REVIEWS New insights into mechanisms of therapeutic effects of antimalarial agents in SLE Daniel J. Wallace, Vineet S. Gudsoorkar, Michael H. Weisman and Swamy R. Venuturupalli Abstract | Antimalarial agents have routinely been used for the treatment of systemic lupus erythematosus (SLE) for over 50 years. These agents continue to enjoy success as the initial pharmacotherapy for SLE even in the era of targeted therapies. Antimalarial agents have numerous biological effects that are responsible for their immunomodulatory actions in SLE. Their inhibitory effect on Toll-like receptor-mediated activation of the innate immune response is perhaps the most important discovery regarding their putative mechanism of action, but some other, previously known properties, such as antithrombotic and antilipidaemic effects, are now explained by new research. In the 1980s and 1990s, these antihyperlipidaemic and antithrombotic effects were demonstrated in retrospective clinical studies, and over the past few years prospective studies have confirmed those findings. Knowledge about the risk–benefit profile of antimalarial agents during pregnancy and lactation has evolved, as has the concept of retinal toxicity. Antimalarial agents have unique disease-modifying properties in SLE and newer iterations of this class of anti-inflammatory agents will have a profound effect upon the treatment of autoimmune disease. Wallace, D. J. et al. Nat. Rev. Rheumatol. 8, 522–533 (2012); published online 17 July 2012; doi:10.1038/nrrheum.2012.106 Introduction As medicinal agents, antimalarial agents have been prescribed for over 500 years, and their potential uses are still being explored. Literary references to quinine —the first effective treatment for malaria—date back to the 16th century, although Payne reported its evidencebased use for cutaneous lupus for the first time in 1894.1 Quinacrine (also marketed as mepacrine or atabrine) was patented in 1930, and was used by four million US and allied soldiers during World War II. Studies in this cohort of subjects yielded observational evidence that quinacrine ameliorated inflammatory arthritis and rash. This discovery led to further research that marked the transition of antimalarial agents to the treatment of rheumatic diseases. Chloroquine was first synthesized in 1943 and hydroxychloroquine came on the market in 1955.1 This Review presents new insights based on recent advances in our understanding of the mechanisms of action of antimalarial agents. Pharmacology of antimalarial agents Division of Rheumatology, Cedars– Sinai Medical Center, 8700 Beverly Boulevard, Becker B‑131, Los Angeles, CA 90048, USA (D. J. Wallace, V. S. Gudsoorkar, M. H. Weisman, S. R. Venturupalli). Chloroquine and hydroxychloroquine are weakly basic 4‑aminoquinoline compounds. Hydroxychloroquine differs from chloroquine by a hydroxyl group attached to a side chain. Quinacrine is an acridine compound that differs from chloroquine by the presence of an extra benzene ring (Figure 1).2 The clinically important aspects of pharmacology and metabolism of antimalarial agents are summarized in Box 1.3,4 Correspondence to: D. J. Wallace [email protected] Competing interests The authors declare no competing interests. 522 | SEPTEMBER 2012 | VOLUME 8 In general, chloroquine is 2–3 times more potent than hydroxychloroquine,5 and the risk of retinopathy is lower with the latter.6 For these reasons, hydroxy chloroquine is preferred over chloroquine in rheumatology practice. Commercially available tablet preparations of hydroxychloroquine sulfate contain 155 mg base equivalent in a 200 mg tablet, and tablets of chloroquine phosphate are available as 500 mg (300 mg base equivalent) and 250 mg (150 mg base equivalent) doses. Chloroquine hydrochloride is available in some countries as 100 mg tablets containing 80 mg base equivalent. Quinacrine is not commercially manufactured in the USA but can be ordered from pharmacies that compound it. Blood concentrations of hydroxychloroquine can independently predict exacerbation in patients with SLE;7 therefore, measurements of hydroxychloroquine levels in whole blood could help to identify ‘at-risk’ patients and optimize treatment. Owing to hydroxychloroquine’s long elimination half-life, routine measurements of serum hydroxychloroquine concentration can also serve as a marker of treatment adherence.8 Although these tests are not yet routinely ordered by rheumatologists in the USA, many laboratories around the world perform these tests using high-performance liquid chromatography. Mechanisms of action Antimalarial agents exert their effects via multiple molecular pathways. Some well-known effects are listed in Box 2,9 and the principal modes of action with new insights into their mechanisms are discussed below. www.nature.com/nrrheum © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS Interference with lysosome function Chloroquine and hydroxychloroquine are weak bases that have an affinity for the acidic lysosome (‘lysosomo tropic’ action).10 These agents alter the endolysosomal pH and interfere with acidification of lysosomes. In T cells, lysosomes perform the dual function of degradation of endocytosed material and participation in apoptosis of antigen-presenting cells (APCs). These functions, if impaired, can affect the cellular immune response and APC function, and lead to several downstream effects such as inhibition of cytokine production, especially IL‑1, IL‑ 6 and TNF. IL‑6 is an important inflammatory mediator that stimulates B‑cell differentiation leading to subsequent antibody response, and is considered an important pathogenic mediator in SLE. Our group has demonstrated that hydroxychloroquine exerts a long-lasting suppressive effect on levels of IL‑6 possibly of macrophage–monocyte origin as opposed to lymphocytic origin.11 This effect has been confirmed independently by other studies.12,13 The exact nature and reason for a preferential effect of hydroxychloroquine on IL‑6 is not clear. Possibly, in addition to inhibition of antigen presentation, antimalarial agents also downregulate mRNA expression of cytokines at the transcriptional level.14 TLR-associated mechanisms Perhaps the most important advance in our understanding of antimalarial agents is knowledge of their antagonistic effect on the nucleic-acid sensing Toll-like receptors (TLRs). The presence of antibodies against nucleic acids is a hallmark of many autoimmune diseases, including SLE. Ineffective clearance of apoptotic cellular material exposes intracellular contents such as nucleic acids to the immune system and invokes an autoimmune response, as evident from animal models of DNase-deficient mice being prone to developing antinuclear antibodies and SLE-like disease,15 and from the observation of reduced activity of serum DNase‑I in patients with active SLE.16 TLRs are type‑I transmembrane receptors that form the early defense mechanism against foreign organisms. These receptors are germline coded during the evolution of species and are geared to recognize specific molecular patterns associated with pathogenic species. Upon exposure to a pathogen, TLRs activate more-specific pathways of the acquired immune response. Nucleic acid-sensing TLRs (TLR3, TLR7, TLR8 and TLR9) are located in the intracellular compartments to minimize accidental exposure to self-nucleic material; these TLRs are activated only when foreign nuclear material is presented to them by specialized intermediate molecules that facilitate the delivery of antigens to the intracellular compartment, such as Fcγ receptors on dendritic cells or B‑cell receptors on the surface of B cells. Activated TLRs, via adaptor molecules such as MyD88 (for TLR7 and TLR9) and TRIF (for TLR3), stimulate the production of type I interferons and proinflammatory cytokines (Figure 2).17 Plasmacytoid dendritic cells (pDCs) have a unique ability to couple the signaling pathways of TLR7 and Key points ■■ Antimalarial agents are the cornerstone agents in the clinical management of systemic lupus erythematosus ■■ Toll-like receptor (TLR)-antagonism has emerged as an important mechanism of action of antimalarial agents ■■ The antilipidaemic, photoprotective and antiproliferative effects of chloroquine, hydroxychloroquine and quinacrine are in part explained by TLR antagonism ■■ Antimalarial agents also act by several additional molecular mechanisms, the understanding of which continues to evolve ■■ Antimalarial agents are generally safe, effective and clinically useful in almost all patients with systemic lupus erythematosus ■■ These drugs offer considerable promise for treating a variety of immune-mediated as well as nonimmune diseases, and have exciting potential N Cl NH H3C N H3C NH H3C Chloroquine H3C N Cl Hydroxychloroquine N H3C H3C OH N H3C Quinacrine H3C NH O Cl CH3 N Figure 1 | Chemical structures of antimalarial agents.2 TLR9, which leads to the production of large quantities of type I interferons and substantially increases transcription of type I interferon genes.18 IFN‑α has a crucial role in the pathogenesis of SLE. IFN‑α levels are high in patients with SLE and IFN genes have been known to be upregulated in this population, an effect referred to as the ‘interferon signature’ of SLE. 19 pDCs are considered the main source of IFN‑α. 20 The novel concept of NETosis (Box 3) hypothesizes that neutrophils might play an equally important part in interferon secretion by stimulating pDCs.21 Importantly, TLRs are central to both these interferon pathways. In pDCs, TLR activation increases IFN‑α synthesis, as described above. In the case of neutrophils, evidence suggests that not only is pDC activation by neutrophil extracellular traps (NETs) mediated via TLRs, but also the inhibition of TLR9 reduces activation of pDCs by NETs.21 Evidence that antimalarial agents act by blocking the activation of TLRs comes from many sources. Unmethylated CpG motifs are seen in bacterial DNA whereas vertebrate DNA contains more of the methy lated fraction, 22 and unmethylated motifs evoke a stronger immune response than their methylated counterparts.22 Antimalarial agents have been shown in animal studies and in vitro models to antagonize immune stimulation by CpG-DNA (a ligand for TLR9) NATURE REVIEWS | RHEUMATOLOGY VOLUME 8 | SEPTEMBER 2012 | 523 © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS Box 1 | Pharmacology and metabolism of antimalarial agents Absorption Chloroquine, hydroxychloroqine and quinacrine all have very good bioavailability after oral administration. Quinacrine can be given intralesionally. The drugs are distributed extensively in tissues; they have an affinity for pigmented tissues and are not retained in adipose tissue. Metabolism Hepatic metabolism by cytochrome P450 enzymes is the primary mode of metabolism and results in S‑enantiomers and R‑enantiomers. Of the two types, the S‑enantiomer of hydroxychloroquine has a shorter half-life and lower blood levels. 21–47% of the dose is excreted without being metabolized. Excretion 40–50% of the dose is excreted renally, and small amounts are excreted in faeces, through the skin, and in breast milk. Antimalarial agents have a long elimination half-life (approximately 40 days) but are detected in tissues for prolonged periods (up to 5 years).103 Box 2 | Well-known effects of antimalarial agents9 ■■ ■■ ■■ ■■ ■■ ■■ ■■ ■■ ■■ ■■ ■■ Inhibit phospholipase A2 and phospholipase C Stabilize lysosome membranes Decrease production of oestrogen Hypoglycaemic Quinidine-like cardiac actions Antimicrobial effects Block graft-versus-host reaction* Induce apoptosis* Antioxidant actions: block superoxide release* Antiproliferative effects* Dissolve circulating immune complexes* *These effects occur at levels higher than those reached with routine clinical use in rheumatology practice. and inhibit CpG-DNA-induced synthesis of IL‑6 and TNF.24,25 Additionally, chloroquine inhibits immune stimulation by small nuclear RNA (a ligand for TLR7) and subsequent production of IFN‑α.26 Taken together, this evidence suggests that antimalarial agents antagonize TLR-mediated immune responses and synthesis of interferon and inflammatory cytokines (Box 4). The exact nature of this antagonism is not clear and could be either noncompetitive (by pH alteration) or competitive (by structural binding). In order to function, intracellular TLRs require an acidic pH,27 probably because they are proteolysed by acid-dependent proteases28 that function normally within the acidic endoplasm of lysosomes. Antimalarial agents, by altering the lysosomal pH, possibly prevent functional transformation of intracellular TLRs and inhibit their activation. Research published in 2011 proposed that TLR antagonism by antimalarial agents might not be driven entirely by endosomal pH alterations but by mechanical inhibition, whereby antimalarial agents structurally bind to nucleic acids and mask their TLR-binding epitopes.29 Other actions relevant to SLE In addition to the effects of antimalarial agents on TLRs, these compounds have several additional actions that are relevant to SLE (Figure 3). Recent discoveries regarding the mechanisms of these actions, concentrating on those made in the period 2000–2012, are discussed below. 524 | SEPTEMBER 2012 | VOLUME 8 Ultraviolet light absorption A well-described action of antimalarial agents is ultra violet light absorption, and exposure to ultraviolet light is a proven risk factor for cutaneous lupus lesions. 30 Ultraviolet radiation can induce local inflammation and cell injury to keratinocytes, leading to cell death and possibly TLR activation. Antimalarial agents might act by negating this ultraviolet radiation-induced inflammation. Chloroquine concentration in the epidermis is 5–15 times higher than the dermis,31 probably owing to its affinity for melanin. This high concentration facilitates local anti-inflammatory actions of antimalarial agents such as inhibition of antigen presentation and cytokine synthesis. Additionally, antimalarial agents activate transcription of the c‑Jun gene, which is postulated to be a component of the early protective response to ultraviolet light.32 Anti-lipidaemic effects Lipid-lowering actions of antimalarial agents in patients with SLE were demonstrated by our group in the 1980s and 1990s in retrospective studies.33 Antimalarial agents have been suggested to inhibit hydrolysis of internalized cholesterol esters through their lysosomotropic action.34 Chloroquine upregulates transcription of LDL-receptor genes and probably affects the activity of HMG-CoA reductase, a rate limiting enzyme in lipid metabolism.35 Furthermore, over the past decade it has become increasingly apparent that TLRs play a major part in lipid metabolism. TLR9-mediated stimulation of perilipin‑3 increases lipid accumulation inside macrophages. 36 Other studies have shown that TLRs have an important role in atherogenesis.37 These discoveries provide a possible mechanistic explanation for the anti-lipidaemic effects of antimalarial agents. Anti-angiogenic effects Lesiak et al.38 demonstrated in vivo that chloroquine inhibits angiogenesis by reducing expression of vascular endothelial growth factor and by interfering with CD34 glycoprotein, thereby improving cutaneous lesions. TLRs (specifically TLR2, TLR4, TLR7 and TLR9) strongly upregulate expression of vascular endothelial growth factor upon stimulation with unmethylated CpG motifs in the presence of adenosine receptors which then promote angiogenesis.39 Thus, although no evidence yet suggests that antimalarial agents block angiogenesis by antagonizing TLRs, this putative mechanism of action remains a possibility. Antithrombotic effects Antiphospholipid syndrome frequently coexists with SLE. In this condition, antiphospholipid antibodies (aPL) disrupt the protective covering over the phospholipid bilayer of the cell membrane formed by the natural anticoagulant annexin A5. This loss of protection exposes the phospholipids to coagulation factors, leading to inappropriate activation of the coagulation pathway. Hydroxychloroquine abrogates the stimulatory effect of aPL on platelet aggregation even in the presence of a thrombin agonist,40 and reverses the antibody-mediated www.nature.com/nrrheum © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS Antimalarial agent masks TLR-binding epitopes of nucleic acids pDC Fc receptor Nucleic acid bound antibody Lysosome Endocytosis 3 ssRNA 2 Acid pH activation Antimalarial agent DNA TLR7 TLR9 Endocytosis MyD88 1 Inactive TLR Nucleic acid bound antibody 4 IRF5–IRF7 Nucleus Vesicle 5 Positive feedback amplification Positive feedback amplification Golgi ER Positive feedback amplification (dendritic cell maturation) Anti-dsDNA antibodies 6 IFN-α secretion NETs IFN-α/βR2 8 7 IFN-α/βR2 9 IFN-α/βR2 B cell Antibody secretion and immune complex formation T cell PMN cell Antigen presentation IL-10 production TH1 cells TREG cells NETosis Figure 2 | Role of antimalarial agents in TLR-mediated innate immune pathways in SLE. TLRs exit Golgi complex in an inactive state (1), and are cleaved and activated in endosomes by acid-dependent proteases (2), thus interacting with nucleic acids presented to endosomal compartments by specialized receptors such as Fcγ receptors (3). Putative actions of antimalarial agents involve blocking (2) or (3), or both. Upon interaction with the respective ligands, TLRs stimulate the synthesis of type I interferon (4,5). The release of IFN‑α (6) has widespread effects on both the innate and adaptive immune systems; most importantly, it stimulates gene expression of TLRs as well as feedback activation of more pDCs, perpetuating a vicious cycle.104 IFN‑α promotes T‑cell survival, upregulation of the TH1 response, proliferation of CD8+ cells and suppression of TREG cells (7). IFN‑α affects B cells by causing maturation of plasmablasts, immunoglobulin classswitching, and increased antibody secretion (8). IFN‑α also stimulates the development of memory B cells and induces BAFF, a B‑cell maturation and survival factor.104 In the presence of immune complexes and interferons, PMNs undergo NETosis and NETs in turn stimulate pDCs—possibly via TLRs (9). IFN‑α also promotes maturation of monocytes into dendritic cells, which are more efficient in antigen processing and presentation. It upregulates expression of co-stimulatory molecules and HLA, and stimulates synthesis of IL‑10105 and IL‑12 by dendritic cells.106 Abbreviations: dsDNA, doublestranded DNA; ER, endoplasmic reticulum; IRF, interferon regulatory factor; NET, neutrophil extracellular trap; pDC, plasmacytoid dendritic cell; PMN, polymorphonuclear cell; SLE, systemic lupus erythematosus; ssRNA, single-stranded RNA; TH1 cell, type 1 T helper cell; TLR, Toll-like receptor; TREG cell, regulatory T cell. disruption of the annexin A5 shield.41 These observations provide a new rationale for the use of antimalarial agents as preventive agents in aPL-positive patients. MMP–TIMP modulation Matrix metalloproteinases (MMPs) are a group of enzymes involved in extracellular matrix remodeling. NATURE REVIEWS | RHEUMATOLOGY VOLUME 8 | SEPTEMBER 2012 | 525 © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS Box 3 | NETosis NETs are chromatin structures containing antimicrobial peptides formed by neutrophils and are capable of trapping microorganisms, thus constituting a form of innate immunity. A process of neutrophil cell death, distinct from apoptosis and necrosis, results in release of these NETs and is hence referred to as NETosis. As these NETs are rich in antigenic structures, including self-nucleic acids, it has been suggested that activation of plasmacytoid dendritic cells by NETs is a putative pathogenic mechanism in systemic lupus erythematosus, and NET formation has been implicated in progression of endothelial damage and renal disease.107 Abbreviation: NET, neutrophil extracellular trap. Box 4 | TLR-dependent effects in SLE ■■ TLR3, TLR7 and TLR9 are nucleic acid-sensing TLRs that have been implicated in the pathogenesis of SLE ■■ By activating the immune system, TLRs upregulate production of inflammatory cytokines including IFN‑α ■■ Activation of TLRs is a controlled step requiring an acidic pH ■■ Antimalarial agents antagonize TLR activation possibly by altering pH or via competitive inhibition ■■ TLR antagonism results in inhibition of IFN‑α expression and thereby prevents activation of multiple IFN‑α-mediated pathways and resultant widespread inflammatory damage Abbreviations: SLE, systemic lupus erythematosus; TLR, Toll-like receptor. These enzymes are secreted by monocytes, macrophages, neutrophils, fibroblasts, endothelial cells and various tumour cells. Tissue inhibitors of metalloproteinases (TIMPs) are counter-regulatory enzymes that inhibit MMPs and maintain homeostasis of the extracellular matrix. An imbalance between MMPs and TIMPs has been suggested to have a pathogenic role in autoimmune diseases. 42 Quinacrine and chloroquine have been shown to inhibit MMPs in vitro and in vivo, respectively. Stuhlmeier et al.43 demonstrated that quinacrine, but not chloroquine, downregulates the transcription of mRNA thereby inhibiting the synthesis of MMP‑1, MMP‑2 and MMP‑8. Lesiak et al.44 found that blood levels of MMP‑9 and TIMP‑1 were significantly elevated in 25 patients with SLE in comparison with 25 healthy individuals; following 3 months of treatment with chloroquine, MMP‑9 levels were significantly reduced and TIMP levels were increased in the patients with SLE, suggesting that chloroquine modulated MMP–TIMP inter actions. Studies have suggested that activation of TLR9 (and TLR4) promotes MMP production.45 Chloroquine possibly acts through a TLR-mediated pathway whereas quinacrine acts at the mRNA transcription level to achieve similar effects on MMP–TIMP modulation. Other mechanistic effects of quinacrine Similar to other, weakly basic antimalarial agents, quinacrine also shows tropism for lysosomes.46 Additionally, 526 | SEPTEMBER 2012 | VOLUME 8 quinacrine exerts an inhibitory effect on B‑cell-activating factor (also known as BLyS and TNF ligand superfamily, member 13b)—a survival factor for B cells and an important therapeutic target in SLE.47 Quinacrine stabilizes the cell membrane and inhibits the enzymatic activity of cytosolic phospholipase A2, thereby inhibiting the arachido nic acid pathway and eventually resulting in inhibition of inflammatory processes such as chemotaxis, cell adhesion and platelet aggregation.48 Clinical effects in SLE Antimalarial agents have both disease-modifying effects and benefits for specific outcome measures in SLE (Box 5). The effects of these agents on various disease outcomes are discussed in the following sections. Effects on SLE onset, progression and survival In 1991, the Canadian Hydroxychloroquine Study Group demonstrated that discontinuation of hydroxychloro quine increased the relative risk of a clinical flare by 2.5 times over a 6‑month period, compared with maintenance of hydroxychloroquine therapy. Also, the risk of severe disease exacerbation was 6.1 times higher for patients on placebo as compared with those who continued hydroxychloroquine.49 Several subsequent clinical trials over the past decade have confirmed the clinical benefits of hydroxychloroquine in SLE patients (Table 1).50–55 In 2002, Molad et al.50 reported that, in a cohort of 151 patients with SLE, hydroxychloroquine use corre lated negatively with Systemic Lupus International Collaborating Clinics–American College of Rheumato logy (SLICC–ACR) damage index score, and positively with damage-free survival measured over a period of 3.5 years. In the LUMINA (Lupus in Minorities, Nature versus Nurture) cohort, hydroxychloroquine use was associated with reduced accrual of new damage. Patients with no evidence of prior damage benefited the most,51 which seems to suggest that early initiation of hydroxychloroquine potentially maximizes its benefits. Hydroxychloroquine also improves overall survival: in 608 patients, 5% of deaths occurred in patients using hydroxychloroquine as compared with 17% in patients not receiving hydroxychloroquine.52 Ruiz-Irastroza et al.53 also reported a survival benefit associated with use of antimalarial agents in 232 patients with SLE from Spain. In 2007, a retrospective chart review of 130 US military personnel demonstrated that hydroxychloroquine use in patients before diagnosis of SLE was associated with a delay in the onset of SLE from the time of appearance of initial symptoms (median time 1.08 years), as compared with no use of hydroxychloroquine (median time 0.29 years). The same paper reported that hydroxy chloroquine use before SLE diagnosis was associated with lesser likelihood of developing proteinuria, leucopenia or lymphopenia.54 In the longitudinal GLADEL (Grupo Latino Ameri cano de Estudio del Lupus Eritematoso) cohort of 1,480 patients with SLE, antimalarial agents were determined to have a beneficial effect on survival, possibly in a www.nature.com/nrrheum © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS TLR-independent mechanisms of antimalarial therapy UV protection Antilipidaemic effects ■ Local anti■ Act at the lipid inflammatory receptor level to effects and regulate enzyme upregulation of activity and possibly the protective also through TLRs c-Jun-encoding gene ■ Reduce LDL, VLDL ■ Control of photoand cholesterol, sensitivity and and increase cutaneous lupus HDL levels Antiangiogenic effects ■ Reduce epidermal expression of VEGF ■ In vitro antiproliferative and apoptotic effects on ECs ■ Possible mode of action in discoid lupus MMP−TIMP modulation PLA2 inhibition BAFF inhibition ■ Inhibit expression of MMP-1, MMP-2, MMP-8, MMP-9 ■ Regulate ECM homeostasis ■ Inhibit excess ECM breakdown ■ Cell membrane stabilization ■ Inhibit arachidonic acid pathway and downstream synthesis of inflammatory mediators ■ Reduce maturation and survival of B cells, including autoreactive B cells Antithrombotic effects ■ Inhibit platelet aggregation ■ Block interaction between platelets and coagulation factors ■ Reduce thrombotic events in patients with SLE ■ Possible role in primary thromboprophylaxis in APS and SLE Figure 3 | Summary of TLR-independent mechanisms of antimalarial agents. Antimalarial agents have several actions that are relevant to SLE and independent of their effects on TLRs. Abbreviations: APS, antiphospholipid syndrome; BAFF, B-cell activating factor; EC, endothelial cell; ECM, extracellular matrix; MMP, matrix metalloproteinase; PLA2, phospholipase A2; SLE, systemic lupus erythematosus; TIMP, tissue inhibitor of metalloproteinases; TLR, Toll-like receptor; UV, ultraviolet; VEGF, vascular endothelial growth factor. time-dependent manner.55 In other words, patients who used antimalarial agents for a longer time had lower mortality than patients who used them for a shorter time. Thus, early initiation of antimalarial agents in symptomatic patients delays the onset and progression of SLE and improves overall survival. Effects on specific outcome measures In addition to its benefits for survival, hydroxychloroquine has been shown to improve morbidity in lupus by affecting specific outcome measures such as integument damage, thrombosis, lipid profile, glycaemic status, infections and renal failure (summarized in Table 2). Data from 580 SLE patients in the LUMINA cohort suggests that hydroxychloroquine use is associated with less integument damage. The cumulative probability of developing integument damage at 5 years was 5% for patients on hydroxychloroquine as opposed to 24% for those not taking hydroxychloroquine.56 Since our initial report in 1987,57 numerous studies have seemingly confirmed that hydroxychloroquine is thromboprotective in SLE.53,58–62 The lipid-lowering propert ies of hydroxychloroquine, chloroquine and quinacrine have been demonstrated in retrospective as well as blinded, prospective study settings.33,63–65 Data from the Baltimore lupus cohort demonstrated that the mean blood glucose level in hydroxychloroquine users was significantly lower than patients not taking hydroxychloroquine.66 Hydroxychloroquine use is also protective against renal damage 67,68 and major infections in SLE. 69 Additionally, evidence suggests that antimalarial agents may prevent bone mass loss, subclinical atherosclerosis and development of cancer in SLE (moderate-to-low level evidence).70 Special considerations in SLE Although antimalarial agents have been in widespread clinical use for decades, they are often used suboptimally in SLE.71 In this section, we present an update on the clinical use of these agents along with ‘clinical pearls’ we have employed over the years in using these agents. Box 5 | Summary of effects of antimalarial agents in SLE ■■ Hydroxychloroquine has sustained beneficial effects on overall survival, disease-free survival and damage accrual ■■ Hydroxychloroquine delays onset of SLE and reduces the number of and severity of clinical flares ■■ Early use of hydroxychloroquine maximizes these benefits ■■ Antimalarial agents improve survival in a time-dependent manner ■■ Use of hydroxychloroquine protects against thrombosis, even in aPL-positive patients ■■ All three antimalarial agents have lipid-lowering properties that are also apparent in patients taking corticosteroids ■■ Antimalarial agents have beneficial effects on glycaemic status in patients with SLE, and this benefit possibly increases with duration of use ■■ Antimalarial agents have a protective effect against renal damage and major infections in SLE Abbreviations: aPL, antiphospholipid antibody; SLE, systemic lupus erythematosus. Antimalarial agents for cutaneous lupus Hydroxychloroquine, chloroquine and quinacrine have all been mainstays in the treatment of cutaneous lupus and are considered first-line systemic agents for the treatment of widespread skin manifestations. Hydroxychloroquine is most often used as the initial agent, and more than 50% of the patients respond to therapy with hydroxychloroquine alone.72 If hydroxychloroquine monotherapy fails, the addition of quinacrine could be beneficial. If the response is still inadequate, we recommend a combination of chloroquine and quinacrine. A combination of hydroxy chloroquine and quinacrine improves Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) score, enhances response rate in nonresponders and possibly reduces mean daily dose of steroids.73,74 Antimalarial agents and smoking Tobacco smoking is thought to reduce the efficacy of antimalarial agents in lupus. This interference is dose- dependent (that is, the number of cigarettes smoked per day is inversely proportional to the degree of clinical NATURE REVIEWS | RHEUMATOLOGY VOLUME 8 | SEPTEMBER 2012 | 527 © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS Table 1 | Beneficial clinical effects of antimalarial agents for patients with SLE Reference Study design Population Findings Canadian Hydroxychloroquine Study Group (1991)49 Double-blind, randomized, placebo-controlled 47 patients with clinically stable SLE assigned to continue hydroxychloroquine therapy (n = 25) or to receive placebo (n = 22) Discontinuation of hydroxychloroquine increased the relative risk of a clinical flare Risk of severe disease exacerbation was 6.1 times higher for patients on placebo as compared with those who continued hydroxychloroquine Molad et al. (2002)50 Cohort 151 patients with SLE Hydroxychloroquine use correlated negatively with damage index score, and positively with damage-free survival Fessler et al. (2005)51 Observational cohort (LUMINA) 518 patients with SLE Hydroxychloroquine use associated with reduced accrual of new damage Patients with no evidence of prior damage benefited the most Alarcon et al. (2007)52 Case–control study within the LUMINA cohort 608 patients with SLE (of whom 61 were deceased) Hydroxychloroquine use increases overall survival (5% of hydroxychloroquine users died vs 17% of non-users) Ruiz-Irastorza et al. (2006)53 Observational prospective cohort 232 patients with SLE from Spain Survival benefit associated with use of antimalarial agents James et al. (2007)54 Retrospective chart review 130 US military personnel Use of hydroxychloroquine before diagnosis associated with delayed onset of SLE, and with lesser likelihood of developing proteinuria, leucopenia or lymphopaenia Shinjo et al. (2010)55 Longitudinal cohort (GLADEL) 1,480 patients with SLE Beneficial effect of antimalarial agents on survival, with greater benefit associated with longer duration of use Abbreviation: SLE, systemic lupus erythematosus. response to antimalarial therapy) and this effect is especially pronounced in cutaneous lupus.75,76 Conversely, patients who quit smoking seem to respond better to treat ment with antimalarial agents.77 The exact mechanism by which smoking interferes with the efficacy of antimalarial agents is not well understood. Tobacco smoke is a potent inducer of cytochrome p450 enzymes (Box 1);78 one hypo thesis is that it interferes with metabolism of antimalarial agents. However, Leroux et al.78 did not find any correlation between blood concentrations of the metabolites of hydroxychloroquine and smoking habits in 223 patients, which suggests that the interference might involve other, indirect pathways. Other possible pathways common to the mechanisms of antimalarial agents include MMP– TIMP modulation via IL‑10 and TNF,79,80 and induction of cytokines such as IL‑6 by tobacco smoke.81 On the other hand, a few reports found no associ ation between smoking and response to antimalarial agents.82,83 Also, smoking is independently associated with cutaneous lupus,84 thus confounding the picture. Smoking is also associated with skin lesions of lupus that are refractory to conventional therapies not limited to antimalarial agents,85 suggesting the possibility that the interference caused by smoking is not restricted to antimalarial agents. However, this perception was challenged in 2012 by a prospective study in patients with cutaneous lupus, which reported that smokers, surprisingly, had a better response when treated with antimalarial agents alone.86 Clearly, more prospective clinical studies free of confounders and biases, as well as better biological models, are required to understand the exact relationship between smoking and antimalarial agents. Regardless, we 528 | SEPTEMBER 2012 | VOLUME 8 advise that all patients, irrespective of their therapeutic regimen, should be encouraged to quit smoking. Antimalarial agents in pregnancy and lactation Whilst we recommend a detailed conversation with each individual patient about the potential risks of any therapeutic agent in pregnancy, several studies have assessed the effect of hydroxychloroquine on pregnancies in women with SLE. In case series published in the 1980s and 1990s, Parke87,88 suggested that hydroxychloroquine was safe to use in pregnant patients with SLE. Buchanan89 reported that exposure to hydroxychloroquine during pregnancy did not have any teratogenic effects. Data regarding 257 pregnancies from the Hopkins cohort showed no fetal abnormalities directly attributable to hydroxychloroquine; this study also reported that stopping hydroxychloroquine during or just before pregnancy resulted in increased disease activity.90 Hydroxychloroquine use during pregnancy by patients positive for anti-Ro/SSA or anti-La/SSB antibodies is associated with reduced risk of developing the cardiac manifestations of neonatal lupus.91 Thus, hydroxychloroquine use during pregnancy results in favourable maternal outcomes and there has been no evidence of visual or auditory abnormalities in the foetus, either congenital or developmental.92 Hydroxychloroquine is secreted in breastmilk. Hydroxyc hloroquine ingestion by breastfed infants corresponds to 0.06–0.20 mg/kg per day after adjustment for body weight. Compared with the adult therapeutic dose of 6.5 mg/kg per day, the amount transferred can be considered a low dose unlikely to cause any significant toxic effects.93 www.nature.com/nrrheum © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS Table 2 | Beneficial clinical effects of antimalarial agents on specific SLE outcomes Reference Study design Population Findings Ho et al. (2005)58 Observational cohort (LUMINA) 442 patients with SLE and known aPL status Hydroxychloroquine use protective against thrombosis in univariate analysis (OR 0.536) Ruiz-Irastroza et al. (2006)53 Observational prospective cohort 232 patients with SLE Antimalarial use protective against thrombosis (HR 0.28) Sisó et al. (2008)59 Cohort 206 patients with lupus nephritis, 56 of whom previously used antimalarial agents Previous hydroxychloroquine use protective against thrombosis (5% in hydroxychloroquine users vs 17% in non-users) Kaiser et al. (2009)60 Cohort 1,930 patients with SLE Hydroxychloroquine use thromboprotective after propensity analysis (OR 0.62) Tektonidou et al. (2009)61 Longitudinal case–control 144 aPL positive patients with SLE matched with 144 aPL-negative patients with SLE Hydroxychloroquine use protective against thrombosis in both groups (aPL positive patients: HR per month 0.99; aPL negative patients: HR per month 0.98) Jung et al. (2010)62 Nested case– control 162 patients with SLE, 54 of whom had a history of thrombotic event Ever-use of antimalarial agents thromboprotective (OR 0.32) Overall 68% reduction in thrombotic events in users of antimalarial drugs Wallace et al. (1990)33 Retrospective 155 patients with SLE or RA subdivided according to exposure to hydroxychloroquine and/or steroids Addition of hydroxychloroquine to steroids reduced levels of LDL cholesterol and triglyceride by 15% compared with steroids alone Petri et al. (1994)63 Longitudinal cohort (Hopkins) 264 patients with SLE Hydroxychloroquine use associated with an 8.94mg% reduction in serum total cholesterol level Kavanaugh et al. (1997)64 Double blind, prospective 17 patients with SLE Hydroxychloroquine associated with a mean decrease of 11.6 mg/dl in serum total cholesterol level Borba et al. (2001)65 Case–control 60 patients with SLE (subdivided according to exposure to hydroxychloroquine and steroids) and 30 healthy controls Chloroquine use associated with elevated level of HDL cholesterol as compared with no therapy, and with lower level of VLDL cholesterol in the group taking chloroquine plus steroids Pons-Estel et al. (2009)67 Prospective study in a longitudinal cohort (LUMINA) 203 patients with lupus nephritis without renal damage at baseline Hydroxychloroquine use associated with lower frequency of WHO class IV glomerulonephritis, lower disease activity and lower steroid requirement Hydroxychloroquine protective against renal damage occurrence in full (HR 0.12) and reduced (HR 0.29) models after adjustment for confounding factors Pons-Estel et al. (2012)68 Nested case– control study within the GLADEL cohort 265 patients with SLE and renal disease and 530 controls without renal disease Use of antimalarial agents negatively associated with risk of development of renal disease (OR 0.39) Protective effect persisted after adjustment for confounding factors (OR 0.38) Longitudinal cohort (LUMINA) 580 patients with SLE Hydroxychloroquine use associated with longer time to integument damage (HR 0.23) Cumulative probability of developing integumental damage at 5 years lower in patients taking hydroxychloroquine than those not taking the drug (5% vs 24%) Longitudinal cohort 71 patients with SLE who had serial cohort visits both on and off hydroxychloroquine Mean blood glucose level whilst taking hydroxychloroquine was 84.9 ± 15.2 mg/dl, significantly lower than level whilst off hydroxychloroquine (89.0 ± 21.5 mg/dl) Nested case– control within a prospective cohort 86 patients with SLE and major infections and 166 controls (without major infections) After logistic regression, treatment with antimalarial agents had an independent protective effect against major infections in SLE (OR 0.06) Thrombosis* Lipid profile* Renal damage Integument damage Pons-Estel et al. (2010)56 Glycemic status Petri (1996)66 Major infections Ruiz-Irastorza et al. (2009)69 *Studies included are evidence-based level A or B studies with various study designs. Abbreviations: aPL, antiphospholipid antibody; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus. NATURE REVIEWS | RHEUMATOLOGY VOLUME 8 | SEPTEMBER 2012 | 529 © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS Box 6 | Non-ophthalmologic toxic effects ■■ Common*: nausea, vomiting, pruritus, maculopapular rash, skin & mucosal pigmentation, insomnia, nightmares, nervousness ■■ Less common‡: tinnitus, vestibular changes, neuropathy, psychosis, seizures ■■ Rare: leucopenia, anaemia, porphyria, depigmentation of hair, hair loss, anorexia, diarrhoea, liver dysfunction *Frequency 1–20% with chloroquine use, 1–10% with hydroxychloroquine use. ‡Frequency <5% with chloroquine use, <1% with hydroxychloroquine use. Box 7 | New insights into ophthalmologic toxicity ■■ Retinopathy can be missed in early stages and might be irreversible later ■■ Baseline screening at the beginning of treatment should be followed by annual screening after 5 years of treatment ■■ Pre-existing macular disease should be considered a contraindication for antimalarial therapy; if antimalarial agents must be used, annual screening should start from the onset of treatment ■■ Special consideration should be given to elderly patients and those with kidney or liver disease, entailing annual screening from the onset of treatment; short-statured and obese patients also deserve careful monitoring ■■ Recognize the possibility of ‘outlier’ cases, in which retinopathy occurs even within the safe limits of antimalarial treatment; patients should be made aware of this possibility and provided with risk–benefit counselling ■■ Quinacrine can be used relatively safely in the presence of pre-existing retinopathy Thus, from the available evidence, we opine that hydroxychloroquine is generally safe for use during pregnancy and lactation and should be continued throughout this period since its withdrawal may be associated with worsening of the disease and adverse outcome of the preg nancy. Hydroxychloroquine might also reduce the risk of heart block associated with neonatal lupus. Update on the safety of antimalarial agents Antimalarial agents are generally considered relatively safe and non-toxic as compared with other disease-modifying agents for SLE. Non-ophthalmologic adverse effects of antimalarial agents are listed in Box 6. Advances since 2000 have increased our understanding of the ocular toxicity of antimalarial agents, and these are discussed in detail below and summarized in Box 7. Ocular adverse effects of antimalarial agents include keratopathy in the form of corneal deposits, bull’s eye maculopathy, cycloplegia and posterior cataracts. The incidence rate of keratopathy is more than 50% (up to 90%) with chloroquine, 10% with hydroxychloroquine and approximately 5% with quinacrine.94 Keratopathy is almost always completely reversible without any residual corneal damage and by itself is not an indication for stopping treatment. Retinopathy is seen in 10% of patients receiving chloroquine and in approximately 1% of those 530 | SEPTEMBER 2012 | VOLUME 8 receiving hydroxychloroquine after 7 years;94 it is not a reported consequence of quinacrine use. The adverse effect profile of quinacrine is generally similar to the other two antimalarial agents, but some important differences exist. First, quinacrine does not cause retinopathy or haemolysis in patients deficient in g lucose‑6-phosphate dehydrogenase. Second, quinacrine use has been associated with aplastic anaemia, but infrequently in patients receiving modern, lower doses of quinacrine, thus making aplastic anaemia a very rare event.95 The causal association between use of antimalarial agents and retinopathy has been recognized for many years, but the precise incidence of retinopathy has been a topic of debate. Although it is argued that retinopathy is a rare adverse effect of antimalarial therapy, the sheer number of patients undergoing long-term treatment with these agents makes it an important issue. Much has been speculated about the mechanism of retinal toxicity of antimalarial agents—theories put forward include melanin binding, phototropic effect and alteration of retinal pigment metabolism—but no theories have been conclusively proven. The clinical picture of chronic eye toxicity is bull’s eye maculopathy, which often occurs bilaterally. The patient might have excellent visual acuity in the initial stages despite underlying foveal changes. These changes are almost always reversible if monitoring is performed in accordance with updated guidelines on ophthalmologic monitoring for patients on anti malarial agents published by the American Academy of Ophthalmology in 2011.96 The cumulative dose of antimalarial agents has recently been recognized as an independent risk factor for retinopathy,96 a consideration that is prominent in the American Academy of Ophthalmology guidelines.96 Advanced age, impaired liver and/or renal function and pre-existing macular disease have been identified as additional risk factors.96 Reflecting advances since publication of the previous guidelines in 2002, the new recommendations call for the use of more-advanced and objective tests for eye monitoring. In addition to dilated eye examination, the guidelines recommend automated visual field testing, multifocal electro retinogram, spectral domain coherence tomography and fundus autofluorescence; by contrast, use of Amsler grid testing, fluorescein angiography and fundus photo graphy are no longer recommended. A cumulative dose of 1,000 g hydroxychloroquine is considered the typical safety limit above which caution is advised. This limit is reached within 7 years for patients receiving the usual hydroxychloroquine dose of 400 mg per day. The guidelines advise that dosing should be adjusted according to ideal (lean) body weight and not by actual body weight because antimalarial agents are not retained in adipose tissue; this adjustment is particularly important considering the global rise in the incidence of obesity and the weight gain associated with steroids. Finally, it should be noted that patients with kidney or liver disease and elderly patients carry an increased risk of earlier development of retinopathy.96 www.nature.com/nrrheum © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS Table 3 | Community-based use of antimalarial agents in patients with lupus: authors’ perspective Agent Administration Monitoring Hydroxychloroquine Once‑a-day dose of 5 mg/kg can be considered optimal in most cases, but can be used at 7 mg/kg per day for up to 3 months This dose should be reduced back to 5 mg/kg or less for maintenance therapy This regimen gives a response rate of up to 80% for non-organ-threatening disease and cutaneous lupus In our experience, anti-inflammatory and photoprotective effects are seen by 6–12 weeks Monitor CBC and liver and kidney functions every 3–4 months for the duration of use Perform eye examinations as per AAO guidelines96 Chloroquine Can be used for refractory cutaneous disease at 500 mg per day for 30–60 days followed by 250 mg per day Transition to hydroxychloroquine is advised once a response is seen; however, if transition is not possible, tapering should be achieved by reducing the frequency of administration by one day every week, keeping the dose (mg per day) the same Monitor CBC and perform comprehensive metabolic panel every 3 months Perform eye examinations as per AAO guidelines96 Quinacrine Add-on treatment for refractory skin disease (synergistic with hydroxychloroquine or chloroquine) or as monotherapy for patients with contraindications to these two drugs (for example, those with glucose‑6phosphate dehydrogenase deficiency or pre-existing retinopathy) Starting dose 50 mg per day and then can be adjusted to 25–100 mg per day according to the response observed Should be stopped if no response is seen by 6 weeks or if cytopaenia or lichen planus develops Monitor CBC and perform comprehensive metabolic panel every month for the first 3 months and every 3 months thereafter Eye examination is not required Abbreviation: AAO, American Academy of Ophthalmology. How to use antimalarial agents in SLE As antimalarial agents have disease-modifying properties and can affect inflammatory activity as well as favourably influence quality of life, morbidity and prognosis, most patients with SLE should be treated with one of these agents. Table 3 summarizes how we use antimalarial agents in our patients with SLE. Potential translational insights TLRs have been implicated in a variety of rheumatologic disorders, and both laboratory and clinical research seem to show a promising role for antimalarial agents in the treatment of these diseases.97 Attempts are being made to synthesize small molecule TLR antagonists that can be administered orally or subcutaneously. 98 The enantiospecific metabolism of antimalarial agents provides an opportunity to develop a pure formation of the S‑enantiomer, which could be less toxic, especially to the retina, than current formulations owing to its preferential metabolism and elimination (Box 1). Topical preparations of hydroxychloroquine could be useful in treating cutaneous lupus.99 It has been suggested that patients with genotypes associated with expression of low levels of IL‑10 and high levels of TNF respond best to antimalarial therapy.100 As additional data from genome-wide association studies becomes available, this area of individualized tailoring of medications on the basis of genotype will become more important. Quinacrine has also generated 1. 2. 3. Wallace, D. J. The history of antimalarials. Lupus 5 (Suppl. 1), S2–S3 (1996). Knox, C. et al. DrugBank 3.0: a comprehensive resource for ‘omics’ research on drugs. Nucleic Acids Res. 39, D1035–D1041 (2011). Tett, S. E., McLachlan, A. J., Cutler, D. J. & Day, R. O. Pharmacokinetics and pharmacodynamics of hydroxychloroquine enantiomers in patients with rheumatoid 4. 5. interest in non-immune disorders owing to its anti-prion and multiple anti-tumorigenic actions.101,102 Conclusions Antimalarial agents exert beneficial effects in rheumatic disease through a variety of molecular pathways and have rekindled strong interest within the lupus community. Their antagonist effects on TLRs and cytokines are potentially clinically relevant. In SLE, not only can these drugs control active inflammation, but they also improve survival, prevent organ-specific damage and reduce morbidities. When properly monitored, antimalarial agents have a good safety profile in routine use as well as in special conditions such as pregnancy and lactation. We opine that most patients with SLE should be on antimalarial agents unless contraindicated. Review criteria The MEDLINE database was searched to identify papers published in the English language from 2000 to 2011 using the MeSH terms “(antimalarials OR chloroquine OR hydroxychloroquine OR quinacrine) AND (lupus erythematosus, systemic)”. This search identified studies related to clinical use of antimalarial agents and outcomes. An additional manual search was conducted of the reference lists of these studies to identify other relevant publications. arthritis receiving multiple doses of racemate. Chirality 6, 355–359 (1994). Furst, D. E. Pharmacokinetics of hydroxychloroquine and chloroquine during treatment of rheumatic diseases. Lupus 5 (Suppl. 1), S11–S15 (1996). Scherbel, A. L., Schuchter, S. L. & Harrison, J. W. Comparison of effects of two antimalarial agents, hydroxychloroquine sulfate and chloroquine NATURE REVIEWS | RHEUMATOLOGY 6. 7. phosphate, in patients with rheumatoid arthitis. Cleve. Clin. Q. 24, 98–104 (1957). Block, J. A. Hydroxychloroquine and retinal safety. Lancet 351, 771 (1998). Costedoat-Chalumeau, N. et al. Low blood concentration of hydroxychloroquine is a marker for and predictor of disease exacerbations in patients with systemic lupus erythematosus. Arthritis Rheum. 54, 3284–3290 (2006). VOLUME 8 | SEPTEMBER 2012 | 531 © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. Costedoat-Chalumeau, N. et al. Very low blood hydroxychloroquine concentration as an objective marker of poor adherence to treatment of systemic lupus erythematosus. Ann. Rheum. Dis. 66, 821–824 (2007). Wallace, D. J. Antimalarials—the ‘real’ advance in lupus. Lupus 10, 385–387 (2001). Fox, R. I. Mechanism of action of hydroxychloroquine as an antirheumatic drug. Semin. Arthritis Rheum. 23, 82–91 (1993). Wallace, D. J., Linker-Israeli, M., Hyun, S., Klinenberg, J. R. & Stecher, V. The effect of hydroxychloroquine therapy on serum levels of immunoregulatory molecules in patients with systemic lupus erythematosus. J. Rheumatol. 21, 375–376 (1994). Wozniacka, A., Lesiak, A., Narbutt, J., McCauliffe, D. P. & Sysa-Jedrzejowska, A. Chloroquine treatment influences proinflammatory cytokine levels in systemic lupus erythematosus patients. Lupus 15, 268–275 (2006). Jang, C. H., Choi, J. H., Byun, M. S. & Jue, D. M. Chloroquine inhibits production of TNF-α, IL‑1β and IL‑6 from lipopolysaccharide-stimulated human monocytes/macrophages by different modes. Rheumatology (Oxford) 45, 703–710 (2006). Wozniacka, A. et al. The influence of antimalarial treatment on IL‑1β, IL‑6 and TNF-α mRNA expression on UVB-irradiated skin in systemic lupus erythematosus. Br. J. Dermatol. 159, 1124–1130 (2008). Napirei, M. et al. Features of systemic lupus erythematosus in DNase1-deficient mice. Nat. Genet. 25, 177–181 (2000). Chitrabamrung, S., Rubin, R. L. & Tan, E. M. Serum deoxyribonuclease I and clinical activity in systemic lupus erythematosus. Rheumatol. Int. 1, 55–60 (1981). Marshak-Rothstein, A. Toll-like receptors in systemic autoimmune disease. Nat. Rev. Immunol. 6, 823–835 (2006). Blasius, A. L. & Beutler, B. Intracellular Toll-like receptors. Immunity 32, 305–315 (2010). Vallin, H., Blomberg, S., Alm, G. V., Cederblad, B. & Ronnblom, L. Patients with systemic lupus erythematosus (SLE) have a circulating inducer of interferon-α (IFN-α) production acting on leucocytes resembling immature dendritic cells. Clin. Exp. Immunol. 115, 196–202 (1999). Ronnblom, L. & Alm, G. V. The natural interferon-α producing cells in systemic lupus erythematosus. Hum. Immunol. 63, 1181–1193 (2002). Garcia-Romo, G. S. et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra20. (2011). Bird, A. P., Taggart, M. H., Nicholls, R. D. & Higgs, D. R. Non-methylated CpG-rich islands at the human α-globin locus: implications for evolution of the α-globin pseudogene. Embo J. 6, 999–1004 (1987). Cornelie, S. et al. Direct evidence that Toll-like receptor 9 (TLR9) functionally binds plasmid DNA by specific cytosine–phosphate–guanine motif recognition. J. Biol. Chem. 279, 15124–15129 (2004). Hong, Z. et al. Chloroquine protects mice from challenge with CpG ODN and LPS by decreasing proinflammatory cytokine release. Int. Immunopharmacol. 4, 223–234 (2004). Macfarlane, D. E. & Manzel, L. Antagonism of immunostimulatory CpG-oligodeoxynucleotides by quinacrine, chloroquine, and structurally related compounds. J. Immunol. 160, 1122–1131 (1998). 532 | SEPTEMBER 2012 | VOLUME 8 26. Vollmer, J. et al. Immune stimulation mediated by autoantigen binding sites within small nuclear RNAs involves Toll-like receptors 7 and 8. J. Exp. Med. 202, 1575–1585 (2005). 27. de Bouteiller, O. et al. Recognition of doublestranded RNA by human Toll-like receptor 3 and downstream receptor signaling requires multimerization and an acidic pH. J. Biol. Chem. 280, 38133–38145 (2005). 28. Ewald, S. E. et al. The ectodomain of Toll-like receptor 9 is cleaved to generate a functional receptor. Nature 456, 658–662 (2008). 29. Kuznik, A. et al. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J. Immunol. 186, 4794–4804 (2011). 30. Furukawa, F., Kashihara-Sawami, M., Lyons, M. B. & Norris, D. A. Binding of antibodies to the extractable nuclear antigens SS‑A/Ro and SS‑B/La is induced on the surface of human keratinocytes by ultraviolet light (UVL): implications for the pathogenesis of photosensitive cutaneous lupus. J. Invest. Dermatol. 94, 77–85 (1990). 31. Shaffer, B., Cahn, M. M. & Levy, E. J. Absorption of antimalarial drugs in human skin; spectroscopic and chemical analysis in epidermis and corium. J. Invest. Dermatol. 30, 341–345 (1958). 32. Nguyen, T. Q., Capra, J. D. & Sontheimer, R. D. 4‑aminoquinoline antimalarials enhance UV‑B induced c‑Jun transcriptional activation. Lupus 7, 148–153 (1998). 33. Wallace, D. J., Metzger, A. L., Stecher, V. J., Turnbull, B. A. & Kern, P. A. Cholesterol-lowering effect of hydroxychloroquine in patients with rheumatic disease: reversal of deleterious effects of steroids on lipids. Am. J. Med. 89, 322–326 (1990). 34. Goldstein, J. L., Brunschede, G. Y. & Brown, M. S. Inhibition of proteolytic degradation of low density lipoprotein in human fibroblasts by chloroquine, concanavalin A, and Triton WR 1339. J. Biol. Chem. 250, 7854–7862 (1975). 35. Lange, Y., Duan, H. & Mazzone, T. Cholesterol homeostasis is modulated by amphiphiles at transcriptional and post-transcriptional loci. J. Lipid Res. 37, 534–539 (1996). 36. Gu, J. Q. et al. A Toll-like receptor 9‑mediated pathway stimulates perilipin 3 (TIP47) expression and induces lipid accumulation in macrophages. Am. J. Physiol. Endocrinol. Metab. 299, E593–E600 (2010). 37. Tobias, P. & Curtiss, L. K. Thematic review series: the immune system and atherogenesis. Paying the price for pathogen protection: Toll receptors in atherogenesis. J. Lipid Res. 46, 404–411 (2005). 38. Lesiak, A. et al. Systematic administration of chloroquine in discoid lupus erythematosus reduces skin lesions via inhibition of angiogenesis. Clin. Exp. Dermatol. 34, 570–575 (2009). 39. Pinhal-Enfield, G. et al. An angiogenic switch in macrophages involving synergy between Toll-like receptors 2, 4, 7, and 9 and adenosine A2A receptors. Am. J. Pathol. 163, 711–721 (2003). 40. Espinola, R. G., Pierangeli, S. S., Gharavi, A. E. & Harris, E. N. Hydroxychloroquine reverses platelet activation induced by human IgG antiphospholipid antibodies. Thromb. Haemost. 87, 518–522 (2002). 41. Rand, J. H. et al. Hydroxychloroquine protects the annexin A5 anticoagulant shield from disruption by antiphospholipid antibodies: evidence for a novel effect for an old antimalarial drug. Blood 115, 2292–2299 (2010). 42. Woessner, J. F. Jr. Matrix metalloproteinases and their inhibitors in connective tissue remodeling. Faseb J. 5, 2145–2154 (1991). 43. Stuhlmeier, K. M. & Pollaschek, C. Quinacrine but not chloroquine inhibits PMA induced upregulation of matrix metalloproteinases in leukocytes: quinacrine acts at the transcriptional level through a PLA2-independent mechanism. J. Rheumatol. 33, 472–480 (2006). 44. Lesiak, A. et al. Effect of chloroquine phosphate treatment on serum MMP‑9 and TIMP‑1 levels in patients with systemic lupus erythematosus. Lupus 19, 683–688 (2010). 45. Merrell, M. A. et al. Toll-like receptor 9 agonists promote cellular invasion by increasing matrix metalloproteinase activity. Mol. Cancer Res. 4, 437–447 (2006). 46. Lullmann-Rauch, R., Pods, R. & von Witzendorff, B. The antimalarials quinacrine and chloroquine induce weak lysosomal storage of sulphated glycosaminoglycans in cell culture and in vivo. Toxicology 110, 27–37 (1996). 47. Toubi, E. et al. The reduction of serum B‑lymphocyte activating factor levels following quinacrine add-on therapy in systemic lupus erythematosus. Scand. J. Immunol. 63, 299–303 (2006). 48. Ehsanian, R., Van Waes, C. & Feller, S. M. Beyond DNA binding—a review of the potential mechanisms mediating quinacrine’s therapeutic activities in parasitic infections, inflammation, and cancers. Cell Commun. Signal. 9, 13 (2011). 49. [No authors listed] A randomized study of the effect of withdrawing hydroxychloroquine sulfate in systemic lupus erythematosus. The Canadian Hydroxychloroquine Study Group. N. Engl. J. Med. 324, 150–154 (1991). 50. Molad, Y. et al. Protective effect of hydroxychloroquine in systemic lupus erythematosus. Prospective long-term study of an Israeli cohort. Lupus 11, 356–361 (2002). 51. Fessler, B. J. et al. Systemic lupus erythematosus in three ethnic groups: XVI. Association of hydroxychloroquine use with reduced risk of damage accrual. Arthritis Rheum. 52, 1473–1480 (2005). 52. Alarcon, G. S. et al. Effect of hydroxychloroquine on the survival of patients with systemic lupus erythematosus: data from LUMINA, a multiethnic US cohort (LUMINA L). Ann. Rheum. Dis. 66, 1168–1172 (2007). 53. Ruiz-Irastorza, G. et al. Effect of antimalarials on thrombosis and survival in patients with systemic lupus erythematosus. Lupus 15, 577–583 (2006). 54. James, J. A. et al. Hydroxychloroquine sulfate treatment is associated with later onset of systemic lupus erythematosus. Lupus 16, 401–409 (2007). 55. Shinjo, S. K. et al. Antimalarial treatment may have a time-dependent effect on lupus survival: data from a multinational Latin American inception cohort. Arthritis Rheum. 62, 855–862 (2010). 56. Pons-Estel, G. J. et al. Possible protective effect of hydroxychloroquine on delaying the occurrence of integument damage in lupus: LXXI, data from a multiethnic cohort. Arthritis Care Res. (Hoboken) 62, 393–400 (2010). 57. Wallace, D. J. Does hydroxychloroquine sulfate prevent clot formation in systemic lupus erythematosus? Arthritis Rheum. 30, 1435–1436 (1987). 58. Ho, K. T. et al. Systemic lupus erythematosus in a multiethnic cohort (LUMINA): XXVIII. Factors predictive of thrombotic events. Rheumatology (Oxford) 44, 1303–1307 (2005). www.nature.com/nrrheum © 2012 Macmillan Publishers Limited. All rights reserved REVIEWS 59. Sisó, A. et al. Previous antimalarial therapy in patients diagnosed with lupus nephritis: influence on outcomes and survival. Lupus 17, 281–288 (2008). 60. Kaiser, R., Cleveland, C. M. & Criswell, L. A. Risk and protective factors for thrombosis in systemic lupus erythematosus: results from a large, multi-ethnic cohort. Ann. Rheum. Dis. 68, 238–241 (2009). 61. Tektonidou, M. G., Laskari, K., Panagiotakos, D. B. & Moutsopoulos, H. M. Risk factors for thrombosis and primary thrombosis prevention in patients with systemic lupus erythematosus with or without antiphospholipid antibodies. Arthritis Rheum. 61, 29–36 (2009). 62. Jung, H. et al. The protective effect of antimalarial drugs on thrombovascular events in systemic lupus erythematosus. Arthritis Rheum. 62, 863–868 (2010). 63. Petri, M., Lakatta, C., Magder, L. & Goldman, D. Effect of prednisone and hydroxychloroquine on coronary artery disease risk factors in systemic lupus erythematosus: a longitudinal data analysis. Am. J. Med. 96, 254–259 (1994). 64. Kavanaugh, A., Adams-Huet, B., Jain, R., Denke, M. & McFarlin, J. Hydroxychloroquine effects on lipoprotein profiles (the HELP trial): a double-blind, randomized, placebo-controlled, pilot study in patients with systemic lupus erythematosus. J. Clin. Rheumatol. 3, 3–8 (1997). 65. Borba, E. F. & Bonfa, E. Longterm beneficial effect of chloroquine diphosphate on lipoprotein profile in lupus patients with and without steroid therapy. J. Rheumatol. 28, 780–785 (2001). 66. Petri, M. Hydroxychloroquine use in the Baltimore Lupus Cohort: effects on lipids, glucose and thrombosis. Lupus 5 (Suppl. 1), S16–S22 (1996). 67. Pons-Estel, G. J. et al. Protective effect of hydroxychloroquine on renal damage in patients with lupus nephritis: LXV, data from a multiethnic US cohort. Arthritis Rheum. 61, 830–839 (2009). 68. Pons-Estel, G. J. et al. Anti-malarials exert a protective effect while Mestizo patients are at increased risk of developing SLE renal disease: data from a Latin-American cohort. Rheumatology (Oxford) http://dx.doi.org/ 10.1093/rheumatology/ker514. 69. Ruiz-Irastorza, G. et al. Predictors of major infections in systemic lupus erythematosus. Arthritis Res. Ther. 11, R109 (2009). 70. Ruiz-Irastorza, G., Ramos-Casals, M., BritoZeron, P. & Khamashta, M. A. Clinical efficacy and side effects of antimalarials in systemic lupus erythematosus: a systematic review. Ann. Rheum. Dis. 69, 20–28 (2010). 71. Schmajuk, G., Yazdany, J., Trupin, L. & Yelin, E. Hydroxychloroquine treatment in a communitybased cohort of patients with systemic lupus erythematosus. Arthritis Care Res. (Hoboken) 62, 386–392 (2010). 72. Ruzicka, T., Sommerburg, C., Goerz, G., Kind, P. & Mensing, H. Treatment of cutaneous lupus erythematosus with acitretin and hydroxychloroquine. Br. J. Dermatol. 127, 513–518 (1992). 73. Cavazzana, I. et al. Treatment of lupus skin involvement with quinacrine and hydroxychloroquine. Lupus 18, 735–739 (2009). 74. Chang, A. Y. et al. Response to antimalarial agents in cutaneous lupus erythematosus: 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. a prospective analysis. Arch. Dermatol. 147, 1261–1267 (2011). Rahman, P., Gladman, D. D. & Urowitz, M. B. Smoking interferes with efficacy of antimalarial therapy in cutaneous lupus. J. Rheumatol. 25, 1716–1719 (1998). Jewell, M. L. & McCauliffe, D. P. Patients with cutaneous lupus erythematosus who smoke are less responsive to antimalarial treatment. J. Am. Acad. Dermatol. 42, 983–987 (2000). Kreuter, A. et al. Lupus erythematosus tumidus: response to antimalarial treatment in 36 patients with emphasis on smoking. Arch. Dermatol. 145, 244–248 (2009). Leroux, G. et al. Relationship between blood hydroxychloroquine and desethylchloroquine concentrations and cigarette smoking in treated patients with connective tissue diseases. Ann. Rheum. Dis. 66, 1547–1548 (2007). Wright, J. L., Tai, H., Wang, R., Wang, X. & Churg, A. Cigarette smoke upregulates pulmonary vascular matrix metalloproteinases via TNF-α signaling. Am. J. Physiol. Lung Cell. Mol. Physiol. 292, L125–L133 (2007). Lim, S. et al. Balance of matrix metalloprotease‑9 and tissue inhibitor of metalloprotease‑1 from alveolar macrophages in cigarette smokers. Regulation by interleukin‑10. Am. J. Respir. Crit. Care Med. 162, 1355–1360 (2000). Bermudez, E. A., Rifai, N., Buring, J. E., Manson, J. E. & Ridker, P. M. Relation between markers of systemic vascular inflammation and smoking in women. Am. J. Cardiol. 89, 1117–1119 (2002). Lardet, D. et al. Effect of smoking on the effectiveness of antimalarial drugs for cutaneous lesions of patients with lupus: assessment in a prospective study [French]. Rev. Med. Interne 25, 786–791 (2004). Wahie, S. et al. Clinical and pharmacogenetic influences on response to hydroxychloroquine in discoid lupus erythematosus: a retrospective cohort study. J. Invest. Dermatol. 131, 1981–1986 (2011). Turchin, I., Bernatsky, S., Clarke, A. E., St-Pierre, Y. & Pineau, C. A. Cigarette smoking and cutaneous damage in systemic lupus erythematosus. J. Rheumatol. 36, 2691–2693 (2009). Moghadam-Kia, S. et al. Cross-sectional analysis of a collaborative Web-based database for lupus erythematosus-associated skin lesions: prospective enrollment of 114 patients. Arch. Dermatol. 145, 255–260 (2009). Piette, E. W. et al. Impact of smoking in cutaneous lupus erythematosus. Arch. Dermatol. 148, 317–322 (2012). Parke, A. Antimalarial drugs and pregnancy. Am. J. Med. 85, 30–33 (1988). Parke, A. & West, B. Hydroxychloroquine in pregnant patients with systemic lupus erythematosus. J. Rheumatol. 23, 1715–1718 (1996). Buchanan, N. M. et al. Hydroxychloroquine and lupus pregnancy: review of a series of 36 cases. Ann. Rheum. Dis. 55, 486–488 (1996). Clowse, M. E., Magder, L., Witter, F. & Petri, M. Hydroxychloroquine in lupus pregnancy. Arthritis Rheum. 54, 3640–3647 (2006). Izmirly, P. M. et al. Evaluation of the risk of antiSSA/Ro-SSB/La antibody-associated cardiac manifestations of neonatal lupus in fetuses of mothers with systemic lupus erythematosus NATURE REVIEWS | RHEUMATOLOGY exposed to hydroxychloroquine. Ann. Rheum. Dis. 69, 1827–1830 (2010). 92. Levy, R. A. et al. Hydroxychloroquine (HCQ) in lupus pregnancy: double-blind and placebocontrolled study. Lupus 10, 401–404 (2001). 93. Costedoat-Chalumeau, N., Amoura, Z., Huong, D. L., Lechat, P. & Piette, J. C. Safety of hydroxychloroquine in pregnant patients with connective tissue diseases. Review of the literature. Autoimmun. Rev. 4, 111–115 (2005). 94. Wallace, D. J. in Dubois Lupus Erythematosus 7th edn (eds Wallace, D. J. & Hahn, B. H.) 1152–1176 (Lippincott Williams & Wilkins, Philadelphia, 2007). 95. Van Beek, M. J. & Piette, W. W. Antimalarials. Dermatol. Clin. 19, 147–160, ix (2001). 96. Marmor, M. F., Kellner, U., Lai, T. Y., Lyons, J. S. & Mieler, W. F. Revised recommendations on screening for chloroquine and hydroxychloroquine retinopathy. Ophthalmology 118, 415–422 (2011). 97. Venuturupalli, S. R., Gudsoorkar, V. S. & Wallace, D. J. Revisiting antimalarials in systemic lupus erythematosus: developments of translational clinical interest. J. Rheumatol. (in press). 98. Wallace, D. J. Advances in drug therapy for systemic lupus erythematosus. BMC Med. 8, 77 (2010). 99. Wozniacka, A., Carter, A. & McCauliffe, D. P. Antimalarials in cutaneous lupus erythematosus: mechanisms of therapeutic benefit. Lupus 11, 71–81 (2002). 100.Lopez, P., Gomez, J., Mozo, L., Gutierrez, C. & Suarez, A. Cytokine polymorphisms influence treatment outcomes in SLE patients treated with antimalarial drugs. Arthritis Res. Ther. 8, R42 (2006). 101.Collinge, J. et al. Safety and efficacy of quinacrine in human prion disease (PRION‑1 study): a patient-preference trial. Lancet Neurol. 8, 334–344 (2009). 102.Gurova, K. New hopes from old drugs: revisiting DNA-binding small molecules as anticancer agents. Future Oncol. 5, 1685–1704 (2009). 103.Adelusi, S. A. & Salako, L. A. Tissue and blood concentrations of chloroquine following chronic administration in the rat. J. Pharm. Pharmacol. 34, 733–735 (1982). 104.Ronnblom, L., Eloranta, M. L. & Alm, G. V. The type I interferon system in systemic lupus erythematosus. Arthritis Rheum. 54, 408–420 (2006). 105.Aman, M. J. et al. Interferon-α stimulates production of interleukin‑10 in activated CD4+ T cells and monocytes. Blood 87, 4731–4736 (1996). 106.Ronnblom, L., Alm, G. V. & Eloranta, M. L. The type I interferon system in the development of lupus. Semin. Immunol. 23, 113–121 (2011). 107.Villanueva, E. et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J. Immunol. 187, 538–552 (2011). Author contributions D. J. Wallace, V. S. Gudsoorkar and S. R. Venuturupalli researched the data for the article. All authors provided a substantial contribution to discussions of the content and contributed equally to writing the article and to review and/or editing of the manuscript before submission. VOLUME 8 | SEPTEMBER 2012 | 533 © 2012 Macmillan Publishers Limited. All rights reserved
© Copyright 2024