Artritis Inflamtoria en La Practica 2015
Artritis Inflamtoria en La Practica 2015
Artritis Inflamtoria en La Practica 2015
Scott
James B. Galloway
David L. Scott
Inflammatory Arthritis
in Clinical Practice
Second Edition
123
Inflammatory Arthritis
in Clinical Practice
Ian C. Scott James B. Galloway
David L. Scott
Inflammatory Arthritis
in Clinical Practice
Second Edition
Ian C. Scott David L. Scott
Molecular and Cellular Biology Rheumatology
of Inflammation Kings College Hospital
Kings College London London
London UK
UK
James B. Galloway
Rheumatology
Kings College Hospital
London
UK
vii
viii Contents
3 Clinical Features. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Synovitis of Peripheral Joints . . . . . . . . . . . . . . . . . . . . 24
Symptoms. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Joint Swelling and Tenderness. . . . . . . . . . . . . . . . . 24
Peripheral Joints Involved . . . . . . . . . . . . . . . . . . . . . . 25
Rheumatoid Arthritis . . . . . . . . . . . . . . . . . . . . . . . . 25
Psoriatic Arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
Other Forms of Spondyloarthropathies. . . . . . . . . 29
Enthesitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Spinal Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
Ankylosing Spondylitis . . . . . . . . . . . . . . . . . . . . . . 30
Other Types of Spondyloarthritis . . . . . . . . . . . . . . 31
Rheumatoid Arthritis . . . . . . . . . . . . . . . . . . . . . . . . 31
Systemic Features. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
Clinical Course . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
Onset of Rheumatoid Arthritis. . . . . . . . . . . . . . . . 32
Established Rheumatoid Arthritis . . . . . . . . . . . . . 33
Other Inflammatory Arthropathies . . . . . . . . . . . . 33
Undifferentiated Early Arthritis. . . . . . . . . . . . . . . 33
Extra-articular Disease . . . . . . . . . . . . . . . . . . . . . . . . . 34
Rheumatoid Arthritis . . . . . . . . . . . . . . . . . . . . . . . . 34
Other Inflammatory Arthropathies . . . . . . . . . . . . 36
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
Rheumatoid Factor. . . . . . . . . . . . . . . . . . . . . . . . . . 51
Antibodies to Citrullinated Protein Antigens. . . . 52
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
X-Rays in Rheumatoid Arthritis . . . . . . . . . . . . . . 53
Ultrasound in Rheumatoid Arthritis . . . . . . . . . . . 55
Magnetic Resonance Imaging in Rheumatoid
Arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
Imaging in Psoriatic Arthritis . . . . . . . . . . . . . . . . . 56
Imaging in Ankylosing Spondylitis
and Reactive Arthritis . . . . . . . . . . . . . . . . . . . . . . . 57
Combined Measures . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
Core Data Set in Rheumatoid Arthritis . . . . . . . . 57
Composite Disease Activity Indices: Disease
Activity Score . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
Simplified Indices . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
American College of Rheumatology (ACR)
Response Criteria . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
Other Forms of Arthritis . . . . . . . . . . . . . . . . . . . . . 60
Outcomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
Rheumatoid Arthritis . . . . . . . . . . . . . . . . . . . . . . . . 61
Psoriatic Arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
Ankylosing Spondylitis . . . . . . . . . . . . . . . . . . . . . . 63
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
Conventional NSAIDs . . . . . . . . . . . . . . . . . . . . . . . 75
Cox-2 Drugs (COXIBs) . . . . . . . . . . . . . . . . . . . . . . 75
Adverse Reactions to NSAIDs . . . . . . . . . . . . . . . . . . 78
Gastrointestinal Adverse Effects . . . . . . . . . . . . . . 79
Cardiovascular Adverse Effects . . . . . . . . . . . . . . . 81
Use in Specific Forms of Arthritis . . . . . . . . . . . . . . . . 82
Rheumatoid Arthritis . . . . . . . . . . . . . . . . . . . . . . . . 82
Psoriatic Arthritis and Seronegative
Spondyloarthritis. . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Ankylosing Spondylitis . . . . . . . . . . . . . . . . . . . . . . 83
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
Abatacept . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
Clinical Use . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
Dosing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
Clinical Efficacy. . . . . . . . . . . . . . . . . . . . . . . . . . . . 148
Adverse Reactions . . . . . . . . . . . . . . . . . . . . . . . . . 149
Ustekinumab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
Clinical Use . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
Adverse Events . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
Anakinra . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
Clinical Use . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
Side-Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 151
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 151
10 Steroids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155
Background. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155
Pharmacological Effects . . . . . . . . . . . . . . . . . . . . . . . 156
Effects on Joint Inflammation . . . . . . . . . . . . . . . . . . 157
Erosive Progression . . . . . . . . . . . . . . . . . . . . . . . . . . . 157
Early Rheumatoid Arthritis . . . . . . . . . . . . . . . . . . . . 158
Treating Flares with Systemic Steroids . . . . . . . . . . . 159
Psoriatic Arthritis and Ankylosing Spondylitis . . . . 159
Side-Effects. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159
Local Steroids. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 160
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
Abbreviations
Introduction
The inflammatory arthropathies are a group of disorders
characterised by joint pain and swelling. Their similar treat-
ments, which include disease-modifying anti-rheumatic drugs
(DMARDs) and biologic agents, mean they are often consid-
ered together. Their serological and extra-articular manifes-
tations help to differentiate them. This chapter will provide
an overview of the inflammatory arthropathies, with a
specific focus on their subtypes and how they are diagnosed.
Historical Perspectives
Most types of inflammatory arthritis were identified in the
nineteenth century, though their exact classification was only
finalised in the last 50 years. Prior to 1850 there was consider-
able uncertainty about how to differentiate different forms of
arthritis, and a number of complex, somewhat confusing
names were used, such as chronic rheumatic gout. Rheumatic
fever, which was commonplace at that time, also caused con-
siderable uncertainly.
The concept of rheumatoid arthritis dates from Victorian
times and the term was introduced by Sir Archibald Garrod,
4 Chapter 1. An Overview of Inflammatory Arthritis
Diagnosis
Rheumatoid Arthritis
Ankylosing Spondylitis
than 1 h before improvement better explained by another pathology and Score of 6/10 from the
following four domains:
2. Arthritis involving three or more 1. Joint involvement
joint areas
1 large joint = 0 points
210 large joints = 1 point
13 small joints = 2 points
410 small joints = 3 points
>10 joints (at least 1 small joint) = 5 points
3. Arthritis of the hand joints 2. Serology
Negative RF and negative ACPA = 0 points
Low-positive RF or low-positive ACPA = 2 points
An Overview of Inflammatory Arthritis
To diagnose RA criteria 14 must To diagnose RA patients must satisfy all three criteria. Low positive
have been present for at least 6 weeks; serological tests refer to values 3 times the upper limit of normal; high
4 or more criteria must be present. The positive refers to values >3 times the upper limit of normal. RF = Rheumatoid
diagnosis of RA should not be made Factor; ACPA = Antibodies to Citrullinated Protein Antigens
by criteria alone if another systemic
disease associated with arthritis is
definitely present
Table adapted from 1987 ACR and 2010 ACR/EULAR RA classification criteria [1, 2]
Diagnosis
7
8 Chapter 1. An Overview of Inflammatory Arthritis
Psoriatic Arthritis
AND
Other Spondyloarthropathies
References
1. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF,
Cooper NS, et al. The American Rheumatism Association 1987
revised criteria for the classification of rheumatoid arthritis.
Arthritis Rheum. 1988;31:31524.
2. Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham 3rd
CO, et al. 2010 rheumatoid arthritis classification criteria: an American
College of Rheumatology/European League Against Rheumatism
collaborative initiative. Arthritis Rheum. 2010;62:256981.
3. van der Linden S, Valkenburg HA, Cats A. Evaluation of diagnos-
tic criteria for ankylosing spondylitis. A proposal for modification
of the New York criteria. Arthritis Rheum. 1984;27:3618.
4. Rudwaleit M, van der Heijde D, Landewe R, Listing J, Akkoc N,
Brandt J,et al.The development ofAssessment of SpondyloArthritis
international Society classification criteria for axial spondyloar-
thritis (part II): validation and final selection. Ann Rheum Dis.
2009;68:77783.
5. Moll JM, Wright V. Psoriatic arthritis. Semin Arthritis Rheum.
1973;3:5578.
6. Taylor W, Gladman D, Helliwell P, Marchesoni A, Mease P,
Mielants H, et al. Classification criteria for psoriatic arthritis:
development of new criteria from a large international study.
Arthritis Rheum. 2006;54:266573.
Chapter 2
Epidemiology and Pathology
Females
5
4.5
4
Prevalence (%) ofRA
3.5
3
2.5
2
1.5
1
0.5
0
1644 4564 6574 Over 75
Age (years)
Males
1.8
1.6
1.4
Prevalence (%) ofRA
1.2
0.8
0.6
0.4
0.2
0
4564 Over 65
Age (years)
Lawrence's study Norfolk study
Rheumatoid Arthritis
Seronegative Spondyloarthropathies
Synovium
Cartilage
Bone
Normal
Inflammatory
Pannus
Inflammatory effusion
Thickened synovium
Lymphocytes
Co-stimulation
Recruitment
T cell
Antigen
presenting Activated T
cell cells
Lymph node
Cytokines
Chemokines
Metalloproteinases
References
1. Kalla AA, Tikly M. Rheumatoid arthritis in the developing world.
Best Pract Res Clin Rheumatol. 2003;17:86375.
References 21
Symptoms
Rheumatoid Arthritis
Hand involvement is particularly characteristic of RA
(Fig. 3.1). It usually involves the metacarpophalangeal, proxi-
mal interphalangeal, thumb interphalangeal and wrist joints.
The distal interphalangeal joints can be affected when there
is coexisting disease in other hand joints.
Involvement of the tendons is also characteristic.
Tenosynovitis of flexor tendons reduces finger flexion and
strength. Nodular thickenings in tendon sheaths may result in
a trigger finger.
Damage to the wrists causes compaction of bone at the
small wrist joints. In late disease this damage may progress to
bony ankylosis. Historically in late RA characteristic defor-
mities developed in the hands. There was ulnar deviation of
the fingers, subluxation of the metacarpophalangeal joints,
hyperextension of the proximal interphalangeal with flexion
of the distal joints (swan-neck deformity), flexion of the
26 Chapter 3. Clinical Features
Psoriatic Arthritis
PsA has highly variable clinical features and there are several
different patterns of joint involvement [5]. These tend to
change over time.
Conventionally PsA is divided into five different clinical
subtypes. These clinical subtypes comprise:
1. Polyarthritis: many joints are involved and the clinical pat-
tern is very similar to RA
2. Oligoarthritis: in most patients with PsA only two or three
joints are involved. As a consequence the arthritis seems
very asymmetric.
3. Monoarthritis: as the classification indicates only one joint
is involved. Over time the pattern may change with the
involvement of more joints.
4. Distal interphalangeal joint arthritis: this is a highly char-
acteristic arthritis of the distal interphalangeal joints of the
hands. It can be very destructive.
Peripheral Joints Involved 29
Enthesitis
The entheses are the insertion of tendons into bone [6]. These
are frequently inflamed in spondyloarthritis. Such inflamma-
tion is known as an enthesitis. It involves sites such as the
30 Chapter 3. Clinical Features
Spinal Disease
Involvement of the sacroiliac joints, other cartilaginous joints
like the pubic symphysis, and the spine is the central feature
of AS and other seronegative spondyloarthropathies [7]. It is
less common, and very different, in RA.
Ankylosing Spondylitis
Rheumatoid Arthritis
Systemic Features
Arthritis causes general ill health in addition to the synovitis.
Patients feel tired and lack energy. They also often have sys-
temic features of a flu-like illness. These include loss of
appetite, inability to sleep, some weight loss and, in some
cases a low-grade fever. These systemic features are most
noticeable at the onset of arthritis, especially in cases with an
explosive onset. It is likely they represent the non-specific
inflammatory response of immunity.
32 Chapter 3. Clinical Features
Clinical Course
Extra-articular Disease
Rheumatoid Arthritis
References
1. van der Heijde DM, van t Hof MA, van Riel PL, Theunisse LA,
Lubberts EW, van Leeuwen MA, et al. Judging disease activity in
clinical practice in rheumatoid arthritis: first step in the develop-
ment of a disease activity score. Ann Rheum Dis. 1990;49:91620.
38 Chapter 3. Clinical Features
Role of Assessments
The presence of an inflammatory arthritis is relatively easy to
diagnose. The presence of painful, swollen joints is usually
self-evident to both patients and clinicians. However, dividing
patients into specific diagnostic groups can be challenging.
Equally difficult is determining changes in the severity of
arthritis over time. Clinical and laboratory assessments are
vital to making informed decisions about diagnosis classifica-
tion and disease severity.
Assessments used in patients with arthritis include joint
counts, assessments of global health, patient self-assessments
of disability and quality of life, laboratory assessments of
disease activity and autoantibody status and imaging assess-
ments of joint inflammation and joint damage [1]. Often a
number of these various assessments are linked together to
give composite scores of disease activity.
Clinical Assessments
Joint Counts
40 %
35 %
30 %
25 %
Tender joints
20 %
15 %
10 %
5%
0%
0 2 4 6 8 10 12 14 16 18 20 22 24 26 28
40 %
35 %
30 %
25 %
20 %
15 % Swollen joints
10 %
5%
0%
0 2 4 6 8 10 12 14 16 18 20 22 24 26 28
Global Assessments
Global or overall assessments by patients and by clinicians
are often used to assess disease activity. They are convention-
ally recorded using visual analogue scales (VAS). These score
responses on 100 mm scales. Descriptive Likert scores have
also been used; these group responses into distinct categories.
Typically there are five categories which range from strongly
44 Chapter 4. Clinical and Laboratory Assessments
16 %
14 %
12 %
10 %
8%
6%
4%
2%
0%
0 10 20 30 40 50 60 70 80 90
Pain Scores
Functional Assessments
SF-36
EuroQol
Laboratory Assessments
Quantitative laboratory markers like the erythrocyte sedi-
mentation rate (ESR) are useful for monitoring because they
are a consequence of systemic disease. Qualitative markers
like RF indicate prognosis and may have pathogenic
relevance.
16 %
14 %
12 %
10 %
8%
6%
4%
2%
0%
0 10 20 30 40 50 60 70 80 90
Rheumatoid Factor
These are antibodies directed against the Fc portion of
immunoglobulin G (IgG). All classes of immunoglobulin can
form RF. RF and IgG combine together as immune com-
plexes. The immune disturbance of RA is influenced by these
immune complexes.
Most patients with RA have RF in their synovial fluid and
blood [9]. The exact frequency varies between centres.
Overall 6080 % of RA patients possess significantly ele-
vated levels of RF. They are termed seropositive while
those who are persistently RF-negative are termed sero-
negative patients. Measuring RF is an essential part of diag-
nosing RA, and RF tests are incorporated in all classification
criteria for the disease.
The first RF tests used agglutination methods.This includes
the classical Rose-Waaler tests based on sheep red cells and
latex tests. These mainly detect IgM RF.
52 Chapter 4. Clinical and Laboratory Assessments
Imaging
The commonly used imaging approaches are plain X-rays,
ultrasound and MRI [12]. Other methods that are used less
often include computerised tomography (CT), which has lim-
ited value in peripheral joints, and radio-isotope scans, which
are now rarely used to diagnose or assess arthritis.
Figure 4.5 Erosive X-ray changes in the hands and feet in rheuma-
toid arthritis patients seen at Kings College Hospital, London
a b
Combined Measures
Simplified Indices
Outcomes
Rheumatoid Arthritis
Psoriatic Arthritis
Ankylosing Spondylitis
References
1. Anderson J, Caplan L, Yazdany J, Robbins ML, Neogi T,
Michaud K, et al. Rheumatoid arthritis disease activity measures:
American College of Rheumatology recommendations for use
in clinical practice. Arthritis Care Res. 2012;64:6407.
64 Chapter 4. Clinical and Laboratory Assessments
Introduction
Pain is the dominant symptom in arthritis. It is present from
the earliest stages of synovitis and persists throughout the
course of the disease. In early inflammatory arthritis pain is
predominantly related to the activity of the synovitis. In late
disease it is influenced by the development of joint damage
and failure.
Simple Analgesics
Simple analgesics should be used in all patients with inflam-
matory arthritis as an adjunct to non-steroidal anti-
inflammatory drugs (NSAID) and DMARD therapy. The
available drugs include paracetamol, co-proxamol and trama-
dol (Table 5.1). Although there is some evidence from clinical
trials that analgesics reduce pain in RA, the amount of data
Simple Analgesics 69
Buprenorphine
Paracetamol
Opioids
Tramadol
700
600
Controls
Per 1,000 patients
500
Opioids
400
300
200
100
0
Good patient- Withdrew for lack Withdrew for
assessed of effect adverse effect
improvement
Buprenorphine
Compound Analgesics
Classification of NSAIDs
Conventional NSAIDs
Baseline 6 Months
25
Tender joint count
20
15
10
0
Celecoxib Diclofenac
Baseline 6 Months
60
50
Pain scores
40
30
20
10
0
Celecoxib Diclofenac
Haematological Thrombocytopenia
Haemolytic anaemia
Adverse Reactions to NSAIDs 79
These side effects are the main problem with NSAIDs. The
range of adverse effects includes dyspepsia, gastric erosions,
peptic ulceration, bleeding, perforation, haematemesis or
melaena, small bowel inflammation, occult blood loss and
anaemia.
Between 10 and 20 out of every 1,000 RA patients taking
NSAIDs for 1 year will have serious gastrointestinal compli-
cations. The mortality risks attributable to NSAID-related GI
adverse effects are four times that for those not using
NSAIDs. There is some evidence that gastro-intestinal mor-
tality from NSAIDs is similar in its extent to other major
causes of death such as melanoma and leukaemia. The most
serious problems are perforations, ulcers and bleeds. There is
some evidence that the risk of GI adverse events differs
across NSAIDs (Fig. 5.3) [5].
Many patients who have serious gastro-intestinal compli-
cations do not have prior dyspepsia. In the absence of warn-
ing signs there is no way to ascertain if a patient is on the
point of developing serious problems. Therefore if NSAID
use is unavoidable, some protective strategy is needed, par-
ticularly in those patients at greatest risk. There are several
potential options.
80 Chapter 5. Symptomatic Drug Treatment
4
Relative risk
0
Ibuprofen Naproxen Diclofenac Celecoxib
1.75
1.50
Odds ratio
1.25
1.00
0.75
Any Naproxen Celecoxib Ibuprofen Diclofenac
NSAID
Rheumatoid Arthritis
Ankylosing Spondylitis
References
1. Hazlewood G, van der Heijde DM, Bombardier C. Paracetamol
for the management of pain in inflammatory arthritis: a system-
atic literature review. J Rheumatol Suppl. 2012;90:116.
2. Whittle SL, Richards BL, Husni E, Buchbinder R. Opioid ther-
apy for treating rheumatoid arthritis pain. Cochrane Database
Syst Rev. 2011;(11):CD003113.
3. Vane JR, Botting RM. Anti-inflammatory drugs and their mech-
anism of action. Inflamm Res. 1998;47 Suppl 2:S7887.
4. Emery P, Zeidler H, Kvien TK, Guslandi M, Naudin R, Stead H,
et al. Celecoxib versus diclofenac in long-term management of
rheumatoid arthritis: randomised double-blind comparison.
Lancet. 1999;354:210611.
5. Castellsague J, Riera-Guardia N, Calingaert B, Varas-Lorenzo C,
Fourrier-Reglat A, Nicotra F, et al. Individual NSAIDs and
upper gastrointestinal complications: a systematic review and
meta-analysis of observational studies (the SOS project). Drug
Saf. 2012;35:112746.
6. Trelle S, Reichenbach S, Wandel S, Hildebrand P, Tschannen B,
Villiger PM, et al. Cardiovascular safety of non-steroidal anti-
inflammatory drugs: network meta-analysis. BMJ. 2011;342:c7086.
7. Scott PA, Kingsley GH, Smith CM, Choy EH, Scott
DL. Non-steroidal anti-inflammatory drugs and myocardial
infarctions: comparative systematic review of evidence from
observational studies and randomised controlled trials. Ann
Rheum Dis. 2007;66:1296304.
References 85
Background
DMARDs are a diverse range of drugs. They form a single
group because they both improve symptoms and also, to a
greater or lesser extent, modify the course of the disease. This
Starting DMARDs
In patients with definite rheumatoid arthritis DMARDs are
started as soon as the diagnosis has been made. The evidence
in favour of early DMARDs, though incomplete, is compel-
ling. Most experts recommend starting with methotrexate.
Monitoring DMARDs
The risk of blood and hepatic toxicity means that most
DMARDs require monthly blood monitoring tests. This
practice began because it was possible to predict patients at
risk of marrow toxicity with gold injections by prospective
monitoring; this approach substantially improved safety
risks. With modern treatment the risks are less clear cut and
the benefits of monitoring are more questionable. However,
in some patients adverse events involving the blood and
liver can be detected on monitoring and such reactions
carry substantial risks for patients. Consequently monitor-
ing has become part of the standard approach to DMARD
treatment [4].
90 Chapter 6. Disease-Modifying Anti-Rheumatic Drugs
Stopping DMARDs
DMARDs are stopped for toxicity and for loss of effect.
Often these overlap. In patients who have entered remission
or a state of low activity the benefit of continuing DMARDs
is often questioned. The evidence suggests that stopping
treatment in such patients often results in a disease flare;
consequently it is not generally recommended [5].
Early and Late Disease 91
All
Leflunomide
Hydroxychloroquine
Sulfasalazine
Methotrexate
0 10 20 30 40 50
Median retention time (months)
Intensive Treatment
Historically DMARDs were given singly as DMARD
monotherapy but there has been a shift in practice over the
last 2 years to using then in combination with each other.
There are two ways of giving combination therapy:
Step-down: starting combinations together and then stop-
ping one or more DMARDs
Step-up: starting one DMARD and then adding another if
needed.
In addition combinations can involve two DMARDs,
DMARDs and steroids, and DMARDs and biologics.
Ideally patients with the worst prognosis would be identified
rapidly and then given intensive treatment to control their
arthritis as soon as possible. However, this has proved problem-
atic to do as at present it is difficult to identify patients with poor
prognoses with sufficient accuracy to justify a policy of starting
intensive DMARD combinations in specific groups of patients.
Treat to Target
The concept of treat to target builds on intensive manage-
ment approaches in rheumatoid arthritis [7]. It sets a goal of
reducing disease activity to very low levels or remission. With
modern treatments this appears increasingly realistic. It should
reduce long-term structural damage and improve quality of
life. Treat to target involves setting a clear end point, which in
this case is the target of remission, and establishing a specific
treatment algorithm, which simplifies the many complex treat-
ment sequences available to treat arthritis. However, there are
some complexities. The definition of treat to target varies and
a range of different data is used to justify its benefits. Some
patients are reluctant to receive more intensive treatment due
to concern over adverse effects. Finally, the evidence that
remission is generally achievable in active rheumatoid arthritis
is incomplete no matter how patients are treated.
Changes in Clinical Trials 93
Seronegative Arthritis
Seronegative rheumatoid arthritis responds similarly to sero-
positive disease. Patients with other forms of inflammatory
arthritis such as psoriatic arthritis and reactive arthritis are
treated in the same way as patients with rheumatoid arthritis.
The evidence for using DMARDs in seronegative spondylo-
arthritis, particularly psoriatic arthritis, is weaker than that
for rheumatoid arthritis. However, the evidence that
DMARDs have different effects in any form of inflammatory
arthritis is incomplete. Most rheumatologists consider they
provide comparable results [8], though some experts are
unconvinced about the role of methotrexate in psoriatic
arthritis. One important point is that DMARDs do not
appear to improve either the spinal disease or enthesopathy
of seronegative spondyloarthritis [9].
Weeks
0 4 12 24
0
4
Placebo
6
8 SZP
10 Leflunomide
Figure 6.2 Falls in tender joint counts with placebo, sulfasalazine and
leflunomide (Figure adapted using data reported by Smolen et al. [10])
Improved Disability
12 Methotrexate
10
6 Year 1
4 Year 2
0
Tender Swollen ESR C-reactive
joint count joint count protein
12 Leflunomide
10
6 Year 1
4 Year 2
0
Tender Swollen ESR C-reactive
joint count joint count protein
Figure 6.3 Falls in joint counts and acute phase reactants with
methotrexate and leflunomide treatment (Figure adapted using data
reported by Cohen et al. [11])
Radiological Damage
60 %
40 %
20 %
0%
ACR20 ACR50
Methotrexate
Background
Percent patients
0% 20 % 40 % 60 % 80 % 100 %
Leflunomide (a)
Leflunomide (b)
Treatment Placebo
Methotrexate
Chloroquine (a)
Chloroquine (b)
Hydroxychloroquine
ACR70
ACR50
ACR20
0% 20 % 40 % 60 % 80 %
Oral Subcutaneous
Adverse Reactions
Leflunomide
Background
Leflunomide is the only new DMARD to have been intro-
duced in the last two decades. It was developed as an immu-
nosuppressant and acts as a pyrimidine synthesis inhibitor
with consequential anti-proliferative activity. Leflunomide is
a prodrug and is rapidly converted in the gastrointestinal
tract and plasma to its active metabolite, a malononitrilamide,
which is responsible for its activity in vivo.
The active metabolite of leflunomide has a range of
effects, which include:
Inhibiting pyrimidine synthesis: inhibits dihydroorotate
dehydrogenase
Inhibiting nuclear factor kappa-B activation
Reducing cell-cell contact
Reducing bone marrow cell proliferation
Inhibiting synovial nitric oxide production
The effect on pyrimidine synthesis is mediated by the inhi-
bition of dihydroorotate dehydrogenase, the rate-limiting
Leflunomide 101
Adverse Events
Sulfasalazine
Sulfasalazine combines an anti-inflammatory agent
(5-aminosalicylic acid) with a sulfonamide antibiotic (sulfa-
pyridine), which if often thought to be the active component.
Other DMARDs 103
Other DMARDs
Injectable Gold
Gold salts are the oldest DMARD and are also the most
toxic. They take months before showing evidence of efficacy.
Gold is usually given as gold sodium thiomalate, though gold
sodium thioglucose can also be given.
104 Chapter 6. Disease-Modifying Anti-Rheumatic Drugs
Hydroxychloroquine
This anti-malarial drug was used in rheumatology to treat sys-
temic lupus erythematosus. Subsequent placebo-controlled
trials confirmed its efficacy in rheumatoid arthritis. It is
thought to be most useful in mild early disease. It is usually
given at a dose of 400 mg/day. It is less effective than metho-
trexate or sulfasalazine. It has little effect on erosive damage.
Common adverse effects include rash, abdominal cramps
and diarrhoea. Its main toxicity is the rare but highly unpleas-
ant complication of retinopathy. Some form of retinal screen-
ing is recommended for patients starting hydroxychloroquine.
Patients at higher risk of retinopathy, who are over 60 years
of age, have over 5 years treatment or use doses greater than
6.5 mg/kg/day, need regular review, with annual ophthalmo-
logical examination.
Other DMARDs 105
Ciclosporin
Azathioprine
Minor DMARDs
Oral gold, penicillamine and cyclophosphamide are rarely
used to treat inflammatory arthritis. Oral gold has limited effi-
cacy and the other drugs have excessive toxicity. They are used
in occasional patients in whom other options are limited.
106 Chapter 6. Disease-Modifying Anti-Rheumatic Drugs
Combining DMARDs
Rationale
Effective Combinations
Ineffective Combinations
Methotrexate vs
methotrexate/doxycycline
DMARD monotherapy vs
DMARD combination therapy
Methotrexate vs
methotrexate/ciclosporin
Methotrexate vs
methotrexate/ciclosporin
Methotrexate vs
methotrexate/ciclosporin
0% 20 % 40 % 60 %
Percent patients
Seronegative Spondyloarthritis
References
1. Gaujoux-Viala C, Nam J, Ramiro S, Landewe R, Buch MH,
Smolen JS, et al. Efficacy of conventional synthetic disease-
modifying antirheumatic drugs, glucocorticoids and tofacitinib: a
systematic literature review informing the 2013 update of the
EULAR recommendations for management of rheumatoid
arthritis. Ann Rheum Dis. 2014;73:5105.
2. van Gestel AM, Haagsma CJ, Furst DE, van Riel PL. Treatment of
early rheumatoid arthritis patients with slow-acting anti-rheumatic
drugs (SAARDs). Baillieres Clin Rheumatol. 1997;11:6582.
3. Donahue KE, Gartlehner G, Jonas DE, Lux LJ, Thieda P, Jonas
BL, et al. Systematic review: comparative effectiveness and
harms of disease-modifying medications for rheumatoid arthri-
tis. Ann Intern Med. 2008;148:12434.
4. Chakravarty K, McDonald H, Pullar T, Taggart A, Chalmers R,
Oliver S, et al. BSR/BHPR guideline for disease-modifying anti-
rheumatic drug (DMARD) therapy in consultation with the
British Association of Dermatologists. Rheumatology (Oxford).
2008;47:9245.
5. OMahony R, Richards A, Deighton C, Scott D. Withdrawal of
disease-modifying antirheumatic drugs in patients with rheuma-
toid arthritis: a systematic review and meta-analysis. Ann Rheum
Dis. 2010;69:18236.
6. Agarwal S, Zaman T, Handa R. Retention rates of disease-
modifying anti-rheumatic drugs in patients with rheumatoid
arthritis. Singapore Med J. 2009;50:68692.
7. Solomon DH, Bitton A, Katz JN, Radner H, Brown EM,
Fraenkel L. Review: treat to target in rheumatoid arthritis: fact,
fiction, or hypothesis? Arthritis Rheum. 2014;66:77582.
8. Huynh D, Kavanaugh A. Psoriatic arthritis: current therapy and
future approaches. Rheumatology (Oxford). 2015;54:208.
9. Yang Z, Zhao W, Liu W, Lv Q, Dong X. Efficacy evaluation of
methotrexate in the treatment of ankylosing spondylitis using
meta-analysis. Int J Clin Pharmacol Ther. 2014;52:34651.
110 Chapter 6. Disease-Modifying Anti-Rheumatic Drugs
10. Smolen JS, Kalden JR, Scott DL, Rozman B, Kvien TK, Larsen
A, et al. Efficacy and safety of leflunomide compared with pla-
cebo and sulphasalazine in active rheumatoid arthritis: a double-
blind, randomised, multicentre trial. European Leflunomide
Study Group. Lancet. 1999;353:25966.
11. Cohen S, Cannon GW, Schiff M, Weaver A, Fox R, Olsen N, et al.
Two-year, blinded, randomized, controlled trial of treatment of
active rheumatoid arthritis with leflunomide compared with
methotrexate. Utilization of Leflunomide in the Treatment of
Rheumatoid Arthritis Trial Investigator Group. Arthritis Rheum.
2001;44:198492.
12. Strand V, Cohen S, Schiff M, Weaver A, Fleischmann R, Cannon G,
et al. Treatment of active rheumatoid arthritis with leflunomide
compared with placebo and methotrexate. Leflunomide Rheumatoid
Arthritis Investigators Group. Arch Intern Med. 1999;159:254250.
13. Scott DL, Strand V. The effects of disease-modifying anti-
rheumatic drugs on the Health Assessment Questionnaire score.
Lessons from the leflunomide clinical trials database.
Rheumatology (Oxford). 2002;41:899909.
14. Jones G, Halbert J, Crotty M, Shanahan EM, Batterham M,
Ahern M. The effect of treatment on radiological progression in
rheumatoid arthritis: a systematic review of randomized placebo-
controlled trials. Rheumatology (Oxford). 2003;42:613.
15. Favalli EG, Biggioggero M, Meroni PL. Methotrexate for the
treatment of rheumatoid arthritis in the biologic era: still an
anchor drug? Autoimmun Rev. 2014;13:11028.
16. Braun J, Kastner P, Flaxenberg P, Wahrisch J, Hanke P, Demary
W, et al. Comparison of the clinical efficacy and safety of subcu-
taneous versus oral administration of methotrexate in patients
with active rheumatoid arthritis: results of a six-month, multi-
center, randomized, double-blind, controlled, phase IV trial.
Arthritis Rheum. 2008;58:7381.
17. Pincus T, Castrejon I. Evidence that the strategy is more impor-
tant than the agent to treat rheumatoid arthritis. Data from clini-
cal trials of combinations of non-biologic DMARDs, with
protocol-driven intensification of therapy for tight control or
treat-to-target. Bull Hosp Jt Dis. 2013;71 Suppl 1:S3340.
18. Bijlsma JW. Disease control with glucocorticoid therapy in rheu-
matoid arthritis. Rheumatology (Oxford). 2012;51 Suppl 4:iv913.
19. Ma MH, Kingsley GH, Scott DL. A systematic comparison of
combination DMARD therapy and tumour necrosis inhibitor
therapy with methotrexate in patients with early rheumatoid
arthritis. Rheumatology (Oxford). 2010;49:918.
Chapter 7
Biologics An Overview
Introduction
A biologic therapy, simply put, is a treatment that has been
derived from a biological process rather than being manufac-
tured chemically. In medical terms, this includes blood prod-
ucts, vaccinations and, more recently, monoclonal antibodies
(often called biologics).
Historically, long term outcomes in RA were considered
not to be modifiable [1]. The introduction of biologic drugs
has resulted in a complete shift of the goal posts of ther-
apy. Conventional anti- rheumatic drugs such as DMARDs
are small molecules that are designed to bind to and inter-
fere with a target receptor. However, a key challenge with
a small molecule agent is a lack of specificity. Methotrexate,
for example, targets multiple immune pathways including
neutrophils, B cells and T cells. The idea of a magic bullet
approach to target one specific immune process has long
been the goal of drug development in RA.
In the late 1990s Elliot et al. published a phase 2 trial of the
compound cA2, a monoclonal antibody later to be named
infliximab, targeting tumour necrosis factor (TNF) in patients
with RA [2]. Elliots study revealed results that were unparal-
leled in the field, with 2/24 patients in the placebo responding,
compared to 19/24 in those treated with 10 mg/kg of the study
drug. Infliximab moved into phase three trials producing
equally optimistic results [3]. Since the success of the first
anti-TNF, multiple other monoclonal antibodies have been
licensed for the treatment of RA. There are now numerous
anti-TNF agents, as well as monoclonal antibodies targeting
interleukins, B cells and T cells.
The biologics used in inflammatory arthritis are geneti-
cally engineered proteins derived from human genes. They
inhibit specific components of the immune system which play
pivotal roles in driving or inhibiting inflammation in arthritis.
Unlike conventional drugs, that modify the immune system
as a whole, biologics affect specific components of the
immune system. Theoretically this targeted approach is both
more specific in its effects and causes fewer adverse events.
Introduction 113
Rheumatoid Arthritis
B-cell inhibition
Agent: Rituximab. Given with methotrexate unless
contra-indicated
Rituximab
failure/intolerance/contra-
indication
Psoriatic Arthritis
Ankylosing Spondylitis
References
1. Scott DL, Symmons DP, Coulton BL, Popert AJ. Long-term out-
come of treating rheumatoid arthritis: results after 20 years.
Lancet. 1987;1:110811.
2. Elliott MJ, Maini RN, Feldmann M, Kalden JR, Antoni C, Smolen
JS, et al. Randomised double-blind comparison of chimeric
monoclonal antibody to tumour necrosis factor alpha (cA2) ver-
sus placebo in rheumatoid arthritis. Lancet. 1994;344:110510.
3. Lipsky PE, van der Heijde DM, St Clair EW, Furst DE, Breedveld
FC, Kalden JR, et al. Infliximab and methotrexate in the treat-
ment of rheumatoid arthritis. Anti-Tumor Necrosis Factor Trial in
Rheumatoid Arthritis with Concomitant Therapy Study Group.
N Engl J Med. 2000;343:1594602.
4. National Institute for Health and Care Excellence. Adalimumab,
etanercept and infliximab for the treatment of rheumatoid arthri-
tis. NICE Website. 2007. http://www.nice.org.uk/TA130. Accessed
20 Apr 2014.
5. Scott IC. Risk prediction in rheumatoid arthritis. Unpublished
thesis, Kings College London; 2014.
6. Breedveld FC, Weisman MH, Kavanaugh AF, Cohen SB, Pavelka
K, van Vollenhoven R, et al. The PREMIER study: a multicenter,
randomized, double-blind clinical trial of combination therapy
with adalimumab plus methotrexate versus methotrexate alone
or adalimumab alone in patients with early, aggressive rheuma-
toid arthritis who had not had previous methotrexate treatment.
Arthritis Rheum. 2006;54:2637.
Chapter 8
Anti-Tumour Necrosis
Factor-Alpha (TNF-)
Treatment
Background
Early research in the 1980s and 1990s highlighted the
importance of cytokines in inflammatory arthritis. It
showed many different pro- and anti-inflammatory cyto-
kines were over-expressed in the rheumatoid synovium.
Although there was debate about whether IL-1 was the
major driver in inflammatory arthritis, research suggested
that TNF- might have a central role. The proof of principle
for inhibiting this cytokine came from an open label clinical
study in which patients with RA received a single infusion
of a TNF- inhibitor. This showed a rapid response, includ-
ing an early fall in CRP levels [1]. However, the anti-
inflammatory effect lasted only 612 weeks. It was followed
by a return of active disease. As a result patients were
retreated with further infusions and they showed responses
of a similar magnitude and duration. The scene was set for
a major clinical development programme. One concern was
that the TNF- inhibitor, which included mouse protein,
might lose efficacy due to immunogenicity. Up to 60 % of
Infliximab treated patients develop anti-drug antibodies
[2]. This was solved by giving methotrexate as an additional
treatment. This has become a standard approach with these
biologics.
There are currently five TNF- inhibitors available to treat
inflammatory arthropathies (Table 8.1). These can be subdi-
vided into first generation agents (comprising etanercept,
infliximab and adalimumab) and second generation agents
(comprising certolizumab and golimumab). In RA all TNF-
inhibitors are nationally approved for use in routine clinical
care. There is no evidence that one of these agents is superior
to another, and practical and financial issues determine which
is chosen. In other forms of inflammatory arthritis, in particu-
lar in AS, there is evidence that some biologics may be less
effective than others.
Table 8.1 Currently available TNF- inhibitors for the treatment of inflammatory arthritis
TNF- inhibitor Site of action Dosing schedule Methotrexate
Infliximab Binds soluble and transmembrane TNF- and IV administration Essential to
inhibits binding of TNF- to TNF receptors every 48 weeks co-prescribe
Etanercept Soluble TNF-receptor fusion protein that binds Subcutaneous once Optional to
TNF- and TNF- preventing them from interacting or twice weekly co-prescribe
with their receptors
Adalimumab Binds soluble and transmembrane TNF- and Subcutaneous Optional to
inhibits binding of TNF- to TNF receptors fortnightly co-prescribe
Certolizumab Binds soluble and transmembrane TNF- and Subcutaneous Optional to
inhibits binding of TNF- to TNF receptors fortnightly co-prescribe
Golimumab Binds soluble and transmembrane TNF- and Subcutaneously Optional to
inhibits binding of TNF- to TNF receptors monthly co-prescribe
Background
121
122 Chapter 8. Anti-Tumour Necrosis Factor Treatment
Etanercept
Etanercept is a soluble TNF-receptor fusion protein. It com-
prises two dimers. Each dimer has an extracellular, ligand-
binding portion of the higher-affinity type 2 TNF receptor
(p75) which is linked to the Fc portion of human IgG1. This
fusion protein binds to both TNF- and TNF- and prevents
them from interacting with their receptors. It is given by sub-
cutaneous administration at a dose of 25 mg twice a week or
50 mg once a week. This dosing reflects its half-life of approx-
imately 4 days. Interestingly etanercept has never received a
license for inflammatory bowel disease because of lack of
efficacy in clinical trials [3].
Infliximab
Infliximab is a chimeric IgG1 antiTNF- antibody in which
the antigen-binding region is derived from a mouse antibody
and the constant region from a human antibody. It binds to
soluble and membrane bound TNF- with high affinity. This
binding impairs the binding of TNF- to its receptor. Infliximab
also kills cells that express TNF- through antibody- depen-
dent and complement-dependent cytotoxicity. There are con-
siderable differences between patients in the pharmacokinetics
of infliximab. The intravenous dosing method of infliximab
results in much greater initial peak concentrations along with
higher peak to trough ratios when compared to other anti-
TNF agents [4]. Trough concentrations, seen 8 weeks after
intravenous administration of 3 mg/kg of infliximab vary
enormously between patients. Shortening the interval between
doses may be more effective in increasing the trough levels
than increasing the dose. In RA patients the dose given is
3 mg/kg every 8 weeks. In PsA and AS the dose is 5 mg/kg
every 8 weeks. Some patients have higher doses, and in certain
circumstances the intervals between doses can be reduced.
Golimumab 123
Adalimumab
This is a recombinant human IgG1 monoclonal antibody. It
binds human TNF- with a high affinity, and as a conse-
quence inhibits this cytokine binding to its receptors. It also
lyses cells that express TNF-a on their surface. It is given by
subcutaneous injection and is then absorbed slowly. Peak
concentrations are achieved after 120 h. Although there are
wide variations between patients, it is given fortnightly at a
dose of 40 mg.
Certolizumab
Certolizumab pegol is a recombinant humanised Fab frag-
ment (which is the antigen-binding domain) of a TNF-
antibody, which has been coupled to polyethylene glycol
(PEG). This coupling prolongs its plasma half-life to
approximately 2 weeks. It binds and neutralises mem-
brane-bound and soluble human TNF-. It is the only
TNF- inhibitor without an Fc region. It is given by subcu-
taneous injection with an initial loading dose of 400 mg
every 2 weeks for 6 weeks, followed by 200 mg once
fortnightly.
Golimumab
Golimumab is a human IgG1 monoclonal antibody against
TNF-, which is produced in a transgenic mouse. It targets
and neutralises both soluble and membrane bound TNF-. It
has a variable half-life of between 7 and 20 days. It is given as
a monthly subcutaneous injection at dose of 50 mg, which can
be increased to 100 mg a month if the patients body-weight
is more than 100 kg.
124 Chapter 8. Anti-Tumour Necrosis Factor Treatment
Indications
Rheumatoid Arthritis
Psoriatic Arthritis
Ankylosing Spondylitis
Clinical Effects
Rheumatoid Arthritis
16
14
Number needed to treat
Recommended
12
Higher dose
10 Lower dose
8
6
4
2
0
ACR20 ACR50 ACR70
12
10
6 26 weeks
52 weeks
4 104 weeks
0
Methotrexate Adalimumab Methotrexate +
adalimumab
(Fig. 8.1) [5]. Clinical features like joint counts, disability and
joint damage were all improved or progressed less with
TNF- inhibitor use. There was no reason to prefer one
TNF- inhibitor over another. There was limited evidence
that giving lower or higher doses changes the effect of biolog-
ics, though lower doses reduced the chance of developing an
ACR70 response.
There is growing evidence that TNF- inhibitors slow
radiographic progression in RA (Fig. 8.2) [6]. In some
patients they completely halt it. The evidence is stronger in
early disease. It is also stronger with combined therapy
using methotrexate with a biologic. The relationship
between clinical and radiological response is incomplete.
However, the long term implications of slowing radiologi-
cal damage are uncertain, and benefits on radiological
damage alone should not influence clinical decision
making.
Safety and Adverse Effects 127
Psoriatic Arthritis
Ankylosing Spondylitis
Biologics Registries
Hypersensitivity Responses
Minor symptoms such as headache and nausea are common
in patients during infliximab infusions. Symptoms suggesting
an immediate hypersensitivity response with infliximab
infusions, such as urticaria, are uncommon but well described;
serious anaphylaxis is rare. Antihistamines, steroids and
adrenaline should be available while infusions are being
given, though they are rarely needed.
Infections
Demyelinating-like Disorders
Haematological Disorders
Heart Failure
Malignancy
Auto-immune Disorders
Psoriasis
References
1. Elliott MJ, Maini RN, Feldmann M, Kalden JR, Antoni C,
Smolen JS, et al. Randomised double-blind comparison of
chimeric monoclonal antibody to tumour necrosis factor alpha
(cA2) versus placebo in rheumatoid arthritis. Lancet. 1994;344:
110510.
2. Maini R, St Clair EW, Breedveld F, Furst D, Kalden J, Weisman M,
et al. Infliximab (chimeric anti-tumour necrosis factor alpha
monoclonal antibody) versus placebo in rheumatoid arthritis
patients receiving concomitant methotrexate: a randomised
phase III trial. ATTRACT Study Group. Lancet. 1999;354:
19329.
3. Sandborn WJ, Hanauer SB, Katz S, Safdi M, Wolf DG, Baerg RD,
et al. Etanercept for active Crohns disease: a randomized,
double-blind, placebo-controlled trial. Gastroenterology.
2001;121:108894.
4. Nestorov I. Clinical pharmacokinetics of tumor necrosis factor
antagonists. J Rheumatol Suppl. 2005;74:138.
5. Alonso-Ruiz A, Pijoan JI, Ansuategui E, Urkaregi A, Calabozo
M, Quintana A. Tumor necrosis factor alpha drugs in rheumatoid
arthritis: systematic review and metaanalysis of efficacy and
safety. BMC Musculoskelet Disord. 2008;9:52.
6. Breedveld FC, Weisman MH, Kavanaugh AF, Cohen SB,
Pavelka K, van Vollenhoven R, et al. The PREMIER study: a
multicenter, randomized, double-blind clinical trial of combina-
tion therapy with adalimumab plus methotrexate versus metho-
trexate alone or adalimumab alone in patients with early,
aggressive rheumatoid arthritis who had not had previous
methotrexate treatment. Arthritis Rheum. 2006;54:2637.
134 Chapter 8. Anti-Tumour Necrosis Factor Treatment
7. Saad AA, Symmons DP, Noyce PR, Ashcroft DM. Risks and
benefits of tumor necrosis factor-alpha inhibitors in the manage-
ment of psoriatic arthritis: systematic review and metaanalysis of
randomized controlled trials. J Rheumatol. 2008;35:88390.
8. Kavanaugh A, van der Heijde D, McInnes IB, Mease P,
Krueger GG, Gladman DD, et al. Golimumab in psoriatic
arthritis: one-year clinical efficacy, radiographic, and safety
results from a phase III, randomized, placebo-controlled trial.
Arthritis Rheum. 2012;64:250417.
9. Escalas C, Trijau S, Dougados M. Evaluation of the treatment
effect of NSAIDs/TNF blockers according to different domains
in ankylosing spondylitis: results of a meta-analysis.
Rheumatology (Oxford). 2010;49:131725.
10. Galloway JB, Hyrich KL, Mercer LK, Dixon WG, Fu B,
Ustianowski AP, et al. Anti-TNF therapy is associated with an
increased risk of serious infections in patients with rheumatoid
arthritis especially in the first 6 months of treatment: updated
results from the British Society for Rheumatology Biologics
Register with special emphasis on risks in the elderly.
Rheumatology (Oxford). 2011;50:12431.
11. Dixon WG, Hyrich KL, Watson KD, Lunt M, Galloway J,
Ustianowski A, et al. Drug-specific risk of tuberculosis in patients
with rheumatoid arthritis treated with anti-TNF therapy: results
from the British Society for Rheumatology Biologics Register
(BSRBR). Ann Rheum Dis. 2010;69:5228.
12. Bozkurt B, Torre-Amione G, Warren MS, Whitmore J, Soran OZ,
Feldman AM, et al. Results of targeted anti-tumor necrosis
factor therapy with etanercept (ENBREL) in patients with
advanced heart failure. Circulation. 2001;103:10447.
13. Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT,
Anti TNFTACHFI. Randomized, double-blind, placebo-
controlled, pilot trial of infliximab, a chimeric monoclonal
antibody to tumor necrosis factor-alpha, in patients with moder-
ate-to-severe heart failure: results of the anti-TNF Therapy
Against Congestive Heart Failure (ATTACH) trial. Circulation.
2003;107:313340.
14. Mariette X, Reynolds AV, Emery P. Updated meta-analysis of
non-melanoma skin cancer rates reported from prospective
observational studies in patients treated with tumour necrosis
factor inhibitors. Ann Rheum Dis. 2012;71:e2.
References 135
Introduction
Although most emphasis has been placed on tumour necrosis
factor inhibitors, there are many other biological agents that
are effective in patients with inflammatory arthritis. Currently
Rituximab
Background
B Cells in RA
The mechanism or mechanisms by which B cell depletion
improves RA are unclear [2]. B cells have potentially important
Rituximab 139
Mechanism of Action
Clinical Indications
Clinical Efficacy
4
Relative risk
0
ACR20 ACR50 ACR70 Adverse Serious
events adverse
events
Adverse Reactions
Infection
Anti-biologic Antibodies
Tocilizumab
Background
Clinical Use
Dosing
Clinical Efficacy
Adverse Events
The overall safety profile of Tocilizumab is acceptable. Most
adverse events are mild to moderate in severity. Common
adverse events with tocilizumab include infection, often naso-
pharyngitis, followed by gastrointestinal disturbance,
stomatitis, rash, and headache.
Tocilizumab 145
Control Tocilzumab
600
Per 1,000 patients
400
200
0
ACR50 Remission Improved
improvements function
Infection
Elevated Lipids
Anti-biologic Antibodies
Abatacept
Background
Clinical Use
Dosing
Abatacept
Certolizumab
Golimumab
Adalimumab
Etanercept
Infliximab
Placebo
Clinical Efficacy
Adverse Reactions
Ustekinumab
Background
Clinical Use
Adverse Events
Anakinra
Background
Clinical Use
Side-Effects
References
1. Tsokos GC. B cells, be goneB-cell depletion in the treatment of
rheumatoid arthritis. N Engl J Med. 2004;350:25468.
2. Shaw T, Quan J, Totoritis MC. B cell therapy for rheumatoid
arthritis: the rituximab (anti-CD20) experience. Ann Rheum Dis.
2003;62 Suppl 2: ii559.
3. Lee YH, Bae SC, Song GG. The efficacy and safety of rituximab
for the treatment of active rheumatoid arthritis: a systematic
review and meta-analysis of randomized controlled trials.
Rheumatol Int. 2011;31:14939.
152 Chapter 9. B-Cell Inhibition and Other Biologics
4. Isaacs JD, Cohen SB, Emery P, Tak PP, Wang J, Lei G, et al. Effect
of baseline rheumatoid factor and anticitrullinated peptide anti-
body serotype on rituximab clinical response: a meta-analysis.
Ann Rheum Dis. 2013;72:32936.
5. van Vollenhoven RF, Emery P, Bingham 3rd CO, Keystone EC,
Fleischmann RM, Furst DE, et al. Long-term safety of rituximab
in rheumatoid arthritis: 9.5-year follow-up of the global clinical
trial programme with a focus on adverse events of interest in RA
patients. Ann Rheum Dis. 2013;72:1496502.
6. Dhillon S. Intravenous tocilizumab: a review of its use in adults
with rheumatoid arthritis. BioDrugs. 2014;28:75106.
7. Singh JA, Beg S, Lopez-Olivo MA. Tocilizumab for rheumatoid
arthritis: a Cochrane systematic review. J Rheumatol. 2011;38:1020.
8. Al-Shakarchi I, Gullick NJ, Scott DL. Current perspectives on
tocilizumab for the treatment of rheumatoid arthritis: a review.
Patient Prefer Adherence. 2013;7:65366.
9. Burmester GR, Rubbert-Roth A, Cantagrel A, Hall S,
Leszczynski P, Feldman D, et al. A randomised, double-blind,
parallel-group study of the safety and efficacy of subcutaneous
tocilizumab versus intravenous tocilizumab in combination with
traditional disease-modifying antirheumatic drugs in patients
with moderate to severe rheumatoid arthritis (SUMMACTA
study). Ann Rheum Dis. 2014;73:6974.
10. Genovese MC, Tena CP, Covarrubias A, Leon G, Mysler E,
Keiserman M, et al. Subcutaneous abatacept for the treatment of
rheumatoid arthritis: longterm data from the ACQUIRE trial. J
Rheumatol. 2014;41:62939.
11. Maxwell LJ, Singh JA. Abatacept for rheumatoid arthritis: a
Cochrane systematic review. J Rheumatol. 2010;37:23445.
12. Guyot P, Taylor PC, Christensen R, Pericleous L, Drost P,
Eijgelshoven I, et al. Indirect treatment comparison of abatacept
with methotrexate versus other biologic agents for active rheu-
matoid arthritis despite methotrexate therapy in the United
kingdom. J Rheumatol. 2012;39:1198206.
13. Atzeni F, Sarzi-Puttini P, Mutti A, Bugatti S, Cavagna L, Caporali
R. Long-term safety of abatacept in patients with rheumatoid
arthritis. Autoimmun Rev. 2013;12:11157.
14. Weitz JE, Ritchlin CT. Ustekinumab: targeting the IL-17 path-
way to improve outcomes in psoriatic arthritis. Expert Opin Biol
Ther. 2014;14:51526.
15. McKeage K. Ustekinumab: a review of its use in psoriatic arthri-
tis. Drugs. 2014;74:102939.
References 153
Background
Steroids are given locally into joints or soft-tissues to treat
synovitis, tenosynovitis and other local problems. They are
given systemically as either high or low dose oral steroids by
Pharmacological Effects
Steroids have complex anti-inflammatory and immunomodu-
latory effects. The term glucocorticoid reflects their involve-
ment in glucose metabolism, which is a central effect of all
glucocorticoids. Their anti-inflammatory and immunosup-
pressive effects involve a range of different mechanisms [2].
Steroids affect almost all immune cells. They inhibit white cell
traffic and access to the sites of inflammation, interfere with
the function of immune cells and suppress the production
and actions of humoral factors. Many of their anti-
inflammatory effects are mediated by cytosolic glucocorti-
coid receptors. These regulate proteins involved in
inflammation and interfere with the function of transcription
factors like nuclear factor-B. Steroids also have rapid, non-
genomic mechanisms, which are implicated in many of their
immediate impacts.
In inflammatory arthritis steroids have impacts on the
clinical symptoms of inflammation, which is partly due to
infiltration of the synovium by lymphocytes. They also effect
erosive progression, a process which is partly due to synovial
infiltration by macrophages. Steroids influence the inflamma-
tory process especially during the first months of treatment.
They also have effects on erosions which are only evident
after more prolonged treatment.
Erosive Progression 157
Erosive Progression
There is extensive evidence that steroids reduce erosive
damage, particularly in early arthritis when given in combi-
nation with disease modifying anti-rheumatic drugs. Even
low dose steroids achieve this goal. A systematic review
of the impact of steroids on erosive progression in RA
included 1,414 patients enrolled in 15 trials [4]. All the trials
except one showed treatment benefits in favour of steroids.
The proportion of benefit from steroids in reducing the
158 Chapter 10. Steroids
30 %
Percent with new erosions
20 %
10 %
0%
Methotrexate Methotrexate Methotrexate Methotrexate,
and steroids ciclosporin
ciclosporin and steroids
Side-Effects
The disadvantages of systemic steroid use are their frequent
and serious side-effects [10]. These problems, which are
multiple, have been known for many years. Patients are con-
cerned by general changes such as weight gain and oedema.
Cardiovascular risks, especially accelerated atherosclerosis,
are one major drawback. Other adverse events include hyper-
tension, increased risk for diabetes and its complications,
development of cataracts and increased incidence of glau-
coma, increased susceptibility to infections, hyperlipidaemia,
160 Chapter 10. Steroids
Local Steroids
Steroid injections are used in individual joints to control local
synovitis [11]. Patients show improvements in symptoms last-
ing a few weeks to a few months. This approach is more
commonly used for large joints such as the knee.
Other sites that can be injected include entheses where
tendons are inserted into bones and areas of compression, such
as the carpal tunnel when there is median nerve compression.
Usually steroid injections are given without any imaging,
but there is an increasing tendency to give steroids under
ultrasound guidance. The evidence supporting this change is
incomplete.
Injection of the sacroiliac joints may be beneficial for
patients with seronegative arthritis, who have sacroiliac joint
pain as part of a spondylo-arthropathy. This is best carried
out under imaging control.
Adverse effects of local steroid injections are uncommon.
Iatrogenic infection is potentially serious but rare. It occurs
in less than one in 10,000 cases. More common, but less
References 161
References
1. Bijlsma JW, Boers M, Saag KG, Furst DE. Glucocorticoids in the
treatment of early and late RA. Ann Rheum Dis. 2003;
62:10337.
2. Spies CM, Bijlsma JW, Burmester GR, Buttgereit F. Pharmacology
of glucocorticoids in rheumatoid arthritis. Curr Opin Pharmacol.
2010;10:3027.
3. Gotzsche PC, Johansen HK. Short-term low-dose corticosteroids
vs placebo and nonsteroidal antiinflammatory drugs in rheuma-
toid arthritis. Cochrane Database Syst Rev. 2005;(1):CD000189.
4. Kirwan JR, Bijlsma JW, Boers M, Shea BJ. Effects of glucocorti-
coids on radiological progression in rheumatoid arthritis.
Cochrane Database Syst Rev. 2007;(1):CD006356.
5. Choy EH, Smith CM, Farewell V, Walker D, Hassell A, Chau L,
et al. Factorial randomised controlled trial of glucocorticoids and
combination disease modifying drugs in early rheumatoid arthri-
tis. Ann Rheum Dis. 2008;67:65663.
6. Gorter SL, Bijlsma JW, Cutolo M, Gomez-Reino J, Kouloumas M,
Smolen JS, et al. Current evidence for the management of rheu-
matoid arthritis with glucocorticoids: a systematic literature
review informing the EULAR recommendations for the manage-
ment of rheumatoid arthritis. Ann Rheum Dis. 2010;69:10104.
7. Choy EH, Kingsley GH, Khoshaba B, Pipitone N, Scott DL,
Intramuscular Methylprednisolone Study G. A two year ran-
domised controlled trial of intramuscular depot steroids in
patients with established rheumatoid arthritis who have shown
an incomplete response to disease modifying antirheumatic
drugs. Ann Rheum Dis. 2005;64:128893.
8. Fendler C, Baraliakos X, Braun J. Glucocorticoid treatment in
spondyloarthritis. Clin Exp Rheumatol. 2011;29:S13942.
9. Haibel H, Fendler C, Listing J, Callhoff J, Braun J, Sieper
J. Efficacy of oral prednisolone in active ankylosing spondylitis:
162 Chapter 10. Steroids
Background
Treating inflammatory arthritis is not just a matter of giving
medications. Patients require many non-drug approaches and
treatments. These include education and advice, physiother-
apy, occupational therapy, podiatry, psychological support
and a range of orthopaedic interventions.
Non-drug treatments can be considered in terms of which
type of therapist is involved or by the overall aim of the treat-
ment. This latter approach is preferable in that it does not
specify that treatments should be given by one specific group
of clinicians and that overlapping skills are needed.
Multidisciplinary Teams
Patients with inflammatory arthritis should be treated by
a multidisciplinary team rather than by an individual clini-
cian. The exact make-up of the team will differ between
units depending on a range of local circumstances [3]. Key
members of the team include rheumatologists, specialist
nurses, physiotherapists, occupational therapists and podia-
trists. Patients are often involved in making decisions about
the care they receive and therefore may sometimes be con-
sidered part of the team. Ideally primary care clinicians and
surgeons would also be involved. Although many clinicians
are involved in the team, one member needs to have over-
all responsibility for coordinating care between the various
health professionals involved. Often this role will be taken
by a specialist nurse.
Promoting Mobility, Function and Participation 165
Promoting Coping
It is essential to help patients deal with their arthritis. A range
of approaches can be used. These include promoting self-
management approaches and attempting to instil a readiness
to change. Cognitive-behavioural approaches, which can be
employed by a range of clinicians, can help promote health
behaviour change. Other psychological therapies may also be
useful. These include relaxation training and stress and mood
management as well as formal cognitive behavioural therapy.
Promoting coping can usually be combined with patient edu-
cation and exercise programmes [5].
Physiotherapy
Physiotherapy uses a range of physical approaches with the
aim of reducing pain, preventing deformity and maximising
function. These aims are achieved by using a range of inter-
ventions. Some are active interventions and others are more
passive (Table 11.1).
A number of guidelines for physiotherapy in inflam-
matory arthritis exist and these have been evaluated by
an expert group [6]. They found that recommendations on
exercise therapy and patient education were included in all
guidelines. Other treatment modalities including transcuta-
neous nerve stimulation and thermotherapy were recom-
mended in most but not all guidelines and a few treatment
approaches such as ultrasound were only recommended
occasionally. Recommendations on physiotherapy interven-
tions were variable and often, lacked detail concerning mode
of delivery, intensity, frequency and duration.
Educating patients about what they can do to improve
their arthritis and exercise to maintain function are key
issues. Until recently there were concerns that exercise would
Physiotherapy 167
Occupational Therapy
The goal of occupational therapy is improving patients abili-
ties to perform daily activities and participate in life activities
at work, in the home and socially. In addition therapists help
adapt lifestyles and minimise functional and psychological
problems. Comprehensive programmes include a wide range
of interventions. Occupational therapists are trained in both
Occupational Therapy 169
Podiatry
Feet are commonly involved in inflammatory arthritis.
Involvement of the joints of the feet causes reduced mobility
and impaired function. In about three quarters of people with
inflammatory arthritis of the feet contributes to difficulty
with walking; it is the main problem in many people.
Regrettably the area is relatively overlooked.
Podiatrists can help people with arthritis in four main ways.
Firstly they can provide education about foot problems and
give advice on footwear. Secondly, they can help provide
orthoses and footwear. Thirdly they can arrange general foot
care, which will include support with nail cutting, corn and cal-
lus reduction and providing padding. Finally in patients with
risks of vasculitic or ulcerative foot involvement they can
provide help in reducing the risks of progression. The evidence
for benefit is strongest with insoles and footwear [9].
Orthopaedic Surgery
The surgical treatment of arthritis is a subject in its own right.
It is impractical to provide detailed guidance on this complex
area in a short book on medical treatments. At the same time
it is inappropriate to entirely ignore this important issue for
many patients. It is also an area in which there have been
dramatic changes in approach [10].
The most important contribution of surgery is the replace-
ment of failed joints. The knee and hip are the most impor-
tant examples, but shoulder and elbow replacements are also
important. Equally effective is the prevention of damage to
tendons by timely intervention to prevent rupture or to repair
ruptured tendons at an early stage. Finally a small minority
of patients have major problems with their cervical spine and
may require surgical intervention to prevent cord damage.
References 171
References
1. Forestier R, Andre-Vert J, Guillez P, Coudeyre E, Lefevre-Colau
MM, Combe B, et al. Non-drug treatment (excluding surgery) in
rheumatoid arthritis: clinical practice guidelines. Joint Bone
Spine. 2009;76:6918.
2. Braun J, van den Berg R, Baraliakos X, Boehm H, Burgos-
Vargas R, Collantes-Estevez E, et al. 2010 update of the ASAS/
EULAR recommendations for the management of ankylosing
spondylitis. Ann Rheum Dis. 2011;70:896904.
3. Vliet Vlieland TP. Multidisciplinary team care and outcomes in
rheumatoid arthritis. Curr Opin Rheumatol. 2004;16:1536.
4. Fautrel B, Pham T, Gossec L, Combe B, Flipo RM, Goupille P,
et al. Role and modalities of information and education in the
management of patients with rheumatoid arthritis: development
of recommendations for clinical practice based on published
evidence and expert opinion. Joint Bone Spine. 2005;72:16370.
5. Manning VL, Hurley MV, Scott DL, Coker B, Choy E, Bearne
LM. Education, self-management, and upper extremity exercise
training in people with rheumatoid arthritis: a randomized con-
trolled trial. Arthritis Care Res. 2014;66:21727.
6. Hurkmans EJ, Jones A, Li LC, Vliet Vlieland TP. Quality
appraisal of clinical practice guidelines on the use of physio-
therapy in rheumatoid arthritis: a systematic review.
Rheumatology (Oxford). 2011;50:187988.
172 Chapter 11. Non-drug Treatments
Introduction
Inflammatory arthritis is usually a long-term condition. It can
persist for years and decades. Two different decisions need to
be made about the timing of treatment. The first decision is
when to start treatment. The second decision is when to stop
it. This chapter deals with the latter clinical problem. This
problem is important as patients are often enthusiastic to
have their treatment minimised [1].
Recommendations in Guidelines
Biological Treatments
Steroids
Intramuscular steroids and intra-articular steroids are invari-
ably given intermittently. Their use is designed to ensure
patients do not receive long-term treatment. Consequently,
tapering and stopping are not relevant considerations.
Oral steroids bring short-term benefits. However, the
overwhelming balance of evidence suggests in the long-term
their use leads to more adverse events than benefits. As a
consequence there is a strong rationale for stopping
treatment.
In early arthritis most studies show that both short-term
high dose and medium term intermediate dose steroids can
be tapered and then stopped without major disadvantages. In
established arthritis the evidence is incomplete [13].
When patients have taken steroids for prolonged periods
of time treatment should gradually be tapered by about
Conclusions 179
Conclusions
There is nothing worse than agreeing to discontinue treat-
ment in a patient with inflammatory arthritis who is in sus-
tained clinical remission and then to discover over time that
the disease flares and it is impossible to regain control. Most
experienced clinicians will have come across this scenario at
some time or other. It has a major and potentially dispropor-
tional impact on future practice.
Such an occasional negative occurrence must be weighed
against the substantial weight of evidence that shows it is pos-
sible to successfully taper DMARD, steroid and biologic
treatments in patients who have achieved sustained remis-
sions in their arthritis. Stopping treatment completely seems
less practical. When patients do stop treatment, it is usually
possible to regain control by restarting treatment. Patients
with RA may do better if they are left on a single DMARD. As
DMARDs are relatively ineffective in AS, the likelihood of
stopping biologic treatments in these patients seems less.
Leaving patients on long-term treatments that are over
and above those needed to maintain disease control has two
negative features. Firstly, it risks more adverse events.
Secondly, it is unlikely to be cost-effective, and this latter
point is particularly important with high cost biologic treat-
ments. Therefore on balance when patients achieve sustained
remission the best advice appears to be to taper and minimise
but not entirely stop treatment.
180 Chapter 12. Stopping Treatments
References
1. Markusse IM, Akdemir G, Huizinga TW, Allaart CF. Drug-free
holiday in patients with rheumatoid arthritis: a qualitative study
to explore patients opinion. Clin Rheumatol. 2014;33:11559.
2. Maetzel A, Wong A, Strand V, Tugwell P, Wells G, Bombardier
C. Meta-analysis of treatment termination rates among rheuma-
toid arthritis patients receiving disease-modifying anti-rheumatic
drugs. Rheumatology (Oxford). 2000;39:97581.
3. Scott IC, Kingsley GH, Scott DL. Can we discontinue synthetic
disease-modifying anti-rheumatic drugs in rheumatoid arthritis?
Clin Exp Rheumatol. 2013;31:S48.
4. Boers M, van Tuyl L, van den Broek M, Kostense PJ, Allaart CF.
Meta-analysis suggests that intensive non-biological combina-
tion therapy with step-down prednisolone (COBRA strategy)
may also disconnect disease activity and damage in rheumatoid
arthritis. Ann Rheum Dis. 2013;72:4069.
5. OMahony R, Richards A, Deighton C, Scott D. Withdrawal of
disease-modifying antirheumatic drugs in patients with
rheumatoid arthritis: a systematic review and meta-analysis. Ann
Rheum Dis. 2010;69:18236.
6. van der Woude D, Young A, Jayakumar K, Mertens BJ, Toes RE,
van der Heijde D, et al. Prevalence of and predictive factors for
sustained disease-modifying antirheumatic drug-free remission
in rheumatoid arthritis: results from two large early arthritis
cohorts. Arthritis Rheum. 2009;60:226271.
7. National Institute for Health and Care Excellence. NICE
technology appraisal guidance 130 adalimumab, etanercept
and infliximab for the treatment of rheumatoid arthritis. 2007.
http://www.nice.org.uk/guidance/ta130. Accessed 29 Oct 2014.
8. Smolen JS, Landewe R, Breedveld FC, Buch M, Burmester G,
Dougados M, et al. EULAR recommendations for the manage-
ment of rheumatoid arthritis with synthetic and biological
disease-modifying antirheumatic drugs: 2013 update. Ann
Rheum Dis. 2014;73:492509.
9. Singh JA, Furst DE, Bharat A, Curtis JR, Kavanaugh AF,
Kremer JM, et al. 2012 update of the 2008 American College of
Rheumatology recommendations for the use of disease-
modifying antirheumatic drugs and biologic agents in the treat-
ment of rheumatoid arthritis. Arthritis Care Res. 2012;64:
62539.
References 181
10. van Herwaarden N, den Broeder AA, Jacobs W, van der Maas A,
Bijlsma JW, van Vollenhoven RF, et al. Down-titration and
discontinuation strategies of tumor necrosis factor-blocking
agents for rheumatoid arthritis in patients with low disease
activity. Cochrane Database Syst Rev. 2014;(9):CD010455.
11. Moverley AR, Coates LC, Helliwell PS. Can biologic therapies
be withdrawn or tapered in psoriatic arthritis? Clin Exp
Rheumatol. 2013;31:S513.
12. Lee J, Noh JW, Hwang JW, Oh JM, Kim H, Ahn JK, et al.
Extended dosing of etanercept 25 mg can be effective in patients
with ankylosing spondylitis: a retrospective analysis. Clin
Rheumatol. 2010;29:114954.
13. Boers M. The COBRA trial 20 years later. Clin Exp Rheumatol.
2011;29:S4651.
14. Bacon PA, Myles AB, Beardwell CG, Daly JR. Corticosteroid
withdrawal in rheumatoid arthritis. Lancet. 1966;2:9357.
Chapter 13
Emerging Therapies
in Rheumatoid Arthritis
Introduction
The last decade has seen an unprecedented expansion in the
therapeutic options available to treat RA. The growing num-
ber of effective biologic drugs highlights the complex nature
of the human immune system. It has become apparent that
similar clinical responses can be obtained by blocking any
number of immune pathways. It therefore seems likely, given
the enormous number of potential immunologic mechanisms
at play in RA that many more therapeutic targets exist. The
game change that biologics have brought about is the appre-
ciation of the success of highly specific drugs.
However, biologic agents face several challenges: first, they
are very expensive, and only marketed by a limited number
of pharmaceutical companies; second, they are large proteins,
which means they are both subject to immunogenicity and
also are unable to be used to target intracellular immunologic
pathways. The solutions to these problems are imminent.
Biosimilars
The pharmaceutical regulators define a biosimilar as a biologi-
cal product that is highly similar to an existing licensed biologic,
allowing for minor differences in clinically inactive components,
and for which there are no clinically meaningful differences
between the biological product and the reference product in
terms of the safety, purity, and potency of the product.
The concept of a biosimilar is not a new one, with such
products available for drugs such as insulin or colony stimu-
lating factors having been available for many years. The bio-
logics used in rheumatic diseases differ from existing
biosimilars though, being far more intricate entities, compris-
ing large proteins with complex tertiary and quarternary
structures that are challenging to duplicate. As such, it is not
feasible to produce truly identical products, as subtle changes
in structure, such as those that occur in post-translational
modification, can never be entirely consistent: hence the use
Interchangeability of Biosimilars with Existing Agents 185
Drugs in Development
The advent of biologic therapies has highlighted the impor-
tance of delivering drugs that have a specific target. However,
biologics are also limited by a requirement for parenteral
administration, an inability to enter cells and also the risk of
immunogenicity. In recent years there has been a surge in the
development of highly targeted small molecules that block
pathways downstream of where the current biologics are act-
ing. Such agents would be available orally, and not run the
risk of immunogenicity.
JAK Inhibitors
The Janus Kinase (JAK) inhibitor class is already licensed in
the US and at the time of writing is currently awaiting a deci-
sion from the regulators in Europe. The JAK inhibitors spe-
cifically bind and block members of the JAK enzyme family
(JAK1, JAK2, JAK3 or TYK2). The resultant effect is to sup-
press the downstream production of inflammatory mediators.
Under normal circumstances, JAK phosphorylates a trans-
membrane cytokine receptor in response to an extracellular
signal (for example activation by IL-6). The signal is then
transmitted to the nucleus of the cell via a number of routes
including the Signal Transducer and Activator of Transcription
(STAT) pathway. Subsequent nuclear effects result in increase
production of new inflammatory proteins (including TNF).
Therefore, given the knowledge of success in blocking both
IL-6 and TNF pathways, JAK inhibition represents an
obvious target in RA.
JAK Inhibitors 187
60
50
% (SE) ACR50
40
30
20
10 Methotrexate
Tofacitinib 5 mg
Tofacitinib 10 mg
0
0 6 12 24
Time (months)
PDE4 Inhibitors
Phosphodiesterase 4 (PDE4) is a member of the larger family
of phosphodiesterases. PDE4 is an intracellular molecule that
is involved in the hydrolysis of cyclic AMP within immune
cells, enabling subsequent signaling pathway activation.
Blocking PDE4 has the potential to suppress a wide range of
inflammatory pathways including specific Th1 and Th2
responses. Inhibitors of the PDE pathways have been avail-
able for many years: theophylline is an example of a non-
selective phosphodiesterase inhibitor. Such agents have been
limited in use because of a very narrow therapeutic window
and a high rate of adverse events. However, newer com-
pounds that selectively target phosphodiesterase subfamilies
are emerging as important anti-inflammatory agents.
References 189
Future Targets
The examples mentioned already represent pathways where a
drug is already in a late stage of development. However there
are numerous other drugs in various stages of development that
will filter through in the coming years. In particular the rela-
tively recent discovery of a new subset of T cells, the Th17 class,
that appear to be specifically implicated in autoimmune disease
is of great interest [8]. Two corresponding cytokines, IL-17 and
IL-22 have been identified as circulating potential targets to
inhibit the Th17 pathway. Several blocking agents are currently
under study across a wide range of autoimmune diseases.
References
1. Dranitsaris G, Amir E, Dorward K. Biosimilars of biological drug
therapies: regulatory, clinical and commercial considerations.
Drugs. 2011;71:152736.
2. Scheinberg MA, Kay J. The advent of biosimilar therapies in
rheumatologyO brave new world. Nat Rev Rheumatol.
2012;8:4306.
3. Schiestl M, Stangler T, Torella C, Cepeljnik T, Toll H, Grau
R. Acceptable changes in quality attributes of glycosylated bio-
pharmaceuticals. Nat Biotechnol. 2011;29:3102.
4. Radstake TR, Svenson M, Eijsbouts AM, van den Hoogen FH,
Enevold C, van Riel PL, et al. Formation of antibodies against
infliximab and adalimumab strongly correlates with functional
drug levels and clinical responses in rheumatoid arthritis. Ann
Rheum Dis. 2009;68:173945.
5. Finckh A, Simard JF, Gabay C, Guerne PA, Physicians S. Evidence
for differential acquired drug resistance to anti-tumour necrosis
factor agents in rheumatoid arthritis. Ann Rheum Dis. 2006;65:
74652.
190 Chapter 13. Emerging Therapies in Rheumatoid Arthritis
Serology
RF and ACPA are established predictors of RA severity.
Their presence associates with a more severe disease course
characterised by higher rates of radiological damage and
extra-articular manifestations. In one observational study of
135 early RA patients, individuals with a persistently positive
RF had greater X-ray damage, disease activity and disability
and required more aggressive treatment [3]. Similarly in a
cohort study of 93 early RA patients identified amongst
blood donors, the presence of ACPA prior to and at disease
onset associated with worse radiological outcomes [4].
Smoking
Alcohol
Social Deprivation
Several studies have observed a correlation between social
deprivation and poor RA outcomes. In 869 patients from the
Early Rheumatoid Arthritis Study (ERAS) higher social
deprivation scores associated with higher HAQ scores and
joint counts [8]. This effect may be driven by other lifestyle
factors that associate with low socioeconomic status such as
smoking.
Gender
5
Percentage of variance in x-ray progression explained
4.5
3.5
2.5
1.5
0.5
Shared Epitope
rs11908352 (A)
rs7607479 (C)
rs7667746 (G)
rs7665842 (G)
rs4371699 (A)
rs6821171 (C)
rs1485305 (T)
rs1896368 (G)
rs1896367 (A)
rs1528873 (A)
rs2104286 (C)
rs8192916 (A)
rs1119132 (A)
rs4810485 (T)
rs451066 (A)
rs26232 (T)
SPAG16 IL15 IL15 IL15 IL15 C5orf30 HLA-DRB1 OPG DKK1 DKK1 DKK1 IL2RA GRZB RAD51L1, IL4R CD40 MMP6
ZFP36L1
2 4 4 4 4 5 6 8 10 10 10 10 14 14 16 20 20
Genetics
Predicting RA Severity
Several research groups have attempted to combine these
predictive factors in prediction models, which can be used
to predict which patients are likely to have rapid radiological
196 Chapter 14. Stratified Medicine in Inflammatory Arthritis
Gender
Smoking
Serology
12 12
Triple therapy ACPA Triple therapy ACPA
10 *** *** 10
8 *** 8
Larsen
Larsen
6 6
**
4 4
2 2
0 0
0 6 12 18 24 Triple 0 6 12 18 24
Mono
Time(months) Time(months)
Disease Duration
It is established that treating RA in its very early stages
improves patient outcomes. This concept, known as the win-
dow of opportunity, is demonstrated in a meta-analysis of 14
RCTs evaluating predictors of DMARD responses [19]. The
percentages of patients attaining an ACR 20 response with
active treatment comprised 53, 43, 44, 38 and 35 % for
Prediction Models for Treatment Responses in RA 199
Genetics
Attempts to identify genetic predictors of methotrexate
response have focussed on the methotrexate cellular path-
way. Their results have been inconsistent and no clear genetic
marker that predicts methotrexate responses has been identi-
fied [20]. One validated genetic marker that associates with
anti-TNF response has been identified. In this study of nearly
3,000 RA patients one SNP (rs6427528) at the 1q23 locus
associated with changes in DAS28 in patients receiving etan-
ercept (P = 8 108) [21].
Psoriatic Arthritis
Ankylosing Spondylitis
Conclusion
Stratified medicine is an area of emerging importance in
the management of inflammatory arthritis patients. Many
poor prognostic factors have been identified, particularly
in RA patients, which can be used to identify patients
requiring more aggressive treatment. This knowledge is
reflected in current International guidelines for managing
RA and PsA patients, which advocate reserving intensive
treatment for poor prognosis patients with active disease.
By contrast knowledge of which factors predict responses
to specific medications is more limited. Further research
is required to better define relevant predictive factors in
inflammatory arthritis patients and to develop and vali-
date prediction models that combine these factors to
identify patient groups likely to respond to certain
therapies.
References
1. Medical Research Council.MRC website.2014.[Internet] Available
at: http://www.mrc.ac.uk/Ourresearch/ResearchInitiatives/
StratifiedMedicine/index.htm. Accessed 19 Apr 2014.
2. Rubbert-Roth A, Finckh A. Treatment options in patients with
rheumatoid arthritis failing initial TNF inhibitor therapy: a criti-
cal review. Arthritis Res Ther. 2009;11 Suppl 1:S1.
3. van Zeben D, Hazes JM, Zwinderman AH, Cats A, van der
Voort EA, Breedveld FC. Clinical significance of rheumatoid
factors in early rheumatoid arthritis: results of a follow up study.
Ann Rheum Dis. 1992;51:102935.
4. Berglin E, Johansson T, Sundin U, Jidell E, Wadell G, Hallmans
G, et al. Radiological outcome in rheumatoid arthritis is pre-
dicted by presence of antibodies against cyclic citrullinated pep-
tide before and at disease onset, and by IgA-RF at disease onset.
Ann Rheum Dis. 2006;65:4538.
5. Manfredsdottir VF, Vikingsdottir T, Jonsson T, Geirsson AJ,
Kjartansson O, Heimisdottir M, et al. The effects of tobacco
smoking and rheumatoid factor seropositivity on disease activity
and joint damage in early rheumatoid arthritis. Rheumatology
(Oxford). 2006;45:73440.
204 Chapter 14. Stratified Medicine in Inflammatory Arthritis
Anti-tumour necrosis B
factor-alpha (TNF-) Bath ankylosing spondylitis
treatment (cont.) disease activity index
psoriatic arthritis, 127 (BASDAI), 6061
radiographic progression, B-cell inhibition and other
126 biologics
rheumatoid arthritis, abatacept
125126 adverse reactions, 149
etanercept, 122 clinical efficacy, 148
golimumab, 123 clinical use, 147
immune responses, 132 definition, 147
indications, 124 dosing, 147148
inflammatory arthropathies, anakinra
120121 clinical use, 150151
infliximab, 122 definition, 150
primary/secondary failure, side-effects, 151
132133 rituximab
safety and adverse effects adverse reactions, 141142
auto-immune anti-biologic antibodies,
disorders, 131 143
biologics registries, clinical efficacy, 139140
127128 clinical indications, 139
demyelinating-like definition, 138
disorders, 130 infection, 142143
haematological initial and repeat courses,
disorders, 130 141
heart failure, 130 mechanism of action, 139
hypersensitivity RA, 138139
responses, 128 tocilizumab
infections, 128129 adverse events, 144146
interstitial lung disease, anti-biologic antibodies, 146
131132 clinical efficacy, 144
local injection site clinical use, 143
reactions, 128 definition, 143
malignancy, 131 dosing, 144
psoriasis, 132 ustekinumab
Arthritis mutilans, 29 adverse events, 150
Assessment of Spondyloarthritis clinical use, 149
International definition, 149
Society (ASAS) Biologics
study, 89 anti-rheumatic drugs, 112
Auto-immune anti-TNF, 112
disorders, 131 classes of, 113114
Azathioprine, 105 cost-utility analyses, 117
Index 209
Disease-modifying G
anti-rheumatic drugs Genetics
(DMARDs) (cont.) disease course, 195
intensive treatment, 92 treatment responses, 199
leflunomide Golimumab, 123
active metabolite, 100 Greyscale ultrasound, 5556
adverse events, 101102
clinical use and efficacy,
101 H
definition, 100 Haematological disorders, 130
pyrimidine synthesis Health assessment questionnaire
effect, 100101 (HAQ), 4647
methotrexate Heart failure, 130
adverse reactions, 99100 Hydroxychloroquine, 104
clinical use and efficacy, Hypersensitivity responses, 128
9799 Hypertension, 102
definition, 9697
minor, 105
monitoring, 89 I
seronegative arthritis, 93 Inflammatory arthritis
starting, 89 ACR response criteria, 60
stopping, 9091 adhesion molecules in, 20
sulfasalazine, 102103 aetiology, 1617
treat to target, 92 assessment role, 40
use of, 88 BASDAI, 6061
Distal interphalangeal joint biological treatments
arthritis, 28 ankylosing spondylitis, 178
DMARDs. See Disease-modifying psoriatic arthritis, 178
anti-rheumatic drugs rheumatoid arthritis, 177
(DMARDs) steroids, 178179
Doppler ultrasound, 5556 stopping and tapering,
176177
CDAI, 59
E chemokines, 20
Elliots study, 112 classification criteria, 5
Enthesitis, 2930 clinical assessments, 4043
Enzyme-linked immunosorbent clinical course, 33
assays (ELISA), 52 core data set, 5758
Erythrocyte sedimentation rate diagnosis
(ESR), 4950 ankylosing spondylitis,
Etanercept, 122 5, 78
European League Against psoriatic arthritis, 810
Rheumatism rheumatoid arthritis, 5
(EULAR), 5, 7, spondyloarthropathies, 10
201202 disease activity score, 5859
EuroQol, 4849 disease outcomes, 6163
Index 211
N P
Neutropenia, 146 Paracetamol, 6970
Non-drug treatments Peripheral joints, synovitis of
definition, 163 joint swelling and tenderness,
education and support, 165 2425
multidisciplinary teams, 164 symptoms, 24
occupational therapy, 168170 Phosphodiesterase 4 (PDE4)
orthopaedic surgery, 170171 inhibitors, 188189
overview of, 164 Physiotherapy
physiotherapy aerobic exercise programmes,
aerobic exercise 168
programmes, 168 benefits, 168
benefits, 168 interventions, 166, 167
interventions, 166, 167 Podiatry, 170
podiatry, 170 Polyarthritis, 28
promote mobility and Psoriasis, 132
enhance function, Psoriatic arthritis (PsA), 200201
165166 biological treatments, 116, 178
promoting coping, 165 classification criteria for, 810
Non-steroidal anti-inflammatory disease outcomes, 63
drugs (NSAIDs) imaging, 5657
adverse reactions NSAID, 83
cardiovascular, 8182 peripheral joints involvement,
central nervous system, 78 2829
gastrointestinal, 7981 steroids, 159
renal side effects, 7879 TNF- inhibitors
ankylosing spondylitis, 8384 clinical effects, 127
classification of, 7475 indications, 124
Cox-2 drugs, 7577 Psoriatic arthritis response
Cox I/Cox II effects, 7374 criteria (PsARC), 60
mechanism of action, 7374 Pulmonary interstitial
psoriatic arthritis, 83 disease, 99
rheumatoid arthritis, 8283
seronegative
spondyloarthritis, 83 R
Norfolk study, 14, 15 Rapid radiological progression
(RRP), 195196
Reactive arthritis, 57
O Rheumatoid arthritis (RA)
Occupational therapy, 168170 aetiology of, 16
Oligoarthritis, 28 biological treatments,
Omeprazole, 80 114115, 177
Opioids, 7071 biosimilars, 184186
Orthopaedic surgery, 170171 BTK inhibitors, 188
Index 213
Steroids Tocilizumab
ankylosing spondylitis, 159 adverse events
definition, 155156 infection, 145
early rheumatoid arthritis, infusion and
158 hypersensitivity
erosive progression, 157158 reactions, 145
intramuscular steroid lipid elevations, 146
injections, 159 liver function test
joint inflammation effects, 157 abnormalities, 146
local steroids injections, neutropenia and
160161 thrombocytopenia, 146
pharmacological effects, anti-biologic antibodies, 146
156157 clinical efficacy, 144
psoriatic arthritis, 159 clinical use, 143
side-effects, 159160 definition, 143
Stratified medicine dosing, 144
ACR guidelines, 201202 Tramadol, 7172
disease course, variability, 192 Tuberculosis, 129
EULAR guidelines, 201202 Tumour necrosis factor-alpha
non-RA inflammatory (TNF-), 19
arthropathies
ankylosing spondylitis, 201
psoriatic arthritis, 200201 U
RA (see Rheumatoid arthritis Ultrasound, 5556
(RA)) Ustekinumab
research, 193 adverse events, 150
treatment responses, clinical use, 149
variability, 192 definition, 149
Sulfasalazine, 102103
Swollen joint count, 4243
V
Visual analogue scales (VAS),
T 43, 45
T-cell modulation, 138
Tender joint count, 4243
Tenosynovitis, 25 X
Thrombocytopenia, 146 X-rays, rheumatoid arthritis, 5355