Adverse Cardiovascular Effects of Antirheumatic Drugs: Implications For Clinical Practice and Research

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Current Pharmaceutical Design, 2012, 18, 1543-1555 1543

Adverse Cardiovascular Effects of Antirheumatic Drugs: Implications for Clinical


Practice and Research

Armen Yuri Gasparyan1,*, Lilit Ayvazyan2, Giuseppe Cocco3, and George D. Kitas1,4

1
Department of Rheumatology, Clinical Research Unit, Dudley Group NHS Foundation Trust (A Teaching Trust of University of Bir-
mingham, UK), Russell's Hall Hospital, Dudley, West Midlands, United Kingdom; 2Department of Medical Chemistry, Yerevan State
Medical University, Yerevan, Armenia; 3Medical Practice, Cardiology and Rheumatology Office, Rheinfelden, Switzerland; 4Arthritis
Research UK Epidemiology Unit, University of Manchester, Manchester, United Kingdom

Abstract: Clinical manifestations of most rheumatic diseases have changed over the past few decades, largely due to advances in thera-
pies targeting autoimmune and (auto)inflammatory pathways. Improvements in the management of rheumatic diseases have also now
brought to the fore the issue of comorbidities. It has become evident that the burden of cardiovascular morbidity and mortality is in-
creased in rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and the spondyloarthropathies, amongst other conditions. As a
result, efforts have switched toward investigating the effects of conventional antirheumatic and new biologic agents on inflammation-
induced atherothrombosis. Evidence is accumulating suggesting a beneficial cardiovascular profile of some antirheumatic drugs, such as
methotrexate and hydroxychloroquine, but it also indicates the possibility of a variety of adverse events developing in the short- and
long-term. The aim of this review is to highlight cardiovascular adverse effects of the drugs widely used in the treatment of rheumatic
diseases. The literature search was performed through PubMed, the Cochrane Library, Scopus, and Web of Science databases using the
following terms: “antirheumatic drugs”, “inflammation”, “rheumatic diseases”, “cardiovascular diseases”, “adverse events”, “toxicity”,
“drug design”, and “drug interactions”. Adverse events ranging from infusion-related hypertension and myocardial ischemia, to restric-
tive cardiomyopathy and congestive heart failure have been reported in large trials and case series on most antirheumatic drugs. Clini-
cians should be alert of the wide variety of cardiovascular adverse effects of individual antirheumatic drugs, and should carefully monitor
blood pressure and markers of inflammation, thrombosis, myocardial ischemia, electrolytes, and lipid disturbances while administering
these drugs. Future prospective studies should specifically investigate the cardiovascular safety of most antirheumatic drugs as part of
mono- or combination therapy in relation to different dosage regimens, duration of therapy, age, and gender.
Keywords: Antirheumatic agents, monoclonal antibodies, rheumatic diseases, cardiovascular diseases, inflammation, risk factors, adverse
events.

INTRODUCTION morbidity and mortality in inflammatory rheumatic diseases [5-7].


Clinical manifestations and complications of rheumatic diseases To tackle the issue of cardiovascular comorbidities, a great deal of
have changed over the past few decades, owing to the advances in effort has been invested towards investigating the effects of conven-
diagnostics and discovery of numerous drugs with targeted action tional antirheumatic and new biologic agents on inflammation-
on autoimmune and inflammatory factors implicated in the patho- induced atherothrombosis [8]. Preliminary evidence has accumu-
physiology of these diseases. Subsequently, quality of life, func- lated suggesting a beneficial effect of the pharmacological suppres-
tional activity and long-term outcomes in patients with most in- sion of inflammation on atherosclerotic cardiovascular comorbidity
flammatory rheumatic diseases have greatly improved. In fact, con- [9, 10] and distinguishing methotrexate as a relatively safe antir-
ventional disease-modifying antirheumatic drug (DMARD) therapy heumatic drug and a promising antiatherosclerotic agent [11, 12]. In
has substantially decreased the prevalence of destructive rheuma- a large, prospectively followed cohort of 41,885 patients with RA,
toid arthritis (RA) and inflammatory amyloidosis [1]. Further im- methotrexate monotherapy and even tumor necrosis factor alpha
provements in the course of RA are expected with the advent of antagonists (which are contraindicated in advanced heart failure),
biological agents suppressing synovial and systemic inflammation substantially reduced risk of hospitalization for congestive heart
poorly controlled by DMARDs [2]. Another relevant example is the failure (Rate Ratio [RR] 0.8, 95% Confidence Interval [CI] 0.6-1.0
successful case of colchicine therapy in familial Mediterranean and 0.5, 95%CI 0.2-0.9, respectively) [13]. The list of antirheumatic
fever (FMF), a common autoinflammatory disorder in the Mediter- drugs with pleiotropic effects on atherogenesis in rheumatic dis-
ranean region, which was frequently associated with renal amyloi- eases is rapidly expanding [14], pending confirmation of their im-
dosis and premature death due to the end-stage renal failure before pact on cardiovascular events in large, specifically designed trials.
introducing colchicine for prevention of flares in FMF [3, 4]. In the mean time, data are accumulating on toxicity and diverse,
including cardiovascular, adverse effects of antirheumatic drugs
The changes in the landscape of antirheumatic therapies and [15-18], which are not always reported in research studies and
improvements in the management of rheumatic diseases with asso- sometimes are overlooked in clinical practice due to insufficient
ciated aging of the patient populations have raised the issue of co- awareness of their specific signs. A variety of causes are believed to
morbidities. In particular, it has been shown that accelerated athero- underlie these adverse effects, with polypharmacy, overdosage, and
sclerosis, sharing pathophysiological features with classic inflam- unjustifiably long pharmacotherapy most commonly reported [19-
matory rheumatic diseases, such as RA and systemic lupus erythe- 21].
matosus (SLE), leads to an increased burden of cardiovascular
The aim of this review is to highlight cardiovascular adverse
effects of drugs widely used in the treatment of rheumatic diseases.
*Address correspondence to this author at the Dudley Group NHS Trust,
Russell’s Hall Hospital, North Block, Clinical Research Unit, Dudley, West SEARCH STRATEGY
Midlands, DY1 2HQ, United Kingdom; Tel: +44-1384-244842; Fax: +44-
1384-244808; E-mails: a.gasparyan@gmail.com; We performed a literature search through PubMed, the Coch-
armen.gasparyan@dgh.nhs.uk rane Library, Scopus, and Web of Science databases for English-
language sources using the following terms: “antirheumatic drugs”,

1873-4286/12 $58.00+.00 © 2012 Bentham Science Publishers


1544 Current Pharmaceutical Design, 2012, Vol. 18, No. 11 Gasparyan et al.

“inflammation”, “rheumatic diseases”, “cardiovascular diseases”, on 113 patients with RA treated with infliximab reported on hy-
“adverse events”, “toxicity”, “drug design”, and “drug interac- potension, hypertension, and tachycardia in 3% of cases; these ef-
tions”. Preference was given to the sources published in the past 5 fects resolved within several hours following cessation of the infu-
years. Information on the currently used antirheumatic drugs and sion [48]. Sporadic cases of acute coronary syndromes during in-
their cardiovascular adverse effects was also obtained from the fliximab infusion have also been reported, warning that chest pain
open-access sources of the US Food and Drug Administration and dyspnea during the infusion should be carefully evaluated to
Agency (FDA) [22]. rule out ischemic origin, even in the young patients [49, 50]. Addi-
tionally, case series of life-threatening tachy- and bradyarrhythmias
BIOLOGIC AGENTS and complete heart block developing during or immediately after
Tumor Necrosis Factor (TNF)- blockers infliximab infusion have been published, requiring ECG monitoring
Elucidation of the critical role of TNF- in the development and and vigilance in patients with preexistent arrhythmias [51, 52].
progression of RA and other diseases has substantially altered Moreover, an earlier study on 150 patients with NYHA class III and
treatment strategies in current rheumatology practice [23, 24]. It has IV heart failure revealed that high doses of infliximab (10 mg/kg)
been shown that TNF-, a potent immunoregulatory and proin- can substantially increase the risk of adverse cardiovascular events
flammatory cytokine produced in large quantities by monocytes, in the short-term [53].
macrophages, and T-lymphocytes, activates nuclear factor kappaB It should be acknowledged that there are differences in the level
and other transcription factors of adhesion molecules, proteolytic of evidence on adverse effects of different biologic agents. More
enzymes and other cytokines, and stimulates chemotaxis of neutro- robust is the evidence on the cardiovascular profile of infliximab,
phils, thereby facilitating inflammation in the synovium and in the oldest agent in use. A recent systematic review, largely based on
other tissues [25-27]. Remarkably, the agents targeting the TNF- observational studies on infliximab therapy in RA, found a deterio-
cascade are now recommended for administration even in early RA ration of lipid profile due to anti-TNF- therapies: a statistically
and in conditions with low severity of inflammatory arthritis [28]. significant increase of total cholesterol (TC), low-density lipopro-
Anti-TNF- agents neutralize soluble and/or membrane-bound tein-cholesterol (LDL-C), high-density lipoprotein-cholesterol
TNF, acting as monoclonal antibodies (human-mouse chimeric (HDL-C), and triglycerides (TG) [54].
antibody infliximab and fully humanized antibodies adalimumab, Infliximab-induced immunosuppression may reactivate oppor-
golimumab, and certolizumab-pegol) or soluble receptor (fully tunistic infections (eg, Listeria monocytogenes) causing rare but
humanized TNF receptor-IgG1 fusion protein etanercept) [29]. fatal myocarditis [55]. Infliximab and other anti-TNF- drugs also
Over the past decade, five anti-TNF- agents have been approved alter the clearance of apoptotic cells and production of autoantibod-
in the US and Europe for the treatment of RA: infliximab, etaner- ies, raising levels of anticardiolipin and triggering arterial and ve-
cept, adalimumab, certolizumab-pegol, and golimumab [30]. Some nous thrombogenesis [56]. The risk of thrombotic events is further
of these drugs have also been successfully applied in the treatment increased in those on concomitant glucocorticoid, methotrexate, or
of psoriasis and psoriatic arthritis [31], ankylosing spondylitis [32], COX-2 inhibitor therapy [57].
juvenile idiopathic arthritis [33], inflammatory bowel disease [34, Short-term safety of the subcutaneously administered etanercept
35], non-infectious ocular inflammation, pyoderma gangrenosum, is more favorable than that of infliximab [58]. A placebo-controlled
Behçet disease (BD), and other vasculitides [36-38]. study of a 16-week etanercept therapy in 535 patients with RA re-
Based on the outcomes of numerous experimental and clinical ported a slightly higher incidence of serious cardiovascular events,
studies, TNF- is viewed as a cytokine playing a crucial role in including MI and heart failure, in the etanercept vs. the placebo
rheumatic disease-associated endothelial dysfunction and acceler- group (4.9% vs. 2.9%), occurring on the background of multiple
ated atherosclerosis [39, 40], justifying the use of anti-TNF- cardiovascular risk factors [59].
agents for both suppressing activity of rheumatic diseases and de- Adalimumab, a recombinant humanized IgG1 monoclonal anti-
creasing cardiovascular burden. Most notably, evidence derived body with minimized antigenicity and high affinity to TNF-, has
from observational cohort studies and randomized controlled trials also demonstrated an enhanced profile. Large studies on adalimu-
has accumulated indicating benefits of anti-TNF- therapies in mab therapy in psoriatic arthritis distinguished hypertriglyceridemia
terms of reducing the incidence of MI, heart failure and cerebrovas- (2.3%) [60] and hypertension (0.9%) [61] as most frequent adverse
cular events in RA [41-43]. cardiovascular events. It is, however, suggested that adalimumab
However, despite the optimism over the cardioprotective effects may have a more favorable lipid profile than infliximab, particu-
of anti-TNF- agents, there have also been reports necessitating a larly in the long-term [62].
cautious approach to anti-TNF- therapy in patients with estab- Golimumab is a new fully human anti-TNF- monoclonal anti-
lished cardiovascular disease and mild heart failure (New York body approved by the US FDA for the treatment of RA, psoriatic
Heart Association [NYHA] I and II functional classes) and contra- arthritis, and ankylosing spondylitis in 2009 [63]. Its main advan-
indicating the therapy in those with NYHA III and IV classes [44, tage is the subcutaneous administration once monthly, with efficacy
45]. Large prospective studies are warranted to distinguish determi- and safety comparable to that of other anti-TNF agents [64]. In one
nants of heart failure in RA cohorts exposed to different anti-TNF- of the several recent phase III studies on RA, golimumab demon-
agents. This is particularly important in view of the data from the strated relatively safe profile, and was even considered a first-line
large cohort study on 2,757 patients with RA treated with inflixi- drug for the treatment of methotrexate-naïve patients with early RA
mab, etanercept, or adalimumab indicating that the incidence of [65]. It was also shown to be effective in patients with RA previ-
heart failure over a 3-year follow-up substantially increases in those ously treated with one or more anti-TNF agents [66]. A systematic
with high disease activity, concomitant glucocorticoid or cyclooxy- review of the trials with 1,231 patients with RA treated with go-
genase (COX)-2 inhibitor therapy, while anti-TNF therapy itself limumab proved its efficacy, compared with placebo [67]. When
contributes to the risk non-significantly (adjusted Hazard Ratio safety of golimumab, golimumab plus DMARDs, and placebo plus
[HR] 1.66, 95%CI 0.67-4.1, P=0.28) [46]. DMARDs arms of the recent phase III trials compared, the com-
Factors affecting cardiovascular safety of different anti-TNF- bined incidence of MI, angina, heart failure, hypertension, and car-
agents vary. A prime factor, in this regard, is the route of the drug diac arrest was less frequent in RA patients treated with golimumab
administration, with acute infusion-related reactions constituting alone, though there were no statistically significant differences
rare but sometimes life-threatening events (eg, anaphylactic reac- between the arms [68].
tions due to intravenously administered infliximab) [47]. A study
Adverse Cardiovascular Effects of Antirheumatic Drugs Current Pharmaceutical Design, 2012, Vol. 18, No. 11 1545

Certolizumab-pegol is a novel subcutaneously administered tuximab in combination with cyclophosphamide for the treatment
PEGylated human anti-TNF antibody, consisting of a Fab fragment of refractory lupus, which yielded conflicting results [86, 87]. Even
and lacking Fc region, which yield the prolonged plasma half-life of more uncertain are issues of long-term efficacy of rituximab, par-
2 weeks and enhanced safety in terms of autoimmune reactions. It ticularly in patients with lupus nephritis [88].
has demonstrated clinical efficacy and safety in patients with RA Preliminary longitudinal studies have shown beneficial effects
and Crohn disease [69-71]. In a multicenter phase III trial with of rituximab on lipid profile, surrogate markers of atherosclerosis
more than 600 patients with RA, hypertension was among the most (i.e., common carotid artery intimal-medial thickness) and endothe-
frequently reported adverse events, particularly in those exposed to lial dysfunction assessed by flow-mediated dilation of the brachial
high dose of certolizumab-pegol (2.4% and 3.7% in groups with artery in RA [89, 90]. However, these studies are limited in statisti-
200 mg and 400 mg, respectively) [72]. It was, however, empha- cal power and duration (4-6 months).
sized that these events are transient and closely related to preexis-
tent hypertension. A recent analysis of the results of placebo- Given the crucial role of B-cell-mediated autoimmune reactions
controlled phase III trials proved that certolizumab plus and inconclusive evidence on safety, clinical efficacy, and vascular
methotrexate and certolizumab monotherapy are safer in terms of effects, larger scale and longer prospective studies on rituximab are
cardiovascular events than respective comparators [73]. warranted [91, 92].
Importantly, cardiovascular events frequently occurring during
T-cell Costimulatory Modulation with Abatacept rituximab infusions, such as hypotension (10%) and peripheral
Abatacept is a recombinant, fully human soluble fusion protein, edema (8%) [93], prompted premedication with methylprednisolone
comprised of the cytotoxic T-lymphocyte associated antigen-4 and regulation of the infusion rate [94]. The US FDA issued a
(CTLA-4, CD152) immunoglobulin and a fragment of the Fc do- boxed warning indicating the risk of severe infusion-related cardio-
main of human IgG-1. It is the first in a new class of intravenously vascular events, usually occurring within the first 24 hours. These
administered selective costimulation modulators [74]. The drug may include fatal MI, ventricular fibrillation, other life-threatening
modulates costimulation of T-lymphocytes and disrupts subsequent arrhythmias, and cardiogenic shock [95]. Therefore, rituximab
activation of B-lymphocytes and macrophages, with a resultant should be used with caution in patients with established cardiovas-
decrease of proinflammatory cytokines and complement fixation cular or pulmonary diseases, in whom close cardiovascular moni-
[75]. Abatacept is approved by the US FDA for the treatment of toring is required throughout the infusion at the slowest possible
patients with moderate to severe RA who failed to respond to one rate.
or more DMARDs or anti-TNF agents [76]. Its use is also recom-
mended in methotrexate-naïve patients with early RA and aggres- Interleukin (IL)-1 Inhibitors
sive course of the disease [77], as well as in case of inadequate IL-1 is a proinflammatory cytokine released by activated
response to several biologic agents, including rituximab [78]. A monocytes/macrophages after the cleavage of its amino-terminal
meta-analysis of 7 randomized controlled trials with 2,908 patients region by caspase-1. The cytokine takes part in the inflammatory
showed that it had been efficacious in reducing pain and rheuma- pathways of a wide range of rheumatic diseases by stimulating
toid activity and improving physical activity at 6 and 12 months of matrix metalloproteinases and other destructive enzymes, adhesion
treatment, compared with placebo [79]. molecules, cytokines, and other inflammatory agents in the synovial
The recent integral analysis of the phase III trials on abatacept and cartilaginous tissues [96, 97]. Animal and human studies on
in RA, compared with abatacept plus DMARDs and placebo plus arthritis have also suggested IL-1 involvement in bone resorption
DMARDs, proved its relatively safe cardiovascular profile in terms [98]. Improved understanding of the role of IL-1 in inflammation
of the combined incidence of angina, cardiac arrest, MI, hyperten- has enabled using the IL-1 inhibitors anakinra, rilonacept, and ca-
sion, and heart failure (no significant difference between the arms) nakinumab in several rheumatic diseases.
[68]. Serious adverse events were more likely to develop when Initially, anakinra, a recombinant human IL-1 receptor antago-
other biologic agents were concurrently used [79]. Of mild to mod- nist with binding affinity similar to that of IL-1, was tested in large
erate adverse cardiovascular events, hypertension was the most cohorts of patients with RA, and was approved by the US FDA for
frequent (6.6%) [80]. the treatment of moderate to severe RA unresponsive to DMARDs
in 2001 [99]. Subsequent comparative studies with anti-TNF
CD20-targeted Therapy with Rituximab agents, however, revealed less potent antirheumatoid activity of IL-
Rituximab is a human-murine chimeric monoclonal antibody 1 inhibitors [100] and prompted the search for other clinical condi-
against B-lymphocyte-specific CD20 surface protein that depletes tions where these agents can be more efficacious [101, 102]. As a
these cells and suppresses cytotoxic-antibody and immune-complex result, a series of studies have proved efficacy and relative safety of
mediated reactions. Rituximab therapy holds promise for the treat- IL-1 inhibitors in autoinflammatory disorders, characterized by the
ment of autoimmune rheumatic disorders, such as SLE, RA, an- absence of high-titer autoantibodies and antigen-specific T-cells
tiphospholipid syndrome, Sjögren syndrome, and some vasculitides, and ranging from periodic fever syndromes, such as FMF, BD,
particularly those refractory to conventional antirheumatic drugs Tumour Necrosis Factor Receptor-Associated Periodic Syndrome
[81, 82]. In fact, a recent systematic review of randomized placebo- (TRAPS), Cryopyrin-Associated Periodic Syndrome (CAPS), to
controlled trials with a total of 938 RA patients resistant or intoler- gout, type 2 diabetes, and chronic heart failure [103-109]. Anakinra
ant to DMARDs or anti-TNF agents demonstrated efficacy in terms has been particularly useful in patients with autoinflammatory dis-
of reducing disease activity and safety of rituximab (two infusions orders unresponsive to conventional drug therapies (eg, colchicine
of 1,000 mg each) in combination with methotrexate [83]. Serious in FMF) [110, 111].
adverse events of rituximab were comparable to those of placebo Large trials on anakinra have suggested that this and possibly
over 24-48 weeks of follow-up. other IL-1 inhibitors are much safer than anti-TNF agents, with the
Safety and efficacy of rituximab have also been tested in lupus only common mild and transient injection site reaction developing
patients with or without nephritis enrolled in phase III randomized in 67% of cases [100], and the absence of strong evidence on wors-
controlled trials [84, 85]. Even though serious adverse events in ening heart failure or other cardiovascular events [112]. Actually,
patients treated with rituximab were not more frequent than in the cardiopulmonary arrest [113] and death because of preexistent heart
placebo arm and rituximab had a greater positive effect on lupus- failure [114] were reported in rare cases of anakinra administration
specific antibodies, clinical efficacy of the drug was not superior to in RA.
that of placebo. Several small studies have assessed safety of ri-
1546 Current Pharmaceutical Design, 2012, Vol. 18, No. 11 Gasparyan et al.

On the contrary, a pilot study suggested that anakinra, adminis- also to antiatherogenic and cardioprotective effects [139]. In fact, it
tered in the post-infarction period, prevents adverse cardiac remod- has been shown that methotrexate retards atherogenesis [140, 141]
eling [115]. Another 1-month longitudinal study with RA patients and dampens the heightened cardiometabolic risk in RA [142]. Not
treated with anakinra demonstrated a significant improvement of surprisingly, in a large prospective cohort study with 107,908 pa-
endothelial function, cardiac contractility and diastolic function tients with RA, methotrexate therapy was associated with a signifi-
[116], thus providing a pathophysiological justification of its safety cantly reduced rate of MI (RR 0.81, 95%CI 0.60-1.08) [143]. Addi-
in patients with RA, failing heart, diabetes, and other comorbidities tionally, recent systematic reviews on cardiovascular mortality in
[117]. RA provided solid evidence on cardioprotection due to the long-
Recently, studies on CAPS and FMF have also demonstrated term low-dose methotrexate therapy [12, 144].
the short-term cardiovascular safety of rilonacept, a fusion protein Although numerous reports confirm the efficacy of methotre-
consisting of the extracellular fragments of IL-1 receptor and Fc of xate, the issue of its toxicity and the associated hyperhomocys-
IgG1, and canakinumab, a fully human anti-IL-1 monoclonal anti- teinemia are still subjects of ongoing clinical investigations [145].
body [118, 119]. Both drugs are now approved by the US FDA for Most experts strongly advocate folic acid supplementation on top of
the treatment of CAPS [120-122]. To date, the raise of total choles- methotrexate to treat hyperhomocysteinemia and to decrease car-
terol, triglycerides, HDL-cholesterol, and LDL-cholesterol as a diovascular morbidity and mortality in arthritides [146-148].
result of the anti-inflammatory effect of rilonacept is the only re- Cardiovascular adverse events rarely develop in the course of
ported adverse cardiovascular event of IL-1 blockade (<1%), re- methotrexate therapy. The US FDA reported that about 3% of those
quiring regular monitoring of lipids and, rarely, lipid-lowering ther- on methotrexate presented with pericarditis, pericardial effusion,
apy [123]. hypotension, myocardial ischemia, and thromboembolic events (ie,
cerebral, deep vein, retinal vein thromboses, and pulmonary embo-
IL-6 receptor Inhibitor Tocilizumab
lus) [149]. Methotrexate-induced immunosuppression, presenting
Tocilizumab is a recombinant human monoclonal antibody, with neutropenia and reactivation of opportunistic infections, may
blocking soluble and membrane-bound IL-6 receptors, thereby trigger myocarditis [149]. The risk of adverse effects is especially
suppressing IL-6-induced immune reactions and inflammation, high due to drug-drug interactions and profound immunosuppres-
particularly at the synovial space and the endothelium. The drug sion in the course of combined antirheumatic therapy (eg,
targets hepatic production of acute-phase proinflammatory proteins, methotrexate plus azathioprine, or cyclosporine, or minocycline, or
T-lymphocyte activation, and immunoglobulin synthesis [124]. In infliximab) [150-152].
the US, EU and Japan, it has been approved for combined use with
methotrexate in the treatment of patients with moderate to severe Antimalarial Drugs
RA with inadequate response or intolerance to DMARDs or other Chloroquine and hydroxychloroquine have been used for the
biologic therapies [125, 126]. It may be also useful as monotherapy treatment of autoimmune manifestations in SLE, RA, primary
and for the treatment of other rheumatic diseases such as adult- Sjögren syndrome and some other rheumatic diseases for decades
onset Still disease [127]. [153-156]. Although the mechanisms of action of both agents are
Several recent phase III studies have proved efficacy and safety not fully clarified, it is suggested that the antirheumatic effects
of tocilizumab (4-8 mg/kg monthly) in combination therapy of RA, largely derive from targeting autoantigen processing in macro-
which yielded profound suppression of systemic inflammation and phages and suppressing T-lymphocytes [157]. Antimalarials offer
activity of arthritis, improvement of general health and remission of significant advantages by disrupting complexes between antiphos-
RA during a 6-12 month period [128-131]. pholipid antibodies and 2-glycoprotein I [158] and annexin A5
One of the frequently reported adverse effects of tocilizumab is anticoagulant [159], thereby neutralizing prothrombotic effects of
dyslipidemia with elevation of TC, LDL-C, HDL-C, and triglyc- antiphospholipid antibodies [160]. These agents also enhance lipid
erides [132], which is presumably the result of blocking the IL-6- and glycemic profiles, elasticity of peripheral arteries and systemic
dependent inflammatory pathway and lipid metabolism [133]. In vascular resistance, and reduce the associated cardiovascular risk in
the Tocilizumab in Combination With Traditional DMARD Ther- SLE and RA [161-163]. A recent systematic review provided evi-
apy (TOWARD) study on 1,220 RA patients, 23% of those on to- dence on varying degrees of protection against venous and arterial
cilizumab had elevated total cholesterol as an adverse event [129]. thromboses, bone loss, and dyslipidemia in patients with moderate
Nonetheless, it has been shown that tocilizumab does not adversely SLE treated with antimalarials [164]. The review also evaluated the
affect the atherogenic index (TC/HDL-C ratio), and there has not, outcomes of prospective studies on cardiotoxicity in patients ex-
thus far, been any report on related increase of cardiovascular posed to antimalarials over a period of 7 months – 7.9 years, which
events during short- and long-term follow-ups [134, 135]. yielded no cases of clinically significant adverse events. Likewise,
another study on 85 patients with connective tissue diseases, with-
Importantly, tocilizumab reverses the suppression of the out cardiovascular disease, treated with hydroxychloroquine for at
CYP450 system induced by IL-6 and modulates effects of some least 1 year, demonstrated that the effects of this particular agent on
cardiovascular drugs metabolized through this system (e.g., simvas- the conduction, evaluated by measuring PR and QT intervals and
tatin) [124]. Cardiovascular events can also develop during intrave- heart rate on standard 12-lead electrocardiograms, are within a safe
nous infusion of tocilizumab. Of these, hypertension was reported range [165].
in 1% of cases [124]. The mechanism of this effect is not entirely
clear. Nonetheless, there have been more than 20 case reports on car-
diotoxicity presenting as potentially life-threatening restrictive car-
DISEASE-MODIFYING ANTIRHEUMATIC DRUGS diomyopathy and conduction disorders in patients on prolonged use
Methotrexate (7 months – 25 years) of either chloroquine or hydroxychloroquine
at large cumulative doses (292g - 4380g) [166, 167]. In most cases,
Methotrexate is an analogue of folic acid, acting as an antifolate discontinuation of the therapy allowed improving myocardial re-
agent. Over the past three decades, it has been widely used in the laxation, contractility, and conduction within several months.
treatment of inflammatory arthritides and RA, in particular [136].
Methotrexate is now recognized as the drug of choice for the treat- Cyclosporine
ment of RA [137]. The drug provides immunosuppression and inhi- Cyclosporine is a cyclic polypeptide immunosuppressive agent
bition of neutrophil chemotaxis and synthesis of proinflammatory which binds to cyclophilin molecules and thus inhibits calcineurin,
cytokines [138], which contribute not only to antirheumatoid but a key protein phosphatase involved in T-lymphocyte activation. The
Adverse Cardiovascular Effects of Antirheumatic Drugs Current Pharmaceutical Design, 2012, Vol. 18, No. 11 1547

results of open-label trials have supported its use as a second-line or Azathioprine


alternative drug in severe psoriatic arthritis [168], early severe RA Azathioprine is a purine analogue, which has a certain role in
[169], lupus nephritis [170], and ocular lesions in BD [171]. Due to the maintenance therapy of lupus nephritis [187]. Cardiovascular
neurotoxicity, the drug should be avoided in patients with neu- adverse events of azathioprine include rare cases of angina, renal
rologic manifestations. The long-term use of cyclosporine is limited vein thrombosis, subclavian vein thrombosis [188], hypotension,
by its nephrotoxicity, presenting as glomerular arteriolopathy and and cardiogenic shock [189].
tubulo-interstitial changes with hyperuricemia and electrolyte dis-
turbances, such as hyperkalemia and hypomagnesemia [172]. Cy- Sulfasalazine
closporine is also known as a drug causing thrombotic thrombocy- Sulfasalazine is a folate antagonist primarily used in the treat-
topenic purpura and hemolytic uremic syndrome [173]. Distur- ment of inflammatory bowel disease, as well as in early RA and
bances in glomerular circulation, including vasoconstriction and spondyloarthritides [190, 191]. The drug’s adverse effects include
thrombosis, lead to hypertension (8%-53%) [174] and increase the rare cases of myelosuppression and thrombocytopenia, interfering
risk of arrhythmias, MI, and heart failure [175]. Cardiovascular with antiplatelet and anticoagulant therapies [192].
events may also develop due to the concurrent anticoagulant, potas-
sium-sparing diuretic, statin, calcium channel blocker, amiodarone, COLCHICINE
digoxin, NSAID, and some other drug therapies, all of which re-
Colchicine is an alkaloid derived from Colchicum autumnale
quire close monitoring of blood pressure and kidney functions in a
with a narrow therapeutic window. The drug disrupts polymeriza-
properly equipped facility by doctors experienced in immunosup-
tion of microtubulin in inflammatory cells, predominantly in neu-
pressive therapy [174].
trophils where it accumulates [193]. Its antichemotactic action plays
Leflunomide a crucial role in the treatment of disorders where neutrophils are
target cells (eg, FMF, BD, and gout) [193, 194]. Also, it has been
Leflunomide is an inhibitor of pyrimidine synthesis with a reported that colchicine suppresses inflammatory pathways linked
proven efficacy in RA and psoriatic arthritis [176]. It inhibits pro- to monocytes/macrophages, T-lymphocytes, and provides an antifi-
liferation of CD4+ T-lymphocytes, thereby providing antiinflam- brotic effect, substantially widening its therapeutic indications
matory and immunomodulatory effects. Its efficacy and tolerability [195]. Colchicine is used as first-line therapy in FMF, and in some
are comparable to those of methotrexate and sulfasalazine [177]. cases of gout, pericarditis, and arthritis in BD [193]. Importantly,
Even though preliminary experimental studies suggested some prevention of flares and suppression of subclinical inflammation in
antiatherogenic and antiproliferative properties of leflunomide, FMF by regular 1-2 mg/daily colchicine therapy are believed to
implicated in successful coronary angioplasty with leflunomide- contribute to antiatherosclerotic and cardioprotective effects of the
eluting stents [178], strong evidence is lacking to recommend the drug [10].
drug for cardiovascular risk reduction in rheumatic diseases. On the
contrary, some experts consider leflunomide along with cy- There have been a few reports on colchicine overdosage and
closporine, NSAIDs, and COX-2 inhibitors as drugs worsening poisoning-induced cardiotoxicity [196, 197]. Lethal cases are usu-
cardiovascular risk [179]. ally associated with acute ingestion of colchicine at doses exceed-
ing 0.5 mg/kg [198]. In critical cases, colchicine-induced cardiotox-
The most common cardiovascular adverse effect of leflunomide icity present with hypotension and cardiac arrest [199]. Toxicity,
is hypertension, which was reported in 2.1%-10.6% of cases in however, may develop with even low doses in patients with hepatic
phase II and III trials [180]. A recent cohort study on 325 RA pa- or renal failure and in cases of the concurrent use of drugs interfer-
tients also linked hypertension with leflunomide therapy (HR 1.02, ing with P-glycoprotein (increasing intracellular accumulation of
95%CI 1.00-1.05) [181]. The precise mechanisms of this adverse the drug) and with the cytochrome (CYP) 450 3A4 system (de-
effect remain unknown, although, based on the pharmacokinetics of creasing production of the drug metabolites). In fact, combination
the drug and the pathophysiology of hypertension in RA [182], it is of therapeutic doses of colchicine with cyclosporine, macrolides,
possible to speculate that interactions with concurrently adminis- such as erythromycin and clarithromycin (but not spiramycin), and
tered NSAIDs, sodium and fluid retention, and increased sympa- statins, affecting both P-glycoprotein and CYP450 3A4, was asso-
thetic activity are the major players in leflunomide-induced hyper- ciated with toxicity [200]. Critical pancytopenia may arise in pa-
tension. Blood pressure monitoring and antihypertensive treatment tients on combined treatment with colchicine and macrolides, while
may be needed in some cases (eg, in patients with preexistent hy- myopathy may be a consequence of the combination with lipid-
pertension and combined drug therapy) [183]. lowering drugs (ie, statins and fibrates) [201].
Cyclophosphamide GLUCOCORTICOIDS
Cyclophosphamide is a nitrogen mustard alkylating agent from It has been long known that glucocorticoids exert permissive
the oxazophorine group. It has been successfully used as an induc- effects on other hormones and anti-inflammatory effects, reducing
tion and maintenance therapy for lupus nephritis for many years, vascular permeability and inhibiting proinflammatory cytokines and
though the optimal dosage regimens have not been established adhesion molecules [202]. For more than half a century, these
[153]. In spite of its efficacy in lupus nephritis and systemic vascu- agents have been successfully used in the treatment of cardiovascu-
litides, evidence has emerged indicating the associated high risk of lar and autoimmune disorders to provide anti-inflammatory, immu-
infections, malignancies, and other toxicities, giving, in some in- nosuppressive and, to some extent, cardioprotective effects [203].
stances, preference to the less toxic agents with comparable effi- Evidence has accumulated suggesting benefits of adding low-dose
cacy (eg, mycophenolate mofetil) [184]. glucocorticoids (below 7.5 mg/daily prednisone) to DMARDs in
Rare cases of cardiotoxicity with acute heart failure have been early RA in terms of radiological progression of bone destruction
reported in association with high-dose (50 mg/kg) cyclophos- [204, 205]. A recent meta-analysis of six randomized controlled
phamide therapy in severe autoimmune diseases [185]. High risk of trials with 689 patients with RA, followed for more than 2 years,
cardiotoxicity and acute heart failure in patients on cyclophos- revealed an acceptable safety profile of glucocorticoids compared
phamide requires close monitoring of heart function, with a special with placebo [206]. Nonetheless, glucocorticoids in a dose (mainly
attention to QT dispersion and its prolongation on ECG [186]. above 7.5 mg/daily) and time-dependent (more than 2.7-5 years)
manner increase the incidence of hypertension, hyperglycemia,
1548 Current Pharmaceutical Design, 2012, Vol. 18, No. 11 Gasparyan et al.

diabetes, osteoporosis, and MI throughout various cohorts of rheu- blockers. Cardiovascular safety of both subgroups still warrants
matic patients [207-209]. evaluation in the context of each agent, different age and gender
Importantly, the US-based Rochester population-based study groups, doses of NSAIDs, drug combinations, and duration of the
identified rheumatoid factor seropositivity as a critical contributor treatment.
to the composite risk of MI, heart failure, and cardiovascular mor- An inflammatory polyarthritis inception cohort study with 923
tality in RA patients exposed to glucocorticoids at high cumulative subjects followed from 1990 to 2004 demonstrated an inverse asso-
doses (HR 3.06 vs. 0.85 in patients with and without rheumatoid ciation between NSAIDs (predominantly diclofenac, naproxen, and
factor positivity) [210]. A number of other factors may also in- ibuprofen) and cardiovascular mortality (adjusted Odds Ratio 0.54,
crease the incidence of cardiovascular adverse effects of glucocorti- 95%CI 0.34-0.86) [216]. The majority of other epidemiological
coids, with electrolyte disturbances and fluid retention among the studies, however, indicated the heightened cardiovascular risk asso-
most likely contributors. The role of electrolytes is of critical im- ciated with most nonselective and selective NSAIDs. In a study
portance in case of pulse therapy with glucocorticoids, requiring with 49,711 subjects aged 65 years and older the relative risk of MI
close monitoring of blood electrolytes, electrocardiograms, and within 180 days after the initiation of therapy with nonselective
blood pressure recordings to timely detect various life-threatening (diclofenac, ibuprofen, and naproxen) and selective NSAIDs (ro-
arrhythmias [211]. Rarely, pulse therapy causes supraventricular fecoxib and celecoxib) was the highest for diclofenac and rofecoxib
and ventricular arrhythmias, hypotension, shock, and sudden death and the lowest for naproxen [217]. A nationwide study from Fin-
[212]. land with 33,039 subjects exposed to NSAIDs found a similarly
increased risk of first-time MI within 180 days after the initiation of
NONSTEROIDAL ANTIINFLAMMATORY DRUGS both nonselective and selective agents [218].
(NSAIDS)
Comparative systematic reviews of randomized controlled stud-
NSAIDs form a large group of agents, differing in chemical ies confirmed that diclofenac and rofecoxib have the worst cardio-
structure, pharmacodynamics, and pharmacokynetics, which block vascular safety profiles [219, 220]. Furthermore, these systematic
different isoenzymes of COX and provide antiinflammatory and reviews demonstrated that rofecoxib at either low or high dose tops
analgesic effects in a wide range of rheumatic diseases. Whilst the the list of NSAIDs, with substantially increased risk of thrombotic
blockade of COX-1 brings about a decrease of prostaglandins at events. In fact, the results of the Vioxx Gastrointestinal Outcomes
inflammatory sites and gastrointestinal, renal, and cardiovascular Research (VIGOR) [221] and the Adenomatous Polyp Prevention
adverse events, the blockade of COX-2 is associated with allevia- on Vioxx (APPROVe) trials [222] comparing 50 mg/daily ro-
tion of pain, partly due to reducing levels of prostaglandins in the fecoxib with 1,000 mg/daily naproxen and 25 mg/daily rofecoxib
central nervous system [213]. Mechanisms of adverse events are with placebo, respectively, led to the withdrawal of this COX-2
not fully explored. Yet, it is possible that the imbalance between selective inhibitor from the market, based on the established risk of
by-products of the COX-1 (thromboxane A2) and COX-2 (prosta- thrombotic events.
glandin I2) pathways, with an increase of the former and a decrease
Despite the wealth of evidence on unfavorable cardiovascular
of the latter, may be responsible for vasoconstriction, platelet acti-
profile of most NSAIDs, these agents are still recommended by
vation, subsequent hypertension, and accelerated atherosclerosis
experts for symptomatic treatment of rheumatic diseases after care-
[214]. The inhibition of renal prostaglandins may also lead to the
ful evaluation of the risk/benefit ratio in each individual case (eg, in
sodium retention, peripheral edema, and decompensation of heart
patients without gastrointestinal and cardiovascular risk all cur-
failure [215].
rently marketed NSAIDs can be used, while in those with high
Currently, two subgroups of NSAIDs are marketed: traditional cardiovascular risk - naproxen only) [223].
agents blocking both COX-1 and COX-2 and selective COX-2

Table 1. Cardiovascular Safety Profile of Biologic Agents in Rheumatic Diseases

Drugs Adverse Events

TNF- blockers (infliximab, etanercept, adalimumab, Worsening of heart failure


golimumab, and certolizumab-pegol) Arrhythmias, conduction disorders
Dyslipidemia
Hypertension
Arterial and venous thromboses
Acute coronary syndrome
Cardiomyopathy
Myocarditis

T-cell costimulatory modulator abatacept Hypertension

Anti-CD20 monoclonal antibody rituximab Infusion-related reactions (hypotension, peripheral edema, MI, arrhythmias, and cardiogenic
shock)

IL-1 inhibitor rilonacept Dyslipidemia

IL-6 receptor inhibitor tocilizumab Dyslipidemia


Infusion-related reactions (hypertension)
Adverse Cardiovascular Effects of Antirheumatic Drugs Current Pharmaceutical Design, 2012, Vol. 18, No. 11 1549

Table 2. Cardiovascular Safety Profile of DMARDs

Drugs Adverse Events

Methotrexate Pericarditis
Pericardial effusion
Hypotension
Myocardial ischemia
Arterial and venous thromboses
Pulmonary embolus
Myocarditis

Antimalarials Restrictive cardiomyopathy


Conduction disorders

Cyclosporine Hypertension
Hyperuricemia
Hypokalemia
Hypomagnesemia
Thrombocytopenia

Leflunomide Hypertension
Angina
Thrombocytopenia

Cyclophosphamide Acute heart failure

Azathioprine Hypotension
Angina
Cardiogenic shock
Renal and subclavian veins thromboses

Sulfasalazine Thrombocytopenia

restrictive cardiomyopathy. Cardiotoxicity has also been reported in


CONCLUSION
those treated with colchicine Table 3, the essential drug for target-
The management of most rheumatic diseases has undergone ing activated neutrophils in autoinflammatory disorders, but exhib-
dramatic changes in the recent decades. Better understanding of the iting adverse profile when used in subjects with renal failure or in
autoimmune and autoinflammatory pathways, coupled with ad- those on combined drug therapies.
vances in drug design, has led to more targeted therapies in a vari-
Uncertainties surround the issue of glucocorticoid therapy in
ety of rheumatic diseases. Suppressing diverse mediators of in-
rheumatic diseases. It is particularly unclear which dosage regimens
flammation by DMARDs, biologic agents, and glucocorticoids has
and how long should be used to minimize or avoid events, such as
been shown to be cardioprotective, mostly in retrospective and
hypertension, hyperglycemia, and diabetes Table 3.
short-term prospective studies employing surrogate markers of
atherogenesis. While large prospective studies on the implications Perhaps the most challenging is the symptomatic pain manage-
of most antirheumatic agents on cardiovascular morbidity and mor- ment with NSAIDs Table 3. Preliminary evidence indicates the
tality are still pending, evidence is accumulating suggestive of ad- relative cardiovascular safety of naproxen and strongly contraindi-
verse effects of potent immunosuppressive and anti-inflammatory cates the use of rofecoxib that is no longer marketed due to the well
therapies with biologic agents on lipid profile Table 1. Moreover, documented prothrombotic effects.
some new monoclonal antibodies with proven antirheumatic effi- In conclusion, clinicians should be alert of the wide variety of
cacy may cause infusion-related adverse cardiovascular events, cardiovascular adverse effects of individual antirheumatic drugs,
such as hypotension, peripheral edema, MI, arrhythmias, and car- and should carefully monitor blood pressure and markers of in-
diogenic shock Table 1. flammation, thrombosis, myocardial ischemia, electrolytes, and
For decades, clinicians have been using DMARDs for mono lipids while administering these drugs. Future prospective studies
and combination therapy of rheumatic diseases, which have led to should specifically investigate cardiovascular safety of most antir-
substantial improvements in the quality of life and survival of pa- heumatic drugs as part of mono- or combination therapy in relation
tients with SLE, RA, and systemic vasculitides. These successes, to different dosage regimens, duration of therapy, age, and gender.
however, have come with reports on the “dark” side of DMARD
therapy Table 2. In particular, leflunomide and cyclosporine fre- CONFLICT OF INTEREST
quently cause hypertension, long-term therapy with antimalarials The authors declare that they have no competing interests.
may be associated with very rare but potentially life-threatening
1550 Current Pharmaceutical Design, 2012, Vol. 18, No. 11 Gasparyan et al.

Table 3. Cardiovascular Safety Profile of other Drugs Used in the Treatment of Rheumatic Diseases

Drugs Adverse Events

Colchicine Hypotension and cardiac arrest due to cardiotoxicity

Glucocorticoids Hypertension
Hyperglycemia
Diabetes
Heart failure
Arrhythmias

NSAIDs Hypertension
Decompensated heart failure
Thromboses
Arrhythmias

[15] Vonkeman HE, van de Laar MA. Nonsteroidal anti-inflammatory


ACKNOWLEDGMENT drugs: adverse effects and their prevention. Semin Arthritis Rheum
AYG and GDK thank The Dudley Group NHS Foundation 2010; 39: 294-312.
Trust (A Teaching Trust of the University of Birmingham, UK) for [16] Katchamart W, Trudeau J, Phumethum V, Bombardier C. Efficacy
providing support during the preparation of this review. and toxicity of methotrexate (MTX) monotherapy versus MTX
combination therapy with non-biological disease-modifying antir-
heumatic drugs in rheumatoid arthritis: a systematic review and
REFERENCES meta-analysis. Ann Rheum Dis 2009; 68: 1105-12.
[1] Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet [17] Turnbull J, Harper L. Adverse effects of therapy for ANCA-
2010; 376: 1094-108. associated vasculitis. Best Pract Res Clin Rheumatol 2009; 23:
[2] Møller Døhn U, Boonen A, Hetland ML, et al. Erosive progression 391-401.
is minimal, but erosion healing rare, in patients with rheumatoid ar- [18] Jaksch W, Dejaco C, Schirmer M. 4 years after withdrawal of ro-
thritis treated with adalimumab. A 1 year investigator-initiated fol- fecoxib: where do we stand today? Rheumatol Int 2008; 28: 1187-
low-up study using high-resolution computed tomography as the 95.
primary outcome measure. Ann Rheum Dis 2009; 68: 1585-90. [19] Mangoni AA, Reid DM, Knights KM. Non-steroidal anti-
[3] Onen F. Familial Mediterranean fever. Rheumatol Int 2006; 26: inflammatory drugs and atherothrombotic risk in older patients:
489-96. where do we stand? Age Ageing 2010; 39: 530-3.
[4] Yonem O, Bayraktar Y. Secondary amyloidosis due to FMF. Hepa- [20] Nuki G. Colchicine: its mechanism of action and efficacy in crys-
togastroenterology 2007; 54: 1061-5. tal-induced inflammation. Curr Rheumatol Rep 2008; 10: 218-27.
[5] Schneider S, Schmitt G, Richter W. Prevalence and correlates of [21] Kirwan JR, Bijlsma JW, Boers M, Shea BJ. Effects of glucocorti-
inflammatory arthritis in Germany: data from the First National coids on radiological progression in rheumatoid arthritis. Cochrane
Health Survey. Rheumatol Int 2006; 27: 29-38. Database Syst Rev 2007; (1): CD006356.
[6] John H, Kitas G, Toms T, Goodson N. Cardiovascular co- [22] US Food and Drug Administration. http://www.fda.gov (Accessed
morbidity in early rheumatoid arthritis. Best Pract Res Clin Rheu- April 20, 2011).
matol 2009; 23: 71-82. [23] Feldmann M, Maini RN. Discovery of TNF-alpha as a therapeutic
[7] John H, Toms TE, Kitas GD. Rheumatoid arthritis: is it a coronary target in rheumatoid arthritis: preclinical and clinical studies. Joint
heart disease equivalent? Curr Opin Cardiol 2011; 26: 327-33. Bone Spine 2002; 69: 12-8.
[8] Nurmohamed MT, van Halm VP, Dijkmans BA. Cardiovascular [24] Feldmann M, Maini RN. Anti-TNF therapy, from rationale to stan-
risk profile of antirheumatic agents in patients with osteoarthritis dard of care: what lessons has it taught us? J Immunol 2010; 185:
and rheumatoid arthritis. Drugs 2002; 62: 1599-609. 791-4.
[9] Boyer JF, Cantagrel A, Constantin A. Impact of traditional thera- [25] Ma Y, Pope RM. The role of macrophages in rheumatoid arthritis.
pies and biologics on cardiovascular diseases in rheumatoid arthri- Curr Pharm Des 2005; 11: 569-80.
tis. Curr Vasc Pharmacol 2008; 6: 218-27. [26] Ackermann C, Kavanaugh A. Tumor necrosis factor as a therapeu-
[10] Gasparyan AY, Stavropoulos-Kalinoglou A, Mikhailidis DP, Toms tic target of rheumatologic disease. Expert Opin Ther Targets 2007;
TE, Douglas KM, Kitas GD. The rationale for comparative studies 11: 1369-84.
of accelerated atherosclerosis in rheumatic diseases. Curr Vasc [27] Okamoto H, Hoshi D, Kiire A, Yamanaka H, Kamatani N. Molecu-
Pharmacol 2010; 8: 437-49. lar targets of rheumatoid arthritis. Inflamm Allergy Drug Targets
[11] Ridker PM. Testing the inflammatory hypothesis of atherothrom- 2008; 7: 53-66.
bosis: scientific rationale for the cardiovascular inflammation re- [28] Hyrich KL, Watson KD, Lunt M, Symmons DP. British Society for
duction trial (CIRT). J Thromb Haemost 2009; 7(Suppl 1): 332-9. Rheumatology Biologics Register (BSRBR). Changes in disease
[12] Westlake SL, Colebatch AN, Baird J, et al. The effect of characteristics and response rates among patients in the United
methotrexate on cardiovascular disease in patients with rheumatoid Kingdom starting anti-tumour necrosis factor therapy for rheuma-
arthritis: a systematic literature review. Rheumatology (Oxford) toid arthritis between 2001 and 2008. Rheumatology (Oxford)
2010; 49: 295-307. 2011; 50: 117-23.
[13] Bernatsky S, Hudson M, Suissa S. Anti-rheumatic drug use and [29] Taylor PC. Pharmacology of TNF blockade in rheumatoid arthritis
risk of hospitalization for congestive heart failure in rheumatoid ar- and other chronic inflammatory diseases. Curr Opin Pharmacol
thritis. Rheumatology (Oxford) 2005; 44: 677-80. 2010; 10: 308-15.
[14] Atzeni F, Turiel M, Caporali R, et al. The effect of pharmacologi- [30] Nam JL, Winthrop KL, van Vollenhoven RF, et al. Current evi-
cal therapy on the cardiovascular system of patients with systemic dence for the management of rheumatoid arthritis with biological
rheumatic diseases. Autoimmun Rev 2010; 9: 835-9. disease-modifying antirheumatic drugs: a systematic literature re-
Adverse Cardiovascular Effects of Antirheumatic Drugs Current Pharmaceutical Design, 2012, Vol. 18, No. 11 1551

view informing the EULAR recommendations for the management TNF Therapy Against Congestive Heart Failure (ATTACH) trial.
of RA. Ann Rheum Dis 2010; 69: 976-86. Circulation 2003; 107: 3133-40.
[31] Weger W. Current status and new developments in the treatment of [54] Pollono EN, Lopez-Olivo MA, Lopez JA, Suarez-Almazor ME. A
psoriasis and psoriatic arthritis with biological agents. Br J Phar- systematic review of the effect of TNF-alpha antagonists on lipid
macol 2010; 160: 810-20. profiles in patients with rheumatoid arthritis. Clin Rheumatol 2010;
[32] Goh L, Samanta A. A systematic MEDLINE analysis of therapeu- 29: 947-55.
tic approaches in ankylosing spondylitis. Rheumatol Int 2009; 29: [55] Jessel P, Safdar N, McCune WJ, Saint S, Kaul DR. Clinical prob-
1123-35. lem-solving. Thinking inside the box. N Engl J Med 2010; 363:
[33] Shenoi S, Wallace CA. Tumor necrosis factor inhibitors in the 574-9.
management of juvenile idiopathic arthritis: an evidence-based re- [56] Ferraccioli G, Gremese E. Thrombogenicity of TNF alpha in rheu-
view. Paediatr Drugs 2010; 12: 367-77. matoid arthritis defined through biological probes: TNF alpha
[34] Bosani M, Ardizzone S, Porro GB. Biologic targeting in the treat- blockers. Autoimmun Rev 2004; 3: 261-6.
ment of inflammatory bowel diseases. Biologics 2009; 3: 77-97. [57] Petitpain N, Gambier N, Wahl D, et al. French Network of Phar-
[35] Caprioli F, Pallone F, Monteleone G. Cytokine therapies in Crohn's macovigilance Centers. Arterial and venous thromboembolic
disease: where are we now and where should we go? Inflamm Al- events during anti-TNF therapy: a study of 85 spontaneous reports
lergy Drug Targets 2011; 10: 47-53. in the period 2000-2006. Biomed Mater Eng 2009; 19: 355-64.
[36] Langford CA. Drug insight: anti-tumor necrosis factor therapies for [58] Gisondi P, Girolomoni G. Biologic therapies in psoriasis: a new
the vasculitic diseases. Nat Clin Pract Rheumatol 2008; 4: 364-70. therapeutic approach. Autoimmun Rev 2007; 6: 515-9.
[37] Karampetsou MP, Liossis SN, Sfikakis PP. TNF- antagonists [59] Weisman MH, Paulus HE, Burch FX, et al. A placebo-controlled,
beyond approved indications: stories of success and prospects for randomized, double-blinded study evaluating the safety of etaner-
the future. QJM 2010; 103: 917-28. cept in patients with rheumatoid arthritis and concomitant comor-
[38] Arida A, Fragiadaki K, Giavri E, Sfikakis PP. Anti-TNF Agents for bid diseases. Rheumatology (Oxford) 2007; 46: 1122-5.
Behçet's Disease: Analysis of Published Data on 369 Patients. [60] Gordon KB, Langley RG, Leonardi C, et al. Clinical response to
Semin Arthritis Rheum 2011; 41: 61-70. adalimumab treatment in patients with moderate to severe psoria-
[39] Bacon PA, Stevens RJ, Carruthers DM, Young SP, Kitas GD. sis: double-blind, randomized controlled trial and open-label exten-
Accelerated atherogenesis in autoimmune rheumatic diseases. sion study. J Am Acad Dermatol 2006; 55: 598-606.
Autoimmun Rev 2002; 1: 338-47. [61] Mease PJ, Ory P, Sharp JT, et al. Adalimumab for long-term treat-
[40] McKellar GE, McCarey DW, Sattar N, McInnes IB. Role for TNF ment of psoriatic arthritis: 2-year data from the Adalimumab Effec-
in atherosclerosis? Lessons from autoimmune disease. Nat Rev tiveness in Psoriatic Arthritis Trial (ADEPT). Ann Rheum Dis
Cardiol 2009; 6: 410-7. 2009; 68: 702-9.
[41] Barnabe C, Martin BJ, Ghali WA. Systematic review and meta- [62] Kerekes G, Soltész P, Dér H, et al. Effects of biologics on vascular
analysis: Anti-tumor necrosis factor alpha therapy and cardiovascu- function and atherosclerosis associated with rheumatoid arthritis.
lar events in rheumatoid arthritis. Arthritis Care Res (Hoboken) Ann N Y Acad Sci 2009; 1173: 814-21.
2011; 63: 522-9. [63] Voulgari PV. Golimumab: a new anti-TNF-alpha agent for rheuma-
[42] Westlake SL, Colebatch AN, Baird J, et al. Tumour necrosis factor toid arthritis, psoriatic arthritis and ankylosing spondylitis. Expert
antagonists and the risk of cardiovascular disease in patients with Rev Clin Immunol 2010; 6: 721-33.
rheumatoid arthritis: a systematic literature review. Rheumatology [64] Kay J, Rahman MU. Golimumab: A novel human anti-TNF-alpha
(Oxford) 2011; 50: 518-31. monoclonal antibody for the treatment of rheumatoid arthritis, an-
[43] Cugno M, Ingegnoli F, Gualtierotti R, Fantini F. Potential effect of kylosing spondylitis, and psoriatic arthritis. Core Evid 2010; 4:
anti-tumour necrosis factor-alpha treatment on reducing the cardio- 159-70.
vascular risk related to rheumatoid arthritis. Curr Vasc Pharmacol [65] Emery P, Fleischmann RM, Moreland LW, et al. Golimumab, a
2010; 8: 285-92. human anti-tumor necrosis factor alpha monoclonal antibody, in-
[44] Wolfe F, Michaud K. Heart failure in rheumatoid arthritis: rates, jected subcutaneously every four weeks in methotrexate-naive pa-
predictors, and the effect of anti-tumor necrosis factor therapy. Am tients with active rheumatoid arthritis: twenty-four-week results of
J Med 2004; 116: 305-11. a phase III, multicenter, randomized, double-blind, placebo-
[45] Sarzi-Puttini P, Atzeni F, Shoenfeld Y, Ferraccioli G. TNF-alpha, controlled study of golimumab before methotrexate as first-line
rheumatoid arthritis, and heart failure: a rheumatological dilemma. therapy for early-onset rheumatoid arthritis. Arthritis Rheum 2009;
Autoimmun Rev 2005; 4: 153-61. 60: 2272-83.
[46] Listing J, Strangfeld A, Kekow J, et al. Does tumor necrosis factor [66] Smolen JS, Kay J, Doyle MK, et al. GO-AFTER study investiga-
alpha inhibition promote or prevent heart failure in patients with tors. Golimumab in patients with active rheumatoid arthritis after
rheumatoid arthritis? Arthritis Rheum 2008; 58: 667-77. treatment with tumour necrosis factor alpha inhibitors (GO-AFTER
[47] Maggi E, Vultaggio A, Matucci A. Acute infusion reactions in- study): a multicentre, randomised, double-blind, placebo-
duced by monoclonal antibody therapy. Expert Rev Clin Immunol controlled, phase III trial. Lancet 2009; 374: 210-21.
2011; 7: 55-63. [67] Singh JA, Noorbaloochi S, Singh G. Golimumab for rheumatoid
[48] Wasserman MJ, Weber DA, Guthrie JA, et al. Infusion-related arthritis. Cochrane Database Syst Rev 2010; (1): CD008341.
reactions to infliximab in patients with rheumatoid arthritis in a [68] Storage SS, Agrawal H, Furst DE. Description of the efficacy and
clinical practice setting: relationship to dose, antihistamine pre- safety of three new biologics in the treatment of rheumatoid arthri-
treatment, and infusion number. J Rheumatol 2004; 31: 1912-7. tis. Korean J Intern Med 2010; 25: 1-17.
[49] Abedin M, Scheurich D, Reimold SC, Reimold AM. Acute coro- [69] Duggan ST, Keam SJ. Certolizumab pegol: in rheumatoid arthritis.
nary syndrome after infliximab infusion. Cardiol Rev 2006; 14: 50- BioDrugs 2009; 23: 407-17.
2. [70] Fleischmann R. The clinical efficacy and safety of certolizumab
[50] Panteris V, Perdiou A, Tsirimpis V, Karamanolis DG. Acute coro- pegol in rheumatoid arthritis. Expert Opin Biol Ther 2010; 10: 773-
nary syndrome after infliximab therapy in a patient with Crohn's 86.
disease. World J Gastroenterol 2006; 12: 6235-8. [71] Connock M, Tubeuf S, Malottki K, et al. Certolizumab pegol
[51] Lazzerini PE, Acampa M, Hammoud M, et al. Arrhythmic risk (CIMZIA®) for the treatment of rheumatoid arthritis. Health Tech-
during acute infusion of infliximab: a prospective, single-blind, nol Assess 2010; 14 (Suppl. 2): 1-10.
placebo-controlled, crossover study in patients with chronic arthri- [72] Smolen J, Landewé RB, Mease P, et al. Efficacy and safety of
tis. J Rheumatol 2008; 35: 1958-65. certolizumab pegol plus methotrexate in active rheumatoid arthri-
[52] Sote Y, Green S, Maddison P. Complete heart block after inflixi- tis: the RAPID 2 study. A randomised controlled trial. Ann Rheum
mab therapy. Rheumatology (Oxford) 2008; 47: 227-8. Dis 2009; 68: 797-804.
[53] Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT. Anti- [73] Mease PJ. Certolizumab pegol in the treatment of rheumatoid ar-
TNF Therapy Against Congestive Heart Failure Investigators. Ran- thritis: a comprehensive review of its clinical efficacy and safety.
domized, double-blind, placebo-controlled, pilot trial of infliximab, Rheumatology (Oxford) 2011; 50: 261-70.
a chimeric monoclonal antibody to tumor necrosis factor-alpha, in [74] Hervey PS, Keam SJ. Abatacept. BioDrugs 2006; 20: 53-61.
patients with moderate-to-severe heart failure: results of the anti- [75] Solomon GE. T-cell agents in the treatment of rheumatoid arthritis.
Bull NYU Hosp Jt Dis 2010; 68: 162-5.
1552 Current Pharmaceutical Design, 2012, Vol. 18, No. 11 Gasparyan et al.

[76] Bluestone JA, St Clair EW, Turka LA. CTLA4Ig: bridging the [99] Cohen SB. The use of anakinra, an interleukin-1 receptor antago-
basic immunology with clinical application. Immunity 2006; 24: nist, in the treatment of rheumatoid arthritis. Rheum Dis Clin North
233-8. Am 2004; 30: 365-80.
[77] Schiff M. Abatacept treatment for rheumatoid arthritis. Rheumatol- [100] Thaler K, Chandiramani DV, Hansen RA, Gartlehner G. Efficacy
ogy (Oxford) 2011; 50: 437-49. and safety of anakinra for the treatment of rheumatoid arthritis: an
[78] Mishra R, Singh V, Pritchard CH. Safety of biologic agents after update of the Oregon Drug Effectiveness Review Project. Biologics
rituximab therapy in patients with rheumatoid arthritis. Rheumatol 2009; 3: 485-98.
Int 2011; 31: 481-4. [101] Goldbach-Mansky R. Blocking interleukin-1 in rheumatic diseases.
[79] Maxwell L, Singh JA. Abatacept for rheumatoid arthritis. Cochrane Ann N Y Acad Sci 2009; 1182: 111-23.
Database Syst Rev 2009; (4): CD007277. [102] McDermott MF. Rilonacept in the treatment of chronic inflamma-
[80] Briefing document for abatacept (BMS-188667). Biological license tory disorders. Drugs Today (Barc) 2009; 45: 423-30.
application 125118. 6 September 2005. [103] Simon A, Bodar EJ, van der Hilst JC, et al. Beneficial response to
http://www.fda.gov/ohrms/dockets/ac/05/briefing/2005- interleukin 1 receptor antagonist in traps. Am J Med 2004; 117:
4170B1_01_01-BMS-Abatacept.pdf (Accessed April 25, 2011). 208-10.
[81] Arkfeld DG. The potential utility of B cell-directed biologic ther- [104] Düzgün N, Ayaliolu E, Tutkak H, Aydintu OT. Cytokine inhibi-
apy in autoimmune diseases. Rheumatol Int 2008; 28: 205-15. tors: soluble tumor necrosis factor receptor 1 and interleukin-1 re-
[82] Engel P, Gómez-Puerta JA, Ramos-Casals M, Lozano F, Bosch X. ceptor antagonist in Behçet's disease. Rheumatol Int 2005; 25: 1-5.
Therapeutic Targeting of B Cells for Rheumatic Autoimmune Dis- [105] Rigante D, Ansuini V, Bertoni B, Pugliese AL, Avallone L,
eases. Pharmacol Rev 2011; 63: 127-56. Federico G, Stabile A. Treatment with anakinra in the hyperimmu-
[83] Lee YH, Bae SC, Song GG. The efficacy and safety of rituximab noglobulinemia D/periodic fever syndrome. Rheumatol Int 2006;
for the treatment of active rheumatoid arthritis: a systematic review 27: 97-100.
and meta-analysis of randomized controlled trials. Rheumatol Int [106] Meinzer U, Quartier P, Alexandra JF, Hentgen V, Retornaz F,
2011; 31: 1493-9. Koné-Paut I. Interleukin-1 Targeting Drugs in Familial Mediterra-
[84] Furie R, Looney RJ, Rovin B, et al. Efficacy and Safety of Rituxi- nean Fever: A Case Series and a Review of the Literature. Semin
mab in Subjects with Active Proliferative Lupus Nephritis (LN): Arthritis Rheum 2011; 41: 265-71.
Results From the Randomized, Double-Blind Phase III LUNAR [107] Rigante D, Leone A, Marrocco R, Laino ME, Stabile A. Long-term
Study [abstract]. Arthritis Rheum 2009; 60 (Suppl 10): 1149. response after 6-year treatment with anakinra and onset of focal
[85] Merrill JT, Neuwelt CM, Wallace DJ, et al. Efficacy and safety of bone erosion in neonatal-onset multisystem inflammatory disease
rituximab in moderately-to-severely active systemic lupus erythe- (NOMID/CINCA). Rheumatol Int 2011; 31: 1661-4.
matosus: the randomized, double-blind, phase II/III systemic lupus [108] Dinarello CA. Blocking interleukin-1 in acute and chronic autoin-
erythematosus evaluation of rituximab trial. Arthritis Rheum 2010; flammatory diseases. J Intern Med 2011; 269: 16-28.
62: 222-33. [109] Kuemmerle-Deschner JB, Tyrrell PN, Koetter I, et al. Efficacy and
[86] Smith KG, Jones RB, Burns SM, Jayne DR. Long-term comparison safety of anakinra therapy in pediatric and adult patients with the
of rituximab treatment for refractory systemic lupus erythematosus autoinflammatory Muckle-Wells syndrome. Arthritis Rheum 2011;
and vasculitis: Remission, relapse, and re-treatment. Arthritis 63: 840-9.
Rheum 2006; 54: 2970-82. [110] Botsios C, Sfriso P, Furlan A, Punzi L, Dinarello CA. Resistant
[87] Jones RB, Tervaert JW, Hauser T, et al. European Vasculitis Study Behçet disease responsive to anakinra. Ann Intern Med 2008; 149:
Group. Rituximab versus cyclophosphamide in ANCA-associated 284-6.
renal vasculitis. N Engl J Med 2010; 363: 211-20. [111] Alpay N, Sumnu A, Calıkan Y, Yazıcı H, Türkmen A, Gül A.
[88] Arce-Salinas CA, Rodríguez-García F, Gómez-Vargas JI. Long- Efficacy of anakinra treatment in a patient with colchicine-resistant
term efficacy of anti-CD20 antibodies in refractory lupus nephritis. familial Mediterranean fever. Rheumatol Int 2010; Apr 13 [Epub
Rheumatol Int 2011; Jan 22 [Epub ahead of print]. ahead of print].
[89] Gonzalez-Juanatey C, Llorca J, Vazquez-Rodriguez TR, Diaz- [112] Fleishmann RM. Safety of anakinra, a recombinant interleukin-1
Varela N, Garcia-Quiroga H, Gonzalez-Gay MA. Short-term im- receptor antagonist (r-metHuIL-1ra), in patients with rheumatoid
provement of endothelial function in rituximab-treated rheumatoid arthritis and comparison to anti-TNF-alpha agents. Clin Exp
arthritis patients refractory to tumor necrosis factor alpha blocker Rheumatol 2002; 20 (5 Suppl 27): S35-41.
therapy. Arthritis Rheum 2008; 59: 1821-4. [113] Sharma R, Sharma CL, Mahajan A. Biological agents targeting
[90] Kerekes G, Soltész P, Dér H, et al. Effects of rituximab treatment beyond TNF-alpha. Indian J Crit Care Med 2008; 12: 181-9.
on endothelial dysfunction, carotid atherosclerosis, and lipid profile [114] den Broeder AA, de Jong E, Franssen MJ, Jeurissen ME, Flendrie
in rheumatoid arthritis. Clin Rheumatol 2009; 28: 705-10. M, van den Hoogen FH. Observational study on efficacy, safety,
[91] Looney RJ. B cell-targeted therapies for systemic lupus erythema- and drug survival of anakinra in rheumatoid arthritis patients in
tosus: an update on clinical trial data. Drugs 2010; 70: 529-40. clinical practice. Ann Rheum Dis 2006; 65: 760-2.
[92] Tieng AT, Peeva E. B-cell-directed therapies in systemic lupus [115] Abbate A, Kontos MC, Grizzard JD, et al. VCU-ART Investiga-
erythematosus. Semin Arthritis Rheum 2008; 38: 218-27. tors. Interleukin-1 blockade with anakinra to prevent adverse car-
[93] Gürcan HM, Keskin DB, Stern JN, Nitzberg MA, Shekhani H, diac remodeling after acute myocardial infarction (Virginia Com-
Ahmed AR. A review of the current use of rituximab in autoim- monwealth University Anakinra Remodeling Trial [VCU-ART] Pi-
mune diseases. Int Immunopharmacol 2009; 9: 10-25. lot study). Am J Cardiol 2010; 105: 1371-1377.e1.
[94] Stoll ML, Gotte AC. Biological therapies for the treatment of juve- [116] Ikonomidis I, Tzortzis S, Lekakis J, et al. Lowering interleukin-1
nile idiopathic arthritis: Lessons from the adult and pediatric expe- activity with anakinra improves myocardial deformation in rheu-
riences. Biologics 2008; 2: 229-52. matoid arthritis. Heart 2009; 95: 1502-7.
[95] Rituxan (rituximab) Injection For Intravenous Use. Detailed View: [117] Schiff MH, DiVittorio G, Tesser J, et al. The safety of anakinra in
Safety Labeling Changes Approved By FDA Center for Drug high-risk patients with active rheumatoid arthritis: six-month ob-
Evaluation and Research (CDER) -- January 2008. servations of patients with comorbid conditions. Arthritis Rheum
http://www.fda.gov/Safety/MedWatch/SafetyInformation/Safety- 2004; 50: 1752-60.
RelatedDrugLabelingChanges/ucm106667.htm (Accessed April 25, [118] Hoffman HM, Throne ML, Amar NJ, et al. Efficacy and safety of
2011). rilonacept (interleukin-1 Trap) in patients with cryopyrin-
[96] Pay S, Erdem H, Pekel A, et al. Synovial proinflammatory cytoki- associated periodic syndromes: results from two sequential pla-
nes and their correlation with matrix metalloproteinase-3 expres- cebo-controlled studies. Arthritis Rheum 2008; 58: 2443-52.
sion in Behçet's disease. Does interleukin-1beta play a major role in [119] Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, et al.
Behçet's synovitis? Rheumatol Int 2006; 26: 608-13. Canakinumab in CAPS Study Group. Use of canakinumab in the
[97] Jacques C, Gosset M, Berenbaum F, Gabay C. The role of IL-1 and cryopyrin-associated periodic syndrome. N Engl J Med 2009; 360:
IL-1Ra in joint inflammation and cartilage degradation. Vitam 2416-25.
Horm 2006; 74: 371-403. [120] Church LD, Savic S, McDermott MF. Long term management of
[98] Strand V, Kavanaugh AF. The role of interleukin-1 in bone resorp- patients with cryopyrin-associated periodic syndromes (CAPS): fo-
tion in rheumatoid arthritis. Rheumatology (Oxford) 2004; 43 cus on rilonacept (IL-1 Trap). Biologics 2008; 2: 733-42.
Suppl 3: iii10-iii16. [121] Dhimolea E. Canakinumab. MAbs 2010; 2: 3-13.
Adverse Cardiovascular Effects of Antirheumatic Drugs Current Pharmaceutical Design, 2012, Vol. 18, No. 11 1553

[122] Church LD, McDermott MF. Canakinumab: a human anti-IL-1


[143] Suissa S, Bernatsky S, Hudson M. Antirheumatic drug use and the
monoclonal antibody for the treatment of cryopyrin-associated pe- risk of acute myocardial infarction. Arthritis Rheum 2006; 55: 531-
riodic syndromes. Expert Rev Clin Immunol 2010; 6: 831-41. 6.
[123] Kapur S, Bonk ME. Rilonacept (Arcalyst), an Interleukin-1 Trap [144] Salliot C, van der Heijde D. Long-term safety of methotrexate
for the Treatment of Cryopyrin-Associated Periodic Syndromes. P monotherapy in patients with rheumatoid arthritis: a systematic lit-
T 2009; 34: 138-141. erature research. Ann Rheum Dis 2009; 68: 1100-4.
[124] Patel AM, Moreland LW. Interleukin-6 inhibition for treatment of [145] Morgan SL, Baggott JE. Folate supplementation during
rheumatoid arthritis: a review of tocilizumab therapy. Drug Des methotrexate therapy for rheumatoid arthritis. Clin Exp Rheumatol
Devel Ther 2010; 4: 263-78. 2010; 28 (5 Suppl 61): S102-9.
[125] Oldfield V, Dhillon S, Plosker GL. Tocilizumab: a review of its use [146] van Ede AE, Laan RF, Blom HJ, et al. Homocysteine and folate
in the management of rheumatoid arthritis. Drugs 2009; 69: 609- status in methotrexate-treated patients with rheumatoid arthritis.
32. Rheumatology (Oxford) 2002; 41: 658-65.
[126] Hirabayashi Y, Ishii T, Harigae H. Clinical efficacy of tocilizumab [147] El Bouchti I, Sordet C, Kuntz JL, Sibilia J. Severe atherosclerosis
in patients with active rheumatoid arthritis in real clinical practice. in rheumatoid arthritis and hyperhomocysteinemia: is there a link?
Rheumatol Int 2010; 30: 1041-8. Joint Bone Spine 2008; 75: 499-501.
[127] Thonhofer R, Hiller M, Just H, Trummer M, Siegel C, Dejaco C. [148] Bakan BM, Sivas F, Aktekin LA, Doan YP, Ozoran K, Bodur H.
Treatment of refractory adult-onset still's disease with tocilizumab: Serum homocysteine level in patients with ankylosing spondylitis.
report of two cases and review of the literature. Rheumatol Int Rheumatol Int 2009; 29: 1435-9.
2011; 31: 1653-6. [149] Methotrexate side effects. http://www.drugs.com/sfx/methotrexate-
[128] Smolen JS, Beaulieu A, Rubbert-Roth A, et al. OPTION Investiga- side-effects.html (Accessed April 20, 2011).
tors. Effect of interleukin-6 receptor inhibition with tocilizumab in [150] Elkayam O, Yaron M, Zhukovsky G, Segal R, Caspi D. Toxicity
patients with rheumatoid arthritis (OPTION study): a double-blind, profile of dual methotrexate combinations with gold, hydroxy-
placebo-controlled, randomised trial. Lancet 2008; 371: 987-97. chloroquine, sulphasalazine and minocycline in rheumatoid arthri-
[129] Genovese MC, McKay JD, Nasonov EL, et al. Interleukin-6 recep- tis patients. Rheumatol Int 1997; 17: 49-53.
tor inhibition with tocilizumab reduces disease activity in rheuma- [151] Van Assche G, Vermeire S, Rutgeerts P. Immunosuppression in
toid arthritis with inadequate response to disease-modifying antir- inflammatory bowel disease: traditional, biological or both? Curr
heumatic drugs: the tocilizumab in combination with traditional Opin Gastroenterol 2009; 25: 323-8.
disease-modifying antirheumatic drug therapy study. Arthritis [152] Katchamart W, Trudeau J, Phumethum V, Bombardier C.
Rheum 2008; 58: 2968-80. Methotrexate monotherapy versus methotrexate combination ther-
[130] Emery P, Keystone E, Tony HP, et al. IL-6 receptor inhibition with apy with non-biologic disease modifying anti-rheumatic drugs for
tocilizumab improves treatment outcomes in patients with rheuma- rheumatoid arthritis. Cochrane Database Syst Rev 2010; (4):
toid arthritis refractory to anti-tumour necrosis factor biologicals: CD008495.
results from a 24-week multicentre randomised placebo-controlled [153] Yildirim-Toruner C, Diamond B. Current and novel therapeutics in
trial. Ann Rheum Dis 2008; 67: 1516-23. the treatment of systemic lupus erythematosus. J Allergy Clin Im-
[131] Kremer JL, Blanco R, Brzosko M, et al. Tocilizumab inhibits munol 2011; 127: 303-12.
structural joint damage in rheumatoid arthritis patients with inade- [154] Ben-Zvi I, Kivity S, Langevitz P, Shoenfeld Y. Hydroxychloro-
quate responses to methotrexate at 1 year: The LITHE study. quine: From Malaria to Autoimmunity. Clin Rev Allergy Immunol
Arthritis Rheum 2011; 63: 609-21. 2011; Jan 8 [Epub ahead of print].
[132] Singh JA, Beg S, Lopez-Olivo MA. Tocilizumab for rheumatoid [155] Becker H, Pavenstaedt H, Willeke P. Emerging treatment strategies
arthritis. Cochrane Database Syst Rev 2010; (7): CD008331. and potential therapeutic targets in primary Sjögren's syndrome.
[133] Choy E, Sattar N. Interpreting lipid levels in the context of high- Inflamm Allergy Drug Targets 2010; 9: 10-9.
grade inflammatory states with a focus on rheumatoid arthritis: a [156] Yavuz S, Asfurolu E, Bicakcigil M, Toker E. Hydroxychloroquine
challenge to conventional cardiovascular risk actions. Ann Rheum improves dry eye symptoms of patients with primary Sjogren's
Dis 2009; 68: 460-9. syndrome. Rheumatol Int 2011; 31: 1045-9.
[134] Kawashiri SY, Kawakami A, Yamasaki S, et al. Effects of the anti- [157] Fox RI. Mechanism of action of hydroxychloroquine as an antir-
interleukin-6 receptor antibody, tocilizumab, on serum lipid levels heumatic drug. Semin Arthritis Rheum 1993; 23 (2 Suppl 1): 82-
in patients with rheumatoid arthritis. Rheumatol Int 2011; 31: 451- 91.
6. [158] Rand JH, Wu XX, Quinn AS, Chen PP, Hathcock JJ, Taatjes DJ.
[135] Steiner G, Urowitz MB. Lipid profiles in patients with rheumatoid Hydroxychloroquine directly reduces the binding of antiphosphol-
arthritis: mechanisms and the impact of treatment. Semin Arthritis ipid antibody-beta2-glycoprotein I complexes to phospholipid bi-
Rheum 2009; 38: 372-81. layers. Blood 2008; 112: 1687-95.
[136] Braun J, Rau R. An update on methotrexate. Curr Opin Rheumatol [159] Rand JH, Wu XX, Quinn AS, et al. Hydroxychloroquine protects
2009; 21: 216-23. the annexin A5 anticoagulant shield from disruption by antiphos-
[137] Smolen JS, Landewé R, Breedveld FC, et al. EULAR recommen- pholipid antibodies: evidence for a novel effect for an old antima-
dations for the management of rheumatoid arthritis with synthetic larial drug. Blood 2010; 115: 2292-9.
and biological disease-modifying antirheumatic drugs. Ann Rheum [160] Petri M. Use of hydroxychloroquine to prevent thrombosis in sys-
Dis 2010; 69: 964-75. temic lupus erythematosus and in antiphospholipid antibody-
[138] Swierkot J, Szechi ski J. Methotrexate in rheumatoid arthritis. positive patients. Curr Rheumatol Rep 2011; 13: 77-80.
Pharmacol Rep 2006; 58: 473-92. [161] Tanay A, Leibovitz E, Frayman A, Zimlichman R, Shargorodsky
[139] Moreira DM, Vieira JL, Gottschall CA. The effects of M, Gavish D. Vascular elasticity of systemic lupus erythematosus
METhotrexate therapy on the physical capacity of patients with patients is associated with steroids and hydroxychloroquine treat-
ISchemic heart failure: a randomized double-blind, placebo- ment. Ann N Y Acad Sci 2007; 1108: 24-34.
controlled trial (METIS trial). J Card Fail 2009; 15: 828-34. [162] Wasko MC, Hubert HB, Lingala VB, et al. Hydroxychloroquine
[140] Wållberg-Jonsson S, Ohman M, Rantapää-Dahlqvist S. Which and risk of diabetes in patients with rheumatoid arthritis. JAMA
factors are related to the presence of atherosclerosis in rheumatoid 2007; 298: 187-93.
arthritis? Scand J Rheumatol 2004; 33: 373-9. [163] Morris SJ, Wasko MC, Antohe JL, et al. Hydroxychloroquine use
[141] Reiss AB, Carsons SE, Anwar K, et al. Atheroprotective effects of is associated with improvement in lipid profiles in rheumatoid ar-
methotrexate on reverse cholesterol transport proteins and foam thritis patients. Arthritis Care Res (Hoboken) 2011; 63: 530-4.
cell transformation in human THP-1 monocyte/macrophages. Ar- [164] Ruiz-Irastorza G, Ramos-Casals M, Brito-Zeron P, Khamashta
thritis Rheum 2008; 58: 3675-83. MA. Clinical efficacy and side effects of antimalarials in systemic
[142] Toms TE, Panoulas VF, John H, Douglas KM, Kitas GD. lupus erythematosus: a systematic review. Ann Rheum Dis 2010;
Methotrexate therapy associates with reduced prevalence of the 69: 20-8.
metabolic syndrome in rheumatoid arthritis patients over the age of [165] Costedoat-Chalumeau N, Hulot JS, Amoura Z, et al. Heart conduc-
60- more than just an anti-inflammatory effect? A cross sectional tion disorders related to antimalarials toxicity: an analysis of elec-
study. Arthritis Res Ther 2009; 11: R110. trocardiograms in 85 patients treated with hydroxychloroquine for
1554 Current Pharmaceutical Design, 2012, Vol. 18, No. 11 Gasparyan et al.

connective tissue diseases. Rheumatology (Oxford) 2007; 46: 808- [190] Nandi P, Kingsley GH, Scott DL. Disease-modifying antirheumatic
10. drugs other than methotrexate in rheumatoid arthritis and seronega-
[166] Naqvi TZ, Luthringer D, Marchevsky A, Saouf R, Gul K, tive arthritis. Curr Opin Rheumatol 2008; 20: 251-6.
Buchbinder NA. Chloroquine-induced cardiomyopathy- [191] Goh L, Samanta A. A systematic MEDLINE analysis of therapeu-
echocardiographic features. J Am Soc Echocardiogr 2005; 18: 383- tic approaches in ankylosing spondylitis. Rheumatol Int 2009; 29:
7. 1123-35.
[167] Cotroneo J, Sleik KM, Rene Rodriguez E, Klein AL. Hydroxy- [192] Plosker GL, Croom KF. Sulfasalazine: a review of its use in the
chloroquine-induced restrictive cardiomyopathy. Eur J Echocar- management of rheumatoid arthritis. Drugs 2005; 65: 1825-49.
diogr 2007; 8: 247-51. [193] Cocco G, Chu DC, Pandolfi S. Colchicine in clinical medicine. A
[168] Sarzi-Puttini P, Cazzola M, Panni B, et al. Long-term safety and guide for internists. Eur J Intern Med 2010; 21: 503-8.
efficacy of low-dose cyclosporin A in severe psoriatic arthritis. [194] Bhat A, Naguwa SM, Cheema GS, Gershwin ME. Colchicine re-
Rheumatol Int 2002; 21: 234-8. visited. Ann N Y Acad Sci 2009; 1173: 766-73.
[169] Sarzi-Puttini P, D'Ingianna E, Fumagalli M, et al. An open, ran- [195] Hauber HP, Blaukovitsch M. Current and future treatment options
domized comparison study of cyclosporine A, cyclosporine A + in idiopathic pulmonary fibrosis. Inflamm Allergy Drug Targets
methotrexate and cyclosporine A + hydroxychloroquine in the 2010; 9: 158-72.
treatment of early severe rheumatoid arthritis. Rheumatol Int 2005; [196] Mendis S. Colchicine cardiotoxicity following ingestion of Glo-
25: 15-22. riosa superba tubers. Postgrad Med J 1989; 65: 752-5.
[170] Zavada J, Pesickova S, Rysava R, et al. Cyclosporine A or intrave- [197] Ata B, Caksen H, Tuncer O, Kirimi E, Akgün C, Odaba D. Four
nous cyclophosphamide for lupus nephritis: the Cyclofa-Lune children with colchicine poisoning. Hum Exp Toxicol 2004; 23:
study. Lupus 2010; 19: 1281-9. 353-6.
[171] Hatemi G, Silman A, Bang D, et al. EULAR Expert Committee. [198] Finkelstein Y, Aks SE, Hutson JR, et al. Colchicine poisoning: the
EULAR recommendations for the management of Behçet disease. dark side of an ancient drug. Clin Toxicol (Phila) 2010; 48: 407-14.
Ann Rheum Dis 2008; 67: 1656-62. [199] Miller MA, Hung YM, Haller C, Galbo M, Levsky ME. Colchi-
[172] Krejci K, Tichy T, Bachleda P, Zadrazil J. Calcineurin inhibitor- cine-related death presenting as an unknown case of multiple organ
induced renal allograft nephrotoxicity. Biomed Pap Med Fac Univ failure. J Emerg Med 2005; 28: 445-8.
Palacky Olomouc Czech Repub 2010; 154: 297-306. [200] Bouquié R, Deslandes G, Renaud C, Dailly E, Haloun A, Jolliet P.
[173] Zakarija A, Bennett C. Drug-induced thrombotic microangiopathy. Colchicine-induced rhabdomyolysis in a heart/lung transplant pa-
Semin Thromb Hemost 2005; 31: 681-90. tient with concurrent use of cyclosporin, pravastatin, and azithro-
[174] CycloSPORINE. Drug Information Provided by Lexi-Comp. mycin. J Clin Rheumatol 2011; 17: 28-30.
http://www.merckmanuals.com/professional/lexicomp/cyclosporine [201] [No authors listed] Colchicine: serious interactions. Prescrire Int
.html (Accessed April 27, 2011). 2008; 17: 151-3.
[175] Robert N, Wong GW, Wright JM. Effect of cyclosporine on blood [202] Boumpas DT, Paliogianni F, Anastassiou ED, Balow JE. Glucocor-
pressure. Cochrane Database Syst Rev 2010; (1): CD007893. ticosteroid action on the immune system: molecular and cellular
[176] Kaltwasser JP, Behrens F. Leflunomide: long-term clinical experi- aspects. Clin Exp Rheumatol 1991; 9: 413-23.
ence and new uses. Expert Opin Pharmacother 2005; 6: 787-801. [203] Nussinovitch U, de Carvalho JF, Pereira RM, Shoenfeld Y. Gluco-
[177] Osiri M, Shea B, Robinson V, et al. Leflunomide for treating corticoids and the cardiovascular system: state of the art. Curr
rheumatoid arthritis. Cochrane Database Syst Rev 2003; (1): Pharm Des 2010; 16: 3574-85.
CD002047. [204] Kirwan JR, Bijlsma JW, Boers M, Shea BJ. Effects of glucocorti-
[178] Deuse T, Erben RG, Ikeno F, et al. Introducing the first polymer- coids on radiological progression in rheumatoid arthritis. Cochrane
free leflunomide eluting stent. Atherosclerosis 2008; 200: 126-34. Database Syst Rev 2007; (1): CD006356.
[179] Pieringer H, Pichler M. Cardiovascular morbidity and mortality in [205] Ibañez M, Ortiz AM, Castrejón I, et al. A rational use of glucocor-
patients with rheumatoid arthritis: vascular alterations and possible ticoids in patients with early arthritis has a minimal impact on bone
clinical implications. QJM 2011; 104: 13-26. mass. Arthritis Res Ther 2010; 12: R50.
[180] Rozman B, Praprotnik S, Logar D, et al. Leflunomide and hyper- [206] Ravindran V, Rachapalli S, Choy EH. Safety of medium- to long-
tension. Ann Rheum Dis 2002; 61: 567-9. term glucocorticoid therapy in rheumatoid arthritis: a meta-
[181] Serelis J, Panagiotakos DB, Mavrommati M, Skopouli FN. Cardio- analysis. Rheumatology (Oxford) 2009; 48: 807-11.
vascular disease is related to hypertension in patients with rheuma- [207] Loddenkemper K, Bohl N, Perka C, Burmester GR, Buttgereit F.
toid arthritis: a greek cohort study. J Rheumatol 2011; 38: 236-41. Correlation of different bone markers with bone density in patients
[182] Panoulas VF, Metsios GS, Pace AV, et al. Hypertension in rheuma- with rheumatic diseases on glucocorticoid therapy. Rheumatol Int
toid arthritis. Rheumatology (Oxford) 2008; 47: 1286-98. 2006; 26: 331-6.
[183] Alcorn N, Saunders S, Madhok R. Benefit-risk assessment of le- [208] Panoulas VF, Douglas KM, Stavropoulos-Kalinoglou A, et al.
flunomide: an appraisal of leflunomide in rheumatoid arthritis 10 Long-term exposure to medium-dose glucocorticoid therapy asso-
years after licensing. Drug Saf 2009; 32: 1123-34. ciates with hypertension in patients with rheumatoid arthritis.
[184] Mak A, Cheak AA, Tan JY, Su HC, Ho RC, Lau CS. Mycophe- Rheumatology (Oxford) 2008; 47: 72-5.
nolate mofetil is as efficacious as, but safer than, cyclophos- [209] Hoes JN, Jacobs JW, Hulsmans HM, et al. High incidence rate of
phamide in the treatment of proliferative lupus nephritis: a meta- vertebral fractures during chronic prednisone treatment, in spite of
analysis and meta-regression. Rheumatology (Oxford) 2009; 48: bisphosphonate or alfacalcidol use. Extension of the alendronate or
944-52. alfacalcidol in glucocorticoid-induced osteoporosis-trial. Clin Exp
[185] Tsukamoto H, Nagafuji K, Horiuchi T, et al. A phase I-II trial of Rheumatol 2010; 28: 354-9.
autologous peripheral blood stem cell transplantation in the treat- [210] Davis JM 3rd, Maradit Kremers H, Crowson CS, et al. Glucocorti-
ment of refractory autoimmune disease. Ann Rheum Dis 2006; 65: coids and cardiovascular events in rheumatoid arthritis: a popula-
508-14. tion-based cohort study. Arthritis Rheum 2007; 56: 820-30.
[186] Nakamae H, Tsumura K, Hino M, Hayashi T, Tatsumi N. QT dis- [211] Kumari R, Uppal SS. First report of supraventricular tachycardia
persion as a predictor of acute heart failure after high-dose cyclo- after intravenous pulse methylprednisolone therapy, with a brief
phosphamide. Lancet 2000; 355: 805-6. review of the literature. Rheumatol Int 2005; 26: 70-3.
[187] Bomback AS, Appel GB. Updates on the treatment of lupus nephri- [212] Tvede N, Nielsen LP, Andersen V. Bradycardia after high-dose
tis. J Am Soc Nephrol 2010; 21: 2028-35. intravenous methylprednisolone therapy. Scand J Rheumatol 1986;
[188] Houssiau FA, D'Cruz D, Sangle S, et al. MAINTAIN Nephritis 15: 302-4.
Trial Group. Azathioprine versus mycophenolate mofetil for long- [213] Flower RJ. Non-steroidal anti-inflammatory drugs: back to the
term immunosuppression in lupus nephritis: results from the future. Rheumatology (Oxford) 1999; 38: 693-6.
MAINTAIN Nephritis Trial. Ann Rheum Dis 2010; 69: 2083-9. [214] Joshi GP, Gertler R, Fricker R. Cardiovascular thromboembolic
[189] Demirta-Ertan G, Rowshani AT, ten Berge IJ. Azathioprine- adverse effects associated with cyclooxygenase-2 selective inhibi-
induced shock in a patient suffering from undifferentiated erosive tors and nonselective antiinflammatory drugs. Anesth Analg 2007;
oligoarthritis. Neth J Med 2006; 64: 124-6. 105: 1793-804.
[215] McGettigan P, Han P, Jones L, Whitaker D, Henry D. Selective
COX-2 inhibitors, NSAIDs and congestive heart failure: differ-
Adverse Cardiovascular Effects of Antirheumatic Drugs Current Pharmaceutical Design, 2012, Vol. 18, No. 11 1555

ences between new and recurrent cases. Br J Clin Pharmacol 2008; and randomised controlled trials. Ann Rheum Dis 2007; 66: 1296-
65: 927-34. 304.
[216] Goodson NJ, Brookhart AM, Symmons DP, Silman AJ, Solomon [220] McGettigan P, Henry D. Cardiovascular risk and inhibition of
DH. Non-steroidal anti-inflammatory drug use does not appear to cyclooxygenase: a systematic review of the observational studies of
be associated with increased cardiovascular mortality in patients selective and nonselective inhibitors of cyclooxygenase 2. JAMA
with inflammatory polyarthritis: results from a primary care based 2006; 296: 1633-44.
inception cohort of patients. Ann Rheum Dis 2009; 68: 367-72. [221] Bombardier C, Laine L, Reicin A, et al. VIGOR Study Group.
[217] Schneeweiss S, Solomon DH, Wang PS, Rassen J, Brookhart MA. Comparison of upper gastrointestinal toxicity of rofecoxib and
Simultaneous assessment of short-term gastrointestinal benefits and naproxen in patients with rheumatoid arthritis. VIGOR Study
cardiovascular risks of selective cyclooxygenase 2 inhibitors and Group. 50 mg of rofecoxib daily or 500 mg of naproxen twice daily
nonselective nonsteroidal antiinflammatory drugs: an instrumental N Engl J Med 2000; 343: 1520-8.
variable analysis. Arthritis Rheum 2006; 54: 3390-8. [222] Bresalier RS, Sandler RS, Quan H, et al. Adenomatous Polyp Pre-
[218] Helin-Salmivaara A, Virtanen A, Vesalainen R, et al. NSAID use vention on Vioxx (APPROVe) Trial Investigators. Cardiovascular
and the risk of hospitalization for first myocardial infarction in the events associated with rofecoxib in a colorectal adenoma chemo-
general population: a nationwide case-control study from Finland. prevention trial. N Engl J Med 2005; 352: 1092-102.
Eur Heart J 2006; 27: 1657-63. [223] Burmester G, Lanas A, Biasucci L, et al. on behalf of the European
[219] Scott PA, Kingsley GH, Smith CM, Choy EH, Scott DL. Non- Expert Panel on the Appropriate Use of NSAID. The appropriate
steroidal anti-inflammatory drugs and myocardial infarctions: com- use of non-steroidal anti-inflammatory drugs in rheumatic disease:
parative systematic review of evidence from observational studies opinions of a multidisciplinary European expert panel. Ann Rheum
Dis 2011; 70: 818-22.

Received: December 16, 2011 Accepted: January 10, 2012

You might also like