You are on page 1of 12

REVIEWS

The role of antimalarial agents in the


treatment of SLE and lupus nephritis
Senq‑J Lee, Earl Silverman and Joanne M. Bargman
Abstract | Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease that affects various
organs. Lupus nephritis is one of the most common, and most important, serious manifestations of SLE.
Antimalarial agents are part of the immunomodulatory regimen used to treat patients with SLE; however,
their role in the treatment of patients with lupus nephritis in particular is less well recognized, especially
by nephrologists. Not all antimalarial agents have been used in the treatment of lupus; this Review will
focus on studies using chloroquine and hydroxychloroquine. In addition, this Review will briefly describe
the history of antimalarial drug use in patients with SLE, the theorized mechanisms of action of the agents
chloroquine and hydroxychloroquine, their efficacy in patients with SLE and those with lupus nephritis, their
use in pregnancy, and potential adverse effects. The Review will also cover the latest recommendations
regarding monitoring for hydroxychloroquine-associated or chloroquine-associated retinopathy. Overall,
antimalarial drugs have numerous beneficial effects in patients with SLE and lupus nephritis, and have a
good safety profile.
Lee, S-J. et al. Nat. Rev. Nephrol. 7, 718–729 (2011); published online 18 October 2011; doi:10.1038/nrneph.2011.150

Introduction
Systemic lupus erythematosus (SLE) is a multisystem and safety profile, and describe how patients taking these
autoimmune disease with varying patterns of organ agents should be monitored.
involvement. Lupus nephritis is one of the most common,
and most important, manifestations of SLE, and can lead A history of antimalarial drug use
to permanent renal damage and chronic kidney disease. Antimalarials are among the oldest drugs still used in
The mainstay of therapy for renal involvement in patients practice today. The first use of antimalarial drugs in a
with SLE is corticosteroids, immunosuppressive agents patient with SLE is thought to have occurred in 1630
and antihypertensive medications. when the wife of a Peruvian Viceroy, the Countess of
The role of antimalarial medications in the treatment Chinchon, was treated successfully for ‘tertian fever’
of patients with SLE is, perhaps, underappreciated in the (malaria) with powdered cinchona bark supplied by Jesuit
renal community. A study published in 2010 demon‑ priests. In 1894, J. S. Payne described features of the lupus
strated that the probability of a patient with SLE receiving rash and prescribed quinine to induce pallor, which was
an antimalarial agent was substantially decreased (OR successful. Following the First World War, the formula for
0.51, 95% CI 0.31–0.84) if their primary physician was a quinacrine was turned over to the US military. Atabrine
nephrologist rather than a rheumatologist.1 Antimalarial (the proprietary name for quinacrine) was used by many
Department of
drugs, specifically chloroquine and hydroxychloroquine, soldiers in the Second World War, principally as malaria
Pediatrics, Division of have been gaining increased prominence in the treat‑ prophylaxis; however, soldiers with various rheumatic
Rheumatology, The ment of patients with SLE—either with or without renal complaints (including inflammatory arthritis and cutane‑
Hospital for Sick
Children, 555 involvement—as a result of their excellent safety profile ous lupus) experienced symptomatic improvement while
University Avenue, and increasing evidence of efficacy. A systematic review taking this agent. These observations led to studies on
Toronto, ON M5G 1X8,
Canada (S‑J. Lee,
of antimalarial use in patients with SLE, published in the use of antimalarial drugs in patients with rheumatic
E. Silverman). 2010, showed that treatment with these agents resulted diseases, which demonstrated improvements in arthritis
Department of in improved disease control, reduced accrual of damage, and cutaneous lupus among patients treated with quina‑
Medicine, Division of
Nephrology, University and a beneficial effect on survival.2 crine. Chloroquine was subsequently introduced in 1953,
Health Network, This Review focuses on the role of chloroquine and and hydroxychloroquine in 1955. Both have greater effi‑
Toronto General
Hospital, 200 Elizabeth
hydroxychloroquine in the treatment of patients with cacy and better tolerability than quinacrine,3 and are still
Street, Toronto, SLE, specifically those with lupus nephritis. We discuss the two most commonly used antimalarial medications
ON M5G 2C4, Canada the current knowledge regarding the mechanisms of administered to patients with SLE.
(J. M. Bargman).
action of these drugs, highlight their favorable efficacy
Correspondence to: Metabolism of antimalarial drugs
J. M. Bargman
joanne.bargman@ Competing interests Hydroxychloroquine and chloroquine are both
uhn.ca The authors declare no competing interests. 4‑amino-quinolines; hydroxychloroquine is an analog

718  |  DECEMBER 2011  |  VOLUME 7  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

of chloroquine formed by β‑hydroxylation of one of Key points


the N‑ethyl substituents. Both agents are well absorbed, ■■ Antimalarial therapy for patients with systemic lupus erythematosus (SLE) is
with 70–80% bioavailability after oral administration, associated with improved survival and reduced disease activity, as well as
and half-lives of 40–50 days. Owing to this long half- cardioprotective and anticancer effects
life, 96% of steady-state levels are not achieved until ■■ In lupus nephritis, antimalarial therapy is associated with reduced
after approximately 6 months of continuous treatment.4 corticosteroid use, reduced disease activity, extended time to end-stage renal
After similar doses given to healthy individuals (volun‑ disease, and, with adjunctive immunomodulatory treatment, improved duration
teers) and to patients with rheumatoid arthritis, drug of renal remission
concentrations vary between indivi­duals up to 11-fold.5 ■■ Treatment with antimalarial agents should be continued in pregnant women
Both drugs are deposited in tissue at concentrations of with SLE; the beneficial effects may include a reduction in the risk of cardiac
200–20,000 times above blood levels, with the highest manifestations of neonatal SLE
concentrations found in pigmented cells of the skin and ■■ Antimalarial drugs have a good safety profile; gastrointestinal symptoms are
retina, but also in mono­nuclear cells and muscle.6 In the most common adverse effect
animal models, chloroquine is 2–3 times more toxic ■■ Baseline monitoring for retinopathy is required, but regular monitoring is
than hydroxychloroquine, whereas the latter drug is recommended by the American Academy of Ophthalmology guidelines only for
only 60–70% as potent. Three times as much chloro‑ patients who have taken antimalarial agents for >5 years
quine as hydroxychloroquine is excreted in the urine, ■■ In patients with impaired renal function, caution with dosing of antimalarial
and three times as much hydroxychloroquine as chloro‑ agents is recommended and careful monitoring for adverse events should be
quine is excreted in the feces.7 In vivo studies show that undertaken
approximately 25% of hydroxychloroquine is excreted
by the kidneys; the rest is hepatic excretion and there‑ cause substantial suppression of immune responses
fore liver dysfunction may lead to higher concentrations directed against foreign antigens.10
of the drug in vivo. Renal excretion of both hydroxychlo‑
roquine and chloro­quine is augmented by acidification Blockade of Toll-like receptor signaling
of the urine. The extensive sequestration of both drugs TLRs are pattern-recognition cellular receptors that
within tissue limits their removal by hemo­d ialysis. 4 induce inflammatory responses by activating the innate
Decreased glomerular filtration rate increases the risk immune system.6 TLR activation causes dendritic cells
of toxic effects of both of these agents, in particular to produce large amounts of IFN‑α, and stimulates
cardio­toxic effects and retinopathy.8 B cells to increase their production of immunoglobulins
and cyto­kines, and to upregulate their expression of co-
Mechanisms of action stimulatory molecules.11 In vitro and in vivo studies of
The mechanisms of action of antimalarial drugs in antimalarial agents in both rheumatoid arthritis and SLE
reducing inflammation remain unclear, although under‑ show that these drugs reduce inflammatory responses
standing of the underlying pathways is improving. Both by inhibiting TLR activation. The alkalinization of lyso‑
hydroxychloroquine and chloroquine are lipophilic, somes by antimalarial agents also interferes with endo‑
weak bases that easily pass across cell membranes and somal TLR signaling, primarily on antigen-presenting
into acidic intracellular vesicles, including lysosomes. cells, thereby inhibiting TLR activation and reducing
Their immunomodulatory effects are mediated by inflammatory responses resulting from activation of the
mechanisms that are anti-inflammatory, immuno­ innate immune system.6,11 The delay in onset of the clini‑
suppressive and photoprotective. Specifically, these cal actions of antimalarial drugs may be explained by the
agents might alter lysosome stability, suppress antigen observation that the primary immunosuppressive effect is
presentation, inhibit prostaglandin and cytokine synthe‑ on antigen-­presenting cells, and not pre-existing activated
sis, and influence both Toll-like receptor (TLR) signaling T or B cells; this delay may also be caused by the long
and leukocyte activation. duration of time required to achieve steady-state levels.
Another use of antimalarial agents, of particular
interest to nephrologists, is the ability of hydroxychloro‑ Reduced cytokine and prostaglandin synthesis
quine to reduce 1‑hydroxylation of 25-hydroxyvitamin In vitro and in vivo studies of patients with SLE treated
D3, which reduces elevated levels of 1,25 dihydroxy­ with chloroquine show that this agent inhibits produc‑
vitamin D in patients with sarcoidosis and can be useful tion of the cytokines, tumor necrosis factor (TNF), IL‑6,
for the hypercalcemia seen in this condition.9 IFN‑γ, IL‑1β, and IL‑18.12–15 Macrophages and monocytes
are the major cells affected. Some of these effects might be
Suppression of autoantigen presentation lysosome-independent,16,17 and are thought to be caused
Within the lysosomes of antigen-presenting cells, anti‑ by altered phosphorylation of mitogen-­activated protein
malarial agents cause functional alterations by increasing kinases 3 and 1 (also known as MAPK3/ERK1 and
the pH within lysosomal vesicles. This process results in MAPK1/ERK2), and dual-­specificity mitogen-activated
altered peptide loading and decreased binding of auto­ protein kinase kinases 1 and 2 (also known as MAP2K1/
antigenic peptides to class II MHC molecules (a low- MEK1 and MAP2K2/MEK2).18 Finally, anti­malarial
affinity interaction); however, high-affinity binding of agents are prostaglandin antagonists. They inhibit
exogenous antigens (such as bacteria) to these molecules phospho­lipase A2, with resultant alteration of arachidonic
is not affected. Antimalarial agents do not, therefore, acid metabolism and decreased inflammation.19

NATURE REVIEWS | NEPHROLOGY VOLUME 7  |  DECEMBER 2011  |  719


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Antiproliferative effects Clinical benefits in patients with SLE


Hydroxychloroquine promotes apoptosis by activating In 1894, quinine was used in the treatment of discoid
caspase‑3, and might also sensitize cells to Fas-mediated lupus, and in 1928 the beneficial effects of pamaquine on
apoptosis.20,21 Apoptosis of synoviocytes, antiprolifera‑ discoid and subacute cutaneous lupus were suggested.29
tion of endothelial cells and suppression of lymphocyte The first study investigating withdrawal of amodiaquine
function by antimalarial therapy, might lead to immuno­ treatment, published in 1954, showed that all five patients
suppression and decreased angiogenesis. 21 In vitro experienced a flare within 1–3 months of stopping the
studies show that chloroquine has inhibitory effects on medication.30 The first controlled study on the efficacy of
human endothelial cell proliferation and induces apop‑ chloroquine in patients with SLE appeared in 1975. This
tosis of these cells.22 This mechanism might have impor‑ retrospective study compared the symptoms of a group
tant consequences in patients with rheumatic diseases, of patients when they were on and off therapy, respec‑
in whom upregulation of antiangiogenic pathways and tively. An overall benefit of chloroquine use was demon‑
inhibition of pannus formation are important thera‑ strated, as measured by decreased flare rates, improved
peutic approaches. Antimalarials also inhibit neutro‑ skin disease and reduced glucocorticoid doses.31
phil superoxide release through lysosome-independent In 1991, the Canadian Hydroxychloroquine Study
effects.23 Group performed a prospective, randomized, trial,
which showed that compared with patients who contin‑
Photoprotection ued to take hydroxychloroquine, those who discontin‑
Multiple mechanisms have been proposed to explain the ued this treatment had a sixfold greater rate of severe
clinically recognized effect of chloroquine and hydroxy‑ SLE exacerbation requiring withdrawal from the study
chloroquine on dermatological manifestations of SLE. (4% versus 23%, P = 0.06), and a significantly higher rate
Chloroquine reduces levels of mRNA for IL‑1β, IL‑6 and of SLE flare (73% versus 36%, P = 0.02), defined as the
TNF in skin irradiated with ultraviolet‑B (UVB) light. development of specific clinical manifestations of SLE or
This agent might, therefore, have an inhibitory effect on an increase in their severity, as well as a shorter time to
the production of proinflammatory cytokines induced flare (P = 0.02).32 In 1996, a placebo-controlled Spanish
by UVB radiation.22 The intracellular mechanism pro‑ study demonstrated that patients on chloroquine had
posed for photoprotection is activation of the transcrip‑ significantly lower flare rates and prednisone doses, and
tion factor AP‑1, which subsequently transactivates the lower overall disease activity, as scored by the Systemic
immediate-early genes, including JUN (which encodes Lupus Erythematosus Disease Activity Index (SLEDAI)
AP‑1 itself ).24 Treatment with chloroquine decreases at 1 year.33 Subsequent cohort studies have continued
serum levels of IL‑6, IL‑18 and TNF, and increases the to reinforce these findings. In the USA, the Hopkins
minimal erythemal dose (the amount of UVB light Lupus Cohort and the LUMINA (Lupus in Minorities:
required to induce cutaneous reddening) in patients Nature Versus Nurture) nested case–control study,
with SLE.14 Hence, chloroquine and hydroxychloroquine which was conducted in adults of Hispanic (Mexican
have immunomodulatory properties that could lead to and Central American), African American and white
protection against the skin damage associated with expo‑ European popula­t ions, showed that hydroxychloro‑
sure to ultraviolet light. However, accumulation of these quine use was associated with a long-term protective
agents within keratinocytes can lead to the sun-induced effect on end-organ damage and improved survival.34,35
pigmenta­tion changes associated with their use. The improvement in survival associated with the use of
these medications was still evident even after account‑
Decreased metalloproteinase activity ing for confounding factors, including disease severity,
Metalloproteinases are involved in both inflammatory major organ involvement and socioeconomic class.34 In
and immune responses. Chloroquine reduces serum both studies,34,35 the researchers attributed the increased
levels of matrix metalloproteinase‑9 in patients with survival to beneficial cardioprotective effects of the anti‑
SLE, and increases levels of metalloproteinase inhibitor 1, malarial used and a reduction in SLE flares, accrual of
which inhibits the activity of various metalloproteinases chronic damage and neoplasms. Hydroxychloroquine
and also maintains the balance between extracellular use is associated with reduced overall damage scores
matrix formation and destruction. 25 Altered produc‑ in patients either with or without damage before
tion of multiple metalloproteinases might also occur starting this drug, and also increased time to accrual
via decreased expression of mitogen-activated protein of new damage. Researchers who studied a cohort of
kinase 3 and p38 mitogen-activated protein kinase within 151 Israeli patients also found that those on hydroxy­
the TLR or TNF pathways.26 chloroquine had reduced scores on the Systemic Lupus
International Collaborating Clinics–American College
Decreased leukocyte activation of Rheumatology Damage Index for SLE.36
Serum levels of the leukocyte activation markers, soluble The GLADEL study, which involved a large observa‑
CD8 and soluble IL‑2 receptors are decreased after tional cohort of 1,480 patients from 34 centers in nine
6 weeks of hydroxychloroquine treatment.27 Mean serum Latin-American countries, demonstrated that the use of
levels of B‑cell activating factor are also significantly hydroxychloroquine and/or chloroquine for >6 months
reduced (6.3 ± 0.5 mg/ml to 3.0 ± 0.56 mg/ml, P = 0.0001) led to a 38% reduction in overall mortality (hazard ratio
following hydroxychloroquine use.28 [HR] 0.62, 95% CI 0.39–0.99).37 The greatest survival

720  |  DECEMBER 2011  |  VOLUME 7  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Overall and organ-specific effects of antimalarial drugs in patients with SLE and lupus nephritis
Study Design Population and treatment Outcomes in users
Fessler et al. Longitudinal 518 patients: 291 HCQ users, 227 nonusers Reduction in accrual of new disease damage (HR 0.68, 95% CI
(2005)35 cohort 0.53–0.93, P = 0.014)
LUMINA
Ruiz-Irastorza Prospective 232 patients: 62 HCQ users, 46 CQ users, 42 HCQ Increased cumulative 15-year survival in drug users vs nonusers
et al. (2006)63 cohort and CQ users, 82 nonusers (0.95 vs 0.6, HR 0.14, 95% CI 0.04–0.48, P <0.001)
Calvo-Alén Nested 32 patients with SLE and osteonecrosis vs 59 Possible protection against osteoporosis
et al. (2006)94 case–control controls without osteonecrosis matched for age, SLE Cases: less % HCQ exposure time (40% vs 59%, P = 0.026); higher
LUMINA duration, ethnicity and center; all exposed to HCQ, mean glucocorticoid dose (22.7 mg vs 14.8 mg, P = 0.011);
glucocorticoids and cytotoxic drugs cytotoxic drugs received more often (59% vs 32%, P = 0.015)
Wozniacka Cohort 25 patients with SLE treated with CQ vs 25 sex and Improved reduction in SLAM scores (9.47 to 4.92 after CQ,
et al. (2006)14 age-matched healthy controls P <0.001)
Costedoat- Longitudinal 143 patients with SLE: 120 inactive disease vs 23 Reduced serum HCQ levels in patients with flares (OR 0.4, 95% CI
Chalumeau cohort active disease; HCQ users 0.18–9.85, P = 0.01)
et al. (2006)93
Alarcón et al. Case–control 608 patients with SLE: 61 deaths (cases), 547 live Increased survival with HCQ use (OR 0.128, 95% CI 0.054–0.300,
(2007)34 (controls); HCQ users P <0.0001)
LUMINA
Ruiz-Irastorza Observational 235 patients with SLE: 156 HCQ and/or CQ users vs Protection against neoplasia (1.3% vs 13%, P <0.001); improved
et al. (2007)61 prospective 79 nonusers cumulative cancer-free survival (OR 0.98 vs 0.73, P <0.001)
cohort
James et al. Retrospective 130 US military patients with SLE: 26 patients Time between onset of first clinical signs or symptoms and SLE
(2007)95 chart review treated with HCQ before SLE classification classification was prolonged in patients treated with HCQ before
diagnosis vs not pretreated group (Wilcoxon signed-rank test,
P = 0.018)
Sisó et al. Cohort 206 patients with lupus nephritis: 56 previously Protection against infection (11% vs 29%, P = 0.006); increased
(2008)62 taking HCQ, 150 nonusers survival (2% vs 13%, P = 0.029)
Ruiz-Irastorza Nested 249 patients with SLE: 83 patients with infections Protection against infection (OR 0.06, 95% CI 0.02–0.18)
et al. (2009)96 case–control (cases) vs 166 with no infections (controls);
antimalarial users (agent not specified)
Shinjo et al. Retrospective 57 patients with SLE ≥65 years old: 43 with disease Disease remission strongly associated with CQ use (OR 12.9,
(2009)97 cohort remission, 14 with disease activity; CQ users 95% CI 2.9–58.1, P <0.001)
Pons-Estel Longitudinal 580 patients; HCQ users Possible delayed onset of integument damage (HR 0.23, 95% CI
et al. (2010)98 observational 0.12–0.47, P <0.0001)
LUMINA cohort
Shinjo et al. Longitudinal 1,480 patients: 1,141 CQ and/or HCQ users vs 339 Increased survival (4.4% vs 11.5%, HR 0.62, 95% CI 0.39–0.99,
(2010)37 cohort nonusers P <0.001)
GLADEL
Abbreviations: CQ, chloroquine; HCQ, hydroxychloroquine; SLAM, systemic lupus activity measure; SLE, systemic lupus erythematosus.

benefit was noted with prolonged antimalarial agent Administration of these drugs also leads to significantly
use. 37 Some researchers have hypothesized that the reduced levels of apo­lipoprotein B in patients with either
widespread use of chloroquine to treat and/or prevent rheumatoid arthritis or SLE, and to increased levels of
malaria in West Africa might at least partially explain the apolipoprotein A in patients with rheumatoid arthritis.
dramatically lower prevalence of SLE in this region than Hydroxychloroquine treatment might decrease the
in people of West African origin who migrated to Europe risk of atherosclerosis by improving binding (and inhib‑
or North America.38 A systematic review 2 of articles pub‑ iting dissociation) of insulin to its receptor, thereby
lished from 1982 to 2007 found a number of beneficial improving glucose tolerance (Table 2).45 In patients with
effects of antimalarial use in patients with SLE, includ‑ rheumatoid arthritis and SLE, use of hydroxychloroquine
ing improved survival, reduced disease activity, and was found to improve glucose profiles, lower fasting
organ-specific effects (Table 1). In the following subsec‑ insulin levels, lower insulin resistance, and lower hemo­
tions we will focus on literature published from 2005 to globin A1c (HbA1c) levels.46–48 The favorable metabolic
2010 and discuss the effects of use of chloroquine and effect of hydroxychloroquine was also found in non-
hydroxychloroquine in patients with SLE. insulin-dependent diabetic patients without rheumatic
disease.49 The use of hydroxychloroquine is also associ‑
Dyslipidemia, glycemia and atherosclerosis ated with a reduced frequency of metabolic syndrome
The use of chloroquine and hydroxychloroquine is among patients with SLE.50
associ­ated with significantly decreased levels of tri­ Data on the effects of antimalarial therapy on vascu‑
glycerides, LDL cholesterol and/or VLDL choles‑ lar disease are conflicting (Table 2). In a mouse model of
terol, and increased levels of HDL cholesterol. 39–44 atherosclerosis, chloroquine therapy decreased plaque

NATURE REVIEWS | NEPHROLOGY VOLUME 7  |  DECEMBER 2011  |  721


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 2 | The effects of antimalarial agents on cardiovascular disease, thrombosis and glycemic control in patients with SLE and lupus nephritis
Study Design Population and treatment Outcomes in users
Ruiz-Irastorza Prospective 232 patients with SLE: 62 HCQ users, 46 CQ Protection against thrombosis (HR 0.28, 95% CI 0.08–0.90)
et al. (2006)63 cohort users, 42 HCQ and CQ users, 82 nonusers
de Leeuw et al. Cross- 38 patients with SLE taking HCQ: 7 with CVD, 31 No significant change in cardiovascular risk (54% with CVD vs 53% without
(2006)99 sectional without CVD CVD, P = NS)
Sachet et al. In vivo 30 individuals: 10 patients with SLE taking CQ, Improved clearance of total cholesterol (CQ group 156 ± 16 mg/dl;
(2007)43 10 patients with SLE on no antimalarial therapy, no antimalarial group 174 ± 15 mg/dl; control group 200 ± 24 mg/dl,
10 healthy controls P <0.001) and low-density lipoproteins (CQ group 88 ± 16 mg/dl;
no antimalarial group 108 ± 17 mg/dl; control group 118 ± 23 mg/dl,
P <0.001)
Choojitarom Cohort 67 antiphospholipid-antibody-positive patients Decreased risk of thrombosis (OR 0.18, 95% CI 0.04–0.88, P = 0.034);
et al. (2008)100 with SLE treated with CQ or HCQ the subgroup of patients with lupus nephritis had an increased risk of
venous thrombosis (OR 6.2, P = 0.005)
Sisó et al. Cohort 206 patients with lupus nephritis: 56 had Reduced risk of developing hypertension (32% vs 50%, P = 0.027);
(2008)62 previously taken CQ or HCQ; 150 were nonusers protection against thrombosis (5% vs 17%, P = 0.039)
Kaiser et al. Cohort 1,930 patients with SLE: 1,534 (80%) HCQ users Protection against thrombosis (OR 0.67, 95% CI 0.5–0.9, P = 0.008)
(2009)101
Tektonidou Longitudinal 288 patients with SLE: 144 antiphospholipid- Protection against thrombosis in both groups
et al. (2009)102 cohort antibody-positive patients, matched with 144 Antibody-positive: HR per month of treatment 0.99, 95% CI 0.98–1.0,
antiphospholipid-antibody-negative patients; HCQ P = 0.05
users Antibody-negative: HR per month of treatment 0.98, 95% CI 0.95–0.99,
P = 0.04
Jung et al. Nested 54 patients with SLE who experienced Protection against thrombosis (OR 0.32, 95% CI 0.14–0.74, P <0.01)
(2010)64 case– thromboembolic events vs 108 event-free
control patients with SLE (controls); HCQ users
Penn et al. Cross- 149 patients with SLE and 177 patients with Reduced fasting glucose levels in nondiabetic women (potentially
(2010)48 sectional rheumatoid arthritis; HCQ users vs nonusers improved glycemic control)
SLE: 85.9 mg/dl vs 89.3 mg/dl, P = 0.04; rheumatoid arthritis: 82.5 mg/dl
vs 86.6 mg/dl, P = 0.05
Abbreviations: CQ, chloroquine; CVD, cardiovascular disease; HCQ, hydroxychloroquine; NS, not significant; SLE, systemic lupus erythematosus.

burden.51 Treatment with hydroxychloroquine has been Thrombosis


associated with a lower prevalence of carotid artery Antimalarial drug use is thromboprotective (Table 2).
plaques compared with not taking this agent in some, A significantly reduced risk of thrombosis is evident
but not all studies.52,53 However, in three cohort studies, in patients on chloroquine and hydroxychloroquine
neither the use of nor the duration of use of chloroquine versus the risk in those not receiving this treatment.62,63
or hydroxychloroquine correlated with plaque forma‑ In another study, a 68% reduction in the risk of thrombo­
tion.54–56 Measures of arterial stiffness were significantly embolism was found in patients with SLE on hydroxy‑
lower in patients taking hydroxychloroquine than in chloroquine therapy, as compared to individuals with
patients who were not taking this drug.57,58 The results of SLE who were not taking these medications.64
a case–control study of 29 patients with SLE demonstrated
that individuals with cardiovascular diseases were signifi‑ Clinical benefits in lupus nephritis
cantly less likely than those without such diseases to have Antimalarial therapy has several potential benefits in
received hydroxychloroquine.59 However, a 2010 report patients with lupus nephritis (Table 3). Antimalarial
from the Systemic Lupus Erythematosus International therapy is recommended in patients with lupus nephri‑
Collaborating Clinics international inception cohort study, tis with preserved renal function, even though they may
which included 1,249 patients, did not find any difference have a risk of renal impairment later on. Strategies such
in the rate of atherosclerotic vascular events after 2 years of as dose reduction and monitoring for adverse effects
follow-up between patients who were taking antimalarials of antimalarial treatment should be implemented in
and those who were not on this treatment. However, such patients who have renal impairment.
events occurred in only 1.8% of the whole cohort.60 The Canadian Hydroxychloroquine Study Group was
one of the first to examine the effects of antimalarial drug
Malignancy use on renal outcomes. This randomized withdrawal
Antimalarial drug use in patients with SLE could reduce study with 3-year follow-up demonstrated that contin‑
the risk of malignancy. In one study, the prevalence of ued hydroxychloroquine treatment was associated with
neoplasia was lower in patients treated with anti­malarials a 74% reduction in the risk of nephritic flares compared
(1.3%) than in those not taking such medications (13%).61 with withdrawal (placebo) (4% in the hydroxychloro‑
In another study, the prevalence of malignancy in chloro­ quine group versus 14% in the placebo group, relative
quine or hydroxychloroquine users was 0%, versus 3% risk 0.26, 95% CI 0.03–2.54). Owing to the small cohort
in nonusers.62 of 47 patients included in this study, its statistical power

722  |  DECEMBER 2011  |  VOLUME 7  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 3 | The effects of antimalarial drug use in patients with lupus nephritis
Study Design Population and treatment Outcomes in users
Tsakonas et al. Cohort 47 patients: 25 continued HCQ, Possible reduction in risk of and time to renal disease flare (RR 0.26, 95% CI
(1998)65 (randomized 22 withdrew 0.03–2.54, P = 0.25)
drug withdrawal)
Fessler et al. Longitudinal 518 patients: 291 started HCQ at Lower incidence of renal disease at baseline (25% vs 53%, P <0.0001)
(2005)35 LUMINA observational study enrollment, 227 were nonusers
cohort
Kasitanon et al. Cohort 29 patients: 11 used both HCQ and Patients with membranous lupus nephritis who were taking both drugs had
(2006)69 Hopkins MMF, 18 used MMF only improved rates of renal remission within 12 months (64% vs 22%, P = 0.036)
Lupus Cohort
Barber et al. Retrospective 35 patients with lupus nephritis: Improved sustained remission rates (93.8% vs 52.6%, P = 0.01)
(2006)103 cohort 15 out of 16 patients with sustained
remission taking HCQ vs 10 out of
19 patients with no sustained
remission (controls) taking HCQ
Sisó et al. Cohort 206 patients: 56 were taking CQ or Reduced percentage of patients with creatinine elevations >354 µmol/l (2%
(2008)62 HCQ before diagnosis of lupus vs 11%, P = 0.029); prolonged time to end-stage renal failure (2% vs 11%,
nephritis, 150 were nonusers P = 0.044); reduced frequency of hypertension (32% vs 50%, P = 0.027);
reduced mortality (2% vs 13%, P = 0.029)
Pons-Estel et al. Longitudinal 203 patients: 161 HCQ users, Reduced frequency of class IV glomerulonephritis (9.9% vs 33.3%, P <0.01);
(2009)66 LUMINA observational 42 nonusers protection against ESRD and/or diminished GFR (HR 0.38, 95% CI 0.13–
cohort 1.06, P = 0.065 [full model]; HR 0.38, 95%CI 0.16–0.86, P = 0.0206 [reduced
(prospective) model]); decreased glucocorticoid (prednisone) dose (11.3 ± 12.0 mg vs
16.8 ± 20.5 mg, P = 0.025); protection against renal damage (HR 0.12, 95%
CI 0.02–0.97, P = 0.0464 [full model]; HR 0.29, 95% CI 0.13–0.68,
P = 0.0043 [reduced model]); reduced cumulative probability of renal damage
(HCQ users 20% [5 years] or 38% [10 years]; nonusers 47% [5 years] or 70%
[10 years], P <0.0001)
Shinjo et al. Observational 1,480 patients: 1,141 CQ and/or Reduced prevalence of renal disease (28.4% vs 42.8%, P <0.001)
(2010)37 GLADEL inception cohort HCQ users, 339 nonusers
Abbreviations: CQ, chloroquine; ESRD, end-stage renal disease; GFR, glomerular filtration rate; HCQ, hydroxychloroquine; MMF, mycophenolate mofetil; RR, relative risk.

is low. Nevertheless, these findings provided early evi‑ P = 0.02).68 However, these findings are difficult to gen‑
dence that hydroxychloroquine treatment might reduce eralize to other populations, as the cohort consisted of
nephritic flares.65 only three ethnic groups.
In 2005, the investigators of the LUMINA study A substudy of 29 patients from the Hopkins Lupus
reported that hydroxychloroquine use was associ‑ Cohort who had either membranous nephritis or mixed
ated with a reduced risk of developing renal disease in membranous nephritis and proliferative nephritis treated
patients with SLE.35 In the subgroup of 203 patients with with mycophenolate mofetil, showed that concurrent use
lupus nephritis, 79.3% of patients had received hydroxy‑ of hydroxychloroquine led to a statistically significant
chloroquine treatment. Those who received hydroxy‑ improvement in the rate of renal remission at 12 months.
chloroquine had a lower frequency of World Health This finding persisted after controlling for the presence
Organization class IV glomerulonephritis, lower SLE of antibodies to double-stranded DNA.69
disease activity scores, and lower glucocorticoid doses A Spanish long-term, observational, cohort study of
than those patients not taking hydroxychloroquine. 206 patients showed that those with biopsy-proven lupus
Hydroxychloroquine treatment was also associated with nephritis taking chloroquine or hydroxy­chloroquine
a reduced cumulative probability of renal damage.66 The had a reduced incidence of elevated creatinine levels
magnitude of these effects was remarkable, leading to (>354 μmol/l), hypertension, infections, thrombotic events,
suggestions that the study results reflected confound‑ and death, as well as a prolonged time to develop­ment of
ing by indication, immortal person-time bias (meaning end-stage renal disease, compared to the incidence in
that the time between study enrollment and initiation those not taking antimalarial agents.62 These findings were
of hydroxychloroquine treatment should have been, signifi­cant despite the fact that only 27% of the 206 patients
but was not, excluded from the follow-up period, even were taking antimalarial agents. Although this study
though patients who developed renal damage during included a large cohort of patients, the analysis demon­
this period were not treated with hydroxychloroquine), strates potential methodological limitations, including
and other potential (unknown) sources of bias.67,68 The confounding bias and immortal person-time bias.
authors of the 2005 paper subsequently responded by The investigators of the GLADEL study reported that
performing time-dependent analyses using a longitudi‑ 77% of the participants had received chloroquine and/or
nal approach, and still found that hydroxychloroquine hydroxychloroquine, defined as use of these agents for >6
use reduced the occurrence of renal disease, albeit to consecutive months. In addition to prolonged survival,
a lesser extent (adjusted OR 0.51, 95% CI 0.29–0.91, the researchers demonstrated that renal disease was less

NATURE REVIEWS | NEPHROLOGY VOLUME 7  |  DECEMBER 2011  |  723


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 4 | An overview of studies on antimalarial drug use in pregnant patients with SLE
Study Study type Population and treatment Outcomes of users
Buchanan et al. Cohort 36 pregnant women with SLE taking HCQ No significant difference in live birth rates with and without HCQ (86% vs
(1996)104 and 53 pregnant women with SLE not 83%); no significant difference in rates of fetal death or spontaneous abortion
taking HCQ with and without HCQ (14% vs 17%); no significant difference in premature
births with and without HCQ (55% vs 48%); no significant difference in
intrauterine growth failure with and without HCQ (19% vs 41%)
Levy et al. Prospective 20 pregnant women with SLE randomized Lower SLEPDAI scores at delivery (P = 0.0356); may reduce flares (0% vs 30%,
(2001)75 cohort with 10 to treatment with HCQ and 10 to P = 0.21); reduced prednisone requirements (6.0 ± 6.2 mg vs 15.5 ± 24.5 mg
placebo at 18 weeks, P = 0.07; 4.5 ± 4.3 mg vs 13.7 ± 27.9 mg, P <0.05 at delivery); no
congenital abnormalities, and no retinopathy or ototoxic effects in children
1.5–3.0 years of age born to mothers who used antimalarial drugs
Costedoat- Nested 90 women with SLE (133 pregnancies) No difference in live-birth rates with and without HCQ (88% vs 84%); no
Chalumeau case–control taking HCQ and 53 women with SLE difference in rate of congenital abnormalities with HCQ compared with rates
et al. (2003)105 (70 pregnancies) not taking HCQ in the general population (2.26% vs 2.3%); no visual, hearing, growth, or
developmental abnormalities associated with HCQ treatment
Clowse et al. Prospective 257 pregnancies in 3 groups: (1) 56 No difference in rates of miscarriage, stillbirth, pregnancy loss, congenital
(2006)74 cohort pregnancies in women with SLE taking abnormalities with and without HCQ; stillbirths: 6% vs 8% vs 9%, P = 0.85;
HCQ; (2) 163 pregnancies in women with preterm (20–28 weeks): 12% vs 10% vs 6%, P = 0.83; preterm (28–
SLE not taking HCQ; (3) 38 women 37 weeks): 27% vs 31% vs 47%, P = 0.87; full-term: 61% vs 59% vs 47%,
stopped taking HCQ either in the P = 0.98; reduced overall SLE disease activity; SLEDAI score >4: 52% vs 62%
3 months before pregnancy or during the vs 84%, P = 0.0075; flare rates: 30% vs 36% vs 55%, P = 0.053; reduced
first trimester doses of prednisone during pregnancy (16 ± 12 mg vs 23 ± 19 mg vs
21 ± 16 mg, P = 0.056)
Carvalheiras Retrospective 43 women with SLE who had 51 No cases of maternal mortality; no cases of fetal malformations
et al. (2010)106 cohort pregnancies over 14 years; 20 patients
treated with HCQ
Izmirly et al. Case–control 50 women with SLE whose infants HCQ use might reduce the risk of fetal development of cardiac NLE in
(2010)76 developed NLE (14% HCQ users) vs 151 pregnant women with SLE and either anti-Ro/SSA or anti-La/SSB antibodies
women with SLE whose infants did not (OR 0.46, 95% CI 0.18–1.18, P = 0.10)
develop NLE (controls: 37.1% HCQ users)
Abbreviations: HCQ, hydroxychloroquine; NLE, neonatal lupus erythematosus; SLE, systemic lupus erythematosus; SLEDAI, Systemic Lupus Erythematosus Disease Activity Index; SLEPDAI,
Systemic Lupus Erythematosus Pregnancy Disease Activity Index.

frequent in antimalarial drug users than in nonusers Pregnancy increases disease activity in many patients
(28.4% versus 42.8%).37 with SLE. Flares can potentially worsen renal function,
Antimalarial drugs are postulated to reduce the hypertension and/or proteinuria, leading to an increased
severity of renal disease through immunomodulatory, risk of maternal and fetal complications.73 These adverse
anti-inflammatory and antithrombotic effects. All such outcomes can also occur as a consequence of stopping
effects act beneficially on the vascular endothelium commonly used immuno­suppressive therapies, includ‑
and reduce renal inflammation. The studies discussed ing antimalarial drugs, owing to fears of potential fetal
above provide intriguing evidence that chloroquine and or neonatal complica­tions. Two studies in 2001 and 2006
hydroxy­chloroquine might retard the progression of suggested that hydroxychloroquine use in pregnant
renal disease, increase the duration of renal remission patients with lupus is associated with decreased overall
when combined with other immunosuppressive medica­ disease activity and flare rates.74,75 The 2006 study found
tions, and reduce cumulative dose of glucocorticoids. that women with SLE who stopped hydroxychloroquine
However, caution must be exercised with generalizing when they became pregnant had worse disease activity
these results to specific populations, as the majority and higher glucocorticoid requirements than women
of studies included only Hispanic or Latin American, with SLE who continued taking hydroxychloroquine
African American, and Caucasian populations. during the pregnancy, or women who were not taking
this drug at the time of conception. 74 These results
Pregnancy and fetal outcomes suggest that withdrawal of hydroxychloroquine during
Antimalarials are safe and effective for pregnant patients pregnancy exacerbates the risk of developing increased
with lupus (Table 4). Hydroxychloroquine readily crosses disease activity, as occurs in nonpregnant patients. The
the placenta, and blood levels of this drug are similar results of a case–control study that analyzed data from
between mother and fetus. 6 However, studies have the American Neonatal Lupus Registry suggest that the
revealed no increased risk of retinopathy or ototoxic use of hydroxychloroquine during pregnancy in mothers
effects in infants born to women taking hydroxychloro‑ with antinuclear antibodies (anti-Ro/SSA and/or anti-La/
quine at the recommended (reduced) dose of <4 mg/kg SSB antibodies) decreases the risk of cardiac manifesta­
(lean body weight) per day during the pregnancy.70,71 A tions of neonatal lupus erythematosus in a multi­variable
review published in 2005 of >250 pregnancies also sup‑ analysis (OR 0.46, 95% CI 0.18–1.18, P = 0.10). The
ported the lack of teratogenic effects associated with researchers concluded that use of anti­malarial drugs
hydroxychloroquine use.72 resulted in decreased TLR signaling, which led to

724  |  DECEMBER 2011  |  VOLUME 7  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 5 | Potential adverse effects associated with antimalarial agents


Adverse effect Agent Supporting studies Reported frequency
Retinopathy CQ Leecharonen et al. (2007)8 Retinopathy: 0.18–19%
Marmor et al. (2002)82 Corneal deposits: 6–7%
Wang et al. (1999)77
Aviña-Zubieta et al. (1998)107
Finbloom et al. (1985)108
Retinopathy HCQ Wolfe et al. (2010)85 Retinopathy: 0–6%
Mavrikakis et al. (2003)83 Corneal deposits: 0.8%
Marmor et al. (2002)82
Wang et al. (1999)77
Aviña-Zubieta et al. (1998)107
Levy et al. (1997)86
Spalton et al. (1993)109
Finbloom et al. (1985)108
Ototoxic effects HCQ Wang et al. (1999)77 0–0.6%
Morand et al. (1992)110
Cardiotoxic CQ and HCQ Costedoat-Chalumeau et al. Heart conduction defect: 0–4%
effects (2007)111,112 Cardiomyopathy: <1%
Wozniacka et al. (2006)113 Atrioventricular block: <1%
Nord et al. (2004)88 Case reports of cardiomyopathy, heart conduction disturbances,
Cervera et al. (2001)114 congestive heart failure
Cutaneous Quinacrine, Kalia et al. (2010)6 Skin rash: 0.6–4.3%
lesions CQ and HCQ Di Giacomo et al. (2009)115 Hyperpigmentation: 10–30% (most frequent with quinacrine)
Puri et al. (2008)89 Urticaria: 12%
Herman et al. (2006)90 Psoriasis: insufficient evidence to suggest these agent cause
Wang et al. (1999)77 flares
Aviña-Zubieta et al. (1998)107
Morand et al. (1992)110
Gastrointestinal CQ and HCQ Bezerra et al. (2005)116 Gastrointestinal (overall): 0–30%
symptoms Van Beek & Piette (2001)117 Nausea or vomiting: 12%
Wang et al. (1999)77 Diarrhea: 18%
Aviña-Zubieta et al. (1998)107 Elevated levels of liver enzymes: 10%
Morand et al. (1992)110
Other CQ and HCQ Casado et al. (2006)118 Headaches: 1.3–12%
Bezerra et al. (2005)116 Myopathy: 0–6.7%
Wang et al. (1999)77
Aviña-Zubieta et al. (1998)107
Morand et al. (1992)110
Rare events CQ and HCQ Kalia et al. (2010)6 Hemolysis in patients with G6PD deficiency; severe leukopenia
(case reports) Collins et al. (2008)119 or aplastic anemia; acute CQ-induced psychosis (similar to
Bracamonte et al. (2006)91 phencyclidine-induced psychosis); pseudo-Fabry disease
Abbreviations: CQ, chloroquine; G6PD, glucose-6-phosphate 1-dehydrogenase; HCQ, hydroxychloroquine.

reduced cardiac inflammation and scarring.76 We con‑ levels in the retinal pigmented epithelium; these agents,
clude that hydroxychloroquine use should be maintained therefore, accumulate disproportionately in these cells
during pregnancy. and are more likely to alter the lysosomal pH of melanin-­
containing cells than those of melanin-free cells.78 The
Adverse effects of antimalarial agents earliest symptom of retinal damage is loss of para­central
Both hydroxychloroquine and chloroquine are well- visual fields, with loss of color vision; these losses prog‑
tolerated medications with good safety profiles. The ress as the lesion spreads into the fovea and over the
most commonly reported adverse effects of treatment fundus. The very early stages of functional loss may be
are gastro­intestinal (nausea, vomiting, diarrhea, anorexia reversible with cessation of antimalarial therapy, but
and, rarely, elevated levels of liver enzymes) or neurologi‑ most cases with maculopathy are irreversible.79
cal (headache and dizziness). Retinopathy is an uncom‑ It was originally suggested that ophthalmological
mon, but very important adverse effect associated with examina­t ions should be performed every 3 months
antimalarial use (Table 5).77 in patients receiving antimalarial agents, but by 1996
examina­tions every 6–12 months were considered ade‑
Retinopathy quate.80,81 The American Academy of Ophthalmology
The retinal toxic effects associated with antimalarial (AAO) published revised recommendations in 2011 after
use are the result of disrupted metabolism of the retinal reviewing literature surrounding screening methods,
pigmented epithelium, which results in the degenera‑ implications of screening, and risk factors for devel‑
tion of photoreceptors. Antimalarial drugs demonstrate oping retinotoxicity.79 Screening is not for prevention,
enhanced binding to melanin, which is present at high but rather to detect early toxicity in order to stabilize

NATURE REVIEWS | NEPHROLOGY VOLUME 7  |  DECEMBER 2011  |  725


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

maculopathy and stop further loss of visual acuity. The cardiotoxic effects of high-dose antimalarial therapy
AAO recommendations are to first perform a baseline include decreased myocardial contractility, hypo­tension
screen within the first year of antimalarial therapy. and conduction abnormalities. Chronic cardiotoxic
Previous literature suggests that the cumulative dose effects of this treatment are manifested by heart block,
of anti­m alarial agents has an important role in the biventricular hypertrophy and/or cardio­myopathy
develop­ment of toxic effects. The AAO recommenda‑ (usually constrictive or restrictive). These complica‑
tions suggest that annual screening may not be necessary tions are most frequently seen in patients receiving high
before 5 years of cumulative therapy, and therefore that doses of antimalarial agents and in those with renal
annual screening should be conducted after 5 years of impairment.88
cumulative therapy. However, caution must be exercised
when factors that increase the risk of retino­pathy are Cutaneous toxic effects
present, including: high cumulative doses of these agents Both hydroxychloroquine and chloroquine sometimes
(hydroxy­chloroquine >1,000 g or chloro­quine >460 g); cause oval patches of yellow-brown to slate-gray hyper‑
high daily doses (hydroxychloroquine >400 mg daily, or pigmentation that might enlarge. The hyperpigmenta‑
>6.5 mg/kg of lean body weight; chloroquine >250 mg tion can begin after 4 months of therapy and occurs in up
daily, or >3.0 mg/kg of lean body weight); older age; renal to 10% of patients treated with these agents. The lesions
or liver dysfunction; and pre-existing visual impair‑ tend to develop on mucosal areas, in particular the hard
ment, such as retinal disease or maculo­pathy. Obesity palate, although any area of the skin may be involved.89
is a risk factor for retinopathy because anti­malarials are Antimalarial drugs might exacerbate psoriasis in a small
not deposited in fatty tissue; dosing should, therefore, be proportion (1–2%) of patients. However, a systematic
based on lean body weight.82,83 review published in 2006 revealed no strong evidence
Ophthalmologic examinations in patients taking anti‑ either supporting or refuting the hypothesis that anti­
malarial drugs should include enquiries about visual malarial use worsens psoriasis. 90 Whether hydroxy­
symptoms, a thorough ophthalmologic examination chloroquine has a role in the treatment of new-onset
and an automated visual field assessment (using the psoriasis in patients with SLE is, therefore, unclear.
Humphrey visual field 10–2 pattern testing).80 Other more
specific objective testing to be considered that may only Iatrogenic phospholipidosis
be available in specialist centers include the multi­focal One case report described a woman with inflammatory
electroretinogram, spectral domain-optical coherence polyarthritis who was treated with hydroxychloroquine
tomography, and fundus autofluorescence.79,84 Evidence and went on to develop iatrogenic phospholipidosis
from large clinical studies of patients with rheumatic resembling Fabry disease. A renal biopsy sample obtained
diseases (enrolling 526–3,995 patients) supports these to investigate proteinuria revealed ‘classic’ Fabry
recommendations, as only 0.4–0.65% of patients devel‑ disease. However, DNA mutational analysis revealed
oped retinal toxic effects associated with anti­malarial no abnormali­ties associated with Fabry disease in this
therapy. The majority of such adverse events occurred patient’s α‑galactosidase A gene. The authors of this report
after 6 years of antimalarial treatment and/or in patients postulated that long-term hydroxychloroquine use caused
taking >6.5 mg/kg hydroxychloroquine daily; some cases iatrogenic phospholipidosis secondary to inhibition
of retinopathy were also seen when the dose of anti­ of the activity of circulating α‑galactosidase.91
malarial agent was based on the patient’s overall weight,
rather than lean body weight.83,85,86 The guidelines pub‑ Drug level monitoring
lished by the British Royal College of Ophthalmologists Poor compliance with antimalarial therapy and other
and the British Society for Rheumatology in 2009 are medica­tions for SLE can be a common problem. A study
similar to the AAO recommendations, stating that annual of hydroxychloroquine pharmacokinetics showed that
screening is not required until after 5 years of cumula‑ serum levels of this agent could be quantified by high-
tive therapy, as clinically significant maculopathy is rare performance liquid chromatography. 92 Five patients
and no reliable test exists for detecting this retinopathy had undetectable levels reflecting poor treatment
within the reversible stage. The guidelines recommend compliance. Patients with active SLE had significantly
careful or earlier screening by an ophthalmologist if the lower serum hydroxy­chloroquine concentrations than
patient has baseline visual impairment or eye disease, if those with inactive disease (694 ± 448 ng/ml versus
visual disturbances are noticed during antimalarial treat‑ 1,079 ± 526 ng/ml, P = 0.001). This may be due to poor
ment, or if the patient is receiving high doses (>6.5 mg/kg compliance, but more a result of large interindividual
daily) of hydroxychloroquine or continuous treatment variations in hydroxychloroquine bioavailability. A
for >5 years.87 low serum concentration of hydroxychloroquine was
a predictor of disease exacerbation (OR 0.4, 95% CI
Neuromyotoxic and cardiotoxic effects 0.18–0.85, P = 0.01), and a hydroxychloroquine concen‑
Neuromyotoxic and cardiotoxic effects are rare but tration threshold of 1,000 ng/ml had a negative predic‑
potentially fatal complications of antimalarial therapy. tive value of 96% for exacerbations.93 Hence, measuring
Patients with neuromyotoxic effects usually present with serum levels of hydroxy­chloroquine could prove bene­
bilateral and progressive muscle weakness of the legs, ficial in assessing compliance, as well as in predicting and
which can be accompanied by a polyneuropathy. Acute potentially reducing disease exacerbations. Serum levels

726  |  DECEMBER 2011  |  VOLUME 7  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

of hydroxychloroquine are not measured in current clini‑ most important adverse effects are retinopathy, cardio‑
cal practice, however, as not all laboratories are able to toxic effects, neuromyopathy, cutaneous hyperpigmenta‑
perform this test. tion, and elevated liver enzyme levels and/or creatinine
levels. Baseline visual examination is required within
Conclusions 1 year of commencing antimalarial therapy, but annual
Antimalarial agents have been the mainstay of treatment screening is not recommended unless the patient has
for SLE, in combination with other immunomodula‑ clinical symptoms or findings of retinopathy or is at
tory drugs, but are underused by nephrologists. Their a high risk of developing it. Annual screening should,
immuno­modulatory and anti-inflammatory effects are however, be implemented after 5 years of cumulative
associated with numerous beneficial effects on the out‑ antimalarial therapy.
comes of patients with SLE, including improvements in Overall, antimalarial drugs are generally safe and
survival and remission rates, and reductions in disease have far more potential benefits than potential risks for
activity, accrual of new disease-related damage, and patients with SLE and lupus nephritis. Nephrologists
infection rates. Antimalarial therapy has antithrombotic should not overlook this important class of drugs in the
and vascular protective effects, and might also have a role management of patients with SLE.
in preventing neoplasia. These drugs are beneficial and
safe to use in pregnant women, in whom they reduce
both disease activity and glucocorticoid requirements. Review criteria
These benefits are crucial for pregnant patients with SLE,
as other immunomodulatory drugs must frequently be The MEDLINE database was searched to identify papers
stopped owing to their potential teratogenic effects. Strong published in English between 2005 and 2011 on the
benefits or efficacy of antimalarial agents in SLE,
evidence also supports the use of antimalarial drugs
using the following MeSH terms: (“antimalarials” OR
in patients with lupus nephritis: treatment with these “chloroquine” OR “hydroxychloroquine” OR “plaquenil”)
agents is associated with reductions in the prevalence AND (“lupus erythematosus, systemic” OR “lupus”).
of renal disease (class IV glomerulonephritis, elevated Papers published in English on the benefits of
serum levels of creatinine and hypertension); disease antimalarial drug use in patients with lupus nephritis were
activity; glucocorticoid requirements; and progression identified by a further MEDLINE search up to 2011 using
to chronic kidney disease. the above MeSH terms in combination with “nephritis”
OR “lupus nephritis” OR “renal” OR “kidney”. A PubMed
Cautious monitoring for potential adverse effects
search was then conducted using all of the above terms
of antimalarial therapy is recommended, especially in to identify additional relevant articles.
patients with concurrent renal or liver impairment. The

1. Schmajuk, G., Yazdany, J., Trupin, L. & Yelin, E. undergoing hemodialysis. Am. J. Med. 82, macrophages. J. Immunol. 158, 4901–4907
Hydroxychloroquine treatment in a community- 1259–1262 (1987). (1997).
based cohort of patients with systemic lupus 10. Fox, R. Anti-malarial drugs: possible 17. Sperber, K. et al. Selective regulation of cytokine
erythematosus. Arthritis Care Res. (Hoboken) 62, mechanisms of action in autoimmune disease secretion by hydroxychloroquine: inhibition of
386–392 (2010). and prospects for drug development. Lupus 5 interleukin 1 alpha (IL‑1‑alpha) and IL‑6 in human
2. Ruiz-Irastorza, G., Ramos-Casals, M., Brito- (Suppl. 1), S4–S10 (1996). monocytes and T cells. J. Rheumatol. 20,
Zeron, P. & Khamashta, M. A. Clinical efficacy 11. Ermann, J. & Bermas, B. L. The biology behind 803–808 (1993).
and side effects of antimalarials in systemic the new therapies for SLE. Int. J. Clin. Pract. 61, 18. Weber, S. M., Chen, J. M. & Levitz, S. M.
lupus erythematosus: a systematic review. Ann. 2113–2119 (2007). Inhibition of mitogen-activated protein kinase
Rheum. Dis. 69, 20–28 (2010). 12. van den Borne, B. E., Dijkmans, B. A., signaling by chloroquine. J. Immunol. 168,
3. Wallace, D. J. The history of antimalarials. Lupus de Rooij, H. H., le Cessie, S. & Verweij, C. L. 5303–5309 (2002).
5 (Suppl. 1), S2–S3 (1996). Chloroquine and hydroxychloroquine equally 19. Löffler, B. M., Bohn, E., Hesse, B. & Kunze, H.
4. Tett, S. E., Cutler, D. J., Day, R. O. & Brown, K. F. affect tumor necrosis factor-alpha, interleukin 6, Effects of antimalarial drugs on phospholipase A
Bioavailability of hydroxychloroquine tablets in and interferon-gamma production by peripheral and lysophospholipase activities in plasma
healthy volunteers. Br. J. Clin. Pharmacol. 27, blood mononuclear cells. J. Rheumatol. 24, membrane, mitochondrial, microsomal and
771–779 (1989). 55–60 (1997). cytosolic subcellular fractions of rat liver.
5. Furst, D. E. Pharmacokinetics of 13. Karres, I. et al. Chloroquine inhibits Biochim. Biophys. Acta 835, 448–455 (1985).
hydroxychloroquine and chloroquine during proinflammatory cytokine release into human 20. Kim, W. U. et al. Hydroxychloroquine potentiates
treatment of rheumatic diseases. Lupus 5 whole blood. Am. J. Physiol. 274, R1058–R1064 Fas-mediated apoptosis of rheumatoid
(Suppl. 1), S11–S15 (1996). (1998). synoviocytes. Clin. Exp. Immunol. 144, 503–511
6. Kalia, S. & Dutz, J. P. New concepts in 14. Wozniacka, A., Lesiak, A., Narbutt, J., (2006).
antimalarial use and mode of action in McCauliffe, D. P. & Sysa-Jedrzejowska, A. 21. Potvin, F., Petitclerc, E., Marceau, F. &
dermatology. Dermatol. Ther. 20, 160–174 Chloroquine treatment influences Poubelle, P. E. Mechanisms of action of
(2007). proinflammatory cytokine levels in systemic antimalarials in inflammation: induction of
7. McChesney, E. W. Animal toxicity and lupus erythematosus patients. Lupus 15, apoptosis in human endothelial cells.
pharmacokinetics of hydroxychloroquine sulfate. 268–275 (2006). J. Immunol. 158, 1872–1879 (1997).
Am. J. Med. 75, 11–18 (1983). 15. Jang, C. H., Choi, J. H., Byun, M. S. & Jue, D. M. 22. Wozniacka, A. et al. The influence of antimalarial
8. Leecharoen, S., Wangkaew, S. & Louthrenoo, W. Chloroquine inhibits production of TNF-alpha, treatment on IL‑1beta, IL‑6 and TNF-alpha mRNA
Ocular side effects of chloroquine in patients IL‑1beta and IL‑6 from lipopolysaccharide- expression on UVB-irradiated skin in systemic
with rheumatoid arthritis, systemic lupus stimulated human monocytes/macrophages by lupus erythematosus. Br. J. Dermatol. 159,
erythematosus and scleroderma. J. Med. Assoc. different modes. Rheumatology (Oxford) 45, 1124–1130 (2008).
Thai. 90, 52–58 (2007). 703–710 (2006). 23. Hurst, N. P., French, J. K., Gorjatschko, L. &
9. Barré, P. E., Gascon-Barré, M., Meakins, J. L. & 16. Jeong, J. Y. & Jue, D. M. Chloroquine inhibits Betts, W. H. Studies on the mechanism of
Goltzman, D. Hydroxychloroquine treatment of processing of tumor necrosis factor in inhibition of chemotactic tripeptide stimulated
hypercalcemia in a patient with sarcoidosis lipopolysaccharide-stimulated RAW 264.7 human neutrophil polymorphonuclear leucocyte

NATURE REVIEWS | NEPHROLOGY VOLUME 7  |  DECEMBER 2011  |  727


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

superoxide production by chloroquine and 41. Tam, L. S., Gladman, D. D., Hallett, D. C., 59. Bessant, R. et al. Prevalence of conventional
hydroxychloroquine. Ann. Rheum. Dis. 46, Rahman, P. & Urowitz, M. B. Effect of antimalarial and lupus-specific risk factors for
750–756 (1987). agents on the fasting lipid profile in systemic cardiovascular disease in patients with
24. Nguyen, T. Q., Capra, J. D. & Sontheimer, R. D. lupus erythematosus. J. Rheumatol. 27, systemic lupus erythematosus: a case-control
4‑Aminoquinoline antimalarials enhance UV‑B 2142–2145 (2000). study. Arthritis Rheum. 55, 892–899 (2006).
induced c‑jun transcriptional activation. Lupus 42. Borba, E. F. & Bonfá, E. Longterm beneficial 60. Urowitz, M. B. et al. Atherosclerotic vascular
7, 148–153 (1998). effect of chloroquine diphosphate on lipoprotein events in a multinational inception cohort of
25. Lesiak, A. et al. Effect of chloroquine profile in lupus patients with and without steroid systemic lupus erythematosus. Arthritis Care
phosphate treatment on serum MMP‑9 and therapy. J. Rheumatol. 28, 780–785 (2001). Res. (Hoboken) 62, 881–887 (2010).
TIMP‑1 levels in patients with systemic lupus 43. Sachet, J. C. et al. Chloroquine increases low- 61. Ruiz-Irastorza, G. et al. Antimalarials may
erythematosus. Lupus 19, 683–688 (2010). density lipoprotein removal from plasma in influence the risk of malignancy in systemic
26. Lim, E. J. et al. Toll-like receptor 9 dependent systemic lupus patients. Lupus 16, 273–278 lupus erythematosus. Ann. Rheum. Dis. 66,
activation of MAPK and NF‑kB is required for (2007). 815–817 (2007).
the CpG ODN-induced matrix 44. Cardoso, C. R., Signorelli, F. V., Papi, J. A. & 62. Sisó, A. et al. Previous antimalarial therapy in
metalloproteinase‑9 expression. Exp. Mol. Med. Salles, G. F. Prevalence and factors associated patients diagnosed with lupus nephritis:
39, 239–245 (2007). with dyslipoproteinemias in Brazilian systemic Influence on outcomes and survival. Lupus 17,
27. Wallace, D. J., Linker-Israeli, M., Metzger, A. L. lupus erythematosus patients. Rheumatol. Int. 281–288 (2008).
& Stecher, V. J. The relevance of antimalarial 28, 323–327 (2008). 63. Ruiz-Irastorza, G. et al. Effect of antimalarials
therapy with regard to thrombosis, 45. Bevan, A. P., Christensen, J. R., Tikerpae, J. & on thrombosis and survival in patients with
hypercholesterolemia and cytokines in SLE. Smith, G. D. Chloroquine augments the binding systemic lupus erythematosus. Lupus 15,
Lupus 2 (Suppl. 1), S13–S15 (1993). of insulin to its receptor. Biochem. J. 311, 577–583 (2006).
28. Toubi, E. et al. The reduction of serum 787–795 (1995). 64. Jung, H. et al. The protective effect of
B‑lymphocyte activating factor levels following 46. Petri, M. Hydroxychloroquine use in the antimalarial drugs on thrombovascular events
quinacrine add-on therapy in systemic lupus Baltimore Lupus Cohort: effects on lipids, in systemic lupus erythematosus. Arthritis
erythematosus. Scand. J. Immunol. 63, glucose and thrombosis. Lupus 5 (Suppl. 1), Rheum. 62, 863–868 (2010).
299–303 (2006). S16–S22 (1996). 65. Tsakonas, E. et al. A long-term study of
29. Dubois, E. L. Antimalarials in the management 47. Rekedal, L. R. et al. Changes in glycosylated hydroxychloroquine withdrawal on
of discoid and systemic lupus erythematosus. hemoglobin after initiation of hydroxychloroquine exacerbations in systemic lupus
Semin. Arthritis Rheum. 8, 33–51 (1978). or methotrexate treatment in diabetes patients erythematosus. The Canadian
30. Dubois, E. L. Quinacrine (atabrine) in treatment with rheumatic diseases. Arthritis Rheum. 62, Hydroxychloroquine Study Group. Lupus 7,
of systemic and discoid lupus erythematosus. 3569–3573 (2010). 80–85 (1998).
AMA Arch. Intern. Med. 94, 131–141 (1954). 48. Penn, S. K. et al. Hydroxychloroquine and 66. Pons-Estel, G. J. et al. Protective effect of
31. Rudnicki, R. D., Gresham, G. E. & glycemia in women with rheumatoid arthritis and hydroxychloroquine on renal damage in patients
Rothfield, N. F. The efficacy of antimalarials in systemic lupus erythematosus. J. Rheumatol. with lupus nephritis: LXV, data from a
systemic lupus erythematosus. J. Rheumatol. 37, 1136–1142 (2010). multiethnic US cohort. Arthritis Rheum. 61,
2, 323–330 (1975). 49. Gerstein, H. C., Thorpe, K. E., Taylor, D. W. & 830–839 (2009).
32. [No authors listed] A randomized study of the Haynes, R. B. The effectiveness of 67. Vlad, S. C. Protective effect of
effect of withdrawing hydroxychloroquine hydroxychloroquine in patients with type 2 hydroxychloroquine on renal damage may be
sulfate in systemic lupus erythematosus. The diabetes mellitus who are refractory to biased: comment on the article by Pons-Estel
Canadian Hydroxychloroquine Study Group. sulfonylureas—a randomzed trial. Diabetes Res. et al. Arthritis Rheum. 61, 1614 (2009).
N. Engl. J. Med. 324, 150–154 (1991). Clin. Pract. 55, 209–219 (2002). 68. Vinet, E., Bernatsky, S. & Suissa, S. Have some
33. Meinão, I. M., Sato, E. I., Andrade, L. E., 50. Bellomio, V. et al. Metabolic syndrome in beneficial effects of hydroxychloroquine been
Ferraz, M. B. & Atra, E. Controlled trial with Argentinean patients with systemic lupus overestimated? Potential biases in
chloroquine diphosphate in systemic lupus erythematosus. Lupus 18, 1019–1025 (2009). observational studies of drug effects: comment
erythematosus. Lupus 5, 237–241 (1996). 51. Razani, B., Feng, C. & Semenkovich, C. F. p53 is on the article by Pons-Estel et al. Arthritis
34. Alarcón, G. S. et al. Effect of required for chloroquine-induced Rheum. 61, 1614–1615 (2009).
hydroxychloroquine on the survival of patients atheroprotection but not insulin sensitization. 69. Kasitanon, N., Fine, D. M., Haas, M.,
with systemic lupus erythematosus: data from J. Lipid Res. 51, 1738–1746 (2010). Magder, L. S. & Petri, M. Hydroxychloroquine
LUMINA, a multiethnic US cohort (LUMINA L). 52. Roman, M. J. et al. Prevalence and correlates of use predicts complete renal remission within
Ann. Rheum. Dis. 66, 1168–1172 (2007). accelerated atherosclerosis in systemic lupus 12 months among patients treated with
35. Fessler, B. J. et al. Systemic lupus erythematosus. N. Engl. J. Med. 349, mycophenolate mofetil therapy for membranous
erythematosus in three ethnic groups: XVI. 2399–2406 (2003). lupus nephritis. Lupus 15, 366–370 (2006).
Association of hydroxychloroquine use with 53. Zhang, C. Y. et al. Evaluation of risk factors that 70. Parke, A. Antimalarial drugs and pregnancy. Am.
reduced risk of damage accrual. Arthritis contribute to high prevalence of premature J. Med. 85, 30–3 (1988).
Rheum. 52, 1473–1480 (2005). atherosclerosis in Chinese premenopausal 71. Parke, A. & West, B. Hydroxychloroquine in
36. Molad, Y. et al. Protective effect of systemic lupus erythematosus patients. J. Clin. pregnant patients with systemic lupus
hydroxychloroquine in systemic lupus Rheumatol. 15, 111–116 (2009). erythematosus. J. Rheumatol. 23, 1715–1718
erythematosus. Prospective long-term study of 54. Souza, A. W., Hatta, F. S., Miranda, F. Jr & (1996).
an Israeli cohort. Lupus 11, 356–361 (2002). Sato, E. I. Atherosclerotic plaque in carotid 72. Costedoat-Chalumeau, N., Amoura, Z.,
37. Shinjo, S. K. et al. Antimalarial treatment may arteries in systemic lupus erythematosus: Huong, D. L., Lechat, P. & Piette, J. C. Safety of
have a time-dependent effect on lupus survival: frequency and associated risk factors. Sao Paulo hydroxychloroquine in pregnant patients with
data from a multinational Latin American Med. J. 123, 137–142 (2005). connective tissue diseases. Review of the
inception cohort. Arthritis Rheum. 62, 55. Von Feldt, J. M. et al. Homocysteine levels and literature. Autoimmun. Rev. 4, 111–115 (2005).
855–862 (2010). disease duration independently correlate with 73. Germain, S. & Nelson-Piercy, C. Lupus nephritis
38. Westlake, S. L. & Edwards C. J. Anti-malarials coronary artery calcification in patients with and renal disease in pregnancy. Lupus 15,
and lupus in West Africa use and lupus in systemic lupus erythematosus. Arthritis Rheum. 148–155 (2006).
Africans. Lupus 18, 193–195 (2009). 54, 2220–2227 (2006). 74. Clowse, M. E., Magder, L., Witter, F. & Petri, M.
39. Hodis, H. N., Quismorio, F. P. Jr, Wickham, E. & 56. Selzer, F. et al. Comparison of risk factors for Hydroxychloroquine in lupus pregnancy. Arthritis
Blankenhorn, D. H. The lipid, lipoprotein, and vascular disease in the carotid artery and aorta Rheum. 54, 3640–3647 (2006).
apolipoprotein effects of hydroxychloroquine in in women with systemic lupus erythematosus. 75. Levy, R. A. et al. Hydroxychloroquine (HCQ) in
patients with systemic lupus erythematosus. Arthritis Rheum. 50, 151–159 (2004). lupus pregnancy: double-blind and placebo-
J. Rheumatol. 20, 661–665 (1993). 57. Selzer, F. et al. Vascular stiffness in women with controlled study. Lupus 10, 401–404 (2001).
40. Petri, M., Lakatta, C., Magder, L. & Goldman, D. systemic lupus erythematosus. Hypertension 76. Izmirly, P. M. et al. Evaluation of the risk of anti-
Effect of prednisone and hydroxychloroquine on 37, 1075–1082 (2001). SSA/Ro-SSB/La antibody-associated cardiac
coronary artery disease risk factors in 58. Tanay, A. et al. Vascular elasticity of systemic manifestations of neonatal lupus in fetuses of
systemic lupus erythematosus: a longitudinal lupus erythematosus patients is associated with mothers with systemic lupus erythematosus
data analysis. Am. J. Med. 96, 254–259 steroids and hydroxychloroquine treatment. Ann. exposed to hydroxychloroquine. Ann. Rheum.
(1994). NY Acad. Sci. 1108, 24–34 (2007). Dis. 69, 1827–1830 (2010).

728  |  DECEMBER 2011  |  VOLUME 7  www.nature.com/nrneph


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

77. Wang, C. et al. Discontinuation of antimalarial objective marker of poor adherence to treatment 107. Aviña-Zubieta, J. A., Galindo-Rodriguez, G.,
drugs in systemic lupus erythematosus. of systemic lupus erythematosus. Ann. Rheum. Newman, S., Suarez-Almazor, M. E. &
J. Rheumatol. 26, 808–815 (1999). Dis. 66, 821–824 (2007). Russell, A. S. Long-term effectiveness of
78. Sundelin, S. P. & Terman, A. Different effects of 93. Costedoat-Chalumeau, N. et al. Low blood antimalarial drugs in rheumatic diseases. Ann.
chloroquine and hydroxychloroquine on concentration of hydroxychloroquine is a marker Rheum. Dis. 57, 582–587 (1998).
lysosomal function in cultured retinal pigment for and predictor of disease exacerbations in 108. F inbloom, D. S., Silver, K., Newsome, D. A. &
epithelial cells. APMIS 110, 481–489 (2002). patients with systemic lupus erythematosus. Gunkel, R. Comparison of hydroxychloroquine
79. Marmor, M. F., Kellner, U., Lai, T. Y., Lyons, J. S. & Arthritis Rheum. 54, 3284–3290 (2006). and chloroquine use and the development of
Meiler, W. F. Revised recommendations on 94. Calvo-Alén, J. et al. Systemic lupus retinal toxicity. J. Rheumatol. 12, 692–694
screening for chloroquine and erythematosus in a multiethnic US cohort (1985).
hydroxychloroquine retinopathy. Ophthalmology (LUMINA): XXIV. Cytotoxic treatment is an 109. Spalton, D. J., Verdon Roe, G. M. &
118, 415–422 (2011). additional risk factor for the development of Hughes, G. R. Hydroxychloroquine, dosage
80. Rynes, R. I. Ophthalmologic considerations in symptomatic osteonecrosis in lupus patients: parameters and retinopathy. Lupus 2, 355–358
using antimalarials in the United States. Lupus results of a nested matched case-control study. (1993).
5 (Suppl. 1), S73–S74 (1996). Ann. Rheum. Dis. 65, 785–790 (2006). 110. Morand, E. F., McCloud, P. I. & Littlejohn, G. O.
81. Spalton, D. J. Retinopathy and antimalarial 95. James, J. A. et al. Hydroxychloroquine sulfate Continuation of long term treatment with
drugs—the British experience. Lupus treatment is associated with later onset of hydroxychloroquine in systemic lupus
5 (Suppl. 1), S70–S72 (1996). systemic lupus erythematosus. Lupus 16, erythematosus and rheumatoid arthritis. Ann.
82. Marmor, M. F., Carr, R. E., Easterbrook, M., 401–409 (2007). Rheum. Dis. 51, 1318–1321 (1992).
Farjo, A. A. & Mieler, W. F. Recommendations on 96. Ruiz-Irastorza, G. et al. Predictors of major 111. Costedoat-Chalumeau, N. et al. Cardiomyopathy
screening for chloroquine and infections in systemic lupus erythematosus. related to antimalarial therapy with illustrative
hydroxychloroquine retinopathy: a report by the Arthritis Res. Ther. 11, R109 (2009). case report. Cardiology 107, 73–80 (2007).
American Academy of Ophthalmology. 97. Shinjo, S. K. Systemic lupus erythematosus in 112. Costedoat-Chalumeau, N. et al. Heart
Ophthalmology 109, 1377–1382 (2002). the elderly: antimalarials in disease remission. conduction disorders related to antimalarials
83. Mavrikakis, I. et al. The incidence of irreversible Rheumatol. Int. 29, 1087–1090 (2009). toxicity: an analysis of electrocardiograms in
retinal toxicity in patients treated with 98. Pons-Estel, G. J. et al. Possible protective effect 85 patients treated with hydroxychloroquine for
hydroxychloroquine: a reappraisal. of hydroxychloroquine on delaying the connective tissue diseases. Rheumatology
Ophthalmology 110, 1321–1326 (2003). occurrence of integument damage in lupus: LXXI, (Oxford) 46, 808–810 (2007).
84. Lyons, J. S. & Severns, M. L. Using multifocal data from a multiethnic cohort. Arthritis Care 113. Wozniacka, A., Cygankiewicz, I., Chudzik, M.,
ERG ring ratios to detect and follow Plaquenil Res. (Hoboken) 62, 393–400 (2010). Sysa-Jedrzejowska, A. & Wranicz, J. K. The
retinal toxicity: a review: review of mfERG ring 99. de Leeuw, K. et al. Traditional and non-traditional cardiac safety of chloroquine phosphate
ratios in Plaqunil toxicity. Doc. Ophthal. 118, risk factors contribute to the development of treatment in patients with systemic lupus
29–36 (2008). accelerated atherosclerosis in patients with erythematosus: the influence on arrhythmia,
85. Wolfe, F. & Marmor, M. F. Rates and predictors of systemic lupus erythematosus. Lupus 15, heart rate variability and repolarization
hydroxychloroquine retinal toxicity in patients 675–682 (2006). parameters. Lupus 15, 521–525 (2006).
with rheumatoid arthritis and systemic lupus 100. Choojitarom, K. et al. Lupus nephritis and 114. Cervera, A., Espinosa, G., Font, J. & Ingelmo, M.
erythematosus. Arthritis Care Res. (Hoboken) 62, Raynaud’s phenomenon are significant risk Cardiac toxicity secondary to long term
775–784 (2010). factors for vascular thrombosis in SLE patients treatment with chloroquine. Ann. Rheum. Dis.
86. Levy, G. D. et al. Incidence of hydroxychloroquine with positive antiphospholipid antibodies. Clin. 60, 301 (2001).
retinopathy in 1,207 patients in a large Rheumatol. 27, 345–351 (2008). 115. Di Giacomo, T. B., Valente, N. Y. & Nico, M. M.
multicenter outpatient practice. Arthritis Rheum. 101. Kaiser, R., Cleveland, C. M. & Criswell, L. A. Risk Chloroquine -induced hair depigmentation.
40, 1482–1486 (1997). and protective factors for thrombosis in Lupus 18, 264–266 (2009).
87. The Royal College of Ophthalmologists. systemic lupus erythematosus: results from a 116. Bezerra, E. L., Vilar, M. J., da Trindade
Hydroxychloroquine and Ocular Toxicity large, multi-ethnic cohort. Ann. Rheum. Dis. 68, Neto, P. B. & Sato, E. L. Double-blind,
Recommendations on Screening [online], http:// 238–241 (2009). randomized, controlled clinical trial of
www.rcophth.ac.uk/core/core_picker/download. 102. Tektonidou, M. G., Laskari, K., clofazimine compared with chloroquine in
asp?id=165&filetitle=Ocular+Toxicity+and+Hydr Panagiotakos, D. B. & Moutsopoulos, H. M. Risk patients with systemic lupus erythematosus.
oxychloroquine%3A+Guidelines+for+Screen factors for thrombosis and primary thrombosis Arthritis Rheum. 52, 3073–3078 (2005).
ing+2009 (2009). prevention in patients with systemic lupus 117. Van Beek, M. J. & Piette, W. W. Antimalarials.
88. Nord, J. E., Shah, P. K., Rinaldi, R. Z. & erythematosus with or without antiphospholipid Dermatol. Clin. 19, 147–160 (2001).
Weisman, M. H. Hydroxychloroquine antibodies. Arthritis Rheum. 61, 29–36 (2009). 118. Casado, E. et al. Antimalarial myopathy:
cardiotoxicity in systemic lupus erythematosus: 103. Barber, C. E., Geldenhuys, L. & Hanly, J. G. an underdiagnosed complication? Prospective
a report of 2 cases and review of the literature. Sustained remission of lupus nephritis. Lupus longitudinal study of 119 patients. Ann.
Semin. Arthritis Rheum. 33, 336–351 (2004). 15, 94–101 (2006). Rheum. Dis. 65, 385–390 (2006).
89. Puri, P. K., Lountzis, N. I., Tyler, W. & Ferringer, T. 104. Buchanan, N. M. et al. Hydroxychloroquine and 119. Collins, G. B. & McAllister, M. S. Chloroquine
Hydroxychloroquine-induced hyperpigmentation: lupus pregnancy: review of a series of 36 cases. psychosis masquerading as PCP: a case
the staining pattern. J. Cutan. Pathol. 35, Ann. Rheum. Dis. 55, 486–488 (1996). report. J. Psychoactive Drugs 40, 211–214
1134–1137 (2008). 105. Costedoat-Chalumeau, N. et al. Safety of (2008).
90. Herman, S. M., Shin, M. H., Holbrook, A. & hydroxychloroquine in pregnant patients with
Rosenthal, D. The role of antimalarials in the connective tissue diseases: a study of one
exacerbation of psoriasis: a systematic review. hundred thirty-three cases compared with a Author contributions
Am. J. Clin. Dermatol. 7, 249–257 (2006). control group. Arthritis Rheum. 48, 3207–3211 S-J. Lee and E. Silverman researched the data for the
91. Bracamonte, E. R., Kowalewska, J., Starr, J., (2003). article. S-J. Lee and J. M. Bargman were the principal
Gitomer, J. & Alpers, C. E. Iatrogenic 106. Carvalheiras, G. et al. Pregnancy and systemic contributors to writing the article and discussing its
phospholipidosis mimicking Fabry disease. Am. lupus erythematosus: review of clinical features content, although E. Silverman was also involved in
J. Kidney Dis. 48, 844–850 (2006). and outcome of 51 pregnancies at a single these aspects of the manuscript. J. M. Bargman, and
92. Costedoat-Chalumeau, N. et al. Very low blood institution. Clin. Rev. Allergy Immunol. 38, to a lesser extent S-J. Lee, participated in the review
hydroxychloroquine concentration as an 302–306 (2010). and/or editing of the manuscript before submission.

NATURE REVIEWS | NEPHROLOGY VOLUME 7  |  DECEMBER 2011  |  729


© 2011 Macmillan Publishers Limited. All rights reserved

You might also like