
IL-18R antibodies, soluble IL-18Rα or their constructs, cytokines, or of other TH2-type cytokines, could lead to
and IL-18BP. As a molecule that induces IFN-γ, IL-1 and a more marked improvement203.
TNF-α, IL-18 is an attractive therapeutic target for RA. IL-11, a member of the IL-6 superfamily, is a
Finally, IL-17, a T-cell-derived cytokine present in haematopoietic growth factor for platelets204 that is
RA188, acts as a pro-inflammatory cytokine, particularly approved for use in chemotherapy-associated thrombo-
in conjunction with IL-1 and TNF-α189. IL-17 induces cytopaenia (as oprelvekin), but which is also an anti-
MMP production and downregulates TIMP190, and inflammatory cytokine. However, no significant clinical
blockade of IL-1 and IL-17 has a synergistic effect on benefit in RA was achieved205.
inflammation and bone destruction in vitro191. So, this In experimental arthritis, IL-13 can also attenuate
cytokine could also be an effective target, possibly in disease206. IL-13 is an anti-inflammatory cytokine that
conjunction with blockade of IL-1 and/or TNF in a signals through a receptor that shares the IL-4Rα chain
combination therapy approach. with IL-4R and the common γ-chain with IL-2R, IL-4R
and IL-15R. IL-13 can markedly reduce IL-1, TNF-α
Blocking chemokines and angiogenesis. CHEMOKINES and and IL-8 levels in synovial tissue explant cultures207.
chemokine receptors (CCRs) have attracted signifi- Similarly to IL-4, it is hardly detectable in the RA syn-
cant interest in RA research192, and a CCR5 polymor- ovial membrane and might therefore not be important
phism seems to be related to the severity of RA 193. in the counter-regulatory mechanisms of the local
Small-molecule inhibitors, as well as biological agents, inflammatory response. Nevertheless, the beneficial
of this and other chemokine receptors194,195 could effects seen in experimental animals indicate that it
become valuable compounds in anti-arthritic therapy, might become an attractive novel therapeutic candidate.
although further investigations are needed. Some of the earliest therapeutic studies of cytokines
Angiogenesis is an important event in the genera- in RA were investigations on the effect of IFN-γ.
tion of RA44. A variety of molecules are engaged in the Although there were initial reports of beneficial effects
process, among them chemokines and other angiogenic in controlled trials208,209, the overall efficacy of IFN-γ
cytokines. The most potent is vascular endothelial therapy was relatively low, was not seen in subsequent
growth factor (VEGF), and anti-VEGF antibody ther- studies210, and significant adverse events, including the
apy ameliorates experimental arthritis196. There are induction of autoimmunity, were observed211. On the
various ways to block the action of VEGF or signal other hand, no deterioration of RA was reported, which
transduction through VEGF receptors (VEGF-R), such is interesting to note given that RA is now considered an
as humanized monoclonal anti-VEGF antibodies, solu- autoimmune disease that might be initiated and driven
ble VEGF-R, or tyrosine receptor kinase inhibitors197, by a TH1 response.
and early-phase trials are under way in several diseases
in which VEGF is implicated. The most potent VEGF Cell-surface antigens: T cells. T cells have been the most
blocker seems to be VEGF-Trap, a construct of the first frequently targeted cells in the history of the biological
three domains of the VEGF-R1 with a constant region therapy of RA. During the past decade, several attempts
of IgG1 (REF. 198). This and other VEGF blockers are to influence the disease by reducing cell number, or
CHEMOKINES
Chemokines are chemotactic candidate therapies for RA, and studies are ongoing. interfering with the function of CD4+ cells in particular,
cytokines and play a pathogenic Other anti-angiogenic compounds include angiostatin have been unsuccessful: several anti-CD4 monoclonal
role in various disorders, and endostatin, both of which are endogeneous pro- antibodies, depleting or not-depleting circulating CD4
including inflammatory teins available in recombinant form that inhibit cells, have been tested and led to initially promising
diseases. Many chemokine
receptors, such as CCRs and
endothelial cell proliferation44. Different kinase results in open trials; however, controlled trials either
CXCRs, have characteristic cell inhibitors might act in a relatively specific manner on failed to show benefit or were too small to allow final
or tissue expression profiles. endothelial cells and so inhibit angiogenesis. judgement103. Aside from prolonged lymphopenia,

482 | JUNE 2003 | VOLUME 2 www.nature.com/reviews/drugdisc

© 2003 Nature Publishing Group


REVIEWS

rashes and vasculitis complicated the application of 58%, 80% and 33% ACR20 responses were observed in
some of these antibodies212. Whether the use of higher patients receiving rituximab alone, rituximab and
doses of non-depleting anti-CD4 antibodies still consti- MTX, and placebo and MTX in inadequate MTX
tutes a feasible option remains to be seen. Like the over- responders, respectively. Remarkably, 50% of the
all results of anti-CD4 therapy and the IL-2 diphtheria patients receiving rituximab in combination with MTX,
toxin conjugate discussed above, an anti-CD5 ricin- compared with 32% on rituximab alone and 10% on
linked immunoconjugate failed to show efficacy213. MTX and placebo, achieved an ACR50 response222.
Anti-CD7 therapy is also no longer being pursued103. When administered in combination with cyclophos-
By contrast, therapy directed at interfering with T-cell phamide, rituximab had similar efficacy as when com-
co-stimulation proved quite effective. CD80/CD86 are bined with MTX. Adverse events were observed at
co-stimulatory molecules on antigen-presenting cells similar levels and types among all groups. The data
that bind to the CD28 receptor of T cells. Once activated, indicate that an INDUCTION THERAPY of rituximab might be
T cells can express CTLA4 (CD 152), another member of highly efficacious, although this, ultimately, will have to
the CD28 receptor family, which binds with higher affin- be proven in Phase III trials. It will also be interesting to
ity to CD80/86 than CD28 and silences T-cell activity214. learn more about the mode of action of this therapy.
A fusion protein, CTLA4Ig, which binds to CD80/86 and Although not aiming at a B-cell surface antigen
prevents its interaction with CD28 and subsequent T-cell directly, the efficacy of therapy targeting B cells and their
activation, has meanwhile been successfully tested in products is also supported by studies on immunoglob-
several Phase II trials. ACR20 responses were observed in ulin elimination. In a controlled Phase II trial, an
53% of patients receiving the highest dose (10 mg/kg), ACR20 response was observed in 29% of the patients,
compared with 31% on placebo215. When tested in com- compared with 11% sham-treated individuals223.
bination with MTX in patients with inadequate In this context, mention should be made of Fc-γ
response to MTX, ACR responses were observed in 60% receptors (Fc-γR), which bind IgG though its Fc por-
of the patients, compared with 35% on placebo216. tion. These exist as activating and inhibitory types, such
Furthermore, a combination of CTLA4Ig at suboptimal as Fc-γR IIb224. Interference with Fc-γR activity might
doses (2 mg/kg) with etanercept in patients with inade- significantly modulate disease expression in experi-
quate response to this TNF blocker resulted in ACR20 mental inflammatory and autoimmune models55,225,
responses in 48% of patients, compared with 28% on and the anti-inflammatory properties of intravenous
placebo217. Phase III trials are in progress. Whether the immunoglobulins might be mediated through
mode of action of CTLA4Ig in RA is merely an interrup- inhibitory Fc receptors.
tion of T-cell activation, or some other mechanism, The complement system, aside from its involvement
remains to be established. in innate immunity, can mediate immune-complex-
Another important co-stimulatory ligand–receptor related cell and tissue injury, and is activated in, and so
pair are CD40 and CD40L (CD154), and monoclonal probably pathogenically involved in, RA53. Eculizumab,
anti-CD40L antibodies have been developed to inter- a humanized monoclonal antibody that prevents the
fere with their interaction. Initial trials in another cleavage of human complement component C5 into its
rheumatic disease, systemic lupus erythematosus, pro-inflammatory components, has been investigated
seemed to be successful with respect to reducing clini- in Phase II trials of RA after achieving ACR20 responses
cal disease activity, but were complicated by throm- in 50% of patients in early-phase studies, and results are
boembolic events218. The importance of CD40L/CD40 pending226. Other possibilities for inhibiting comple-
interactions in T- and B-cell activation219 makes this ment-mediated immune responses include interference
co-stimulatory pathway an attractive target in RA, with complement receptors (CRs) and the application
provided that safety issues can be resolved. of soluble CR1, which blocks complement activation227.
Co-stimulation is also provided by yet another
receptor–ligand pair, inducible T-cell co-stimulator Adhesion molecules. Adhesion molecules play a pivotal
(ICOS)–ICOS-L. Interestingly, in contrast to CD40 and role in cell recruitment to inflammatory sites. Interfering
CD28, ICOS seems to be solely expressed on T cells and with the process of adhesion can, therefore, reduce cellu-
ICOS-deficiency is not accompanied by significant lar accumulation and thereby modify the process of
immunodeficiency220. Moreover, inhibition of this co- inflammation. An important adhesion molecule is
stimulatory pathway ameliorates experimental arthritis. ICAM-1 (CD54)228. The efficacy and safety of a mouse
monoclonal anti-ICAM-1 antibody was evaluated in
Cell-surface antigens: B cells and complement. As dis- several studies. Clinical improvement was observed in an
cussed previously, B cells, and particularly autoantibody initial open-label dose-escalation study, in which adverse
production, seem to be important in the pathogenesis of events were minor and transient229. An open-label trial in
INDUCTION THERAPY
RA. Pilot open trials indicated that anti-CD20 (rituxi- ten patients with early RA revealed a marked or moder-
Therapeutic approach that mab), a monoclonal antibody licensed for the treatment ate clinical response in seven of the patients230. However,
involves the initial application of B-cell lymphoma, might be effective as a monother- when a second course was received by eight patients, no
of an ‘aggressive’ regimen to, apy in patients refractory to DMARDs including MTX significant clinical benefit was seen, but adverse effects
ideally, induce a remission-like
state, followed by maintenance
and TNF blockers221. In a double-blind, randomized — immune complex reactions resulting from anti-
therapy with more traditional controlled trial, patients received intravenous infusions mouse Ig antibodies — were observed, which had not
agents, such as MTX. of rituximab alone or in combination with other drugs: been seen during the initial course231. Taken together,

NATURE REVIEWS | DRUG DISCOVERY VOLUME 2 | JUNE 2003 | 4 8 3

© 2003 Nature Publishing Group


REVIEWS

LFA-1 and VLA-4 can be targeted by small compounds


(including peptides) as well as biological agents. Recently,
the humanized anti-CD11a monoclonal antibody efali-
MMPs zumab has been shown to be efficacious in psoriasis, as
Chemokines well as in initial studies in renal transplant patients236.
Likewise, natalizumab, a humanized monoclonal anti-
body against α4 integrin, has been proven to be highly
effective both in Crohn’s disease and multiple sclero-
sis237,238. Finally, alefacept, a recombinant LFA-3–IgG1
VEGF fusion protein that blocks LFA-3–CD2 interactions, has
IL-17 been used successfully in psoriasis and in initial investiga-
tions on psoriatic arthritis239,240. Given that therapies that
IL-6 are effective in RA are also effective in Crohn’s disease
and psoriasis, including the TNF blockers, it might be
assumed that this is also reciprocally true, a supposition
which will have to be proven in the near future.
IL-15 Other potential targets among the adhesion mole-
IL-18 cules obviously include the selectins and the cadherins241.
Moreover, it is of particular interest that a widely used
TNF
group of therapeutics, the statins, apparently inhibit
IL-1
integrin function; this is unrelated to their inhibition
of HMG-CoA reductase and instead results from bind-
ing to a specific site on the integrin molecule. In fact,
Figure 5 | The inflammatory house of cards. The findings that many cytokines are involved in statins inhibited the inflammatory reaction in an
the generation of the inflammatory and destructive events characteristic of rheumatoid arthritis, experimental model242.
as well as observations in various clinical trials on the significant and similar efficacy of targeting
different cytokines, but the rare occurrence of remissions, suggest a multiplicity, redundancy and
even synergy of these events. This is depicted here as a house of cards: pulling one card — Toll-like receptors. Toll-like receptors (TLRs) have
that is, interference with one target — breaks part of this ‘inflammatory composite’. However, attracted significant interest because of their capacity to
no single card, when eliminated, seems to be sufficient to allow the collapse of the whole recognize pathogen-related molecular patterns and to
inflammatory construct; rather, it might be necessary to interfere with more than one molecule to thereby subserve functions within the innate immune
fully block the events. The indicated cytokines can be replaced by other molecules, such as system. In addition, their involvement as regulators of
co-stimulatory molecules or CD20, autoantibodies, and others, or by different types of cells,
the immune system and their bridging function between
including T cells, B cells, macrophages, mast cells and dendritic cells. IL, interleukin; MMPs, matrix
metalloproteinases; TNF, tumour necrosis factor; VEGF, vascular endothelial growth factor.
the innate and the adaptive defence, as well as their inter-
action with several additional endogenous ligands, add
to their allure. These manifold activities make them
attractive candidates for therapeutic interventions in
these data indicate that the use of mouse monoclonal many diseases, including chronic inflammation, and
antibodies is precluded, but that ICAM-1 might con- possibilities in this area have been recently reviewed243. As
stitute an important therapeutic target. stated above, the activation of TLRs might be the earliest
Another adhesion molecule, integrin αvβ3 (CD51/ event in the pathogenic cascade of RA, and several TLRs
CD61), has been the focus of more recent research. This are expressed in the rheumatoid synovial membrane244.
molecule, also known as the vitronectin receptor, is
expressed at high levels on macrophages, osteoclasts and Osteoclasts. The hallmark of RA is the destruction of car-
angiogenic endothelial cells232. The principal ligands of tilage and bone. As has been detailed previously (FIG. 2),
αvβ3 are extracellular matrix molecules such as this bone destruction is mediated predominantly, if not
fibronectin and osteopontin. Interference with integrin solely, by osteoclasts50–52,245. Osteoclasts require signalling
αvβ3 signalling leads to a defect in bone resorption233, through their receptor RANK, which engages its ligand
and antagonists of the vitronectin receptor, such as SB RANKL on other cells, such as osteoblasts, fibroblasts or
273005, ameliorate experimental arthritis234. A human- T cells, for their differentiation, maturation and activa-
ized monoclonal IgG1 antibody to a conformational epi- tion. RANKL is induced by a variety of signals, among
tope formed by the αv and the β3 subunit, MEDI-522 them the pro-inflammatory cytokines TNF-α and IL-1.
(Vitaxin) has been engineered, and clinical trials in RA A decoy receptor, osteoprotegerin (OPG), is able to bind
are in progress235. RANKL and to consequently prevent RANKL–RANK
Three further important adhesion molecules are interaction246. A fusion protein of recombinant OPG and
leukocyte-function-associated antigen (LFA)-1, which IgG-Fc ameliorates several types of experimental arthritis
consists of the subcomponents CD11a and CD18 (αLβ2 in terms of bone destruction, but not inflammation246–249.
integrin), CD58 (also known as LFA-3), which is a mem- So, interfering with osteoclast activation in RA could pre-
ber of the immunoglobulin superfamily, and very late vent joint damage, while the inflammatory process could
antigen-4 (VLA-4), an α4β1 integrin (CD49d/CD29). be targeted by other means (which would not necessarily
LFA-1 interacts with ICAMs, LFA-3 with CD2 and VLA-4 need to be DMARDs). Such approach could, therefore,
binds to VCAM-1 and fibronectin. Like other integrins, also work in patients in whom traditional DMARDs and

484 | JUNE 2003 | VOLUME 2 www.nature.com/reviews/drugdisc

© 2003 Nature Publishing Group


REVIEWS

biologicals fail to arrest bone destruction. OPG has been become a societal issue. The evolution of efficacious and
investigated in several disorders of bone metabolism250 safe small-molecule compounds, especially if orally
and is being studied in RA246; results from these trials are available, could overcome this concern. Somatic gene
eagerly awaited. therapy is another approach being investigated, although
In this respect, another promising group of molecules there are many challenges to be overcome (ONLINE BOX 2).
are the bisphosphonates251. Although initial clinical trials Even the best available therapies at present do not cure
in RA failed to show retardation of joint destruction252, RA and do not even sufficiently retard disease progres-
this could have resulted from the limitation of dosing sion of this ailment in a majority of the patients. Potential
strategies by oral administration. Recent experimental therapeutic targets (see ONLINE TABLE 4 for a summary of
data from TNF-mediated destructive arthritis indicate novel targets) are as numerous as the still partly enigmatic
that high doses of bisphosphonates, although not inter- pathogenesis of RA is complex. Many therapeutic
fering with inflammation, could be highly effective in the approaches have failed, but many others are successful. In
prevention of joint destruction247. With the availability of a few patients, this success is permanent, whereas in
parenteral bisphosphonates, these findings might war- others it is complete but transient, indicating a break-
rant a renewed look at the efficacy of this class of drugs through of other pathological mechanisms. In yet others
in interfering with bone destruction. it is incomplete or does not occur at all. Interestingly, if
effective, most of the newer therapeutic approaches to RA
Summary and prospects furnish similar degrees of efficacy, both clinically and
During the past 10–15 years, rheumatologists have wit- with respect to inflammatory mediators and surrogates.
nessed a change in the fate of most RA patients that Regardless of the intervention, be it cytokine-, receptor-,
would not have been predicted in earlier years: RA can signal-transduction- or cell-type-directed, the inflamma-
be relatively well controlled in many patients, extra- tory process collapses at least partly, but rarely completely,
articular manifestations, particularly vasculitis, have and never in all patients (FIG. 5), which reflects both the
become rare, and a consequence of the chronic inflam- pathogenetic complexity and redundancy. These findings
matory process — secondary amyloidosis — is also now indicate that targeting individual molecules will not
only rarely seen. This change was brought about, in part, suffice to stop the ongoing events to a sufficient degree in
by the broad application of more efficient therapies, a large number of patients. Consequently, until the
such as MTX and sulphasalazine, at effective doses and, cause(s) of RA is known and causative therapy is avail-
more recently, with the newly approved agents. It also able, the aim will be to interfere with several targets in a
resulted from the recognition that DMARD therapy combined way. Although the right mix has not yet been
should be started immediately with the diagnosis of the determined, the multiplicity of the promising targets, and
disease. In fact, there could even be a therapeutic win- the use of combinations of existing and/or future thera-
dow of opportunity for achieving remission or even peutics, could pave the way to novel therapeutic regi-
cure very early in the course of the disease, and future mens. Although rheumatology, and particularly the
studies will have to evaluate which agents, if any, can therapy of RA, seemed to be approaching a dead-end a
induce full remission at the early stage of RA. decade ago, the developments in basic and clinical
Many approved and investigational agents are research (ONLINE BOX 3) during this period, both in acade-
recombinant proteins whose large-scale production is mia and industry, have not only led to the new therapies
expensive. Given the prevalence and chronic nature of that now complement the traditional ones, but have also
RA, the cumulative costs of treatment are likely to made the outlook for the future of RA therapy bright.

1. Gabriel, S. E. The epidemiology of rheumatoid arthritis. 11. Jaffe, I. A. The treatment of rheumatoid arthritis and by rheumatoid arthritis-specific autoantibodies. J. Clin.
Rheum. Dis. Clin. North Am. 27, 269–282 (2001). necrotizing vasculitis with penicillamine. Arthritis Rheum. Invest. 101, 273–281 (1998).
2. Wolfe, F. E. & Hawley, D. J. The longterm outcome of 13, 436–443 (1970). 20. MacKay, K. et al. Whole-genome linkage analysis of rheuma-
rheumatoid arthritis. Work disability: a prospective 18 year 12. Kloppenburg, M. A. et al. Minocycline in active rheumatoid toid arthritis susceptibility loci in 252 affected sibling pairs in
study of 816 patients. J. Rheumatol. 25, 2108–2117 (1998). arthritis. A double-blind, placebo-controlled trial. Arthritis the United Kingdom. Arthritis Rheum.46, 632–639 (2002).
3. Pincus, T. E., Brooks, R. H. & Callahan, L. F. Prediction of Rheum. 37, 629–636 (1994). 21. Steiner, G. et al. Cytokine production by synovial T cells in
long-term mortality in patients with rheumatoid arthritis 13. Armant, M. A. & Fenton, M. J. Toll-like receptors: a family of rheumatoid arthritis. Rheumatology (Oxford) 38, 202–213
according to simple questionnaire and joInt count measures pattern–recognition receptors in mammals. Genome Biol. 3, (1999).
Ann. Intern. Med. 120, 26–34 (1994). REVIEWS3011 (2002). 22. Smolen, J. S. et al. The role of T-lymphocytes and cytokines
4. Yelin, E. & Wanke, L. A. An assessment of the annual and 14. Dobrovolskaia, M. A. & Vogel, S. N. Toll receptors, CD14, in rheumatoid arthritis. Scand. J. Rheumatol. 25, 1–4 (1996).
long-term direct costs: the impact of poor function and and macrophage activation and deactivation by LPS. 23. Schulze-Koops, H. & Kalden, J. R. The balance of Th1/Th2
functional decline. Arthritis Rheum. 42, 1209–1218 (1999). Microbes. Infect. 4, 903–914 (2002). cytokines in rheumatoid arthritis. Best. Pract. Res. Clin.
5. Pugner, K. M., Scott, D. I., Holmes, J. W. & Hieke, K. The 15. Romagnani, S., Matucci, A. & Rossi, O. in Targeted Rheumatol. 15, 677–691 (2001).
costs of rheumatoid arthritis: an international long-term view. Therapies in Rheumatology (eds Smolen, J. S. & Lipsky, P. E.), 24. Dolhain, R. J. et al. Shift toward T lymphocytes T helper 1
Semin. Arthritis Rheum. 29, 305–320 (2000). 317–328 (Martin Dunitz, London–New York, 2003). cytokine-secretion profile in the joints of patients with
6. Smolen, J. S. et al. Validity and reliability of the twenty-eight- 16. Smolen, J. S. & Steiner, G. Rheumatoid arthritis is more than rheumatoid arthritis. Arthritis Rheum. 39, 1961–1969 (1996).
joInt count for the assessment of rheumatoid arthritis activity. cytokines: autoimmunity and rheumatoid arthritis. Arthritis 25. Dolhain, R. J. et al. Shift toward T lymphocytes with a T
Arthritis Rheum. 38, 38–43 (1995). Rheum. 44, 2218–2220 (2001). helper 1 cytokine-secretion profile in the joints of patients with
7. Silman, A. J. & Pearson, J. E. Epidemiology and genetics of 17. Steiner, G. & Smolen, J. S. Autoantibodies in rheumatoid rheumatoid arthritis. Arthritis Rheum. 39, 1961–1969 (1996).
rheumatoid arthritis. Arthritis Res. 4 (Suppl 3), S265–S272 arthritis and their clinical significance. Arthritis Res. 4, S1–S5 26. Skapenko, A. et al. Altered memory T cell differentiation in
(2002). (2002). patients with early rheumatoid arthritis. J. Immunol. 163,
8. Ebringer, A. & Wilson, C. HLA molecules, bacteria and 18. Fritsch, R. et al. Characterization of autoreactive T cells to 491–499 (1999).
autoimmunity. J. Med. Microbiol. 49, 305–311 (2000). the autoantigens heterogeneous nuclear ribonucleoprotein 27. Santiago-Schwarz, F., Anand, P., Liu, S. & Carsons, S. E.
9. Toivanen, P. From reactive arthritis to rheumatoid arthritis. A2 (RA33) and filaggrin in patients with rheumatoid arthritis. Dendritic cells (DCs) in rheumatoid arthritis (RA): progenitor
J. Autoimmun 16, 369–371 (2001). J. Immunol. 169, 1068–1076 (2002). cells and soluble factors contained in RA synovial fluid yield
10. van der Heijde, D. M. et al. Effects of hydroxychloroquine 19. Schellekens, G. A., de Jong, B. A. W., van den Hoogen, F. a subset of myeloid DCs that preferentially activate TH1
and sulphasalazine on progression of joint damage in H. J., van de Putte, L. B. A. & van Venrooj, W. Citrulline is an inflammatory-type responses. J. Immunol. 167,
rheumatoid arthritis. Lancet 1, 1036–1038 (1989). essential constituent of antigenic determinants recognized 1758–1768 (2001).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 2 | JUNE 2003 | 4 8 5

© 2003 Nature Publishing Group


REVIEWS

28. Isler, P., Vey, E., Zhang, J. H. & Dayer, J. M. Cell surface 56. Elenkov, I. J. & Chrousos, G. P. Stress hormones, 81. Feldmann, M. & Maini, R. N. Anti-TNF alpha therapy of
glycoproteins expressed on activated human T cells induce proinflammatory and antiinflammatory cytokines, and rheumatoid arthritis: what have we learned? Annu. Rev.
production of interleukin 1 beta by monocytes: a possible autoimmunity. Ann. N. Y. Acad. Sci. 966, 290–303 (2002). Immunol. 19, 163–196 (2001).
role of CD69. Eur. Cytokine Netw. 4, 15–23 (1993). 57. Shevach, E. M. et al. Control of autoimmunity by regulatory Summarizes the background, effects and limitations
29. McInnes, I. B., Leung, B. P., Sturrock, R. D., Field, M. & T cells. Adv. Exp. Med. Biol. 490, 21–32 (2001). of TNF-blocking therapies.
Liew, F. Y. Interleukin-15 mediates T cell-dependent 58. ten Wolde, S. et al. Randomised placebo-controlled study of 82. Keffer, J. et al. Transgenic mice expressing human tumour
regulation of tumor necrosis factor-alpha production in stopping second-line drugs in rheumatoid arthritis. Lancet necrosis factor: a predictive genetic model of arthritis.
rheumatoid arthritis. Nature Med. 3, 189–195 (1997). 347, 347–352 (1996). EMBO J. 10, 4025–4031 (1991).
30. Stout, R. D. Macrophage activation by T cells: cognate and 59. ten Wolde, S., Hermans, J., Breedveld, F. C. & Dijkmans, B. A. 83. Elliott, M. J. et al. Randomised double-blind comparison of
non-cognate signals. Curr. Opin. Immunol. 5, 398–403 (1993). Effect of resumption of second line drugs in patients with chimeric monoclonal antibody to tumour necrosis factor
31. Jovanovic, D. V. et al. IL-17 stimulates the production of rheumatoid arthritis that flared up after treatment alpha (cA2) versus placebo in rheumatoid arthritis. Lancet
proinflammatory cytokines, IL-1beta and TNF-alpha, by discontinuation. Ann. Rheum. Dis. 56, 235–239 (1997). 344, 1105–1110 (1994).
human macrophages. J. Immunol. 160, 3513–3521 (1998). 60. Weinblatt, M. E. Efficacy of methotrexate in rheumatoid A landmark paper reporting on the first controlled trial
32. Crowston, J. G., Salmon, M., Khaw, P. T. & Akbar, A. N. arthritis. Br. J. Rheumatol. 34, S43–S48 (1995). of a TNF blocker in rheumatoid arthritis.
T-lymphocyte-fibroblast interactions. Biochem. Soc. Trans 61. Bathon, J. M. et al. A comparison of etanercept and 84. Lipsky, P. E. et al. Infliximab and methotrexate in the
25, 529–531 (1997). methotrexate in patients with early rheumatoid arthritis. treatment of rheumatoid arthritis. Anti-Tumor Necrosis Factor
33. Bombara, M. P. et al. Cell contact between T cells and N. Engl. J. Med. 343, 1586–1593 (2000). Trial in Rheumatoid Arthritis with Concomitant Therapy Study
synovial fibroblasts causes induction of adhesion molecules 62. Smolen, J. S. et al. Efficacy and safety of leflunomide Group. N. Engl. J. Med. 343, 1594–1602 (2000).
and cytokines. J. Leukoc. Biol. 54, 399–406 (1993). compared with placebo and sulphasalazine in active 85. Weinblatt, M. E. et al. A trial of etanercept, a recombinant
34. Dayer, J. M., Graham, R., Russel, G. & Krane, S. M. rheumatoid arthritis: a double-blind, randomised, tumor necrosis factor receptor:Fc fusion protein, in patients
Collagenase production by rheumatoid synovial cells: multicentre trial. Lancet 353, 259–266 (1999). with rheumatoid arthritis receiving methotrexate. N. Engl. J.
stimulation by a human lymphocyte factor. Science 195, 63. Manna, S. K. & Aggarwal, B. B. Immunosuppressive Med. 340, 253–259 (1999).
181–183 (1977). leflunomide metabolite (A77 1726) blocks TNF-dependent 86. Moreland, L. W. et al. Treatment of rheumatoid arthritis with
35. Eastgate, J. A. et al. Correlation of plasma interleukin-1 nuclear factor-κB activation and gene expression. recombinant human tumor necrosis factor receptor (p75)-Fc
levels with disease activity in rheumatoid arthritis. Lancet 2, J. Immunol. 162, 2095–2102 (1999). fusion protein. N. Engl. J. Med. 337, 141–147 (1997).
706–709 (1988). 64. Siemasko, K. F. et al. Inhibition of JAK3 and STAT6 tyrosine 87. Weinblatt, M. E. et al. Adalimumab, a fully human anti-tumor
36. Firestein, G. S., Alvaro-Gracia, J. M. & Maki, R. Quantitative phosphorylation by the immunosuppressive drug necrosis factor alpha monoclonal antibody, for the treatment
analysis of cytokine gene expression in rheumatoid arthritis. leflunomide leads to a block in IgG1 production. J. Immunol. of rheumatoid arthritis in patients taking concomitant metho-
J. Immunol. 144, 3347–3353 (1990). 160, 1581–1588 (1998). trexate: The ARMADA trial. Arthritis Rheum.48, 35–45 (2003).
37. Miossec, P., Dinarello, C. A. & Ziff, M. Interleukin-1 65. Wolfe, F., Hawley, D. J. & Cathey, M. A. Termination of slow 88. Kempeni, J. Update on D2E7: a fully human anti-tumour
lymphocyte chemotactic activity in rheumatoid arthritis acting antirheumatic therapy in rheumatoid arthritis: necrosis factor alpha monoclonal antibody. Ann. Rheum.Dis.
synovial fluid. Arthritis Rheum.29, 461–470 (1986). a 14-year prospective evaluation of 1017 consecutive starts. 59 S1, i44–i45 (2000).
38. Okamoto, H. et al. The synovial expression and serum levels J. Rheumatol. 17, 994–1002 (1990). 89. Furst, D. E. et al. Updated consensus statement on
of interleukin-6, interleukin-11, leukemia inhibitory factor, and 66. Aletaha, D. & Smolen, J. S. Effectiveness profiles and dose biological agents for the treatment of rheumatoid arthritis
oncostatin M in rheumatoid arthritis. Arthritis Rheum.40, dependent retention of traditional disease modifying and other rheumatic diseases (May 2002). Ann. Rheum.Dis.
1096–1105 (1997). antirheumatic drugs for rheumatoid arthritis. An 61 S2, ii2–ii7 (2002).
39. Saxne, T., Palladino, M. A. Jr, Heinegard, D., Talal, N. & observational study. J. Rheumatol. 29, 1631–1638 (2002). 90. Keane, J. et al. Tuberculosis associated with infliximab, a
Wollheim, F. A. Detection of tumor-necrosis factor alpha but 67. Aletaha, D. & Smolen, J. S. The rheumatoid arthritis patient in tumor necrosis factor alpha- neutralizing agent. N. Engl. J.
not tumor necrosis factor beta in rheumatoid arthritis synovial the clinic: comparing more than 1300 consecutive DMARD Med. 345, 1098–1104 (2001).
fluid and serum. Arthritis Rheum. 31, 1041–1045 (1988). courses. Rheumatology (Oxford) 41, 1367–1374 (2002). 91. Mohan, N. et al. Demyelination occurring during anti-tumor
40. Chu, C. Q., Field, M., Feldmann, M. & Maini, R. N. 68. van der Heide, A. et al. The effectiveness of early treatment necrosis factor alpha therapy for inflammatory arthritides.
Localization of tumor-necrosis factor-alpha in synovial with ‘second–line’ antirheumatic drugs: a randomized, Arthritis Rheum.44, 2862–2869 (2001).
tissues and at the cartilage-pannus junction in patients with controlled trial. Ann. Intern. Med. 124, 699–707 (1996). 92. Arend, W. P., Malyak, M., Guthridge, C. J. & Gabay, C.
rheumatoid arthritis. Arthritis Rheum.34, 1125–1132 (1991). First major study to show that early institution of Interleukin-1 receptor antagonist: role in biology. Annu. Rev.
41. Brennan, F. M., Chantry, D., Jackson, A., Maini, R. & (traditional) disease-modifying drugs has significantly Immunol. 16, 27–55 (1998).
Feldmann, M. Inhibitory effect of TNF-alpha antibodies on higher efficacy than delayed therapy. 93. Horai, R. et al. Development of chronic inflammatory
synovial cell interleukin-1 production in rheumatoid arthritis. 69. Nell, V. et al. The benefit of very early refferal and therapy arthropathy resembling rheumatoid arthritis in interleukin 1
Lancet 2, 244–247 (1989). with disease-modifying antirheumatic drugs in patients with receptor antagonist-deficient mice. J. Exp. Med. 191,
42. Dinarello, C. A. et al. Tumor necrosis factor (cachectin) is an early rheumatoid arthritis. Arthritis Rheum. 49, S334 (2002). 313–320 (2000).
endogeneous pyrogen and induces production of 70. Boers, M. et al. Randomised comparison of combined step- Reveals the importance of IL-1 for the development of
interleukin 1. J. Exp. Med. 163, 1433–1450 (1986). down prednisolone, methotrexate and sulphasalazine with erosive arthritis using an experimental model; these
43. Liew, F. Y. & McInnes, I. B. Role of interleukin 15 and sulphasalazine alone in early rheumatoid arthritis: a data are supported by the clinical efficacy of
interleukin 18 in inflammatory response. Ann. Rheum. Dis. randomised trial. Lancet 350, 309–318 (1997). rheumatoid arthritis therapy with recombinant IL-1ra.
61 S2, ii100–ii102 (2002). 71. Mottonen, T. et al. Comparison of combination therapy with 94. Bresnihan, B. et al. Treatment of rheumatoid arthritis with
44. Folkman J. in Targeted Therapies in Rheumatology single-drug therapy in early rheumatoid arthritis: a recombinant human interleukin-1 receptor antagonist.
Smolen J. S. & Lipsky, P. L. (eds) 111–131 (Dunitz, London, randomised trial. FIN-RACo trial group. Lancet 353, Arthritis Rheum.41, 2196–2204 (1998).
2003). 1568–1573 (1999). 95. Cohen, S. et al. Treatment of rheumatoid arthritis with
Reviews the role of angiogenesis and therapeutic 72. O’Dell, J. Treatment of rheumatoid arthritis with anakinra, a recombinant human interleukin-1 receptor
approaches to interfere with this important aspect of methotrexate alone, sulfasalazine and hydroxychloroquine, antagonist, in combination with methotrexate: results of a
chronic inflammatory disorders. or a combination of all three medications. N. Engl. J. Med. twenty-four-week, multicenter, randomized, double-blind,
45. Karin, M. & Lin, A. NF-κB at the crossroads of life and death. 334, 1287–1291 (1996). placebo-controlled trial. Arthritis Rheum. 46, 614–624 (2002).
Nature Immunol. 3, 221–227 (2002). 73. Dougados, M. et al. Combination therapy in early 96. Jiang, Y. et al. A multicenter, double-blind, dose-ranging,
46. Shaulian, E. & Karin, M. AP-1 as a regulator of cell life and rheumatoid arthritis: a randomised, controlled, double blind randomized, placebo- controlled study of recombinant
death. Nature Cell Biol. 4, 131–136 (2002). 52 week clinical trial of sulphasalazine and methotrexate human interleukin-1 receptor antagonist in patients with
47. Aringer, M., Watford, W. & O’Shea, J. J. in Targeted compared with the single components. Ann. Rheum. Dis. rheumatoid arthritis: radiologic progression and correlation
Therapies in Rheumatology Smolen, J. S. & Lipsky, P. E. 58, 220–225 (1999). of Genant and Larsen scores. Arthritis Rheum. 43,
(eds) 404–420 (Martin Dunitz, London–New York, 2003). 74. Felson, D. T., Anderson, J. J. & Meenan, R. F. The efficacy 1001–1009 (2000).
48. Weber, C. Novel mechanistic concepts for the control of and toxicity of combination therapy in rheumatoid arthritis. 97. Konttinen, Y. T. et al. Analysis of 16 different matrix
leukocyte transmigration: specialization of integrins, chemo- A meta-analysis. Arthritis Rheum. 37, 1487–1491 (1994). metalloproteinases (MMP-1 to MMP-20) in the synovial
kines, and junctional molecules. J. Mol. Med. 81, 4–19 (2003). 75. Haagsma, C. J., van Riel, P. L., de Jong, A. J. & van de membrane: different profiles in trauma and rheumatoid
49. Zvaifler, N. J. & Firestein, G. S. Pannus and pannocytes. Putte, L. B. Combination of sulphasalazine and methotrexate arthritis. Ann. Rheum. Dis. 58, 691–697 (1999).
Alternative models of joint destruction in rheumatoid arthritis. versus the single components in early rheumatoid arthritis: a 98. Martel-Pelletier, J., Welsch, D. J. & Pelletier, J. P.
Arthritis Rheum. 37, 783–789 (1994). randomized, controlled, double-blind, 52 week clinical trial. Metalloproteases and inhibitors in arthritic diseases. Best.
50. Gravallese, E. M. et al. Identification of cell types responsible Br. J. Rheumatol. 36, 1082–1088 (1997). Pract. Res. Clin. Rheumatol. 15, 805–829 (2001).
for bone resorption in rheumatoid arthritis and juvenile 76. Mottonen, T. T., Hannonen, P. J. & Boers, M. Combination 99. Tang, B. L. ADAMTS: a novel family of extracellular matrix
rheumatoid arthritis. Am. J. Pathol. 152, 943–951 (1998). DMARD therapy including corticosteroids in early rheumatoid proteases. Int. J. Biochem. Cell Biol. 33, 33–44 (2001).
51. Teitelbaum, S. L. Bone resorption by osteoclasts. Science arthritis. Clin. Exp. Rheumatol. 17, S59–S65 (1999). 100. Goodman, M., Bhumralkar, Jefferson, E. A., Kwak, J. &
289, 1504–1508 (2000). 77. Dougados, M. & Smolen, J. S. Pharmacological Locardi, E. Collagen mimetics. Biopolymers 47, 127–142
52. Redlich, K. et al. Osteoclasts are essential for TNF-α-med- management of early rheumatoid arthritis — does (1998).
iated joint destruction. J. Clin. Invest. 110, 1419–1427 (2002). combination therapy improve outcomes? J. Rheumatol. 101. Hidalgo, M. & Eckhardt, S. G. Development of matrix
Provides unequivocal evidence for the role of osteo- 66, S20–S26 (2002). metalloproteinase inhibitors in cancer therapy. J. Natl
clasts in TNF-mediated joint destruction by investi- 78. Tugwell, P. et al. Combination therapy with cyclosporine and Cancer Inst. 93, 178–193 (2001).
gating arthritis in a double-transgenic model of TNF methotrexate in severe rheumatoid arthritis. The 102. Close, D. R. Matrix metalloproteinase inhibitors in rheumatic
overexpression and deficient osteoclastogenesis. Methotrexate-Cyclosporine Combination Study Group. diseases. Ann. Rheum. Dis. 60 S3, iii62–iii67 (2001).
53. Neumann, E. et al. Local production of complement proteins N. Engl. J. Med. 333, 137–141 (1995). 103. Keystone, E. Treatments no longer in development for
in rheumatoid arthritis synovium. Arthritis Rheum. 46, 79. Kremer, J. M. et al. Concomitant leflunomide therapy in rheumatoid arthritis. Ann. Rheum. Dis. 61 S2, ii43–ii45 (2002).
934–945 (2002). patients with active rheumatoid arthritis despite stable doses 104. Brew, K., Dinakarpandian, D. & Nagase, H. Tissue inhibitors
54. Ishikawa, H., Smiley, J. D. & Ziff, M. Electron microscopic of methotrexate. A randomized, double- blind, placebo- of metalloproteinases: evolution, structure and function.
demonstration of immunoglobulin deposition in rheumatoid controlled trial. Ann. Intern. Med. 137, 726–733 (2002). Biochim. Biophys. Acta 1477, 267–283 (2000).
cartilage. Arthritis Rheum. 18, 563–576 (1975). 80. Maini, R. N. et al. Therapeutic efficacy of multiple intravenous 105. Schett, G. et al. Adenovirus-based overexpression of tissue
55. Kleinau, S., Martinsson, P. & Heyman, B. Induction and infusions of anti-tumor necrosis factor alpha monoclonal inhibitor of metalloproteinases 1 reduces tissue damage in
suppression of collagen-induced arthritis is dependent on antibody combined with low-dose weekly methotrexate in the joints of tumor necrosis factor alpha transgenic mice.
distinct Fcγ receptors. J. Exp. Med. 191, 1611–1616 (2000). rheumatoid arthritis. Arthritis Rheum.41, 1552–1563 (1998). Arthritis Rheum. 44, 2888–2898 (2001).

486 | JUNE 2003 | VOLUME 2 www.nature.com/reviews/drugdisc

© 2003 Nature Publishing Group


REVIEWS

106. Black, R. A. et al. A metalloproteinase disintegrin that 131. Liverton, N. J. et al. Design and synthesis of potent, 157. Matsumoto, I. et al. Arthritis provoked by linked T and B cell
releases tumour-necrosis factor-α from cells. Nature 385, selective, and orally bioavailable tetrasubstituted imidazole recognition of a glycolytic enzyme. Science 286,1732-5
729–733 (1997). inhibitors of p38 mitogen-activated protein kinase. J. Med. (1999).
107. Ohta, S. et al. Tumor necrosis factor-α (TNF-α) converting Chem. 42, 2180–2190 (1999). 158. Jansson, L. & Holmdahl, R. Genes on the X chromosome
enzyme contributes to production of TNF-α in synovial 132. Mclay, L. M. et al. The discovery of RPR 200765A, a p38 affect development of collagen-induced arthritis in mice.
tissues from patients with rheumatoid arthritis. J. Rheumatol. MAP kinase inhibitor displaying a good oral anti-arthritic Clin. Exp. Immunol. 94, 459–465 (1993).
28, 1756–1763 (2001). efficacy. Bioorg. Med. Chem. 9, 537–554 (2001). 159. Tsukada, S. et al. Deficient expression of a B cell
108. Beck, G. et al. (E)-2(R)-[1(S)-(Hydroxycarbamoyl)-4-phenyl- 133. Wadsworth, S. A. et al. RWJ. 67657, a potent, orally active cytoplasmic tyrosine kinase in human X- linked
3-butenyl]-2’-isobutyl-2’- (methanesulfonyl)-4- inhibitor of p38 mitogen-activated protein kinase. agammaglobulinemia. Cell 72, 279–290 (1993).
methylvalerohydrazide (Ro 32-7315), a selective and orally J. Pharmacol. Exp. Ther. 291, 680–687 (1999). 160. Bajpai, U. D., Zhang, K., Teutsch, M., Sen, R. & Wortis, H. H.
active inhibitor of tumor necrosis factor-alpha convertase. 134. Jackson, P. F. & Bullington, J. L. Pyridinylimidazole based Bruton’s tyrosine kinase links the B cell receptor to nuclear
J. Pharmacol. Exp. Ther. 302, 390–396 (2002). p38 MAP kinase inhibitors. Curr. Top. Med. Chem. 2, factor κB activation. J. Exp. Med. 191, 1735–1744 (2000).
109. Conway, J. G. et al. Inhibition of tumor necrosis factor-alpha 1011–1020 (2002). 161. Mukhopadhyay, S. et al. Macrophage effector functions
(TNF-α) production and arthritis in the rat by GW3333, a Summarizes one class of signal-transduction pathway controlled by Bruton’s tyrosine kinase are more crucial than
dual inhibitor of TNF-α-converting enzyme and matrix inhibitors, the p38 MAP kinase inhibitors, that are being the cytokine balance of T cell responses for microfilarial
metalloproteinases. J. Pharmacol. Exp. Ther. 298, 900–908 evaluated for treatment of inflammatory disorders. clearance. J. Immunol. 168, 2914–2921 (2002).
(2001). 135. Collis, A. J. et al. RPR203494 a pyrimidine analogue of the 162. Mahajan, S. et al. Rational design and synthesis of a novel
110. Zhang, Y. et al. Development of a novel dual TACE/MMP p38 inhibitor RPR200765A with an improved in vitro anti-leukemic agent targeting Bruton’s tyrosine kinase (BTK),
inhibitor for treatment of RA. Arthritis Rheum. 46, S136 potency. Bioorg. Med. Chem. Lett. 11, 693–696 (2001). LFM-A13 [α-cyano-β-hydroxy-β-methyl-N-(2, 5-
(2002). 136. Regan, J. et al. Pyrazole urea-based inhibitors of p38 MAP dibromophenyl)propenamide]. J. Biol. Chem. 274,
111. Newton, R. C. et al. Biology of TACE inhibition. Ann. kinase: from lead compound to clinical candidate. J. Med. 9587–9599 (1999).
Rheum.Dis. 60 S3, iii25–iii32 (2001). Chem. 45, 2994–3008 (2002). 163. Backesjo, C. M., Vargas, L., Superti-Furga, G. & Edvard
112. Ruuls, S. R. et al. Membrane-bound TNF supports 137. Pargellis, C. et al. Inhibition of p38 MAP kinase by utilizing a Smith, C. I. Phosphorylation of Bruton’s tyrosine kinase by c-
secondary lymphoid organ structure but is subservient to novel allosteric binding site. Nature Struct. Biol. 9, 268–272 Abl. Biochem. Biophys. Res. Commun. 299, 510–515 (2002).
secreted TNF in driving autoimmune inflammation. (2002). 164. Horwood, N. J. et al. Bruton’s tyrosine kinase is required for
Immunity 15, 533–543 (2001). 138. Cirillo, P. F., Pargellis, C. & Regan, J. The non-diaryl lipopolysaccharide-induced tumour necrosis factor α
113. Alexopoulou, L., Pasparakis, M. & Kollias, G. A murine heterocycle classes of p38 MAP kinase inhibitors. Curr. Top. production. J. Exp. Med. (in the press).
transmembrane tumor necrosis factor (TNF) transgene Med. Chem. 2, 1021–1035 (2002). Reveals the importance of BTK for TNF gene
induces arthritis by cooperative p55/p75 TNF receptor 139. Jackson, J. R. et al. Pharmacological effects of SB 220025, activation, expands its role beyond important effects
signaling. Eur. J. Immunol. 27, 2588–2592 (1997). a selective inhibitor of P38 mitogen-activated protein kinase, in B cells and increases the rationale for targeting
114. Williams, L. M. et al. Paradoxical effects of a synthetic in angiogenesis and chronic inflammatory disease models. BTK therapeutically.
metalloproteinase inhibitor that blocks both p55 and p75 J. Pharmacol. Exp. Ther. 284, 687–692 (1998). 165. Tsubata, T. & Wienands, J. B cell signaling. Introduction. Int.
TNF receptor shedding and TNF α processing in RA 140. Fijen, J. W. et al. Suppression of the clinical and cytokine Rev. Immunol. 20, 675–678 (2001).
synovial membrane cell cultures J. Clin. Invest. 97, response to endotoxin by RWJ- 67657, a p38 mitogen- 166. Choy, E. H. et al. Efficacy of a novel PEGylated humanized
2833–2841 (1996). activated protein-kinase inhibitor, in healthy human anti-TNF fragment (CDP870) in patients with rheumatoid
115. Thornberry, N. A. et al. A novel heterodimeric cysteine volunteers. Clin. Exp. Immunol. 124, 16–20 (2001). arthritis: a phase II double-blinded, randomized, dose-
protease is required for interleukin-1 β processing in 141. Pirvola, U. et al. Rescue of hearing, auditory hair cells, and escalating trial. Rheumatology (Oxford) 41, 1133–1137
monocytes. Nature 356, 768–774 (1992). neurons by CEP-1347/KT7515, an inhibitor of c-Jun (2002).
116. Schonbeck, U., Mach, F. & Libby, P. Generation of N-terminal kinase activation. J. Neurosci. 20, 43–50 (2000). 167. Rutgeerts, P. et al. Treatment of active Crohn’s disease with
biologically active IL-1β by matrix metalloproteinases: a 142. Bodmer, D., Gloddek, B., Ryan, A. F., Huverstuhl, J. & onercept (recombinant human soluble p55 tumour necrosis
novel caspase-1-independent pathway of IL-1β processing. Brors, D. Inhibition of the c-Jun N-terminal kinase signaling factor receptor): results of a randomized, open-label, pilot
J. Immunol. 161, 3340–3346 (1998). pathway influences neurite outgrowth of spiral ganglion study. Aliment. Pharmacol. Ther. 17, 185–192 (2003).
117. Fantuzzi, G., Puren, A. J., Harding, M. W., Livingston, D. J. & neurons in vitro. Laryngoscope 112, 2057–2061 (2002). 168. Coxon, A. et al. Inhibition of interleukin-1 but not tumor
Dinarello, C. A. Interleukin-18 regulation of interferon gamma 143. Han, Z. et al. c-Jun N-terminal kinase is required for necrosis factor suppresses neovascularization in rat models
production and cell proliferation as shown in interleukin-1β- metalloproteinase expression and joint destruction in of corneal angiogenesis and adjuvant arthritis. Arthritis
converting enzyme (caspase-1)-deficient mice. Blood 91, inflammatory arthritis. J. Clin. Invest. 108, 73–81 (2001). Rheum.46, 2604–2612 (2002).
2118–2125 (1998). 144. Pintus, G. et al. Inhibition of the MEK/ERK signaling pathway 169. Abramson, S. B. & Amin, A. Blocking the effects of IL-1 in
118. Perregaux, D. G. & Gabel, C. A. Post-translational by the novel antimetastatic agent NAMI-A downregulates rheumatoid arthritis protects bone and cartilage.
processing of murine IL-1: evidence that ATP-induced c-myc gene expression and endothelial cell proliferation. Rheumatology (Oxford) 41, 972–980 (2002).
release of IL-1α and IL-1β occurs via a similar mechanism. Eur. J. Biochem. 269, 5861–5870 (2002). 170. Charles, P. et al. Regulation of cytokines, cytokine
J. Immunol. 160, 2469–2477 (1998). 145. Mori, T., Wang, X., Aoki, T. & Lo, E. H. Downregulation of inhibitors, and acute-phase proteins following anti-TNF-
119. Randle, J. C., Harding, M. W., Ku, G., Schonharting, M. & matrix metalloproteinase-9 and attenuation of edema via alpha therapy in rheumatoid arthritis. J. Immunol. 163,
Kurrle, R. ICE/Caspase-1 inhibitors as novel anti- inhibition of ERK mitogen activated protein kinase in 1521–1528 (1999).
inflammatory drugs. Expert. Opin. Investig. Drugs 10, traumatic brain injury. J. Neurotrauma 19, 1411–1419 (2002). 171. Alonzi, T. et al. Interleukin 6 is required for the development
1207–1209 (2001). 146. Feldmann, M. et al. Is NF-κB a useful therapeutic target in of collagen-induced arthritis. J. Exp. Med. 187, 461–468
120. Ku, G., Faust, T., Lauffer, L. L., Livingston, D. J. & rheumatoid arthritis? Ann. Rheum.Dis. 61 S2, ii13–ii18 (2002). (1998).
Harding, M. W. Interleukin-1β converting enzyme inhibition 147. Yamamoto, Y. & Gaynor, R. B. Therapeutic potential of 172. Wendling, D., Racadot, E. & Wijdenes, J. Treatment of
blocks progression of type II collagen-induced arthritis in inhibition of the NF-κB pathway in the treatment of severe rheumatoid arthritis by anti-interleukin 6 monoclonal
mice. Cytokine 8, 377–386 (1996). inflammation and cancer. J. Clin. Invest. 107, 135–142 (2001). antibody. J. Rheumatol.20, 259–262 (1993).
121. Pavelka, K. et al. Clinical effects of pralnacasan (PRAL), an Summarizes signal transduction through the NF-κB 173. Choy, E. H. et al. Therapeutic benefit of blocking interleukin-6
orally-active interleukin-1β converting enzyme (ICE) inhibitor, pathway and therapeutic strategies to interfere with activity with an anti- interleukin-6 receptor monoclonal
in a 285 patient PhII trial in rheumatoid arthritis (RA). Abstr. this pathway. antibody in rheumatoid arthritis: a randomized, double-blind,
Am. Coll. Rheumatol. 66th Ann. Sci. Meet. (2002). 148. Umezawa, K., Ariga, A. & Matsumoto, N. Naturally occurring placebo-controlled, dose-escalation trial. Arthritis Rheum.
122. Dell’Antonio, G., Quattrini, A., Cin, E. D., Fulgenzi, A. & and synthetic inhibitors of NF-κB functions. Anticancer Drug 46, 3143–3150 (2002).
Ferrero, M. E. Relief of inflammatory pain in rats by local use Des. 15, 239–244 (2000). Reveals the efficacy of blocking yet another pro-
of the selective P2X7 ATP receptor inhibitor, oxidized ATP. 149. Pahl, H. L. et al. The immunosuppressive fungal metabolite inflammatory cytokine, IL-6, in the treatment of
Arthritis Rheum. 46, 3378–3385 (2002). gliotoxin specifically inhibits transcription factor NF-κB. J. Exp. rheumatoid arthritis.
123. Labasi, J. M. et al. Absence of the P2X7 receptor alters Med. 183, 1829–1840 (1996). 174. Naka, T., Nishimoto, N. & Kishimoto, T. The paradigm of IL-6:
leukocyte function and attenuates an inflammatory 150. Seetharaman, R., Mora, A. L., Nabozny, G., Boothby, M. & from basic science to medicine. Arthritis Res. 4 S3,
response. J. Immunol. 168, 6436–6445 (2002). Chen, J. Essential role of T cell NF-κB activation in collagen- S233–S242 (2002).
124. Karin, M. & Ben-Neriah, Y. Phosphorylation meets induced arthritis. J. Immunol. 163, 1577–1583 (1999). 175. Strom, T. B., Kelley, V. R., Murphy, J. R., Nichols, J. &
ubiquination: the control of NF-κB activity. Annu. Rev. 151. Fitzpatrick, L. R., Wang, J. & Le, T. Gliotoxin, an inhibitor of Woodworth, T. G. Interleukin-2 receptor-directed therapies:
Immunol. 18, 621–663 (2000). nuclear factor-κB, attenuates peptidoglycan-polysaccharide- antibody-or cytokine-based targeting molecules. Annu. Rev.
125. Zhang, M. & Tracey, K. J. In The Cytokine Handbook. induced colitis in rats. Inflamm. Bowel. Dis. 8, 159–167 Med. 44, 343–353 (1993).
Thompson, A. (ed.) 517–548 (Academic Press, San Diego, (2002). 176. Banerjee, S., Wei, B. Y., Hillman, K., Luthra, H. S. &
CA, 1998). 152. Burchat, A. F. et al. Pyrazolo[3, 4-d]pyrimidines containing David, C. S. Immunosuppression of collagen-induced
126. Nagase, H. & Woessner, J. F. Matrix metalloproteinases. an extended 3-substituent as potent inhibitors of Lck — a arthritis in mice with an anti-IL-2 receptor antibody.
J. Biol. Chem. 274, 21491–21494 (1999). selectivity insight. Bioorg. Med. Chem. Lett. 12, J. Immunol. 141, 1150–1154 (1988).
127. Han, Z. et al. AP-1 and NF-κB regulation in rheumatoid 1687–1690 (2002). 177. Shao, R. H., Tian, X., Gorgun, G., Urbano, A. G. & Foss, F. M.
arthritis and murine collagen-induced arthritis. Autoimmunity 153. Sadeghi, R. et al. Differential regulation of CD3- and CD28- Arginine butyrate increases the cytotoxicity of DAB(389)IL-2 in
28, 197–208 (1998). induced IL-2 and IFN-gamma production by a novel tyrosine leukemia and lymphoma cells by upregulation of IL-2Rβ gene.
128. Schett, G. et al. Activation, differential localization, and kinase inhibitor XR774 from Cladosporium cf. Leuk. Res. 26, 1077–1083 (2002).
regulation of the stress-activated protein kinases, cladosporioides. Int. Immunopharmacol. 1, 33–48 (2001). 178. Gottlieb, S. L. et al. Response of psoriasis to a lymphocyte-
extracellular signal-regulated kinase, c-JUN N-terminal 154. Yorgin, P. D. et al. Effects of geldanamycin, a heat-shock selective toxin (DAB389IL-2) suggests a primary immune,
kinase, and p38 mitogen-activated protein kinase, in protein 90-binding agent, on T cell function and T cell but not keratinocyte, pathogenic basis. Nature Med. 1,
synovial tissue and cells in rheumatoid arthritis. nonreceptor protein tyrosine kinases. J. Immunol. 164, 442–447 (1995).
Arthritis Rheum. 43, 2501–2512 (2000). 2915–2923 (2000). 179. Moreland, L. W. et al. Interleukin-2 diphtheria fusion protein
129. Haddad, J. J. VX-745. Vertex Pharmaceuticals. Curr. Opin. 155. Trevillyan, J. M. et al. Inhibition of p56(lck) tyrosine kinase by (DAB486IL-2) in refractory rheumatoid arthritis. A double-
Investig. Drugs 2, 1070–1076 (2001). isothiazolones. Arch. Biochem. Biophys. 364, 19–29 (1999). blind, placebo-controlled trial with open-label extension.
130. Badger, A. M. et al. SB 203580 inhibits p38 mitogen- 156. Kamens, J. S., Ratnofsky, S. E. & Hirst, G. C. Lck inhibitors Arthritis Rheum. 38, 1177–1186 (1995).
activated protein kinase, nitric oxide production, and as a therapeutic approach to autoimmune disease and 180. Cibrik, D. M., Kaplan, B. & Meier-Kriesche, H. U. Role of
inducible nitric oxide synthase in bovine cartilage-derived transplant rejection. Curr. Opin. Investig. Drugs 2, anti-interleukin-2 receptor antibodies in kidney
chondrocytes. J. Immunol. 161, 467–473 (1998). 1213–1219 (2001). transplantation. BioDrugs. 15, 655–666 (2001).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 2 | JUNE 2003 | 4 8 7

© 2003 Nature Publishing Group


REVIEWS

181. McInnes, I. B., Leung, B. P., Sturrock, R. D., Field, M. & double blind placebo controlled study. Ann. Rheum.Dis. 51, 235. Wilder, R. L. Integrin αVβ3 as a target for treatment of
Liew, F. Y. Interleukin-15 mediates T cell-dependent 1039–1043 (1992). rheumatoid arthritis and related rheumatic diseases. Ann.
regulation of tumor necrosis factor-alpha production in 209. Cannon, G. W. et al. Prospective two-year followup of Rheum. Dis. 61 S2, ii96–ii99 (2002).
rheumatoid arthritis. Nature Med. 3, 189–195 (1997). recombinant interferon-γ in rheumatoid arthritis. J. Rheumatol. 236. Dedrick, R. L., Walicke, P. & Garovoy, M. Anti-adhesion
182. Ruchatz, H., Leung, B. P., Wei, X. Q., McInnes, I. B. & 17, 304–310 (1990). antibodies efalizumab, a humanized anti-CD11a monoclonal
Liew, F. Y. Soluble IL-15 receptor alpha-chain administration 210. Cannon, G. W. et al. Prospective 5-year followup of antibody. Transpl. Immunol. 9, 181–186 (2002).
prevents murine collagen-induced arthritis: a role for IL-15 recombinant interferon-γ in rheumatoid arthritis. J. Rheumatol. 237. Ghosh, S. et al. Natalizumab for active Crohn’s disease.
in development of antigen- induced immunopathology. 20, 1867–1873 (1993). N. Engl. J. Med. 348, 24–32 (2003).
J. Immunol. 160, 5654–5660 (1998). 211. Graninger, W. B. et al. Induction of systemic lupus 238. Miller, D. H. et al. A controlled trial of natalizumab for relapsing
183. Butler, D. M., Malfait, A. M., Maini, R. N., Brennan, F. M. & erythematosus by interferon-γ in a patient with rheumatoid multiple sclerosis. N. Engl. J. Med. 348, 15–23 (2003).
Feldmann, M. Anti-IL-12 and anti-TNF antibodies arthritis. J. Rheumatol. 18, 1621–1622 (1991). 239. Kraan, M. C. et al. Alefacept treatment in psoriatic arthritis:
synergistically suppress the progression of murine collagen- 212. Choy, E. H. et al. Repeat-cycle study of high-dose intravenous reduction of the effector T cell population in peripheral blood
induced arthritis. Eur. J. Immunol. 29, 2205–2212 (1999). 4162W94 anti-CD4 humanized monoclonal antibody in and synovial tissue is associated with improvement of clinical
184. Gracie, J. A. et al. A proinflammatory role for IL-18 in rheumatoid arthritis. A randomized placebo- controlled trial. signs of arthritis. Arthritis Rheum. 46, 2776–2784 (2002).
rheumatoid arthritis. J. Clin. Invest. 104, 1393–1401 (1999). Rheumatology (Oxford) 41, 1142–1148 (2002). 240. Krueger, G. G. et al. A randomized, double-blind, placebo-
185. Nakamura, K., Okamura, H., Wada, M., Nagata, K. & 213. Olsen, N. J. et al. A double-blind, placebo-controlled study controlled phase III study evaluating efficacy and tolerability
Tamura, T. Endotoxin-induced serum factor that stimulates of anti-CD5 immunoconjugate in patients with rheumatoid of 2 courses of alefacept in patients with chronic plaque
γ-interferon production. Infect. Immun. 57, 590–595 (1989). arthritis. The Xoma RA Investigator Group. Arthritis Rheum. psoriasis. J. Am. Acad. Dermatol. 47, 821–833 (2002).
186. Wei, X. Q., Leung, B. P., Arthur, H. M., McInnes, I. B. & Liew, F. 39, 1102–1108 (1996). 241. Alahari, S. K., Reddig, P. J. & Juliano, R. L. Biological
Y. Reduced incidence and severity of collagen-induced arthritis 214. Slavik, J. M., Hutchcroft, J. E. & Bierer, B. E. CD28/CTLA-4 aspects of signal transduction by cell adhesion receptors.
in mice lacking IL-18. J. Immunol. 166, 517–521 (2001). and CD80/CD86 families: signaling and function. Immunol. Int. Rev. Cytol. 220, 145–184 (2002).
187. Plater-Zyberk, C. et al. Therapeutic effect of neutralizing Res. 19, 1–24 (1999). 242. Weitz-Schmidt, G. et al. Statins selectively inhibit leukocyte
endogenous IL-18 activity in the collagen-induced model of 215 Moreland, L. W. et al. A multi-center, randomized, double- function antigen-1 by binding to a novel regulatory integrin
arthritis. J. Clin. Invest. 108, 1825–1832 (2001). blind, placebo-controlled study to evaluate the safety and site. Nature Med. 7, 687–692 (2001).
188. Chabaud, M. et al. Human interleukin-17: A T cell-derived preliminary clinical activity of multiple doses of CTLA4Ig and 243. Zuany-Amorim, C., Hastewell, J. & Walker, C. Toll-like
proinflammatory cytokine produced by the rheumatoid LEA294 administered intravenously to subjects with receptors as potential therapeutic targets for multiple
synovium. Arthritis Rheum. 42, 963–970 (1999). rheumatoid arthritis. Arthritis Rheum. 44, S274 (2001). diseases. Nature Rev. Drug Discov. 1, 797–807 (2002).
189. Awane, M., Andres, P. G., Li, D. J. & Reinecker, H. C. NF-κB- 216. Kremer, J. et al. A phase 2B multi-center, randomized, 244. Seibl, R. et al. Expression and regulation of toll-like receptor 2
inducing kinase is a common mediator of IL-17-, TNF-α-, double–blind, placebo controlled study to evaluate the in rheumatoid arthritis synovium. Am. J. Pathol. 162,
and IL-1β-induced chemokine promoter activation in safety and efficacy of two different doses of CTLA4Ig 1221–1227 (2003).
intestinal epithelial cells. J. Immunol. 162, 5337–5344 (1999). administered intravenously to subjects with active 245. Gravallese, E. M. et al. Identification of cell types responsible
190. Jovanovic, D. V. et al. Modulation of TIMP-1 synthesis by rheumatoid arthritis while receiving methotrexate. Arthritis for bone resorption in rheumatoid arthritis and juvenile
antiinflammatory cytokines and prostaglandin E2 in Rheum. 46, S203 (2002). rheumatoid arthritis. Am. J. Pathol. 152, 943–951 (1998).
interleukin 17 stimulated human monocytes/macrophages. 217. Weinblatt, M. E. et al. A pilot, multi-center, randomized, 246. Bolon, B. et al. Osteoprotegerin, an endogenous
J. Rheumatol. 28, 712–718 (2001). double blind, placebo controlled study of a co–stimulation antiosteoclast factor for protecting bone in rheumatoid
191. Chabaud, M. & Miossec, P. The combination of tumor blocker CTLA4Ig (2mg/kg) given monthly in combination arthritis. Arthritis Rheum.46, 3121–3135 (2002).
necrosis factor alpha blockade with interleukin-1 and with etanercept in active rheumatoid arthritis. Arthritis 247. Redlich K et al. Tumor necrosis factor alpha-mediated joint
interleukin-17 blockade is more effective for controlling Rheum. 46 S204 (2002). destruction is inhibited by targeting osteoclasts with
synovial inflammation and bone resorption in an ex vivo 218. Daikh, D. I. & Wofsy, D. Treatment of autoimmunity by osteoprotegerin. Arthritis Rheum.46, 785–792 (2002).
model. Arthritis Rheum. 44, 1293–1303 (2001). inhibition of T cell co-stimulation. Adv. Exp. Med. Biol. 490, 248. Kong, Y. Y. et al. Activated T cells regulate bone loss and
192. Szekanecz, Z., Kim, J. & Koch, A. E. In Targeted Therapies 113–117 (2001). joint destruction in adjuvant arthritis through osteoprotegerin
in Rheumatology. Smolen, J. S. & Lipsky, P. E. (eds.) 219. Schonbeck, U. & Libby, P. The CD40/CD154 ligand. Nature 402, 304–309 (1999).
345-357 (Martin Dunitz, London–New York, 2003). receptor/ligand dyad. Cell Mol. Life Sci. 58, 4–43 (2001). 249. Pettit, A. R. et al. TRANCE/RANKL knockout mice are
193. Zapico,-I. et al. CCR5 (chemokine receptor-5) DNA- 220. Frauwirth, K. A. & Thompson, C. B. Activation and inhibition protected from bone erosion in a serum transfer model of
polymorphism influences the severity of rheumatoid arthritis. of lymphocytes by co-stimulation. J. Clin. Invest. 109, arthritis. Am. J. Pathol. 159, 1689–1699 (2001).
Genes Immun. 1, 288–289 (2000). 295–299 (2002). 250. Bekker, P. J. et al. The effect of a single dose of
194. Onuffer, J. J. & Horuk, R. Chemokines, chemokine 221. De Vita, S. et al. Efficacy of selective B cell blockade in the osteoprotegerin in postmenopausal women. J. Bone Miner.
receptors and small-molecule antagonists: recent treatment of rheumatoid arthritis: evidence for a Res. 16, 348–360 (2001).
developments. Trends Pharmacol. Sci. 23, 459–467 (2002). pathogenetic role of B cells. Arthritis Rheum. 46, 251. Heikkila, P. et al. Bisphosphonates inhibit stromelysin-1
195. Bruhl, H., Cihak, J., Stangassinger, M., Schlondorff, D. & 2029–2033 (2002). (MMP-3), matrix metalloelastase (MMP-12), collagenase-3
Mack, M. Depletion of CCR5-expressing cells with bispecific 222. Edwards, J. C. W. et al. Efficacy and safety of rituximab, (MMP-13) and enamelysin (MMP-20), but not urokinase-
antibodies and chemokine toxins: a new strategy in the a B-cell targeted chimeric monoclonal antibody: type plasminogen activator, and diminish invasion and
treatment of chronic inflammatory diseases and HIV. a randomized, placebo–controlled trial in patients with migration of human malignant and endothelial cell lines.
J. Immunol. 166, 2420–2426 (2001). rheumatoid arthritis. Arthritis Rheum. 46, S197 (2002). Anticancer Drugs 13, 245–254 (2002).
196. Sone, H. et al. Neutralization of vascular endothelial growth 223. Furst, D., Felson, D., Thoren, G. & Gendreau, R. M. 252. Eggelmeijer, F. et al. Increased bone mass with pamidronate
factor prevents collagen- induced arthritis and ameliorates Immunoadsorption for the treatment of rheumatoid arthritis: treatment in rheumatoid arthritis. Results of a three-year
established disease in mice. Biochem. Biophys. Res. final results of a randomized trial. Prosorba Trial randomized, double-blind trial. Arthritis Rheum. 39,
Commun. 281, 562–568 (2001). Investigators. Ther. Apher. 4, 363–373 (2000). 396–402 (1996).
197. Hasan, J. & Jayson, G. C. VEGF antagonists. Expert. Opin. 224. Ravetch, J. V. & Bolland, S. IgG Fc receptors. Annu. Rev. 253. Smolen, J. S. The work of the EULAR Standing Committee
Biol. Ther. 1, 703–718 (2001). Immunol. 19, 275–290 (2001). on International Clinical Studies Including Therapeutic Trials.
198. Holash, J. et al. VEGF-Trap: a VEGF blocker with potent anti- 225. Nabbe, K. C. et al. Coordinate expression of activating Fc Br. J. Rheumatol. 31, 219–220 (1992).
tumor effects. Proc. Natl Acad. Sci. USA 99, 11393–11398 gamma receptors I and III and inhibiting Fc gamma receptor 254. Felson, D. T. et al. The American College of Rheumatology
(2002). type II in the determination of joint inflammation and cartilage preliminary core set of disease activity measures for
199. Tarner, I. H. et al. Retroviral gene therapy of collagen- destruction during immune complex-mediated arthritis. rheumatoid arthritis clinical trials. Arthritis Rheum. 36,
induced arthritis by local delivery of IL-4. Clin. Immunol. 105, Arthritis Rheum. 48, 255–265 (2003). 729–740 (1993).
304–314 (2002). 226. Kaplan, M. Eculizumab (Alexion). Curr. Opin. Investig. Drugs 255. van Gestel, A. M. et al. ACR and EULAR improvement
200. van den Bosch, F. et al. rHu IL-4 in subjects with active 3, 1017–1023 (2002). criteria have comparable validity in rheumatoid arthritis.
rheumatoid arthritis (RA): a phase I dose escalating safety 227. Holers, V. M. In Targeted Therapies in Rheumatology. J. Rheumatol. 26, 705–711 (1999).
study. Arthritis Rheum. 41, S56 (1998). Smolen, J. S. & Lipsky, P. E. (eds.), pp. 167-180 256. Sharp, J. T., Strand, V., Leung, H., Hurley, F. & Loew-
201. Weinblatt, M. E. et al. rHuIL-10 (Tenovil) plus methotrexate (Martin Dunitz, London–New York, 2003). Friedrich, I. Treatment with LEF slows radiographic
(MTX) in active rheumatoid arthritis: A phase I/II study. 228. Hubbard, A. K. & Rothlein, R. Intercellular adhesion progression of rheumatoid arthritis. Results from three
Arthritis Rheum. 42 S170 (1999). molecule-1 (ICAM-1) expression and cell signaling randomized controlled trials of LEF in patients with active
202. Maini, R. N. et al. rHu IL-10 in jubjects with active cascades. Free Radic. Biol. Med. 28, 1379–1386 (2000). rheumatoid arthritis. Arthritis Rheum. 43, 495–505 (2000).
rheumatoid arthritis: A phase I cytokine response study. 229. Kavanaugh, A. F. et al. Treatment of refractory rheumatoid
Arthritis Rheum. 41, S224 (1998). arthritis with a monoclonal antibody to intercellular adhesion
203. van Roon, J. A., Bijlsma, J. W. & Lafeber, F. P. Suppression molecule 1. Arthritis Rheum. 37, 992–999 (1994).
Online links
of inflammation and joInt destruction in rheumatoid arthritis 230. Kavanaugh, A. F. et al. A phase I/II open label study of the
DATABASES
may require a concerted action of Th2 cytokines. Curr. Opin. safety and efficacy of an anti- ICAM-1 (intercellular adhesion
The following terms in this article are linked online to:
Investig. Drugs 3, 1011–1016 (2002). molecule-1; CD54) monoclonal antibody in early rheumatoid
204. Reynolds, C. H. Clinical efficacy of rhIL-11. Oncology arthritis. J. Rheumatol. 23, 1338–1344 (1996). LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink/
(Huntingt) 14, 32–40 (2000). 231. Kavanaugh, A. F., Schulze-Koops, H., Davis, L. S. & αlB2 | αvB3 | BTK | CD40 | CD40L | CD58 | CD80 | CD86 | CTLA4 |
205. Moreland, L. et al. Results of a phase-I/II randomized, Lipsky, P. E. Repeat treatment of rheumatoid arthritis ICAM-1 | ICE | IFN-γ | IL-1β | IL-1RI | IL-2 | IL-2R | IL-2Rβ | IL-4 |
masked, placebo-controlled trial of recombinant human patients with a murine anti-intercellular adhesion molecule 1 IL-5 | IL-10 | IL-12 | IL-13 | IL-15 | IL-15R α-chain | IL-18 |
interleukin-11 (rhIL-11) in the treatment of subjects with monoclonal antibody. Arthritis Rheum.40, 849–853 (1997). IκB | IKK-b | JNK | LCK | NF-κB | OPG | p38-MAPK | RANK |
active rheumatoid arthritis. Arthritis Res. 3, 247–252 (2001). 232. Eliceiri, B. P. & Cheresh, D. A. Adhesion events in RANKL | TACE | TIMP-1 | TNF-RI | VLA-4
206. Bessis, N. et al. Attenuation of collagen-induced arthritis in angiogenesis. Curr. Opin. Cell Biol. 13, 563–568 (2001). Online Mendelian Inheritance in Man:
mice by treatment with vector cells engineered to secrete 233. Chellaiah, M. et al. Gelsolin deficiency blocks podosome http://www.ncbi.nlm.nih.gov/Omim/
interleukin-13. Eur. J. Immunol. 26, 2399–2403 (1996). assembly and produces increased bone mass and strength. Agammaglobulinaemia | Crohn’s disease
207. Woods, J. M. et al. Reduction of inflammatory cytokines and J. Cell Biol. 148, 665–678 (2000).
prostaglandin E2 by IL-13 gene therapy in rheumatoid 234. Badger, A. M. et al. Disease-modifying activity of SB FURTHER INFORMATION
arthritis synovium. J. Immunol. 165, 2755–2763 (2000). 273005, an orally active, nonpeptide αVβ3 (vitronectin Encyclopedia of Life Science: http://www.els.net
208. Machold, K. P., Neumann, K. & Smolen, J. S. Recombinant receptor) antagonist, in rat adjuvant-induced arthritis. Rheumatoid arthritis
human interferon-γ in the treatment of rheumatoid arthritis: Arthritis Rheum.44, 128–137 (2001). Access to this interactive links box is free online.

488 | JUNE 2003 | VOLUME 2 www.nature.com/reviews/drugdisc

© 2003 Nature Publishing Group


Rheumatologie
Prof. Dr. med.
Klaus Krüger Quantensprünge in der Therapie
der rheumatoiden Arthritis
MMW-Schriftleiter
Rheumatologie
Praxisgemeinschaft
München Von K. Krüger

– Die Behandlung chronischer Arthriti- wie eine Handvoll Basistherapeutika – drei Substanzen wegen vermeintlich ge-
den in den letzten 25 Jahren kann in dies war neben nicht medikamentösen fährlicher kardiovaskulärer, dermatolo-
zwei Phasen unterteilt werden: Nach eher Behandlungsformen physikalischer gischer oder hepatischer Nebenwir-
schleppenden und sporadischen Neuent- und operativer Art das therapeutische kungen wieder vom Markt genommen.
wicklungen bis Anfang der 1990er-Jahre, Arsenal vor 25 Jahren. Mit Celecoxib und Etoricoxib können
durch die der zerstörerische Krankheits- jedoch zwei Coxibe z. B. bei GI-Proble-
verlauf der rheumatoiden Arthritis (RA) NSAR men eingesetzt werden. Insgesamt ha-
nur zu verzögern war, haben wir in den Im Bereich der NSAR waren die fol- ben NSAR für die symptomatische Ar-
letzten 15 Jahren eine fast quantensprung­ genden drei Jahrzehnte zunächst durch thritistherapie einen unverändert hohen
artige Entwicklung erlebt, die in das heu- diverse Neuentwicklungen gekennzeich- Stellenwert, werden aber wegen der
tige Therapieziel einer kompletten Re- net, die stets mit dem Anspruch besserer sonstigen Fortschritte heute vorwiegend
mission mündete. Dagegen ist bei der Verträglichkeit auf den Markt gelangten. als Bedarfsmedikamente verwendet.
Arthrosebehandlung trotz kleinerer Fort- Bei der praktischen Anwendung konn-
schritte der Durchbruch hin zur nachhal- ten sie dem dann aber nicht oder kaum Glukokortikoide
tigen Verlaufsbeeinflussung bisher nicht gerecht werden. So entstand eine sehr GC werden seit rund 60 Jahren in der
gelungen. Eine heilende Therapie fehlt, breite Palette ähnlicher Wirkstoffe, von Rheumatologie eingesetzt, sind jedoch
wie für die meisten rheumatischen Er- denen viele wieder verschwunden sind. gleichwohl durch ständige Weiterent-
krankungen, für beide nach wie vor. Einen deutlichen Fortschritt zumindest wicklung des Therapieprinzips charakte-
in Bezug auf die gastrointestinale Ver- risiert. Hierzu zählt die feste Etablierung
Rheumatoide Arthritis träglichkeit stellte die Entwicklung der der intraartikulären Verabreichungsform
Symptomatische Therapie mit nicht selektiven Cyclooxygenase-2-Hemmer in den letzten 20 Jahren, weiterhin die
steroidalen Antiphlogistika (NSAR), (Coxibe) Ende der 1990er-Jahre dar. Aus Erkenntnis, dass GC in niedriger Dosie-
Glukokortikoiden (GC) als „Feuerwehr“ dieser Gruppe wurden mit Rofecoxib, rung (≤ 7,5 mg) von Beginn an eine ba-
und in hoher Dosis als Ultima Ratio so- Valdecoxib und Lumiracoxib jedoch sistherapeutische Wirkung besitzen. Sie
Dieter-Lothar Adam
25 Jahre Fortschritte in der Rheumatologie –

Abbildung 1 wiegend mit Mtx kombiniert. Bei früh-


zeitigem Einsatz ist diese Kombination
Exemplarischer Therapieablauf bei hochaktiver RA in der Lage, bei ca. 50 % der RA-Pati-
enten eine Remission zu erzeugen. Ins-
Sofortiger Beginn einer Basistherapie mit Mtx nach Diagnosesicherung, vorzugs- besondere für die Kombination TNF-
weise parenteraler Beginn, schrittweise Ausdosierung + Low-dose-Kortikoid Blocker + Mtx ist nachgewiesen, dass sie
in den meisten Fällen zu einem kom-
innerhalb von 3 Monaten
pletten Stillstand der Progression von
Kombination Mtx + SSZ + Hcl
Destruktionen führt. Neue Daten zeigen
(alternativ: Mtx + Leflunomid, Mtx + Cyclosporin A) + LDC
zudem substanzielle Verbesserungen der
innerhalb von 3 Monaten Lebensqualität und Arbeitsfähigkeit der
Etanercept + Mtx, Adalimumab + Mtx, Patienten sowie eine drastische Reduzie-
Infliximab + Mtx (+ LDC) rung der krankheitsimmanent gesteiger-
ten Mortalität. Ähnlich gut wirken die
innerhalb von 3–6 Monaten TNF-Blocker bei Psoriasis, Psoriasis-Ar-
Rituximab + Mtx, Abatacept + Mtx (+ LDC) thritis und ankylosierender Spondylitis.
Gemessen an ihrer Wirkstärke ist die
Mtx = Methotrexat, SSZ = Sulfasalazin, Hcl = Hydroxychloroquin, LDC = Low-dose-Corticoid Verträglichkeit der Biologika exzellent,
allerdings ist auf eine verminderte Infekt­
abwehr bei den behandelten Patienten
bremsen die radiologisch sichtbaren De- DMARDs erweitert, so Cyclosporin A zu achten. Limitierend auf den Einsatz
struktionen und gehören deshalb min- und Leflunomid, welches heute nach der Biologika wirken die hohen Thera-
destens im ersten Halbjahr fest zur Kom- Mtx als zweitwichtigstes DMARD gilt. piekosten, die ein Vielfaches der meisten
binationstherapie der RA. Parenterales Gold, vor Mtx der DMARD- DMARDs betragen. Gemessen an ihrer
Goldstandard, ist heute ebenso wie Aza- ausgezeichneten Wirksamkeit werden
Basistherapeutika thioprin eher als Reservepräparat zu se­ Biologika in Deutschland jedoch zu zu-
In der heterogenen Gruppe der Basis- hen. Die Antimalariamittel Chloroquin rückhaltend eingesetzt: Nur 3,8 % der
therapeutika (Disease-modifying Anti- und Hydroxychloroquin finden nur RA-Patienten erhalten diese Therapie,
rheumatic Drugs = DMARDs) kam es noch bei sehr gering aktiver RA und in eine der niedrigsten Raten in ganz Euro-
in den späten 1970er- und 80er-Jahren der Kombinationstherapie Verwendung. pa. Bei Einhaltung der nationalen The-
sporadisch zu Neueinführungen wie Der kombinierte Einsatz mehrerer rapieempfehlungen (Einsatz nach zwei
D-Penicillamin oder Auranofin (orales DMARDs wurde seit den frühen 1990er- unwirksamen DMARDs möglich) müsste
Gold), die sich mittelfristig nicht wirk- Jahren vermehrt erprobt und hat heute diese Rate deutlich höher liegen.
lich als Bereicherungen erwiesen. Zwei einen festen Stellenwert, wenn sich die
Wiederentdeckungen war später ein Monotherapie mit Mtx als unzureichend Qualitätsmanagement
besserer Erfolg beschieden: Sulfasala- erweist. Zu den meist verwendeten Kom- Neben der Entwicklung vielverspre-
zin (SSZ) und Methotrexat (Mtx). binationen zählen heute Mtx + SSZ + chender neuer Substanzen haben auch
SSZ war bereits zu Beginn der 1940er- Hydroxychloroquin, Mtx + Leflunomid neue Anwendungsstrategien zu den
Jahre bei RA erprobt worden und dann und Mtx + Cyclosporin A. Kombina­ großen Fortschritten bei der Behand-
bei dieser Indikation in Vergessenheit tionstherapien sind i. d. R. nicht schlech- lung chronischer Arthritiden beigetra-
geraten. Es erwies sich jetzt als gut wirk- ter verträglich als Monotherapien. gen. So beinhaltet heute das Qualitäts-
sames Basistherapeutikum für leichte management der RA-Therapie schnellst-
Formen der RA und anderer chro- Biologika mögliche Diagnosestellung und Einlei-
nischer Arthritiden. Die bessere Aufklärung der Pathogene- tung einer Basistherapie (im Idealfall
Die Wiedereinführung von Mtx ent- se chronischer Arthritiden im Lauf der innerhalb von zwölf Wochen). Im wei-
wickelte sich sowohl bei der RA wie auch letzten 20 Jahre war Voraussetzung für teren Verlauf sollte die Therapie zumin-
bei der Psoriasis-Arthritis als Erfolgsge- die Entdeckung neuer hochwirksamer dest alle drei Monate mittels standar­
schichte. Die Substanz stellt heute unter Sustanzen mit gut definiertem Wirkan- disierter Verlaufsparameter wie DAS
allen DMARDs den Goldstandard für satz, der Biologika. So wurden Hemm- (Disease Activity Score) überprüft und
aktive chronische Arthritiden dar und stoffe der proinflammatorischen Zyto­ ggf. modifiziert bzw. eskaliert werden,
ist der wichtigste Partner in der Kombi­ kine Tumornekrosefaktor a sowie Inter- bis eine Remission oder zumindest eine
na­tionstherapie sowohl mit anderen leukin 1 und 6 entwickelt, weiterhin gute Response erreicht ist. Zusätzlich
DMARDs als auch mit Biologika (s. u.). Substanzen, die B-Lymphozyten bzw. werden bildgebende Kontrollen zur Ver-
Einige Neueinführungen in den die Aktivierung der T-Lymphozyten hinderung von Destruktionen durchge-
1990er-Jahren haben die Palette der hemmen (Tab. 1). Biologika werden vor- führt (Abb. 1).

MMW-Fortschr. Med. Nr. 48 / 2008 (150. Jg.) 121


– 25 Jahre Fortschritte in der Rheumatologie

– Tabelle 1
Für die RA aktuell oder demnächst zugelassene Biologika

Name Wirkprinzip Verabreichung/Dosierung Bemerkungen Weitere Indikationen


Infliximab Chimärer monoklonaler I.v. Inf. – Startdosis 3 mg/kg Bei RA nur in Kombination Ank. Spondylitis, Psoriasis/Psoriasis-
Antikörper gegen TNF Woche 0, 2, 6, 14, dann alle 8 Wo. mit Mtx zugelassen Arthritis, M. Crohn, Colitis ulcerosa
Adalimumab Humaner monoklonaler S.c. 40 mg alle zwei Monotherapie und Kombin. Ank. Spondylitis, Psoriasis/juven. RA
Antikörper gegen TNF Wochen mit Mtx zugelassen Psoriasis-Arthritis, M. Crohn
Etanercept Löslicher TNF-Rezeptor S.c. 25 mg 2 x wöch. Monotherapie und Kombin. Ank. Spondylitis, Psoriasis/
oder 50 mg 1 x wöch. mit Mtx zugelassen Psoriasis-Arthritis, juvenile RA
Anakinra Interleukin-1-Rezeptor S.c. 100 mg 1 x tgl. Bei RA nur noch
wenig verwendet
Rituximab Chimärer monoklonaler I.v. Infusionen mit Zulassung nach Versagen Auch in der Lymphom-
Antikörper gegen 1000 mg, Tag 0/14, dann von TNF-Blocker-Therapie Therapie verwendet
B-Lymphozyten (Anti-CD20) wieder nach frühest. 6 Mon. in Kombination mit Mtx
Abatacept Hemmstoff der I.v. Infusion – 10 mg/kg Zulassung nach Versagen
T-Zell-Aktiv. (Hemmung Tag 0, 14, 28 von TNF-Blocker-Therapie
des kostimul. Signals) dann alle 4 Wochen in Kombination mit Mtx
Tocilizumab Interleukin-6-Rezeptor- 8 mg/kg als Infusion Zulassung vermutlich
Antagonist alle vier Wochen Anfang 2009
Certolizumab Pegylierter monoklonaler 200 mg als Infusion Zulassung vermutlich
Antikörper gegen TNF alle 2–4 Wochen Mitte 2009
Golimumab Humaner monoklonaler 50–100 mg s.c. Zulassung vermutlich
Antikörper gegen TNF alle 4 Wochen Mitte 2009

Arthrose ben in Studien bei guter Verträglichkeit samkeitsnachweis in Form einer aussa-
Mehr noch als die RA ist die Arthrose einen symptomatischen Effekt gezeigt, gekräftigen Publikation erfolgte.
durch eine breite Palette von therapeu- der etwa mit NSAR in niedriger Dosie- Ein weiterer innovativer Ansatz ist
tischen Ansätzen charakterisiert: Neben rung vergleichbar ist, bei ausgeprägter die sogenannte Knorpelersatztherapie
medikamentöser und operativer Interven- Symptomatik jedoch nicht ausreicht. mit Implantation von im Labor gezüch-
tion sowie Krankengymnastik beinhalten In den letzten zehn Jahren wurde die teten Knorpelzellen. Über eine erfolg-
diese u.a. so heterogene Maßnahmen wie Suche nach strukturell wirksamen reiche Anwendung wurde bisher aber
Akupunktur, Röntgenbestrahlung, Radio- („chondro­protektiven“) Substanzen, so- nur für kleine Knorpeldefekte berichtet.
synoviorthese (unter bestimmten Bedin- genannten DMOADs (Disease-modifying
gungen) oder Elektrotherapie. Doch mit Osteoarthritic Drugs) intensiviert, bisher Korrespondenz:
keiner dieser Maßnahmen gelingt bisher aber nicht zu einem eindeutig Erfolg ver- Prof. Dr. med. Klaus Krüger
St.-Bonifatius-Str. 5, D-81541 München
ein grundlegendes und nachhaltiges Ein- sprechenden Abschluss gebracht.
Tel.: 089/6914222
greifen in den Krankheitsablauf, abgese- E-Mail: klaus.krueger@med.uni-muenchen.de
hen von operativen Eingriffen im Endsta- Verschleißbremse – Ersatzknorpel
dium (Endoprothese, Arthrodese). Gentechnologisch entwickelte Inter-
– Ausblick
ventionen mit dem Ziel, im Gelenk
Da auch auf dem Gebiet der medika-
Therapie gegen Schmerz und Entzündung protektive oder die Knorpeldestruktion mentösen Arthrosetherapie intensiv
Die symptomatische Therapie der Ar- bremsende Mechanismen zu induzie- und vielversprechend geforscht wird,
throse wird im gering symptomatischen ren oder mittels Gentransfer Knorpel- ist in den kommenden Jahren mit In-
novationen zu rechnen, die mittelfristig
Stadium heute durch reine Analgetika zellen und -vorläuferzellen zu generie-
die rasante Entwicklung der Behand-
und im fortgeschritteneren Stadium, ren, werden seit fast zehn Jahren in lungsmöglichkeiten bei RA einholen
bei dem Entzündungsmechanismen ei- vitro intensiv beforscht. Keines dieser könnten. Spannende Neuentwicklun-
ne zunehmende Rolle spielen, durch Verfahren ist jedoch bis jetzt in vivo in gen innerhalb der Glukokortikoide zur
Therapie der RA sind demnächst durch
NSAR sowie bei GI-Risiken durch Coxi- das Stadium einer Erfolg verspre-
ein Modified-Release-Präparat, liposo-
be bestimmt. Bei geringer Symptomatik chenden Anwendung gelangt. male GC und selektive Glukokortikoid-
hat sich auch die topische NSAR-Gabe Eine im weiteren Sinne auf diesem Rezeptor-Agonisten zu erwarten.
als wirksam erwiesen. Die systemische Prinzip beruhende Intervention, das aus
Gabe von GC ist hier generell nicht in- Eigenblut gewonnene Orthokin, wird – Keywords
diziert, eine intraartikuläre Verabrei- seit Jahren intensiv mittels Pressemittei- 25 Years of Progress in Rheumatology:
chung gehört im Stadium der akti- lungen und über das Internet beworben Quantum Jumps in the Treatment of
vierten Arthrose hingegen zum festen und sehr kostenintensiv bei Arthrose­ Rheumatoid Arthritis
Behandlungs-Repertoire. Auch Substan- patienten angewendet, ohne dass im ge- Rheumatoid arthritis – Osteoarthritis–
NSAID – Glucocorticoid – Biologicals
zen wie Oxazeprol und Glucosamin ha- samten Zeitraum ein eindeutiger Wirk-

122 MMW-Fortschr. Med. Nr. 48 / 2008 (150. Jg.)

View publication stats

Das könnte Ihnen auch gefallen