ArticlePDF AvailableLiterature Review

Immune-mediated inflammatory diseases (IMIDs) and biologic therapy: a medical revolution

Authors:

Abstract and Figures

Targeted biologic therapies have revolutionised treatment of immune-mediated inflammatory diseases (IMIDs) due to their efficacy, speed of onset and tolerability. The discovery that clinically unrelated conditions, such as rheumatoid arthritis and Crohn's disease, share similar immune dysregulation has led to a shift in the management of IMIDs from one of organ-based symptom relief to mechanism-based treatment. The fact that anticytokine therapy has been effective in treating multiple orphan inflammatory conditions confirms the IMID paradigm. In this review we examine the biologic agents currently licensed for use in the US and Europe: infliximab, etanercept, adalimumab, rituximab, abatacept, anakinra, alefacept and efalizumab. We also discuss the rationale behind the management of IMIDs using rheumatoid arthritis, Crohn's disease, psoriasis and psoriatic arthritis as examples. For the medical profession, IMID represents a breakthrough in the way pathology is classified. In this burgeoning era of biologic therapy the prospect of complete disease remission is conceivable.
Content may be subject to copyright.
REVIEW
Immune-mediated inflammatory diseases (IMIDs) and biologic
therapy: a medical revolution
Annabel Kuek, Brian L Hazleman, Andrew J K O
¨
sto¨r
...................................................................................................................................
Postgrad Med J 2007;83:251–260. doi: 10.1136/pgmj.2006.052688
Targeted biologic therapies have revolutionised treatment of
immune-mediated inflammatory diseases (IMIDs) due to their
efficacy, speed of onset and tolerability. The discovery that
clinically unrelated conditions, such as rheumatoid arthritis and
Crohn’s disease, share similar immune dysregulation has led to
a shift in the management of IMIDs from one of organ-based
symptom relief to mechanism-based treatment. The fact that
anticytokine therapy has been effective in treating multiple
orphan inflammatory conditions confirms the IMID paradigm. In
this review we examine the biologic agents currently licensed
for use in the US and Europe: infliximab, etanercept,
adalimumab, rituximab, abatacept, anakinra, alefacept and
efalizumab. We also discuss the rationale behind the
management of IMIDs using rheumatoid arthritis, Crohn’s
disease, psoriasis and psoriatic arthritis as examples. For the
medical profession, IMID represents a breakthrough in the way
pathology is classified. In this burgeoning era of biologic
therapy the prospect of complete disease remission is
conceivable.
.............................................................................
See end of article for
authors’ affiliations
........................
Correspondence to:
Dr A J K O
¨
sto¨r,
Rheumatology Research
Unit, Box 194, E6,
Addenbrooke’s Hospital,
Hills Road, Cambridge CB2
2QQ, UK; andrew.ostor@
addenbrookes.nhs.uk
Received 24 August 2006
Accepted
29 November 2006
........................
I
mmune-mediated inflammatory disease (IMID)
is a concept used to collectively describe a group
of ostensibly unrelated conditions that share
common inflammatory pathways. Encompassing
disorders as diverse as ankylosing spondylitis (AS),
type 1 diabetes and multiple sclerosis, the immune
dysregulation may afflict any organ system and
result in significant morbidity, reduced quality of
life (QoL) and premature death. Although the
aetiology of these conditions is unknown,
advances in molecular research have revealed that
an imbalance in inflammatory cytokines is central
to their pathogenesis.
1
The incidence of IMIDs in Western society
approximates 5–7%. Genetic factors are crucial
determinants of susceptibility and animal models
have led to the identification of several genes that
contribute to an autoimmune diathesis when
deleted or overexpressed.
2
Many of these diseases
also share similar environmental precipitants such
as infection and trauma. Frequently, multiple
IMIDs co-exist within the same patient, for
example a large study of AS found that 39% of
patients also developed iritis, 16% psoriasis and 8%
inflammatory bowel disease.
3
Various IMIDs may
also co-exist within the same family.
The most convincing evidence linking the
pathophysiology and treatment of autoimmune
diseases has been demonstrated with the tumour
necrosis factor-a (TNFa) inhibitors. Infliximab, the
first anti-TNFa agent licensed, has shown clinical
benefit in a number of seemingly unrelated
conditions including rheumatoid arthritis (RA),
Crohn’s disease (CD) and psoriasis. In addition,
there have been anecdotal suggestions of efficacy
with anti-TNFa agents in a multitude of orphan
inflammatory diseases. This, together with the
observation that treatment of one condition is
capable of improving multiple organ systems,
further validates the IMID paradigm and has
inspired additional research for common treat-
ment strategies. IMID therapy truly epitomises the
triumph of translational research (bench to bed-
side).
The therapeutic aims for IMIDs are identical: to
gain rapid control of inflammation, prevent tissue
damage, improve QoL and, if possible, achieve
long-term disease remission. As these goals are
rarely met in each patient with traditional treat-
ments, presumably due to failure to address the
underlying immunopathology, targeted biologic
therapy has been revolutionary. In this review we
describe the molecular basis for the IMID concept,
examine the immune dysregulation that many
IMIDs share, and briefly focus on the evidence for
biologic therapies that are currently licensed for
use in immune-mediated conditions.
IMID PATHOGENESIS
Over a decade ago, investigators discovered that
cytokine dysregulation was pivotal to the patho-
physiology of IMIDs. While these molecules are
known to be indispensable mediators of normal
immune function, an imbalance in their produc-
tion can lead to chronic inflammatory conditions.
Commonly, IMIDs are associated with a relative
over-expression of cytokines, such as TNFa in RA,
yet a relative under-expression of cytokines may be
equally important in disease pathogenesis, for
example interleukin 10 (IL10) deficiency in CD.
45
Interestingly, cytokines may have different effects,
Abbreviations: ACR, American College of Rheumatology;
ANA, antinuclear antibodies; AS, ankylosing spondylitis;
CD, Crohn’s disease; CTLA4, cytotoxic T lymphocyte
antigen-4; DMARDs, disease modifying anti-rheumatic
drugs; EMEA, European Medicines Agency; FDA, US Food
and Drug Administration; IL1, interleukin 1; IL6, interleukin
6; IL10, interleukin 10; IMID, immune-mediated
inflammatory disease; JIA, juvenile idiopathic arthritis; MTX,
methotrexate; NICE, National Institute for Health and
Clinical Excellence; PEG, polyethylene glycol; PsA, psoriatic
arthritis; QoL, quality of life; RA, rheumatoid arthritis; RCT,
randomised controlled trial; TB, tuberculosis; TNFa, tumour
necrosis factor-a; UC, ulcerative colitis
251
www.postgradmedj.com
either pro- or anti-inflammatory, depending on the stage of
disease.
2
Although immunoregulatory cytokines are produced by
many different cell types, CD4+ T lymphocytes are thought to
be the main orchestrators of the immune response. These T
helper cells are typically divided into two functionally hetero-
geneous subsets, Th 1 and Th 2, on the basis of the cytokines
they produce (fig 1). Th 1 cells have predominantly pro-
inflammatory effects and have been implicated in the immu-
nopathogenesis of IMIDs. Th 2 cells are thought to have anti-
inflammatory or protective functions in this context.
6
Because
of this central role in IMIDs, modulation of T cell function has
became an attractive target for intervention.
Another area arousing considerable interest is the role of B
lymphocytes in IMIDs. Early concepts focused on the ability of
B cells to produce antibodies, however it is now known that B
cells have much broader functions within the immune system,
including T cell activation, cytokine synthesis, regulation of
lymphoid architecture and maintenance of tolerance.
7
Emerging evidence indicates that disruption of these tightly
regulated B cell processes can potentially lead to autoimmune
disease. Certainly in RA loss of B cell tolerance and inappropri-
ate autoantibody production is a key pathological process, and
this has motivated investigators to attempt B cell depletion as a
novel therapeutic strategy.
8
Preliminary studies with the anti-
CD20 monoclonal antibody, rituximab, have now been com-
pleted for a number of IMIDs with clear evidence of efficacy
(table 1).
An appraisal of the specific pathogenic mechanisms involved
in every IMID is beyond the scope of this article, however this
review will focus on four common IMIDs in which biologic
agents have had a major impact, namely RA, CD, and psoriasis
and psoriatic arthritis (PsA).
RHEUMATOID ARTHRITIS
Rheumatoid arthritis (RA) is a systemic inflammatory condi-
tion that primarily affects the synovial membrane of affected
joints. Extra-articular manifestations are common and may
involve any organ system. RA affects approximately 1% of the
adult population and is associated with chronic pain, disability
and increased mortality.
9
The long-term prognosis is generally
poor with approximately 80% of patients experiencing sig-
nificant functional disability within 20 years of diagnosis.
10
Although the aetiology of RA remains unknown, it is
postulated that an antigen, either exogenous or endogenous,
triggers an abnormal immune response in a genetically
susceptible individual.
11
Th 1 cells, activated by antigen
presenting cells and co-stimulatory pathways, infiltrate the
synovium and stimulate the production of pro-inflammatory
cytokines and destructive proteinases. While many cell types
and mediators are involved in the pathogenesis of RA, the key
drivers of inflammation are thought to be TNFa, interleukin 1
(IL1) and interleukin 6 (IL6).
12
There is clear evidence from
clinical trials that antibodies directed against TNFa and IL1 are
capable of ameliorating disease activity. In contrast, the anti-
inflammatory cytokines IL4 and IL10 may play a protective role
in RA.
13
IL4, for example, is produced by Th 2 cells and has been
shown, in vitro, to inhibit the activation of Th 1 cells and
therefore decrease the production of IL1 and TNFa (fig 1).
14
The exact role of B lymphocytes in the pathogenesis of RA
requires elucidation, however several possible mechanisms
have been proposed. Perhaps the most well described process
is loss of B cell tolerance and the production of pathogenic
autoantibodies, such as rheumatoid factor.
15
Additional experi-
ments in mice have demonstrated that T cell activation within
the rheumatoid synovium is critically dependent on the
presence of B cells.
16
Biologics have significantly expanded the therapeutic options
available for RA patients who remain refractory to traditional
disease modifying anti-rheumatic drugs (DMARDs). Agents
currently licensed for use are the three TNFa inhibitors
(infliximab, etanercept and adalimumab) and the two newer
agents, rituximab (anti-CD20) and abatacept (T cell co-
stimulation blocker). Anakinra, a recombinant IL1 receptor
antagonist is also licensed, however its efficacy in clinical trials
has been less impressive than with the TNFa blockers and thus
the National Institute for Health and Clinical Excellence (NICE)
does not recommend its use in the United Kingdom. The anti-
IL6 receptor antibody tocilizumab is however showing promise
in clinical trials and a US Food and Drug Administration (FDA)
licence application is anticipated for 2007.
CROHN’S DISEASE
Crohn’s disease (CD) is a T cell mediated disorder characterised
by a relapsing inflammatory process in the gut with extra-
intestinal manifestations typically involving the skin, eyes and
joints.
17
Although the cause of CD is unknown, the most
prominent theory implicates a dysfunctional mucosal immune
response to an otherwise innocuous luminal antigen in a
genetically susceptible host.
18
Complications such as perianal
fistulae occur in up to 43% of patients, many of which require
surgery.
19
With a prevalence of around 0.1% in the developed
world,
20
CD places a major burden on health care resources.
Similar to RA, an imbalance in the cytokine milieu is central
to its pathogenesis. Gut inflammation typically begins with
mucosal infiltration of neutrophils and macrophages, which in
turn activate T cells.
18
By stimulating the production of pro-
inflammatory cytokines, Th 1 cells amplify the immune
response and promote tissue destruction.
21
Equally, a relative
underproduction of regulatory cytokines (IL4 and IL10) has
been observed in Crohn’s affected mucosa.
5
This finding has led
to targeted drug therapies focusing on either inhibiting or
augmenting cytokine action. IL10, for example, is thought to be
a potent downregulator of the Th 1 response, and although
Figure 1 Th 1 and Th 2 cell responses. APC, antigen presenting cell; IL,
interleukin; MHC, major histocompatibility complex; Th, T helper cell; TNF,
tumour necrosis factor.
252 Kuek, Hazleman, O
¨
sto¨r
www.postgradmedj.com
animal models have shown a strong link between IL10
deficiency and gut inflammation, the use of IL10 therapy in
clinical trials has been disappointing.
22
Great success however,
has been achieved with certain anti-TNFa agents.
Infliximab is currently the only biologic agent approved for
the treatment of moderate-severe and/or fistulising CD.
Certolizumab (Cimzia, Nektar), a subcutaneous anti-TNFa
agent, has also demonstrated efficacy in CD and an application
has been submitted to the FDA for appraisal.
23
Interestingly, the
TNF receptor fusion protein etanercept has failed to show
significant benefit in clinical trials, presumably due to its
different mechanism of action.
24
Etanercept only binds soluble
TNF, yet it is the transmembrane binding of TNF that is crucial
for inducing apoptosis of T cells in CD.
25
PSORIASIS AND PSORIATIC ARTHRITIS
Psoriasis is a chronic inflammatory disease of uncertain
aetiology that affects approximately 2% of the population.
26
Characterised by scaly erythematous plaques, the disease can
lead to significant physical and psychological distress, and in up
to 30% of patients a debilitating arthropathy may develop.
27
A
genetic predisposition for psoriasis is clear, with concordance of
approximately 70% in monozygotic twins, however a complex
interplay with environmental factors is likely.
28
Akin to RA and CD, psoriasis and PsA are T cell-mediated.
Histologically, skin plaques and inflamed synovial tissue
demonstrate an abundance of T cells and increased vascularity.
The inflammatory cascade is thought to be triggered by the
local activation of CD4+ T cells which generate a series of pro-
inflammatory cytokines such as TNFa that subsequently
activate CD8+ T cells, the main effector cells.
29
High concentra-
tions of TNFa have been detected in psoriatic skin lesions and
in the synovium of affected joints and it is believed that this
cytokine plays a pivotal role in perpetuating inflammation in
addition to stimulating the angiogenesis and keratinocyte
proliferation.
30
In many cases, conventional systemic therapies for both
psoriasis and PsA are either ineffective, are poorly tolerated or
are unable to maintain long-lasting remission.
31
Recently,
however, enormous growth in our understanding of the disease
pathogenesis has led to an expansion in therapeutic options.
Biologic agents that are currently licensed for plaque
psoriasis include the TNFa inhibitors, etanercept and inflix-
imab, as well as the T cell-targeted therapies, efalizumab and
alefacept (FDA only). NICE guidance has recently been issued
for the use of etanercept in severe plaque psoriasis in patients
who have failed conventional therapies, and for efalizumab as a
second line agent to etanercept. For PsA, three anti-TNFa
agents (infliximab, etanercept and adalimumab) are licensed,
however only infliximab and etanercept are NICE approved.
Benefit in psoriasis trials has also been seen with the T cell
costimulation blocker, abatacept.
32
BIOLOGICS CURRENTLY LICENSED FOR IMID
TREATMENT
Infliximab
Infliximab (Remicade, Schering-Plough/Centocor), a mouse-
human chimeric monoclonal antibody directed against TNFa,is
licensed for use in RA, AS, PsA, CD, ulcerative colitis (UC) and
psoriasis (tables 2 and 3). To date, the only NICE-approved
indications for infliximab are RA, CD and PsA, although an
appraisal is in development for AS. The drug is given as a 2 h
intravenous infusion with a dose of 3–5 mg/kg at weeks 0, 2
and 6, and then every 8 weeks thereafter. In the event of
waning efficacy, the dose may be increased up to 10 mg/kg, or
the infusion frequency increased to four to six weekly.
Infliximab is currently approved for active RA in those who
have failed DMARDs, and for severe progressive RA in those not
previously treated with DMARDs. The efficacy of infliximab has
been well documented in randomised controlled trials (RCTs).
In the ATTRACT trial, infliximab was compared with placebo in
428 RA patients taking methotrexate (MTX).
33
The actively
Table 1 IMIDs in which biologic therapy appears promising
Infliximab Etanercept Adalimumab Rituximab
Sjogren’s Sjogren’s Crohn’s disease Polymyositis/dermatomyositis
Polymyositis/dermatomyositis Polymyositis/dermatomyositis UC Wegener’s granulomatosis
Wegener’s vasculitis Wegener’s vasculitis Psoriasis GCA/PMR
Behcet’s Behcet’s JIA Polyarteritis nodosa
GCA/PMR GCA Behcet’s JIA
Takayasu’s arteritis Takayasu’s arteritis Takayasu’s arteritis Graft versus host disease
Polyarteritis nodosa Polyarteritis nodosa Sarcoidosis Cryoglobulinaemic vasculitis
Sarcoidosis Sarcoidosis Adult onset Still’s disease ITP
Adult onset Still’s disease Adult onset Still’s disease Hydradenitis supprativa TTP
JIA Cryoglobulinaemic vasculitis Pyoderma gangrenosum AIHA
Kawasaki disease Relapsing polychondritis Pemphigus Anti-phospholipid syndrome
Cryoglobulinaemic vasculitis Hydradenitis supprativa Idiopathic membranous GN
Relapsing polychondritis Pyoderma gangrenosum Hep C associated GN
Hydradenitis supprativa Graft versus host disease Multiple sclerosis
Coeliac disease Chronic hepatitis C Myasthenia gravis
Myelodysplastic syndrome ITP Pemphigus
Pyoderma gangrenosum Refractory asthma Grave’s disease
Erythema nodosum Amyloidosis CIDP
SAPHO syndrome Multicentric reticulohistiocytosis
Graft versus host disease Pemphigus
Chronic hepatitis B/C Grave’s disease
TTP CIDP
Refractory asthma
SLE
Amyloidosis
Multicentric reticulohistiocytosis
Pemphigus
This list is not exhaustive. AIHA, autoimmune haemolytic anaemia; CIDP, chronic inflammatory demyelinating polyneuropathy; GCA, giant cell arteritis; GN,
glomerulonephritis; JIA, juvenile idiopathic arthritis; PMR, polymyalgia rheumatica; SAPHO, synovitis, acne, pustulosis, hyperostosis, osteitis; SLE, systemic lupus
erythematosus; TTP/ITP, thrombotic/idiopathic thrombocytopaenic purpura; UC, ulcerative colitis.
IMIDs and biologic therapy 253
www.postgradmedj.com
treated subjects showed significant improvements in all
American College of Rheumatology (ACR) responder indices
(box 1). Radiological progression was inhibited and QoL was
improved in the infliximab-MTX groups for up to 2 years.
34
Interestingly, sub-analysis showed reduced radiological pro-
gression even in the absence of clinical response.
The ASPIRE trial examined MTX naive patients with early
active RA.
35
In the group taking MTX alone, radiographic
progression was associated with high inflammatory markers
and persistent disease activity.
36
Infliximab plus MTX, on the
other hand, virtually halted radiographic progression of disease
and improvement was seen in all ACR indices. These data
suggested that early aggressive treatment of RA may avert joint
destruction.
Approval for infliximab in psoriasis was based on two large
RCTs. In the SPIRIT study, 88% of patients treated with
infliximab met the primary end point of PASI 75 (75%
improvement in Psoriasis Area and Severity Index) at week
10 versus 6% receiving placebo.
37
Significant improvements
were also seen in health related QoL. Similarly, in the EXPRESS
study, 80% of patients on infliximab therapy achieved a PASI 75
response compared to 3% on placebo.
38
For PsA, the efficacy of infliximab plus MTX has been studied
in two RCTs.
39 40
At week 16 in the IMPACT trial, almost two
thirds of patients achieved an ACR20 response compared to
10% on MTX alone. Improvements in articular and dermato-
logic manifestations of PsA were sustained until study
completion and significant benefits in health related QoL and
physical function were noted.
41
In an early trial of 108 patients with moderate to severe CD,
81% responded to a single dose of infliximab 5 mg/kg compared
to 17% on placebo. Remarkably, clinical remission was achieved
in almost half of those on active treatment.
42
In the ACCENT I
trial, patients who demonstrated an initial response were found
to be more likely to sustain a clinical response with infliximab
maintenance therapy than those on placebo.
43
Clinical improve-
ment was associated with endoscopic evidence of mucosal
healing.
44
The approval for infliximab maintenance therapy in
fistulising CD was based on the results of the ACCENT II
study.
45
At the end of 1 year, maintenance therapy with
infliximab resulted in a more sustained clinical response, a
higher rate of complete fistula closures and improvement in
QoL. Importantly, infliximab maintenance therapy also reduced
the number of hospitalisations and need for surgery.
46
Table 2 Properties of approved biologics
Biologic agent Infliximab Etanercept Adalimumab Rituximab Abatacept Anakinra
Proprietary name Remicade Enbrel Humira Rituxan Orencia Kineret
Mabthera
Construct Chimeric mAb to TNFa receptor Fully humanised Chimeric mAb to CTLA4Ig IL1 receptor antagonist
TNFa fusion protein mAb to TNFa CD20
Mode of action Binds to soluble and Binds to soluble Binds to soluble Binds to CD20 Blocks T cell co- Binds to IL1 receptor
membrane bound TNFa and TNFb and membrane molecule on B cells stimulation via
TNFa bound TNFa CD28-CD80/86
Half-life 9 days 4 days 14 days Variable 13 days 6 h
Dose 3–5 mg/kg of body 25 mg twice 40 mg every second 1000 mg on day 1 10 mg/kg of body 100 mg daily
weight at 0, 2 and weekly week. For incomplete and day 15 weight at 0, 2 and
6 weeks, and then response, dose may 4 weeks, and then
8 weekly. Dose be given weekly monthly thereafter
can be increased Repeated every 6–
to 10 mg/kg or 9 months
frequency of
injection increased
Administration Intravenous Subcutaneous Subcutaneous Intravenous Intravenous Subcutaneous
CTLA4, cytotoxic T lymphocyte antigen-4; IL, interleukin; mAb, monoclonal antibody; TNF, tumour necrosis factor.
Table 3 Biologics approved for IMIDs: FDA, EMEA and NICE
Biologic agent Action FDA licence EMEA licence NICE approval
Infliximab Anti-TNFa RA RA RA
AS AS PsA
PsA PsA CD
CD CD AS (appraisal)
UC UC UC (submitted)
Paediatric CD Psoriasis Psoriasis
Paediatric CD (appraisal)
Etanercept Anti-TNFa RA RA RA
JIA JIA JIA
AS AS PsA
PsA PsA Psoriasis
Psoriasis Psoriasis AS (appraisal)
Adalimumab Anti-TNFa RA RA RA (appraisal)
PsA PsA PsA
AS AS AS
Rituximab Anti-CD20 RA RA RA (submitted)
Abatacept CTLA4Ig RA RA (submitted) RA (submitted)
Efalizumab Anti-CD11a Psoriasis Psoriasis Psoriasis
Alefacept LFA-3/IgG Fc construct Psoriasis None None
Anakinra Anti-IL1 RA RA None
AS, ankylosing spondylitis; CD, Crohn’s disease; CTLA4, cytotoxic T lymphocyte antigen-4; EMEA, European Medicines Agency; FDA, US Food and Drug
Administration; IL, interleukin; JIA, juvenile idiopathic arthritis; NICE, UK National Institute for Health and Clinical Excellence; PsA, psoriatic arthritis; RA, rheumatoid
arthritis; TNFa, tumour necrosis factor-a; UC, ulcerative colitis.
254 Kuek, Hazleman, O
¨
sto¨r
www.postgradmedj.com
Based on success in RCTs, infliximab was the first biologic
approved for the treatment of UC and a licence has also been
granted for use in AS. Additional case reports have demonstrated
the therapeutic value of infliximab in many other IMIDs (table 1).
Etanercept
Etanercept (Enbrel, Wyeth-Ayerst/Amgen) is a soluble TNF
receptor fusion protein which comprises two TNFa receptors
linked to human IgG and binds soluble TNFa. It is given as a
25 mg subcutaneous injection twice weekly or 50 mg weekly.
Etanercept is licensed for use in RA, psoriasis, PsA, AS and
juvenile idiopathic arthritis (JIA) (tables 2 and 3). Unlike
infliximab, etanercept has not shown benefit in CD as
mentioned previously.
The efficacy of etanercept in RA was initially evaluated as
monotherapy. Significant improvements were found in all ACR
responder indices and QoL measures in the etanercept groups
compared with placebo.
47
The TEMPO study, combining
etanercept with MTX, however, indicated that the two drugs
are significantly more effective in retarding disease progression
than either agent alone.
48
In the ERA trial, etanercept was directly compared with MTX
in patients with RA diagnosed within the previous 3 years.
Although response rates were initially more rapid with
etanercept, responses converged at 12 months.
49
After 2 years,
however, significantly more etanercept-treated patients met the
ACR20 improvement criteria than those on MTX monotherapy,
and the rate of erosive change was also retarded.
50
In a 5 year
open-label extension study, sustained efficacy was observed
with etanercept and erosive change was clearly retarded.
Furthermore, many patients were able to discontinue MTX
and corticosteroid therapy.
51
These findings support prompt
treatment of early aggressive RA.
Approval for the use of etanercept in psoriasis was based on
data from two large phase III trials involving over 1200
patients. Similar results were obtained from both studies, with
almost half of the patients on etanercept achieving a PASI 75
response compared to 4% on placebo. With extended etanercept
therapy, clinical improvement was sustained and patient-
reported outcomes reflected a positive effect on QoL.
52 53
For
PsA, 59% of etanercept-treated patients achieved an ACR20
response compared to 15% on placebo at 12 weeks.
Improvement was sustained with maintenance therapy at
1 year and radiographic progression of disease was halted.
54
Approval for etanercept has also been given for JIA and AS
following positive results in RCTs. Pilot studies have also shown
benefit in many other IMIDs (table 1).
Adalimumab
Adalimumab (Humira, Abbott), a fully human anti-TNFa
monoclonal antibody, is licensed for clinical use in RA, PsA
and AS. NICE appraisals are in development for all these
indications. The drug is administered as a 40 mg subcutaneous
injection every other week.
Adalimumab has been assessed in over 2000 RA patients in
RCTs. In the ARMADA trial, patients taking adalimumab plus
MTX showed significant improvements in all ACR responder
indices at 6 months, with effects seen as early as the first
week.
55
In a similar 12 month study, clinical benefit was
accompanied by marked improvements in radiographic and
functional outcomes.
56
Evidence supporting the use of adali-
mumab as monotherapy has also been demonstrated.
57
Approval has also been granted for adalimumab as first-line
therapy for early progressive RA based on data from the
PREMIER study. Although clinical responses were comparable
when either adalimumab and MTX were used alone, impressive
results were seen when the two drugs were combined. Nearly
50% of patients achieved clinical remission, rates approximately
double those found among those receiving monotherapy.
Furthermore, three-quarters of the patients on combination
therapy showed no radiographic progression of disease for
2 years.
58
Again, these data support the notion that early
treatment of RA may prevent long-term disability.
In a 6 month study of PsA, patients treated with adalimumab
showed a dramatic improvement in all arthritis severity scores
in addition to a reduction in joint damage progression and an
improvement in QoL. Among the adalimumab-treated patients
with plaque psoriasis, 59% achieved a PASI 75 response,
compared with 1% on placebo.
59
Phase III trials for psoriasis are
in progress.
For CD, limited published data indicate that adalimumab is
safe and effective for inducing remission in up to 36% of
patients.
60
The efficacy of adalimumab has also been demon-
strated in AS and phase III clinical trials are underway in UC
and JIA (table 1).
Rituximab
Rituximab (Mabthera, Roche; Rituxan, Genentech/Biogen) is a
chimeric anti-CD20 monoclonal antibody of human and mouse
origin that was initially approved for treatment of non-
Hodgkin’s B cell lymphoma. The drug has since been
investigated in a variety of IMIDs in which B cells have been
suggested to play a role.
Convincing evidence of efficacy comes from trials in RA with
significant benefit being demonstrated when using rituximab
either alone or in combination with MTX or cyclophospha-
mide.
61
The subsequent DANCER trial, involving 645 patients,
confirmed the benefit of rituximab plus MTX and the effect was
independent of glucocorticoids.
62
In another phase III trial, the
REFLEX study, patients with an inadequate response to anti-
TNFa therapy also noted marked clinical improvement with
rituximab therapy.
63
The efficacy was similar to that of the
TNFa inhibitors.
In 2006, the FDA and European Medicines Agency (EMEA)
approved rituximab for the treatment of RA in patients who
have failed anti-TNFa therapy. The recommended dose is
1000 mg given intravenously 2 weeks apart with concomitant
MTX. Although the safest and most effective timing for repeat
treatment has yet to be determined, preliminary data suggest
that the interval will likely be 6–12 months.
The use of rituximab has not been limited to RA. Published
open-label studies have shown efficacy in the treatment of
more than 30 other IMIDs including SLE, dermatomyositis and
Wegener’s granulomatosis (table 1).
Abatacept
Abatacept (Orencia, Bristol Myers Squibb) is a recombinant
fusion protein consisting of the extracellular domain of human
cytotoxic T lymphocyte antigen-4 (CTLA-4) with the Fc domain
of IgG. Classed as a co-stimulation blocker, the drug is designed
to inhibit T cell activation by blocking the interaction between
antigen presenting cells and T cells. This interaction is mediated
Box 1 American College of Rheumatology
response criteria for 20% improvement (ACR20)
20% reduction in the number of tender and swollen joints, plus
20% improvement in 3 of the following 5 parameters:
Physician global assessment of disease
Patient global assessment of disease
Patient assessment of physical function
Patient assessment of pain
Erythrocyte sedimentation rate or C-reactive protein
IMIDs and biologic therapy 255
www.postgradmedj.com
via the CD28-CD80/86 pathway. In 2005, the FDA approved
abatacept for the treatment of moderate to severe RA in
patients who have had an inadequate response to one or more
DMARDs or TNFa antagonists. The recommended dose is
10 mg/kg by intravenous infusion at 0, 2 and 4 weeks initially,
and then monthly thereafter.
The observation that the CD28 and CD80/86 ligands are
highly expressed on cells within the rheumatoid synovium
suggests that co-stimulation may play a direct role in the
pathogenesis as well as progression of RA.
64
Endogenous CTLA4
is also expressed on T cells in the rheumatoid joint and is
thought to function as a regulator of T cell activation by
interrupting the CD28 pathway, in addition to stimulating the
release of immunoregulatory cytokines, such as TGFb.
65 66
CTLA4 can also mediate antigen-specific apoptosis of T cells.
67
In the clinical setting, two pivotal efficacy studies have now
been published for RA. In the first, 339 subjects with an
inadequate response to MTX were randomised to receive
abatacept plus MTX or MTX alone. At 1 year, marked clinical
improvements were observed with abatacept, with almost twice
as many patients achieving an ACR20 response compared to
placebo.
68
Striking radiographic improvements have since been
demonstrated in a subsequent phase III study.
69
Abatacept has
also been evaluated in patients with an inadequate response to
anti-TNFa therapy, and again, significant improvements in all
ACR indices were seen with active treatment compared to
placebo.
70
Success with abatacept has been noted in psoriasis, with
early studies showing a clear clinical and biochemical improve-
ment in addition to a quantitative reduction of T cells
infiltrating the skin.
32
Overall, these findings support the view
that T cell activation plays an essential role in the pathogenesis
of IMIDs, and therefore it is likely that abatacept will have great
utility in the treatment of T cell mediated diseases in the future.
Alefacept and efalizumab
Two additional agents have been approved for the treatment of
plaque psoriasis. Efalizumab (Raptiva, Genentech/Xoma), a
humanised monoclonal antibody, binds to the alpha subunit
(CD11a) of leukocyte-function-associated antigen type-1 (LFA-
1) and inhibits the activation of T cells. Alefacept (Amevive,
Biogen/Idec), a fully human LFA-3/IgG1 fusion protein, also
inhibits T cell activation and selectively reduces memory T cells.
These drugs have shown excellent responses in RCTs, with
approximately a quarter of patients achieving 75% improve-
ment in skin lesions within 3 months.
71 72
Alefacept has also
demonstrated efficacy in RA, in combination with MTX, and in
PsA.
73 74
Certolizumab pegol (CDP870)
A new anti-TNFa agent, certolizumab (Cimzia, Nektar),
previously known as CDP870, has recently been submitted to
the FDA and EMEA for appraisal for the treatment of CD.
Certolizumab consists of the Fab fragment of a humanised anti-
TNFa antibody, coupled to polyethylene glycol (PEG). This
produces a drug that can remain in circulation longer and can
be conveniently administered once a month via subcutaneous
injection.
Data from a 12 week phase II clinical trial demonstrated that
certolizumab produced a significant clinical benefit in CD over
placebo from weeks 2 to 10, but not at week 12.
23
In a more
recent trial, however, certolizumab convincingly induced
clinical remission for up to 6 months.
75
Overall, the drug is
well tolerated with a safety profile similar to other anti-TNFa
agents. Not surprisingly, certolizumab has also demonstrated
efficacy in RA with outcomes comparable to that of etanercept
and infliximab.
76
Studies have also begun in psoriasis.
Anakinra
Anakinra (Kineret, Amgen), a recombinant IL1 receptor
antagonist, was approved for use in RA in 2001. In the UK,
however, NICE did not approve its use in RA as it was deemed
to be cost ineffective. To date, RCTs involving nearly 3000
patients have demonstrated efficacy of anakinra in RA, either
as monotherapy or in combination with MTX.
77 78
Rapid
improvements in functional status reflected radiographic
evidence of reduced joint damage.
SAFETY
Few serious adverse events were documented in the clinical
trials of anti-TNFa agents, however post-marketing surveillance
revealed several important complications. Principle among
these was reactivation of latent mycobacterium tuberculosis
(TB) infection as TNFa plays a key role in the integrity of
granumolata.
79
Serious bacterial infections have also been
described, however a recent report from the British Society of
Rheumatology Biologics Register found that the overall risk of
serious infections in RA patients was not increased by anti-
TNFa therapy when compared to those on standard DMARD
therapy. There were however more opportunistic infections,
such as histoplasmosis, and more skin and soft tissue infections
in the cohort on biologic therapy.
80
Additional complications of TNFa blockade include possible
exacerbation of congestive cardiac failure, demyelinating
disease and lymphoproliferative disorders.
81
For RA patients,
the occurrence of lymphoma secondary to biologic therapy is
controversial as there is a two- to threefold increased incidence
of this disease with RA per se. The possible increased risk for
the development of solid malignancy is far from clear. A further
potential side effect of anti-TNFa therapy is the development of
antinuclear antibodies (ANA), although an SLE-like illness is
rare and ANA development alone does not necessitate stopping
therapy.
The tolerability of rituximab and abatacept in clinical trials
has been very favourable with integrated safety data indicating
that adverse events, such as malignancy and serious infections,
are rare. In relation to rituximab, the experience in lymphoma
patients has been vast (.750 000 patients treated) and no
increased risk of TB or opportunistic infection has been noted.
Infusion reactions are common with rituximab, although these
events may be minimised by the use of peri-infusional
corticosteroids.
62
The commonest side effect of anakinra is injection-site
reaction. Although neutropaenia has been noted in clinical
trials with the IL1 blocker, the rate of serious infection is
comparable to control patients. In an RA study comparing the
combined efficacy of anakinra and etanercept, however, no
added benefit was seen and there was an unacceptably high
incidence of serious infections.
82
Thus treatment with con-
current biologic agents is not advised and a warning has been
issued by the regulatory health bodies.
The precise relationship between several of these complica-
tions and biologic therapy is unknown, partly due to under-
reporting but also due to lack of comparison cohorts. It may be
that several of these comorbidities are associated with the
disease itself, or perhaps due to extended exposure to standard
DMARD therapy. Robust safety data will emerge from the
numerous biologic registries established across Europe and the
US.
83
COST
One of the drawbacks of biologic therapy is the expense; the
TNFa antagonists cost approximately US$15–18 000 per patient
per annum in the US and around J10 000 in Europe.
84 85
The
cost of B cell targeted therapy may be less, however further
256 Kuek, Hazleman, O
¨
sto¨r
www.postgradmedj.com
work on the appropriate dosing regimen is needed. Due to the
infrastructure required to manufacture these agents, the prices
are unlikely to plummet following patent expiry. This cost,
however, must be balanced against the significant economic
burden IMIDs can pose to the individual patient and to
society as a whole. Direct annual costs for IMIDs vary
widely from US$791 for psoriasis
86
to US$5822 for RA
87
and
US$12 417 for CD.
88
The indirect costs however, such loss of
productivity and income, may be several times greater as
many IMIDs affect young people subjecting them to chronic
disability.
1
In 1992, the total annual cost of RA to UK society
was over £1 billion, approximately half of which was attri-
buted to loss of productivity.
89
Clearly, those with severe,
debilitating disease consume greater resources than those
less afflicted. In CD for example, it has been estimated that
2% of the patient population accounts for 34% of the medical
costs.
90
CONCLUSIONS
Biologics have revolutionised the treatment of autoimmune
diseases due to their efficacy, speed of onset and tolerability.
The fact that anti-cytokine therapies, such as infliximab, have
been effective in treating multiple orphan inflammatory
conditions confirms that the IMID concept is valid. For the
medical profession, this represents an important breakthrough
in the way we classify pathology. Traditionally, IMIDs are
treated by doctors who specialise in the organ most affected,
however given that CD shares similar cytokine dysregulation to
both RA and psoriasis, these systemic disorders may in the
future be grouped as one disorder.
Although biologics provide a very useful addition to our
therapeutic armamentarium, evidence suggests that they
should not be considered a panacea. Serious adverse events
have been reported and long-term safety data are lacking.
Furthermore, a substantial number of patients demonstrate a
poor response to these agents, confirming that our under-
standing of IMID immunopathogenesis is far from complete.
The current challenge is to identify exactly when to introduce
biologics into the therapeutic algorithm. Traditionally they have
been used in those least likely to respond, that is, those who
have failed multiple DMARDs and still have active disease.
Certainly in rheumatology, the prevailing philosophy is to treat
as early as possible in order to avoid the potential sequelae of
joint destruction and functional loss.
While RCTs have demonstrated efficacy in early RA with the
combination of MTX and TNFa inhibitors, it may be argued
that evidence is lacking to promote this as initial therapy for all
patients. In up to one third of patients with RA, progressive
joint destruction does not develop and anti-TNFa therapy may
be not only costly but unnecessary.
91
Furthermore, combination
conventional DMARD therapy has shown to be very effective in
reducing disease activity in RA.
92–94
There is however evidence
from early RA trials that infliximab, etanercept and adalimu-
mab, when used in combination with MTX, are more effective
in rapidly suppressing disease activity than MTX alone in
patients with aggressive disease diagnosed within the preceding
3 years.
35 49 58
Clearly further studies are required to determine
exactly who and when to treat. If we could identify the patients
most likely to respond, it may be possible to save time and
expense and to avert potential toxic reactions. The field of
pharmacogenetics will address this.
Overall, biologic therapies represent an exciting advance in
the treatment of autoimmune diseases. For millions of patients,
treatment success may translate to rapid suppression of
inflammation, prevention of disability, improved quality of life,
and the goal of complete disease remission something
unthinkable a decade ago.
MULTIPLE CHOICE QUESTIONS (TRUE (T)/FALSE (F),
ANSWERS AFTER THE REFERENCES)
Choose the best of the five options for each question.
1. Which of the following is true regarding anti-TNFa
therapy?
A. Anti-TNFa therapy can be administered both sub-
cutaneously and orally
B. Infliximab and etanercept are of comparable efficacy
in treating Crohn’s disease
C. Etanercept can be used as first line therapy in psoriasis
Summary box
N
Biologic agents are highly effective disease-modifying
medications.
N
Currently licensed biologics target TNFa, interleukin-1, T
cell activation and B cells.
N
Biologic agents improve the signs and symptoms and
quality of life of patients with rheumatoid arthritis,
Crohn’s disease, psoriasis and many orphan inflamma-
tory conditions.
N
Physicians should be vigilant for the potential side-effects
of biologic agents.
N
Treatment should commence early for optimal outcome.
N
IMIDs represent a breakthrough in the way pathology is
classified.
Key references
N
33 Maini R, St Clair EW, Breedveld F, 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;354:1932–
9.
N
49 Bathon JM, Martin RW, Fleischmann RM, et al.A
comparison of etanercept and methotrexate in patients
with early rheumatoid arthritis. N Engl J Med
2000;343:1586–93.
N
58 Breedveld FC, Weisman MH, Kavanaugh AF, et al.
The PREMIER study: a multicenter, randomized, double-
blind clinical trial of combination therapy with adalimu-
mab plus methotrexate versus methotrexate alone or
adalimumab alone in patients with early, aggressive
rheumatoid arthritis who had not had previous metho-
trexate treatment. Arthritis Rheum 2006;54:26–37.
N
62 Emery P, Fleischmann R, Filipowicz-Sosnowska A, et
al. DANCER Study Group. The efficacy and safety of
rituximab in patients with active rheumatoid arthritis
despite methotrexate treatment: results of a phase IIB
randomized, double-blind, placebo-controlled, dose-
ranging trial. Arthritis Rheum 2006;54:1390–400.
N
94 Goekoop-Ruiterman YP,deVries-BouwstraJK,
Allaart CF, et al. Clinical and radiographic outcomes of
four different treatment strategies in patients with early
rheumatoid arthritis (the BeSt study): a randomized,
controlled trial. Arthritis Rheum 2005;52:3381–90.
IMIDs and biologic therapy 257
www.postgradmedj.com
D. D. Adalimumab is administered subcutaneously every
other week
E. E. Only one anti-TNFa agent is licensed in the UK for
the treatment of rheumatoid arthritis
2. Which of the following is true regarding rituximab?
A. It decreases the number of circulating B cells
B. It is licensed for the treatment of rheumatoid arthritis
and juvenile idiopathic arthritis
C. It is licensed in Europe as a second line agent for
patients who have failed methotrexate
D. The efficacy of rituximab is far greater than inflix-
imab in the treatment of rheumatoid arthritis
E. The recommended dose is 1000 mg given intrave-
nously every month.
3. Which of the following are IMIDs?
A. Systemic lupus erythematosus
B. Type 1 diabetes
C. Osteoarthritis
D. Multiple sclerosis
E. Ankylosing spondylitis
4. In clinical trials which of the following have been shown
to be beneficial?
A. Abatacept for rheumatoid arthritis
B. Etanercept for Crohn’s disease
C. Alefacept for psoriasis
D. Anakinra for rheumatoid arthritis
E. Infliximab for ankylosing spondylitis
5. Which of the following is true regarding the safety of
biologics?
A. Infliximab is safer than adalimumab
B. Anti-TNFa treatment is associated with an increased
risk of lung cancer
C. Patients must be screened for tuberculosis before
commencing anti-TNFa therapy
D. Patients commonly have infusion reactions with
rituximab
E. The development of antinuclear antibodies necessi-
tates stopping therapy
Authors’ affiliations
.......................
Annabel Kuek, Brian L Hazleman, Andrew J K O
¨
sto¨r, Rheumatology
Research Unit, Addenbrooke’s Hospital, Cambridge, UK
Financial support was provided by CARE (Cambridge Arthritis Research
Endeavour) Charity.
Competing interests: Dr Annabel Kuek declares no conflicts of interest. Dr
Andrew O
¨
sto¨r and Dr Brian Hazleman receive sponsorship from Schering-
Plough, Wyeth, Abbott and Roche.
REFERENCES
1 Williams JP, Meyers JA. Immune-mediated inflammatory disorders (I.M.I.D. s):
the economic and clinical costs, Am J Manag Care 2002;21(Suppl):S664–81.
2 Davidson A, Diamond B. Autoimmune diseases. N Engl J Med
2001;345:340–50.
3 Brophy S, Pavy S, Lewis P, et al. Inflammatory eye, skin, and bowel disease in
spondyloarthritis: genetic, phenotypic and environmental factors. J Rheumatol
2001;28:2667–73.
4 O’Shea JJ, Ma A, Lipsky P. Cytokines and autoimmunity. Nat Immunol
2002;2:37–45.
5 Niessner M, Volk BA. Altered Th1/Th2 cytokine profiles in the intestinal mucosa
of patients with inflammatory bowel disease as assessed by quantitative reversed
transcribed polymerase chain reaction (RT-PCR). Clin Exp Immunol
1995;101:428–35.
6 Lucey DR, Cerici M, Shearer GM. Type 1 and type 2 cytokine dysregulation in
human infections, neoplastic, and inflammatory diseases. Clin Microbiol Rev
1996;9:532–62.
7 Youinou P, Hillion S, Jamin C, et al. B lymphocytes on the front line of
autoimmunity. Autoimmun Rev 2006;5:215–21.
8 Edwards JC, Szczepanski L, Szechinski J, et al. Efficacy of B-cell-targeted therapy
with rituximab in patients with rheumatoid arthritis. N Engl J Med
2004;350:2572–81.
9 Alamanos Y, Drosos AA. Epidemiology of adult rheumatoid arthritis. Autoimmun
Rev 2005;4:130–6.
10 Scott DL, Symmons DP, Coulton BL, et al. Long-term outcome of treating
rheumatoid arthritis: results after 20 years. Lancet 1987;1:1108–11.
11 Gregersen PK, Silver J, Winchester RJ. The shared epitope hypothesis: an
approach to understanding the molecular genetics of susceptibility to rheumatoid
arthritis. Arthritis Rheum 1987;30:1205–13.
12 Choy EH, Panayi GS. Cytokine pathways and joint inflammation in rheumatoid
arthritis. N Engl J Med 2001;344:907–16.
13 Sugiyama E, Kuroda A, Taki H, et al. Interleukin 10 cooperates with interleukin 4
to suppress inflammatory cytokine production by freshly prepared adherent
rheumatoid synovial cells. J Rheumatol 1995;22:2020–6.
14 van Roon JA, van Roy JL, Duits A, et al. Proinflammatory cytokine production
and cartilage damage due to rheumatoid synovial T helper-1 activation is
inhibited by interleukin-4. Ann Rheum Dis 1995;54:836–40.
15 Edwards JCW, Cambridge G, Abrahams VM. Do self-perpetuating B
lymphocytes drive human autoimmune disease? Immunology 1999;97:188–96.
16 Takemura S, Klimiuk PA, Braun A, et al. T cell activation in rheumatoid synovium
is B cell dependent. J Immunol 2001;167:4710–18.
17 Shanahan F. Crohn’s disease. Lancet 2002;359:62–9.
18 Hanauer SB. Inflammatory bowel disease: epidemiology, pathogenesis, and
therapeutic opportunities. Inflamm Bowel Dis 2006;12(Suppl 1):S3–9.
19 Schwartz DA, Pemberton JH, Sandborn WJ. Diagnosis and treatment of perianal
fistulas in Crohn’s disease. Ann Intern Med 2001;135:906–18.
20 Loftus EV Jr, Schoenfeld P, Sandborn WJ. The epidemiology and natural history
of Crohn’s disease in population–based patient cohorts from North America: a
systematic review. Ailment Pharmacol Ther 2002;16:51–60.
21 Reinecker HC, Steffen M, Witthoeft T, et al. Enhanced secretion of tumour
necrosis factor-alpha, IL-6, and IL-1 beta by isolated lamina propria
mononuclear cells from patients with ulcerative colitis and Crohn’s disease. Clin
Exp Immunol 1993;94:174–81.
22 Fedorak RN, Ganl A, Elson CO, et al. Recombinant human interleukin-10 in the
treatment of patients with mild to moderate active Crohn’s disease.
Gastroenterology 2000;119:1473–82.
23 Schreiber S, Rutgeerts P, Fedorak RN, et al. CDP870 Crohn’s Disease Study
Group. A randomized, placebo-controlled trial of certolizumab pegol (CDP870)
for treatment of Crohn’s disease. Gastroenterology 2005;129:807–18.
24 Sandborn WJ, Hanauer SB, Katz S, et al. Etanercept for active Crohn’s disease:
a randomized, double-blind, placebo-controlled trial. Gastroenterology
2001;121:1088–94.
25 Van den Brande JM, Bratt H, van den Brink GR, et al. Infliximab but not
etanercept induces apoptosis in lamina propria T-lymphocytes from patients with
Crohn’s disease. Gastroenterology 2003;126:1774–85.
26 Gelfand JM, Weinstein R, Porter SB, et al. Prevalence and treatment of psoriasis
in the United Kingdom: a population-based study. Arch Dermatol
2005;141:1537–41.
27 Zachariae H. Prevalence of joint disease in patients with psoriasis: implications
for therapy. Am J Clin Dermatol 2003;4:441–7.
28 Krueger G, Ellis CN. Psoriasis recent advances in understanding its
pathogenesis and treatment. J Am Acad Dermatol 2005;53(Suppl 1):S94–100.
29 Gudjonsson JE, Johnson A, Sigmundsdottir H, et al. Immunopathogenic
mechanisms in psoriasis. Clin Exp Immunol 2004;135:1–8.
30 Veale DJ, Ritchlin C, Fitzgerald O. Immunopathology of psoriasis and psoriatic
arthritis. Ann Rheum Dis 2005;64(Suppl 2):ii26–9.
31 Naldi L, Griffiths CE. Traditional therapies in the management of moderate to
severe chronic plaque psoriasis: an assessment of the benefits and risks.
Br J Dermatol 2005;152:597–615.
32 Abrams JR, Lebwohl MG, Guzzo CA, et al. CTLA4-Ig-mediated blockade of T-
cell costimulation in patients with psoriasis vulgaris. J Clin Invest
1999;103:1243–52.
33 Maini R, St Clair EW, Breedveld F, 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;354:1932–9.
34 Maini RN, Breedveld FC, Kalden JR, et al. ATTRACT Study Group. Sustained
improvement over two years in physical function, structural damage, and signs
and symptoms among patients with rheumatoid arthritis treated with infliximab
and methotrexate. Arthritis Rheum 2004;50:1051–65.
35 St Clair EW, van der Heijde DM, Smolen JS, et al. Active-Controlled Study of
Patients Receiving Infliximab for the Treatment of Rheumatoid Arthritis of Early
Onset Study Group. Combination of infliximab and methotrexate therapy for
early rheumatoid arthritis: a randomized, controlled trial, Arthritis Rheum
2004;50:3432–43.
36 Smolen JS, van der Heijde DM, St Clair EW, et al. ASPIRE Study Group.
Predictors of joint damage in patients with early rheumatoid arthritis treated with
258 Kuek, Hazleman, O
¨
sto¨r
www.postgradmedj.com
high-dose methotrexate with or without concomitant infliximab: results from the
ASPIRE trial, Arthritis Rheum 2006;54:702–10.
37 Gottlieb AB, Evans R, Li S, et al. Infliximab induction therapy for patients with
severe plaque-type psoriasis - a randomized, double-blind, placebo-controlled
trial. J Am Acad Dermatol 2004;51:534–42.
38 Reich K, Nestle FO, Papp K, et al. EXPRESS study investigators. Infliximab
induction and maintenance therapy for moderate-to-severe psoriasis: a phase III,
multicentre, double-blind trial. Lancet 2005;366:1367–74.
39 Antoni CE, Kavanaugh A, Kirkham B, et al. Sustained benefits of infliximab
therapy for dermatologic and articular manifestations of psoriatic arthritis: results
from the infliximab multinational psoriatic arthritis controlled trial (IMPACT).
Arthritis Rheum 2005;52:1227–36.
40 Antoni C, Krueger GG, de Vlam K, et al. IMPACT 2 Trial Investigators. Infliximab
improves signs and symptoms of psoriatic arthritis: results of the IMPACT 2 trial.
Ann Rheum Dis 2005;64:1150–7.
41 Kavanaugh A, Antoni C, Krueger GG, et al. Infliximab improves health related
quality of life and physical function in patients with psoriatic arthritis. Ann Rheum
Dis 2006;64:471–7.
42 Targan SR, Hanauer SB, van Deventer SJ, et al. A short-term study of chimeric
monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease.
Crohn’s Disease cA2 Study Group. N Engl J Med 1997;337:1029–35.
43 Hanauer SB, Feagan BG, Lichtenstein GR, et al. ACCENT I Study Group.
Maintenance infliximab for Crohn’s disease: the ACCENT I randomised trial.
Lancet 2002;359:1541–9.
44 Goboes K, Rutgeerts P, Opdenakker G, et al. Endoscopic and histologic evidence
of persistent mucosal healing and correlation with clinical improvement following
sustained infliximab treatment for Crohn’s disease. Curr Med Res Opin
2005;21:1741–54.
45 Sands BE, Anderson FH, Bernstein CN, et al. Infliximab maintenance therapy for
fistulizing Crohn’s disease. N Engl J Med 2004;350:876–85.
46 Lichtenstein GR, Yan S, Bala M, et al. Infliximab maintenance treatment reduces
hospitalizations, surgeries, and procedures in fistulizing Crohn’s disease. A
randomized, controlled trial. Gastroenterology 2005;128:862–9.
47 Moreland LW, Schiff MH, Baumgartner SW, et al. Etanercept therapy in
rheumatoid arthritis. A randomized, controlled trial. Ann Intern Med
1999;130:478–86.
48 Klaresog L, van der Heijde D, de Jager JP, et al. TEMPO (Trial of Etanercept and
Methotrexate with Radiographic Patient Outcomes) study investigators.
Therapeutic effect of the combination of etanercept and methotrexate compared
with each treatment alone in patients with rheumatoid arthritis: double-blind
randomised controlled trial. Lancet 2004;363:675–81.
49 Bathon JM, Martin RW, Fleischmann RM, et al. A comparison of etanercept and
methotrexate in patients with early rheumatoid arthritis. N Engl J Med
2000;343:1586–93.
50 Genovese MC, Bathon JM, Martin RW, et al. Etanercept versus methotrexate in
patients with early rheumatoid arthritis: two-year radiographic and clinical
outcomes. Arthritis Rheum 2002;46:1143–50.
51 Genovese MC, Bathon JM, Fleischmann RM, et al. Longterm safety, efficacy, and
radiographic outcome with etanercept treatment in patients with early
rheumatoid arthritis. J Rheumatol 2005;32:1232–42.
52 Leonardi CL, Powers JL, Matheson RT, et al. Etanercept Psoriasis Study Group.
Etanercept as monotherapy in patients with psoriasis. N Engl J Med
2003;349:2014–22.
53 Papp KA, Tyring S, Lahfa M, et al. Etanercept Psoriasis Study Group. A global
phase III randomized controlled trial of etanercept in psoriasis: safety, efficacy,
and effect of dose reduction. Br J Dermatol 2005;152:1304–12.
54 Mease PJ, Kivitz AJ, Burch FX, et al. Etanercept treatment of psoriatic arthritis:
safety, efficacy, and effect on disease progression. Arthritis Rheum
2004;50:2264–72.
55 Weinblatt ME, Keystone EC, Furst DE, et al. Adalimumab, a fully human anti-
tumor necrosis factor alpha monoclonal antibody, for the treatment of
rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA
trial. Arthritis Rheum 2003;48:35–45.
56 Keystone ED, Kavanaugh AF, Sharp JT, et al. Radiographic, clinical, and
functional outcomes of treatment with adalimumab (a human anti-tumor necrosis
factor monoclonal antibody) in patients with active rheumatoid arthritis receiving
concomitant methotrexate therapy: a randomized, placebo-controlled, 52-week
trial. Arthritis Rheum 2004;50:1400–11.
57 van der Putte LB, Atkins C, Malaise M, et al. Efficacy and safety of adalimumab
as monotherapy in patients with rheumatoid arthritis for whom previous disease
modifying antirheumatic drug treatment has failed. Ann Rheum Dis
2004;63:508–16.
58 Breedveld FC, Weisman MH, Kavanaugh AF, et al. The PREMIER study: a
multicenter, randomized, double-blind clinical trial of combination therapy with
adalimumab plus methotrexate versus methotrexate alone or adalimumab alone
in patients with early, aggressive rheumatoid arthritis who had not had previous
methotrexate treatment. Arthritis Rheum 2006;54:26–37.
59 Mease PJ, Gladman DD, Ritchlin CT, et al. Adalimumab effectiveness in Psoriatic
Arthritis Trial Study Group. Adalimumab for the treatment of patients with
moderately to severely active psoriatic arthritis: results of a double-blind,
randomized, placebo-controlled trial. Arthritis Rheum 2005;52:3279–89.
60 Hanauer SB, Sandborn QJ, Rutgeerts P, et al. Human anti-tumor necrosis factor
monoclonal antibody (adalimumab) in Crohn’s disease: the CLASSIC-I trial.
Gastroenterology 2006;130:323–33.
61 Edwards JC, Szcepanski L, Szechinski J, et al. Efficacy of B-cell-targeted therapy
with rituximab in patients with rheumatoid arthritis. N Engl J Med
2004;350:2572–81.
62 Emery P, Fleischmann R, Filipowicz-Sosnowska A, et al. DANCER Study Group.
The efficacy and safety of rituximab in patients with active rheumatoid arthritis
despite methotrexate treatment: results of a phase IIB randomized, double-
blind, placebo-controlled, dose-ranging trial. Arthritis Rheum
2006;54:1390–400.
63 Cohen SB, Greenwald M, Dougados MR, et al. Efficacy and safety of rituximab in
active RA patients who experienced an inadequate response to one or more anti-
TNFa therapies (REFLEX Study). Arthritis Rheum 2005;52(Suppl):S677.
64 Liu MF, Kohsaka H, Sakurai H, et al. The presence of costimulatory molecules
CD86 and CD28 in rheumatoid arthritis synovium. Arthritis Rheum
1996;39:110–14.
65 Verwilghen J, Lovis R, De Boer M, et al. Expression of functional B7 and CTLA4
on rheumatoid synovial T cells. J Immunol 1994;153:1378–85.
66 Salomon B, Bluestone JA. Complexities of CD28/B7: CTLA-4 costimulatory
pathways in autoimmunity and transplantation. Annu Rev Immunol
2001;19:225–52.
67 Gribben JG, Freeman GJ, Boussiotis VA, et al. CTLA4 mediates antigen-specific
apoptosis of human T cells. Proc Natl Acad Sci U S A 1995;92:811–15.
68 Kremer JM, Dougados M, Emery P, et al. Treatment of rheumatoid arthritis with
the selective costimulation modulator abatacept: twelve-month results of a phase
IIb, double-blind, randomized, placebo-controlled trial. Arthritis Rheum
2005;52:2263–71.
69 Kremer JM, Genant HK, Moreland LW, et al. Effects of abatacept in patients with
methotrexate-resistant active rheumatoid arthritis: a randomized trial. Ann Intern
Med 2006;144:865–76.
70 Genovese MC, Becker JC, Schiff M, et al. Abatacept for rheumatoid arthritis
refractory to tumor necrosis factor alpha inhibition. N Engl J Med
2005;353:1114–23.
71 Gordon KB, Papp KA, Hamilton TK, et al. Efalizumab Study Group. Efalizumab
for patients with moderate to severe plaque psoriasis: a randomized controlled
trial, JAMA 2003;290:3073–80.
72 Krueger GG, Papp KA, Stough DB, et al. Alefacept Clinical Study Group. A
randomized, double-blind, placebo-controlled phase III study evaluating efficacy
and tolerability of 2 courses of alefacept in patients with chronic plaque psoriasis.
J Am Acad Dermatol 2002;47:821–33.
73 Schneider M, Stahl H, Scaramucci J, et al. Alefacept in subjects with active
rheumatoid arthritis (abstract 1709). Presented at the American College of
Rheumatology Annual Scientific Meeting, Orlando, FL, October 23–28, 2003.
74 Mease PJ, Gladman DD, Keystone EC. Alefacept in combination with
methotrexate for the treatment of psoriatic arthritis: results of a randomized,
double-blind, placebo-controlled study. Arthritis Rheum 2006;54:1638–45.
75 Schreiber S. Certolizumab pegol, a humanised anti-TNF pegylated Fab’
fragment, is safe and effective in the maintenance of response and remission
following induction in active Crohn’s disease: a phase 3 study (PRECISE) (abstract
OP-G-355). Presented at the 13th United European Gastroenterology Week,
Copenhagen, October 15–19, 2005.
76 Choy EH, Hazleman B, Smith M, et al. Efficacy of a novel PEGylated humanized
anti-TNF fragment (CDP870) in patients with rheumatoid arthritis: a phaseII
double-blinded, randomized, dose-escalating trial. Rheumatology
2002;41:1133–7.
77 Bresnihan B, Alvaro-Gracia JM, Cobby M, et al. Treatment of rheumatoid
arthritis with recombinant human interleukin-1 receptor antagonist. Arthritis
Rheum 1998;41:2196–204.
78 Fleischmann RM, Schechtman J, Bennett R, et al. Anakinra, a recombinant
human interleukin-1 receptor antagonist (r-metHuIl-1ra), in patients with
rheumatoid arthritis: a large, international, multicenter, placebo-controlled trial.
Arthritis Rheum 2003;48:927–34.
79 Gardam MA, Keystone EC, Menzies R, et al. Anti-tumour necrosis factor agents
and tuberculosis risk: mechanisms of action and clinical management. Lancet
Infect Dis 2003;3:148–55.
80 Dixon WG, Watson K, Lunt M, et al. Rates of serious infection, including site-
specific and bacterial intracellular infection, in rheumatoid arthritis patients
receiving anti-tumor necrosis factor therapy: results from the British Society for
Rheumatology Biologics Register. Arthritis Rheum 2006;54:2368–76.
81 Hyrich KL, Silman AJ, Watson KD, et al. Anti-tumour necrosis factor alpha
therapy in rheumatoid arthritis: an update on safety. Ann Rheum Dis
2004;63:1538–43.
82 Genovese MC, Cohen S, Moreland L, et al. 20000223 Study Group.
Combination therapy with etanercept and anakinra in the treatment of patients
with rheumatoid arthritis who have been treated unsuccessfully with
methotrexate. Arthritis Rheum 2004;50:1412–19.
83 Hyrich KL. Assessing the safety of biologic therapies in rheumatoid arthritis: the
challenges of study design. J Rheumatol Suppl 2005;72:48–50.
84 Kavanaugh A. Health economics: implications for novel antirheumatic therapies.
Ann Rheum Dis 2005;64(Suppl 4):iv65–69.
85 Kobelt G, Jonsson L, Young A, et al. The cost-effectiveness of infliximab
(Remicade) in the treatment of rheumatoid arthritis in Sweden and the United
Kingdom based on the ATTRACT study. Rheumatology 2003;42:326–35.
86 Javitz HS, Ward MM, Farber E, et al. The direct cost of care for psoriasis and
psoriatic arthritis in the United States. J Am Acad Dermatol 2002;46:850–60.
87 Cooper NJ. Economic burden of rheumatoid arthritis: a systematic review.
Rheumatology 2000;39:28–33.
88 Bodger K. Cost of illness of Crohn’s disease. Pharmacoeconomics
2002;20:639–52.
89 McIntosh E. The cost of rheumatoid arthritis. Br J Rheumatol 1996;35:781–90.
90 Hay JW, Hay AR. Inflammatory bowel disease: costs-of-illness. J Clin
Gastroenterol 1992;14:309–17.
91 Brook A, Corbett M. Radiographic changes in early rheumatoid disease. Ann
Rheum Dis 1977;36:71–3.
IMIDs and biologic therapy 259
www.postgradmedj.com
92 Boers M, Verhoeven AC, Markusse HM, et al. Randomised comparison of
combined step-down prednisolone, methotrexate and sulphasalazine with
sulphasalazine alone in early rheumatoid arthritis. Lancet 1997;350:309–18.
93 Mottonen T, Hannonen P, Leirisalo-Repo M, et al. Comparison of combination
therapy with single-drug therapy in early rheumatoid arthritis: a randomised
trial. FIN-RACo trial group. Lancet 1999;353:1568–73.
94 Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, et al. Clinical and
radiographic outcomes of four different treatment strategies in patients with early
rheumatoid arthritis (the BeSt study): a randomized, controlled trial. Arthritis
Rheum 2005;52:3381–90.
ANSWERS
1. (A) F, (B) F, (C) F, (D) T, (E) F.
2. (A) T, (B) F, (C) F, (D) F, (E) F.
3. (A) T, (B) T, (C) F, (D) T, (E) T.
4. (A) T, (B) F, (C) T, (D) T, (E) T.
5. (A) F, (B) F, (C) T, (D) T, (E) F.
BMJ Clinical EvidenceCall for contributors
BMJ Clinical Evidence is a continuously updated evidence-based journal available worldwide on
the internet which publishes commissioned systematic reviews. BMJ Clinical Evidence needs to
recruit new contributors. Contributors are healthcare professionals or epidemiologists with
experience in evidence-based medicine, with the ability to write in a concise and structured way
and relevant clinical expertise.
Areas for which we are currently seeking contributors:
N
Secondary prevention of ischaemic cardiac events
N
Acute myocardial infarction
N
MRSA (treatment)
N
Bacterial conjunctivitis
However, we are always looking for contributors, so do not let this list discourage you.
Being a contributor involves:
N
Selecting from a validated, screened search (performed by in-house Information Specialists)
valid studies for inclusion.
N
Documenting your decisions about which studies to include on an inclusion and exclusion form,
which we will publish.
N
Writing the text to a highly structured template (about 15003000 words), using evidence from
the final studies chosen, within 810 weeks of receiving the literature search.
N
Working with BMJ Clinical Evidence editors to ensure that the final text meets quality and style
standards.
N
Updating the text every 12 months using any new, sound evidence that becomes available. The
BMJ Clinical Evidence in-house team will conduct the searches for contributors; your task is to
filter out high quality studies and incorporate them into the existing text.
N
To expand the review to include a new question about once every 12 months.
In return, contributors will see their work published in a highly-rewarded peer-reviewed
international medical journal. They also receive a small honorarium for their efforts.
If you would like to become a contributor for BMJ Clinical Evidence or require more information
about what this involves please send your contact details and a copy of your CV, clearly stating the
clinical area you are interested in, to CECommissioning@bmjgroup.com.
Call for peer reviewers
BMJ Clinical Evidence also needs to recruit new peer reviewers specifically with an interest in the
clinical areas stated above, and also others related to general practice. Peer reviewers are
healthcare professionals or epidemiologists with experience in evidence-based medicine. As a
peer reviewer you would be asked for your views on the clinical relevance, validity and
accessibility of specific reviews within the journal, and their usefulness to the intended audience
(international generalists and healthcare professionals, possibly with limited statistical knowledge).
Reviews are usually 15003000 words in length and we would ask you to review between 25
systematic reviews per year. The peer review process takes place throughout the year, and our
turnaround time for each review is 1014 days. In return peer reviewers receive free access to
BMJ Clinical Evidence for 3 months for each review.
If you are interested in becoming a peer reviewer for BMJ Clinical Evidence, please complete the
peer review questionnaire at www.clinicalevidence.com/ceweb/contribute/peerreviewer.jsp
260 Kuek, Hazleman, O
¨
sto¨r
www.postgradmedj.com
... Immune-mediated inflammatory diseases (IMIDs) are a diverse set of diseases, including rheumatoid arthritis (RA) and Crohn's disease (CD), with some common molecular targets for treatment. 1,2 Among these molecular targets is tumor necrosis factor-alpha (TNF-α), which has been the focus for the development of several successful disease-modifying biologics. TNF-α biologics, such as adalimumab, have been a key part of the treatment algorithms for RA and CD for several decades. ...
... TNF-α biologics, such as adalimumab, have been a key part of the treatment algorithms for RA and CD for several decades. 1,3 Most biologic therapies for IMIDs, including adalimumab, are delivered subcutaneously via devices such as prefilled syringes or autoinjectors, both of which allow for convenient self-administration by patients. 4,5 Previous studies have reported patient preference for autoinjectors over prefilled syringes, in terms of reduced discomfort, ease of use, and improved adherence. ...
Article
Full-text available
Purpose Medication delivery device design impacts treatment satisfaction, adherence, and compliance in patients receiving biologics. This survey assessed autoinjector attributes that are important to patients, and assessed patient perceptions and preferences between an adalimumab biosimilar autoinjector (Hyrimoz® SensoReady® Pen [SDZ-ADL pen]) and the reference adalimumab autoinjector (Humira® Pen [ref-ADL pen]) in patients with rheumatoid arthritis (RA) or Crohn’s disease (CD) in Canada. Patients and Methods In this survey, adult patients were recruited for web-assisted telephone interviews. Patients had ≥ 3 months’ experience with the ref-ADL pen and 1–12 months’ experience with the SDZ-ADL pen. Results The survey included 120 patients with RA (n = 32) or CD (n = 88). Mean experience with the ref-ADL pen was 7 years for RA or 5 years for CD vs 9 months with the SDZ-ADL pen. The most important autoinjector attributes were the ability to use the pen independently and the ease and simplicity of self-injection. When comparing the two autoinjectors, patients significantly preferred the SDZ-ADL pen over the ref-ADL pen for nearly every attribute evaluated, with the greatest differences reported for visual and audible feedback mechanisms, ease of self-injection, and ability to use the device independently. Overall, 82% of patients preferred the SDZ-ADL pen over the ref-ADL pen, with buttonless activation and less injection pain being the main drivers for this preference. Conclusion Patients with RA or CD indicated a preference for the SDZ-ADL pen over the ref-ADL pen, independent of the duration of use of the pen. The preference for a biosimilar device within 1 year of switching provides reassurance of rapid patient acceptance of biosimilars and may simplify the switching process. These results confirm the importance of ensuring autoinjector design supports independent self-administration of medication and align with previous data showing high patient satisfaction with the SDZ-ADL pen.
... Abnormal levels of inflammation may lead to chronic inflammatory conditions, such as rheumatoid arthritis (RA), IBD, psoriasis, diabetes mellitus, chronic kidney disease, cancer, non-alcoholic fatty liver disease, and autoimmune and neurodegenerative disorders. Therefore, diverse research groups continuously search for molecules with anti-inflammatory responses [25][26][27]. ...
... cancer, non-alcoholic fatty liver disease, and autoimmune and neuro disorders. Therefore, diverse research groups continuously search for mo anti-inflammatory responses [25][26][27]. ...
Article
Full-text available
Background: The inflammasome is a cytosolic multiprotein complex associated with multiple autoimmune diseases. Phytochemical compounds in soy (Glycine max) foods, such as isoflavones, have been reported for their anti-inflammatory properties. Aim: the anti-inflammatory activity of DZ (daidzein) and EQ (equol) were investigated in an ex vivo model of LPS-stimulated murine peritoneal macrophages and by molecular docking correlation. Methods: Cells were pre-treated with DZ (25, 50, and 100 µM) or EQ (5, 10, and 25 µM), followed by LPS stimulation. The levels of PGE2, NO, TNF-α, IL-6, and IL-1β were analyzed by ELISA, whereas the expressions of COX-2, iNOS, NLRP3, ASC, caspase 1, and IL-18 were measured by Western blotting. Also, the potential for transcriptional modulation by targeting NF-κB, COX-2, iNOS, NLRP3, ASC, and caspase 1 was investigated by molecular docking. Results: The anti-inflammatory responses observed may be due to the modulation of NF-κB due to the binding of DZ or EQ, which is translated into decreased TNF-α, COX-2, iNOS, NLRP3, and ASC levels. Conclusion: This study establishes that DZ and EQ inhibit LPS-induced inflammatory responses in peritoneal murine macrophages via down-regulation of NO and PGE2 generation, as well as the inhibition of the canonical inflammasome pathway, regulating NLRP3, and consequently decreasing IL-1β and IL-18 activation.
... Immune-mediated inflammatory diseases (IMIDs) affect 3-7% of the developed world's population and include approximately 80 conditions (such as multiple sclerosis, arthritis, inflammatory bowel disease, and psoriasis, amongst others), with increasing prevalence [1]. ...
Article
Full-text available
There is a growing trend among consumers to seek out natural foods and products with natural ingredients. This shift in consumer preferences had a direct impact on both food and pharmaceutical industries, leading to a focus of scientific research and commercial efforts to meet these new demands. The aim of this work is to review recent available scientific data on foods of interest, such as the artichoke, gooseberry, and polygonoideae plants, as well as olive oil and red raspberries. Interestingly, the urgency of solutions to the climate change emergency has brought new attention to by-products of grapevine bunch stem and cane, which have been found to contain bioactive compounds with potential health benefits. There is a pressing need for a faster process of translating scientific knowledge from the laboratory to real-world applications, especially in the face of the increasing societal burden associated with non-communicable diseases (NCDs), environmental crises, the post-pandemic world, and ongoing violent conflicts around the world.
... Immune-mediated inflammatory diseases (IMIDs) affect 3-7% of the developed world's population and include approximately 80 conditions (such as multiple sclerosis, arthritis, inflammatory bowel disease, and psoriasis, amongst others) with increasing prevalence [1]. ...
Preprint
Full-text available
There is a growing trend among consumers to seek out natural foods and products with natural ingredients. This shift in consumer preferences had a direct impact on both food and pharmaceutical industries, leading to a focus of scientific research and commercial efforts to meet these new demands. The aim of this work is to review recent available scientific data on foods of interest, such as artichoke, gooseberry, and polygonoideae plants, as well as olive oil and red raspberries. Interestingly, the urgency of solutions to the climate change emergency has brought new attention to by-products of grapevine bunch stem and cane, which have been found to contain bioactive compounds with potential health benefits. There is a pressing need for a faster process of translating scientific knowledge from the laboratory to real-world applications, especially in the face of the increasing societal burden associated to non-communicable diseases (NCDs), environmental crises, the post-pandemic world, and ongoing violent conflicts around the world.
... About one-quarter of IBD patients have a concomitant IMID, while, in the general population, the total prevalence of these diseases is only about 5-7% [9][10][11]. In IBD patients, IMIDs mostly affect women and people with CD [11]. ...
Article
Full-text available
(1) Background: Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders involving innate and adaptive immune responses. Despite primarily affecting the gut, recent insights highlight systemic implications, expanding our understanding beyond intestinal boundaries. (2) Methods: This retrospective multicentric study explored the association of IBD and immune-mediated inflammatory diseases (IMIDs) and the impact of concurrent IMIDs on the course of IBD. Clinical data were collected from consecutive medical records of patients with IBD. For assessing the impact of concurrent IMIDs, a control group of IBD patients without associated IMIDs was considered. (3) Results: Of 6589 IBD patients, 6.8% exhibited concomitant IMIDs. Notably, 79.8% of these patients had an aggressive disease course. Psoriasis, atopic dermatitis, and type 1 diabetes mellitus prevalence were lower in the IBD population than in the general population. Conversely, multiple sclerosis, primary sclerosing cholangitis, and pyoderma gangrenosum were more prevalent in IBD patients. Among the patients with a concomitant IMID, 79.8% had an aggressive disease course vs. 8.1% in the control group (p < 0.001). (4) Conclusions: This study underscores the frequency of IMIDs in IBD patients and their association with a more aggressive disease course. The recognition of concurrent IMIDs is crucial for comprehensive patient management, influencing therapeutic decisions and potentially improving outcomes.
Article
As chronic autoimmune inflammatory diseases, rheumatoid arthritis (RA) and Crohn disease (CD) are closely associated and display a significant positive correlation. However, the underlying mechanisms and disease markers responsible for their cooccurrence remain unknown and have not been systematically studied. Therefore, this study aimed to identify key molecules and pathways commonly involved in both RA and CD through bioinformatic analysis of public sequencing databases. Datasets for RA and CD were downloaded from the GEO database. Overlapping genes were identified using weighted gene co-expression network analysis and differential analysis crossover, and enrichment analysis was conducted for these genes. Protein-protein interaction networks were then constructed using these overlapping genes to identify hub genes. Expression validation and receiver operating characteristic curve validation were performed for these hub genes using different datasets. Additionally, the immune cell correlation, single-cell expression cluster, and the immune cell expression cluster of the core gene were analyzed. Furthermore, upstream shared microRNAs (miRNA) were predicted and a miRNA-gene network was constructed. Finally, drug candidates were analyzed and predicted. These core genes were found to be positively correlated with multiple immune cells that are infiltrated by the disease. Analysis of gene expression clusters revealed that these genes were mostly associated with inflammatory and immune responses. The miRNA-genes network analysis suggested that hsa-miR-31-5p may play an important role in the common mechanism of RA and CD. Finally, tamibarotene, retinoic acid, and benzo[a]pyrene were identified as potential treatment options for patients with both RA and CD. This bioinformatics study has identified ITGB2, LCP2, and PLEK as key diagnostic genes in patients with both RA and CD. The study has further confirmed that inflammation and immune response play a central role in the development of both RA and CD. Interestingly, the study has highlighted hsa-miR-31-5p as a potential key player in the common mechanism of both diseases, representing a new direction in research and a potential therapeutic target. These shared genes, potential mechanisms, and regulatory networks offer new opportunities for further research and may provide hope for future treatment of patients with both RA and CD.
Article
Full-text available
This observational study evaluated the impact of a sponsor company-provided Patient Support Program (PSP) on discontinuation of adalimumab in adult Australian patients eligible for Pharmaceutical Benefit Scheme (PBS)-reimbursed adalimumab for Rheumatoid Arthritis (RA), Ankylosing Spondylitis (AS), Psoriatic Arthritis (PsA), Crohn’s Disease (CD), Ulcerative Colitis (UC), or Hidradenitis Suppurativa (HS). Patients initiating adalimumab between May 2018 and September 2019 were enrolled into two prospective cohorts based on their decision to opt for or decline the PSP (PSP or non-PSP cohorts). In addition, a historical, retrospective Non-PSP cohort was established from the Services Australia 10% PBS dataset by extracting data of patients initiating adalimumab prior to the introduction of adalimumab PSPs and based on adalimumab PBS listing dates (AS: April 2007 to March 2009; PsA/RA: January 2007 to December 2008; CD: January 2009 to December 2010; HS and UC indications not included). Follow-up for all cohorts was 12 months. The primary endpoint was the time to discontinuation, compared between the prospective PSP cohort and the prospective or retrospective Non-PSP cohort. Inverse probability of treatment weighting was used to balance the cohorts. A Cox proportional hazards model indicated no difference in time to discontinuation between the prospective PSP (n = 162) and non-PSP (n = 65) cohorts (HR [95% CI] = 1.256 [0.616–2.563], p = 0.5304). The 12-month adalimumab persistence rates (95% CI) were 78% (69%, 84%) and 82% (67%, 90%), respectively. In contrast, discontinuation was less likely in the prospective PSP (n = 151) compared with the retrospective non-PSP (n = 297) cohort (HR [95% CI] = 0.44 [0.28–0.68], p<0.001). The 12-month persistence rates (95% CI) were 81% (76%, 90%) and 61% (56%, 67%), respectively. Overall, this study suggests that optimal adalimumab persistence can be achieved with either a structured PSP or healthcare support from other sources, but this was not the case more than a decade ago.
Article
Background The aim of this study was to investigate the association between peri‐implant diseases and systemic inflammation assessed by serum C‐reactive protein (CRP) levels in a sample of patients with hypertension. Methods A total of 151 participants with hypertension were included in a cross‐sectional study. The population was divided into six groups according to their peri‐implant and periodontal status (healthy controls, mucositis, peri‐implantitis, periodontitis, periodontitis and mucositis, periodontitis, and peri‐implantitis). Linear, logistic regression, and correlation analyses were performed. Results CRP levels were statistically significantly higher in participants with periodontitis alone (median 3.2 mg/L, interquartile range [IQR] 1.8, p = 0.012), combined with mucositis (3.10 mg/L, IQR 2.35, p < 0.001) or peri‐implantitis (2.7 mg/L, IQR 2.53, p = 0.002) when compared to the healthy controls (1 mg/L, IQR 1.2). This association was independent of age, sex, smoking status, and adiposity differences. Participants with periodontitis with and without peri‐implant diseases had the greatest odds of exhibiting CRP > 3 mg/L (odds ratio = 7.3, 95% confidence interval 1.6–33.9). Conclusions Peri‐implant diseases are associated with systemic inflammation, but the nature of the association should be further investigated.
Article
Background: The rise of biologic agents has been a major breakthrough in treating immune-mediated inflammatory diseases (IMIDs). However, their high cost underscores the need for strategies to optimize treatment efficiency. Biosimilars offer cost-effective alternatives to biologics. This study aimed to assess biosimilar drug availability's impact on biologic therapy access for IMIDs. Research design and methods: A retrospective observational study in 15 Spanish hospitals analyzed IMID patients (arthropathies, inflammatory bowel disease and psoriasis) initiating biologic therapy with originator or biosimilar drugs (infliximab, etanercept, adalimumab). Time to availability and initiation of biologic therapy were assessed. Results: 267 patients were included, with 58.4% starting on biosimilars. The mean time to availability of the biologic drugs in the hospitals was 15.9 ± 6.7 months, (20.0 ± 12.4 for originator and 11.8 ± 5.2 for biosimilars). Mean time to biologic treatment was 7.7 ± 9.0 years (8.6 ± 8.9 for originators and 7.0 ± 9.0 for biosimilars). Showing statistically significant differences among conditions. Conclusion: The emergence of biosimilar drugs has enhanced market competition and accelerated their adoption into hospitals' therapeutic regimens over original reference drugs. This has significantly improved access to biologic therapy for patients with IMIDs, evidenced by a notable 1.6-year reduction in access time for biosimilar drugs.
Preprint
Full-text available
Introduction The development of certain immune-mediated diseases (IMD) in patients with inflammatory bowel diseases (IBD; Crohn’s disease (CD), ulcerative colitis (UC)) has been linked to treatment of IBD. Hair loss in some patients may be due to immune-mediated alopecia areata (AA). Risk factors and outcomes of AA in patients with IBD have not been previously explored. Methods This was a retrospective, multi-center case-control study. Cases were identified as individuals who developed IBD before AA diagnosis. Controls comprised of those who were never diagnosed with AA and treated contemporaneously. We extracted demographic and IBD treatment history. Severity of Alopecia Tool (SALT) was used to stratify AA severity. AA outcomes and interventions were compared within controls. Results We identified 58 cases and 90 controls. Cases had significantly higher rate of tumor necrosis factor α antagonist (anti-TNF) use compared to controls (40.7% vs. 20.0%, p= 0.006). Both groups had similar IBD disease location, behavior, and related surgery. Majority of cases had endoscopic remission or mild disease activity at AA diagnosis. There was no difference in partial or complete improvement of AA between those who stopped or continued IBD therapy (p=0.57). Those with severe AA were significantly less likely to have complete (0% vs 33.3%, p=0.01) or any improvement (50% vs 84.9%, p=0.02) of AA compared to those with non-severe AA. Discussion Individuals with IBD who later develop AA were more likely to have been on anti-TNF at time of AA onset. Severity of AA was a significant predictor of AA resolution. Fortunately many patients had improvement in their AA despite continuation of IBD therapy.
Article
In the mid-1980s, Mosmann, Coffman, and their colleagues discovered that murine CD4+ helper T-cell clones could be distinguished by the cytokines they synthesized. The isolation of human Th1 and Th2 clones by Romagnani and coworkers in the early 1990s has led to a large number of reports on the effects of Th1 and Th2 on the human immune system. More recently, cells other than CD4+ T cells, including CD8+ T cells, monocytes, NK cells, B cells, eosinophils, mast cells, basophils, and other cells, have been shown to be capable of producing "Th1" and "Th2" cytokines. In this review, we examine the literature on human diseases, using the nomenclature of type 1 (Th1-like) and type 2 (Th2-like) cytokines, which includes all cell types producing these cytokines rather than only CD4+ T cells. Type 1 cytokines include interleukin-2 (IL-2), gamma interferon, IL-12 and tumor necrosis factor beta, while type 2 cytokines include IL-4, IL-5, IL-6, IL-10, and IL-13. In general, type 1 cytokines favor the development of a strong cellular immune response whereas type 2 cytokines favor a strong humoral immune response. Some of these type 1 and type 2 cytokines are cross-regulatory. For example, gamma interferon and IL-12 decrease the levels of type 2 cytokines whereas IL-4 and IL-10 decrease the levels of type 1 cytokines. We use this cytokine perspective to examine human diseases including infections due to viruses, bacteria, parasites, and fungi, as well as selected neoplastic, atopic, rheumatologic, autoimmune, and idiopathic-inflammatory conditions. Clinically, type 1 cytokine-predominant responses should be suspected in any delayed-type hypersensitivity-like granulomatous reactions and in infections with intracellular pathogens, whereas conditions involving hypergammaglobulinemia, increased immunoglobulin E levels, and/or eosinophilia are suggestive of type 2 cytokine-predominant conditions. If this immunologic concept is relevant to human diseases, the potential exists for novel cytokine-based therapies and novel cytokine-directed preventive vaccines for such diseases.
Article
In the mid-1980s, Mosmann, Coffman, and their colleagues discovered that murine CD4+ helper T-cell clones could be distinguished by the cytokines they synthesized. The isolation of human Th1 and Th2 clones by Romagnani and coworkers in the early 1990s has led to a large number of reports on the effects of Th1 and Th2 on the human immune system. More recently, cells other than CD4+ T cells, including CD8+ T cells, monocytes, NK cells, B cells, eosinophils, mast cells, basophils, and other cells, have been shown to be capable of producing "Th1" and "Th2" cytokines. In this review, we examine the literature on human diseases, using the nomenclature of type 1 (Th1-like) and type 2 (Th2-like) cytokines, which includes all cell types producing these cytokines rather than only CD4+ T cells. Type 1 cytokines include interleukin-2 (IL-2), gamma interferon, IL-12 and tumor necrosis factor beta, while type 2 cytokines include IL-4, IL-5, IL-6, IL-10, and IL-13. In general, type 1 cytokines favor the development of a strong cellular immune response whereas type 2 cytokines favor a strong humoral immune response. Some of these type 1 and type 2 cytokines are cross-regulatory. For example, gamma interferon and IL-12 decrease the levels of type 2 cytokines whereas IL-4 and IL-10 decrease the levels of type 1 cytokines. We use this cytokine perspective to examine human diseases including infections due to viruses, bacteria, parasites, and fungi, as well as selected neoplastic, atopic, rheumatologic, autoimmune, and idiopathic-inflammatory conditions. Clinically, type 1 cytokine-predominant responses should be suspected in any delayed-type hypersensitivity-like granulomatous reactions and in infections with intracellular pathogens, whereas conditions involving hypergammaglobulinemia, increased immunoglobulin E levels, and/or eosinophilia are suggestive of type 2 cytokine-predominant conditions. If this immunologic concept is relevant to human diseases, the potential exists for novel cytokine-based therapies and novel cytokine-directed preventive vaccines for such diseases.
Article
Immune-mediated inflammatory disorders (I.M.I.D.s) are a group of diseases that involve an immune response that is inappropriate or excessive, and is caused, signified, or accompanied by dysregulation of the body's normal cytokine milieu. I.M.I.D.s cause acute or chronic inflammatory injury, sometimes severe, in any organ system. Despite strong evidence linking the pathophysiologies and treatments of the diseases that constitute the I.M.I.D. group, providers, payers, employers, and benefits consultants have been slow to adopt the I.M.I.D. concept. As a result, these stakeholders risk underestimating the significant clinical and economic burdens of the I.M.I.D. class. In this review we examine those burdens, specifically analyzing I.M.I.D. prevalence and cost data for a group of large employers. We also describe the scientific rationale for the I.M.I.D. paradigm, examine the cytokine dysregulation that many I.M.I.D.s share, and focus in detail on the pathophysiology of 3 I.M.I.D.s with high morbidity: rheumatoid arthritis, Crohn's disease, and type 1 diabetes mellitus. The review concludes with an evaluation of approved anticytokine I.M.I.D. therapies and those in development.
Article
The original article to which this Erratum refers was published in Arthritis & Rheumatism(2003) 48(1) 35–45 In the article by Weinblatt et al published in the January 2003 issue of Arthritis & Rheumatism (pp 35–45), there was an error in data reported in the first paragraph of the Results section (page 37). The fourth sentence of that paragraph should have read, “Of these 92 rollover patients, 23, 17, and 17 were in the adalimumab 20 mg, 40 mg, and 80 mg groups, respectively, and 35 were in the placebo group.” We regret the error.
Article
Background: The selective co-stimulation modulator abatacept demonstrated efficacy for treating rheumatoid arthritis in early clinical studies. Objective: To evaluate the effects of abatacept in patients with persistent, active rheumatoid arthritis despite methotrexate treatment. Design: One-year, multicenter, randomized, double-blind, placebo-controlled trial (November 2002 to October 2004). Setting: 116 centers worldwide. Patients: 652 patients with active rheumatoid arthritis despite methotrexate treatment. Intervention: Once-monthly infusion of a fixed dose of abatacept, approximately 10 mg/kg of body weight, or placebo. Measurements: Co-primary end points were a 20% improvement in American College of Rheumatology (ACR) response criteria (ACR 20) at 6 months, clinically meaningful improvements in physical function, and change from baseline in joint erosion score at 1 year. Results: Four hundred thirty-three and 219 patients were randomly assigned to abatacept or placebo, respectively, and 385 (89%) and 162 (74%), respectively, completed 1 year of treatment. In a modified intention-to-treat analysis, 6-month ACR 20, ACR 50, and ACR 70 responses were 67.9% for abatacept versus 39.7% for placebo (difference, 28.2 percentage points [95% Cl, 19.8 to 36.7 percentage points]), 39.9% for abatacept versus 16.8% for placebo (difference, 23.0 percentage points [Cl, 15.0 to 31.1 percentage points]), and 19.8% for abatacept versus 6.5% for placebo (difference, 13.3 percentage points [CI, 7.0 to 19.5 percentage points]), respectively. At 1 year, the responses increased to 73.1 % for abatacept versus 39.7% for placebo (difference, 33.4 percentage points [Cl, 25.1 to 41.7 percentage points]), 48.3% for abatacept versus 18.2% for placebo (difference, 30.1 percentage points [Cl, 21.8 to 38.5 percentage points]), and 28.8% for abatacept versus 6.1 % for placebo (difference, 22.7 percentage points [Cl, 15.6 to 29.8 percentage points]), respectively (P < 0.001 for all). Physical function significantly improved in 63.7% versus 39.3% of patients (P< 0.001). At 1 year, abatacept statistically significantly slowed the progression of structural joint damage compared with placebo. Abatacept-treated patients had a similar incidence of adverse events (87.3% vs. 84.0%; difference, 3.3 percentage points [Cl, -2.5 to 9.1 percentage points]) and a higher incidence of prespecified serious infections (2.5% vs. 0.9%; difference, 1.6 percentage points [Cl, -0.3 to 3.6 percentage points]) and infusion reactions (acute, 8.8% vs. 4.1%; difference, 4.7 percentage points [Cl, 0.9 to 8.4 percentage points]; peri-infusional, 24.5% vs. 16.9%; difference, 7.6 percentage points [Cl, 1.2 to 14.0 percentage points]) compared with placebo recipients. Limitations: The study involved only 1 group of patients over 1 year. Conclusions: Abatacept statistically significantly reduced disease activity in patients with rheumatoid arthritis and an inadequate response to methotrexate. Longer treatment in different patient populations is needed to establish its appropriate role in rheumatoid arthritis.
Article
Background: In a phase II study, etanercept (recombinant human tumor necrosis factor receptor [p75]:Fc fusion protein) safely produced rapid, dose-dependent improvement in rheumatoid arthritis over 3 months. Objective: To confirm the benefit of etanercept therapy of longer duration and simplified dosing in patients with rheumatoid arthritis. Design: Randomized, double-blind, placebo-controlled trial with blinded joint assessors. Setting: 13 North American centers. Patients: 234 patients with active rheumatoid arthritis who had an inadequate response to disease-modifying antirheumatic drugs. Intervention: Twice-weekly subcutaneous injections of etanercept, 10 or 25 mg, or placebo for 6 months. Measurements: The primary end points were 20% and 50% improvement in disease activity according to American College of Rheumatology (ACR) responses at 3 and 6 months. Other end points were 70% ACR responses at 3 and 6 months and other measures of disease activity at 3 and 6 months. Results: Etanercept significantly reduced disease activity in a dose-related fashion. At 3 months, 62% of the patients receiving 25 mg of etanercept and 23% of the placebo recipients achieved 20% ACR response (P < 0.001). At 6 months, 59% of the 25-mg group and 11% of the placebo group achieved a 20% ACR response (P < 0.001); 40% and 5%, respectively, achieved a 50% ACR response (P < 0.01). The respective mean percentage reduction in the number of tender and swollen joints at 6 months was 56% and 47% in the 25-mg group and 6% and -7% in the placebo group (P < 0.05). Significantly more etanercept recipients achieved a 70% ACR response, minimal disease status (0 to 5 affected joints), and improved quality of life. Etanercept was well tolerated, with no dose-limiting toxic effects. Conclusions: Etanercept can safely provide rapid, significant, and sustained benefit in patients with active rheumatoid arthritis.