Open access

Cardiovascular Lesions of Kawasaki Disease: From Genetic Study to Clinical Management

Written By

Ho-Chang Kuo and Wei-Chiao Chang

Submitted: 23 November 2011 Published: 18 July 2012

DOI: 10.5772/48341

Chapter metrics overview

3,679 Chapter Downloads

View Full Metrics

1. Introduction

Kawasaki disease (KD) is an acute febrile systemic vasculitis that was first described by Kawasaki et al. [1] in 1967 in Japanese [2] and in 1974 in English. Currently, it is the leading cause of acquired heart disease in children in developed countries; however, its etiology remains unknown. [3-5]KD mainly affects children less than 5 years of age, especially those in Asian countries. In Japan, Korea, and Taiwan, the incidence ranges from 69 to 218 cases per 100,000 children less than 5 years of age. [6-9] The incidence of KD in Taiwan has increased from 66 to 69 cases per 100,000 children aged less than 5 years. [9-12] Its incidence worldwide is increasing, especially in Japan, where, in 2010, Nakamura et al. reported the country’s highest rate of 239.6 cases per 100,000 children aged 0–4 years. [13] An epidemiologic survey of KD in Taiwan spanning 2003–2006 found that 1.5% of all cases was recurrent (having a second episode of KD and receiving intravenous immunoglobulin [IVIG] treatment). [9] In Taiwan, KD occurs most frequently in the summer (April to June) and least frequently in the winter; for unknown reasons, its seasonal occurrence varies in other countries. The most serious complication of KD is the development of coronary artery lesions (CAL), including myocardial infarction, coronary artery fistula formation, [14] coronary artery dilatation, and coronary artery aneurysm. [15]

The most commonly used definition of CAL (also known as coronary artery abnormality [CAA] or CAL) is based on the Japanese Ministryof Health criteria: maximum absolute internal diameter >3 mm in childrenyounger than 5 years of age or >4 mm in children 5 years and older,or a segmental diameter 1.5 times greater than that of an adjacent segment, orthe presence of luminal irregularity. [16-21] Coronary arteries should be corrected relative to body surface area (if available) and expressed as standard deviation units from the mean (Z scores). [22] Several studies analyzed CAL, including aortic root dimension, [23] and transient CAL (the definition of “transient” varies among studies, from 30 days to 6–8 weeks after diagnosis of disease). Thus, KD patients with coronary artery ectasia or dilatation that disappears within the first 8 weeks after disease onset are defined as having transient ectasia or dilatation (transient CAL). Kuo et al. reported the serious CAL analysis that comprised 341 KD patients; [24] 35% of KD patients had dilatation during the acute phase of admission, 17.2% still had dilatation 1 month after disease onset, 10.2% had dilatation at 2 months follow-up, and 4.1% had persistent CAL for more than 1 year. [25, 26]Ectasia or transient dilatations are somewhat considered to be a risk for a subsequent cardiovascular event or inflammation duration, rather than normal status. [27]

Although the clinical features of KD are recognizable, its underlying immunopathogenetic mechanisms are still under investigation, particularly the agent responsible for the development of CAL. KD is regarded as an autoimmune disorder rather than an infectious disease. [23] Kuo et al. reported that persistent monocytosis after IVIG treatment is associated with CAL formation. [28] Eosinophils in KD patients were also higher than that in age-matched febrile controls. In addition, IVIG treatment significantly increased eosinophilsin KD patients. This increase of eosinophils after IVIG treatment is inversely correlated with IVIG treatment failure in KD. [29] Further studies have shown that eosinophil changes after IVIG treatment were positively correlated with changes in interleukin (IL)-5 levels. An increase in eosinophils and IL-5 levels after IVIG treatment is inversely correlated with CAL formation. [30]Recently, we found that incidence of allergic diseases (asthma and allergic rhinitis) after onset of KD were higher than that in age and sex-matched controls in a population cohort in Taiwan.

Advertisement

2. Allergy potential of Kawasaki disease patients

Brosius et al. [31] showed that the incidence of atopic dermatitis among children with KD was 9 times greater than that of controls. Burns et al. [32] reported associations of KD with atopic dermatitis and allergy, elevated serum IgE levels, and eosinophilia and that increased circulating numbers of monocytes/macrophages expressing the low-affinity IgE receptor (FCεR2) may be related to the effects of IL-4. Liew et al. [33] reported that KD may be a risk factor for subsequent allergic disease and postulated that KD occurs more frequently in children at risk of immune disequilibrium, with an initial abnormal inflammatory response, and subsequently, more allergic manifestations. Currently, Webster et al. [34] also reported that KD patients were more likely to have been admitted at least once with asthma/allergy than controls were. From our previous reports, we found that the T-helper (Th) type 2 immune response was elevated in the acute stage of KD, including eosinophils, [29] IL-4, IL-5, [35] and eotaxin. The eosinophil changes were correlated to changes of IL-5 levels but not to eosinophil cationic protein (ECP) levels, suggesting a Th2 immune reaction in KD. There are several lines of evidence pointing to an abnormal Th1/Th2 balance in KD patients. [28, 29, 35-39]Lin et al. [40] reported the comparison of eosinophils in KD and enterovirus (EV) patients with IVIG treatment and demonstrated a more significant eosinophil increase in KD patients. EV patients also had elevated eosinophil levels after IVIG therapy, but not as high as that of the KD patients after IVIG treatment. This may indicate an imbalance of the Th1/Th2 immune response, with a skewed Th2 response in KD.

Advertisement

3. Clinical phenotype and presentation of Kawasaki disease

As shown in Figures 18, the clinical characteristics of KD patients include fever lasting longer than 5 days, diffuse mucosal inflammation, bilateral non-purulent conjunctivitis, dysmorphic skin rashes, indurative angioedema over the hands and feet, and cervical lymphadenopathy. In addition to the diagnostic criteria, there is a broad range of non-specific clinical features, including irritability, uveitis, aseptic meningitis, cough, vomiting, diarrhea, abdominal pain, gallbladder hydrops, urethritis, arthralgia, arthritis, hypoalbuminemia, [5] liver function impairment, and heart failure. [4, 29, 41]

Figure 1.

Dysmorphic skin rash of Kawasaki disease

Figure 2.

Skin rash and neck lymphadenopathy (right side, >1.5 cm in diameter)

Figure 3.

Strawberry tongue

Figure 4.

Face of Kawasaki disease patient exhibiting conjunctivitis, fissured lips, and skin rashes

Figure 5.

BCG injection site indurations

Figure 6.

Fissured lips and swelling of finger joints

Figure 7.

Induration change over foot

Figure 8.

Induration change over palm

3.1. Diagnosis of Kawasaki disease

To date, there is no specific diagnostic laboratory test for KD. Diagnosis is based on the clinical phenotype, i.e., presence of fever lasting longer than 5 days and fulfillment of 4 of 5 specific clinical criteria. In Japan, at least 5 of 6 criteria (fever and 5 other clinical criteria) should be fulfilled for a diagnosis of KD. However, patients with 4 of the principal clinical features can be diagnosed when coronary aneurysm or dilatation is identified. [42] From the Japanese Circulation Society Joint Working Groups criteria (JCS 2008, Guidelines for Diagnosis and Management of Cardiovascular Sequelae in Kawasaki Disease), [43] KD can be diagnosed even when fever lasts less than 5 days. However, according to the American Heart Association (AHA) criteria, [15] fever lasting more than 5 days is essential for the diagnosis of KD.

Some patients who do not fulfill the criteria have been diagnosed with “incomplete” or “atypical” KD, a diagnosis often based on echocardiographic identification of CAL. The term “incomplete” may be preferable to “atypical” because these patients have insufficient criteria instead of atypical presentation. [15]

In countries with a bacillus Calmette-Guérin (BCG) vaccine policy (i.e., Taiwan and Japan), KD with erythematous induration or even ulceration of the BCG scar has been observed in one-third to half of KD patients (the incidence of BCG site induration is higher than that of neck lymphadenopathy in these countries). [3] Ueharaet al. [44] reported that redness or the formation of a crust at the BCG inoculation site is a useful diagnostic sign for KD in children aged 3–20 months. Even if patients exhibit 4 or fewer signs of the clinical criteria for KD, physicians should consider the redness or crust formation at the BCG inoculation site as a possible indicator of KD.

Incomplete cases of KD are not uncommon (up to 15–20%). The incidence of CAL in patients exhibiting 4 principal symptoms of KD is slightly higher than that in patients with 5 to 6 principal symptoms. [45] Presentation of a small number (<4) of principal symptoms does not indicate a milder form of the disease. Patients with at least 4 principal symptoms require the same treatment as patients with complete (typical) presentation of KD, and those with 3 or fewer principal symptoms should be treated similarly when they meet the supplementary criteria. Herein, common supplementary criteria for the diagnosis of incomplete KD are introduced.

Figure 9.

Flowchart of Kawasaki disease management

Incomplete KD is more common in young infants than in older children, making accurate diagnosis and timely treatment especially important in these young patients, who are at substantial risk of developing coronary abnormalities. [46, 47] The incidence of KD is actually higher than that previously reported worldwide, partly because earlier reports did not take incomplete forms into account. The AHA criteria (2004), which incorporate suggestions for laboratory tests and early echocardiography, are helpful for diagnosing incomplete KD. [41, 48] Consultation with an expert (cardiologist, immunologist, or rheumatologist) should be sought whenever assistance in making a diagnosis is needed. Patients with fever for 5 days or more (with 2 or 3 principal clinical features for KD) without other causes should undergo laboratory testing, and if there is evidence of systemic inflammation, an echocardiogram should be obtained even if the patient does not fully meet the clinical criteria for KD. Likewise, infants 6 months or younger with fever for 7 days or more without other causes should undergo laboratory testing, and if evidence of systemic inflammation is found, an echocardiogram should be obtained even if the infant fulfills no clinical criteria for KD. [15]

The 2004 AHA supplemental laboratory criteria include [1] albumin ≤ 3.0 g/dL; [2] anemia for age; [3] elevation of alanine aminotransferase (ALT); [4] platelets after 7 days ≥ 450,000/mm3; [5] white blood cell count ≥ 15,000/mm3; and [6] urine ≥ 10 white blood cells/high-power field. [15] If a patient has more than 3 supplementary criteria, incomplete KD is diagnosed and IVIG should be prescribed before performing echocardiography. [15] The flowchart for incomplete KD diagnosis and treatment are depicted in Figure 9.

Advertisement

4. Treatment for Kawasaki disease

The standard treatment for KD is IVIG (2 g/kg) infusion for 8–12 hours with high-dose aspirin (80–100 mg/ [kg day]). [20, 24, 29] The most serious complication of KD is the development of CAL, including myocardial infarction, coronary artery dilatation, coronary artery aneurysms, and coronary fistula formation. [25, 30, 49]Coronary artery aneurysms occur as a sequela of the vasculitis in 20–25% of untreated children. There are several risk factors for developing coronary arteritis, such as low serum albumin, age younger than 1 year, and long duration of the fever before treatment. Young patients with low albumin run a very high risk for CAL and IVIG treatment resistance. [5, 29] Although the introduction of IVIG therapy has greatly decreased the rate of coronary aneurysm to 3–10% of patients still develop some type of CAL. Durongpisitkul et al. showed that 11.6% patients are unresponsive to initial IVIG (2 g/kg) treatment. The worst prognosis occurs in children with so-called “giant aneurysms of the coronary arteries” (those with a maximal diameter of >8 mm), as thrombosis is promoted both by sluggish blood flow within the massively dilated vascular space and by the frequent development of stenotic lesions later. The treatment for KD are reviewed and introduced as follows.

4.1. Aspirin

Aspirin has been used in the treatment of KD for many years, even before the usage of IVIG. Although aspirin has important anti-inflammatory (high dose) and anti-platelet (low dose) effects, it does not appear to reduce the frequency of CAL formation. During the acute phase of the illness, aspirin is administered in 4 doses of 80–100 mg/kg per day (30–50 mg/ [kg day] in Japan) [50] with IVIG. High-dose aspirin and IVIG appear to possess additive anti-inflammatory effects.

Practices regarding the duration of high-dose aspirin administration vary across countries and centers, many of which reduce the aspirin dose when the patient is afebrile. When high-dose aspirin is discontinued, low-dose aspirin (3–5 mg/ [kg day]) is administered until there is no evidence of CAL and inflammatory markers (including platelets, C-reactive protein [CRP], and erythrocyte sedimentation rate [ESR]) have returned to normal levels, which usually occurs 6–8 weeks after disease onset. For children who develop CAL, low-dose aspirin (or other anti-platelet agents) is continued indefinitely until the inflammatory markers return to the normal range and the echocardiogram does not display abnormalities. Hsieh et al. [50] reported that regardless of timing (before or after day 5 of the illness), single-infusion, high-dose (2 g/kg) aspirin in the acute stage of KD had no effect on the response rate to IVIG therapy, duration of fever, or the incidence of CAL. This review reiterates the recommendation that exposing children to high-dose aspirin therapy in the acute phase of KD is unnecessary because available data show no appreciable benefit to IVIG therapy response, CAL formation, or fever duration.

Our recent study investigated 609 KD patients from 2 medical centers in Taiwan. The patients were divided into Group 1, receiving high-dose aspirin (N = 274), and Group 2, without high-dose aspirin (N = 335). There were no significant differences between Groups 1 and 2 in terms of gender (p = 0.51), IVIG resistance rate (34/274 vs. 26/335, p = 0.06), CAL formation rate (57/274 vs. 74/335, p = 0.64), and total hospital stay (6.3 ± 0.2 vs. 6.7 ± 0.2 days, p = 0.13). There were also no significant differences between total white blood cell counts, hemoglobin levels, platelet counts, and CRP levels before (within 1 day) and after (within 3 days) IVIG treatment of the 2 groups (p > 0.1). These results provide evidence that high-dose aspirin in the acute phase of KD does not affect the treatment results (CAL and IVIG resistance rate) or inflammatory condition. High-dose aspirin treatment in the acute phase of KD appears unnecessary, and further randomized controlled trials are needed.

However, Reye syndrome is a risk in children who receive salicylates while they are experiencing active infection with varicella or influenza and has been reported in patients receiving high-dose aspirin for a prolonged period after KD. [52] Taken together, it seems unnecessary to expose children to high-dose aspirin in acute KD, especially those with G6PD deficiency.However, as reported in the literature, due to the anti-platelet effect, low-dose aspirin has been prescribed for at least 6–8 weeks to prevent thrombocytosis in KD patients. [15] If patients are allergic or intolerant to a particular drug, clinicians must avoid using it and look for alternatives.Aspirin is used in most patients, often in conjunction with dipyridamole. Dipyridamole has been widely used to treat patients with a coronary aneurysm resulting from KD. [43, 53] The relationship between aspirin therapy and hemolytic disorder in G6PD-deficient patients is unclear. There are also no literature regarding usage of low-dose aspirin and the outcome of KD. G6PD deficiency, an X-linked disorder, is the most common enzymatic disorder of red blood cells in humans. The clinical expression of G6PD deficiency encompasses a spectrum of hemolytic syndromes. While affected patients are usually asymptomatic, some have episodic anemia, while a few have chronic hemolysis. With the most prevalent G6PD variants (G6PD A- and G6PD Mediterranean), severe hemolysis is induced by the sudden destruction of older, more deficient erythrocytes after exposure to drugs with a high redox potential or to fava beans, selected infections, or metabolic abnormalities. The likelihood of developing hemolysis and the severity of disease are determined by the magnitude of the enzyme deficiency, which in turn is determined by the biochemical characteristics of the G6PD variant. The World Health Organization has classified the different G6PD variants according to the magnitude of the enzyme deficiency and the severity of hemolysis. [54] Class I variants have severe enzyme deficiency (less than 10% of normal) and are associated with chronic hemolytic anemia. Class II variants also have severe enzyme deficiency, but are usually only intermittently associated with hemolysis. Class III variants have moderate enzyme deficiency (10–60% of normal), with intermittent hemolysis usually associated with infection or drugs. Class IV variants have no enzyme deficiency or hemolysis. Class V variants have increased enzyme activity, and classes IV and V are of no clinical significance. The incidence of hemolysis development in a patient with G6PD deficiency after taking aspirin is dosage-related. [55] G6PD deficiency is commonly considered a contraindication to aspirin intake. However, just few studies [56] have suggested that aspirin can be safely administered in therapeutic doses to G6PD-deficient subjects without nonspherocytic hemolytic anemia. Anti-platelet therapy is most commonly used to prevent thrombotic events for adults with atherosclerotic vascular disease, children with certain types of congenital heart disease, stroke, and KD. [57] Unfortunately, very little data on the efficacy and safety of anti-platelet therapy for pediatric patients, or even G6PD patients, are available. No prospective data exist to guide clinicians in selecting an optimal regimen. Therapeutic regimens used in patients with KD depend on the severity of CAL and include anti-platelet therapy with aspirin, with or without dipyridamole or clopidogrel; anticoagulant therapy with warfarin or low-molecular-weight heparin; or a combination of anticoagulant and anti-platelet therapy. [15]

A few articles have reported G6PD-deficient patients with sustained KD. [58] However, the question of whether aspirin is suitable for KD patients with G6PD deficiency remains

4.2. Intravenous immunoglobulin (IVIG or IVGG)responsiveness

The efficacy of IVIG administered in the acute phase of KD for reducing the incidence of coronary artery abnormalities is well established. [59] The mechanism of IVIG action is still under investigation. IVIG appears to have a generalized anti-inflammatory effect. Possible mechanisms of action include modulation of cytokine production, neutralization of bacterial super-antigens or other etiologic agents, augmentation of regulatory T cell activity (TGF-), [23, 26] suppression of antibody synthesis and inflammatory markers (CD40-CD40L, nitric oxide, and iNOS expression), [60-62]provision of anti-idiotypic antibodies, Fc-gamma receptor, [63] and balancing Th1/Th2 responses. [28-30]

KD patients should be treated with a single 12-hour infusion of 2 g/kg IVIG together with aspirin in the acute phase with fever or inflammation progression without fever. [3, 4, 15] This therapy should be administered within 10 days of illness onset, and if this is not possible, within 7 days of illness onset. Treatment of KD before day 5 of illness appears no more likely to prevent cardiac sequelae than treatment on days 5–9. However, it may be associated with an increased need for repeat IVIG treatment. [64, 65] In the presence of 4 of 5 classic criteria for KD, US and Japanese experts agree that only 4 days of fever are necessary before initiating treatment with IVIG. [15, 66]

The efficacy of treating patients using IVIG after 10 days of illness is unknown; therefore, early diagnosis and treatment is desired. IVIG should be administered to children presenting after day 10 of illness (i.e., children with delayed diagnosis or incomplete KD) if they have either persistent fever without explanation or aneurysms and ongoing systemic inflammation, as manifested by elevated ESR or CRP. [4, 67-69] Burns et al. also suggested that any child with KD who has evidence of persisting inflammation, including fever or high concentrations of inflammatory markers with or without coronary artery abnormalities, should be treated even if the diagnosis is made after 10 days of illness. [4]

4.3. IVIG resistance (or IVIG unresponsiveness, initial IVIG treatment failure)

The incidence of IVIG resistance varies from 9.4% to 23% between centers (but it can be as high as 38%, as reported in one US cohort). [70] Recent studies have identified demographic and laboratory characteristics as predictors of IVIG resistance, including age, illness day, platelet count, ESR, hemoglobin concentration, CRP, eosinophils, lactate dehydrogenase, albumin, and ALT. [5, 29, 71-73] As IVIG-resistant patients are at a higher risk for CAL formation, it is important to identify those who may benefit from more aggressive therapy. As shown in Figure 1 (modified from Newburger et al. [15]), there are no definite treatment principles available for the management of KD patients with initial IVIG resistance or unresponsiveness to other adjuvant therapies. A second dose of IVIG (1 or 2 g/kg), [15, 74] methylprednisolone (MP) pulse therapy, [75] tumor necrosis factor (TNF)- blockade, [76]cytotoxic agents (cyclophosphamide, cyclosporine A [CyA], or MTX [77]), plasmapheresis, [78] and plasma exchange [79] have been reported to benefit KD patients with initial IVIG treatment failure. These other treatment modalities will be discussed.

4.4. Methylprednisolone pulse therapy

At present, the usefulness of steroids in the initial treatment of KD is not well established. [15]Newburger et al. reported that, compared to conventional IVIG therapy for routine primary treatment of KD in children, a single-pulse dose of intravenous MP (IVMP) does not improve treatment outcome. [22] However, IVMP therapy appears to benefit IVIG-resistant KD patients. [80] Miura et al. revealed the effectiveness of IVMP therapy for KD patients that were previously unresponsive to initial IVIG treatment. IVMP suppresses cytokine levels faster, and subsequently, the outcomes are similar to those of IVIG-responsive patients who receive a second dose of IVIG. [81] Furukawa et al. reported similar findings. [82] IVMP appears to have the same effect on IVIG-resistant KD patients compared to an additional IVIG treatment. [83] The cost-benefit differences between IVMP and additional IVIG should be carefully considered, taking into account different medical conditions or health insurance policies among countries. The first dose of IVIG is well established, while IVMP or additional IVIG for IVIG-resistant KD patients requires further investigation. Ogata et al. [83] reported that IVMP was useful for reducing fever duration and medical costs for KD patients with initial IVIG resistance. IVMP (N = 13) and additional IVIG treatment (N = 14) were not significantly different in terms of preventing the development of coronary artery aneurysm. IVMP (30 mg/kg MP per day for 3 days) or a second dose of IVIG (2 g/kg) was prescribed to KD patients with fever and marked inflammation (i.e., non-exudative conjunctival injection, strawberry tongue, fissuredlips, and erythematous change at the BCG inoculation site) 48 hours after initial IVIG treatment. [22, 82-84]

The safety of IVMP therapy in patients with KD is uncertain. Miura et al. [85] reported that IVMP (N = 11) incurred a higher incidence of sinus bradycardia and hyperglycemia when compared with the additional IVIG group (N = 11). Hypertension between IVMP and IVIG groups did not differ significantly. All of the adverse effects were transient. There were no convulsions, gastrointestinal symptoms, infections, malignant arrhythmias, or sudden death in any subject. [85] Taken together, IVMP is safe for KD patients as additional or adjuvant therapy of initial IVIG treatment. [22, 86, 87] After additional IVIG therapy, IVMP is considered for KD patients with persistently poor responses to the second IVIG treatment. [74, 88] Kobayashi et al. [89] reported that the addition of prednisolone (2 mg/ [kg day] administered over 15 days) to the standard regimen of IVIG improves coronary artery outcomes in patients with severe KD in Japan.

4.5. Tumor necrosis factor- blockade

TNF- levels are elevated in children with KD, [90] and the TNF- (–308) genetic polymorphism is associated with KD susceptibility, suggesting a role for TNF- receptor blocking in the treatment of KD, especially for those patients/cases refractory to IVIG. The early administration of TNF- receptor antagonists in KD may provide effective adjunctive therapy. Infliximab, which binds the pro-inflammatory cytokine TNF-, has been evaluated in several studies and shown to have a significant effect in KD patients with IVIG resistance. [91-93] Recently, etanercept, a more suitable TNF- receptor blocker for children with refractory juvenile idiopathic arthritis, [94, 95] was reported to benefit the treatment of IVIG-resistant KD as an adjuvant therapy to initial IVIG. [96, 97]A TNF- receptor blocker may be administered after initial IVIG treatment failure or after a second dose of IVIG therapy.

4.6. Statins

Chronic vascular inflammation and endothelial dysfunction persists in KD patients with CAL, even long after the acute stage. [98, 99] There is currently no specific treatment for ongoing vascular inflammation and endothelial dysfunction. Low-dose aspirin can be prescribed until CAL normalizes, but it does not have an effect on inflammation or endothelial dysfunction. Lipid abnormalities in the acute phase of KD, with decreased triglycerides and high-density lipoprotein cholesterol (HDL-C) levels have been reported in previous studies. [100, 101]

Statins, hydroxymethylglutaryl coenzyme A reductase inhibitors, have been shown to reduce cholesterol levels as well as improve surrogate markers of atherosclerosis and cardiovascular disease. [102] Huang et al. [103] reported that short-term (3 months) statin treatment (simvastatin, 10 mg/day as a single dose at bedtime) in KD patients complicated with CAL (N = 11) can significantly reduce total cholesterol and low-density lipoprotein cholesterol levels and increase HDL-C levels. Chronic vascular inflammation is also significantly improved, as is endothelial dysfunction, with no adverse effects. However, long-term and randomized control trials are needed before further conclusions can be drawn.

Recently, Blankier et al. [104] also reported that atorvastatin is able to inhibit critical steps (T cell activation and proliferation, production of the pro-inflammatory cytokine TNF-, and upregulation of matrix metalloproteinase-9 and an elastolytic protease) known to be important in the development of coronary aneurysms in an animal model of KD (murine model with injection of Lactobacillus casei cell wall extract), suggesting that statins may have therapeutic benefits in KD patients. Taken together, statins may be beneficial as an adjuvant therapy in KD patients with CAL. However, the association between dyslipidemia and atherosclerosis in KD patients is not certain.

4.7. Other treatments

Acute KD can lead to the development of large coronary artery aneurysms that may persist for years. Abciximab, a platelet glycoprotein IIb/IIIa receptor inhibitor, is associated with resolution of thrombi and vascular remodeling in adults with acute coronary syndromes. Williams et al. [105] reported that KD patients who were treated with abciximab demonstrated greater regression in aneurysm diameter at early follow-up than patients who received standard therapy alone. McCandless et al. [106] also reported that abciximab treatment might be associated with vascular remodeling in patients with aneurysms. Abciximab appears to benefit KD patients, especially those who develop aneurysms.

There are still no well-defined treatments for refractory KD. Suzuki et al. [107]reported that CyA treatment is considered safe and well tolerated and may serve as a promising option for patients with refractory KD. Hyperkalemia developed in 9 of 28 (32%) patients 3–7 days after commencing CyA treatment. Adverse effects such as arrhythmias should be monitored with CyA. Kuijpers et al. [108] described a case of mortality, and a review of the literature showed that immunosuppressive medication such as CyA may not influence coronary inflammation and proliferation. Further trials are needed to clarify the optimal dose, safety, and timing of CyA treatment.

Specific changes in inflammatory markers (such as white blood cell count, neutrophil count, CRP, IL-6, soluble IL-2 receptor [sIL-2R] [109], Th17/regulatory T-cell imbalance [110], and IL-1 pathway [111]) have been reported to disrupt immunological functions and result in KD with IVIG resistance and CAL formation. This indicates the possible treatment role of plasma exchange (PE) for KD with IVIG resistance. Mori et al. [79] studied 46 children who had not responded to the second IVIG treatment and subsequently received PE, and compared them with 59 children that received a third dose of IVIG therapy. No complications occurred with PE therapy. CAL developed in 8 of the 46 children (17.3%) who received PE and in 24 of the 59 (40.7%) who received a third course of IVIG (p < 0.001). PE is considered safe and effective in the prevention of CAL in KD that is refractory to IVIG therapy. PE can be performed at an early stage, as soon as fractional increases in inflammatory markers are found after the first or second dosage of IVIG therapy. [79]

Advertisement

5. Genetic association study in Kawasaki disease

The higher incidence of KD in Asia, in conjunction with a higher incidence of the disease in Asian descendants compared with other ethnic populations in the United States and Europe, suggests that genetic predisposition might play an important role in the susceptibility to this disease. [3, 4, 9, 15] There is also evidence that the incidence of KD is higher among siblings than in the general population. [112]A growing number of research reports provide evidence that genetic polymorphisms contribute to the susceptibility to KD. For example, single-nucleotide polymorphisms (SNPs) in the monocyte chemoattractant protein 1 (MCP-1), [113]IL-10, [114-116]CD40L, [117]CD40, [62]IL-4, [32]CASP3, [24] IL-18, [118] IL-1B, [119] HLA-E, [120] C-C chemokine receptor 5 (CCR5), [121-124] and ITPKC [20, 125, 126] and TGF- receptors [23] have been reported to be associated with the development of KD. Although genetic association studies have been widely performed in KD, several studies have produced inconsistent results. Some genes were proposed in one population; however, the findings could not be replicated in another population. In addition, the genes that are responsible for KD susceptibility may not be involved in CAL formation. Thus, studies addressing this question are plagued with inconsistencies. Three possibilities may explain these inconsistencies. First, some studies were performed in a small sample size that may not have been able to provide sufficient power to detect minor genetic effects. Second, it is becoming clear that there are different genetic backgrounds within populations that, due to variations in allele frequencies or heterogeneity of the phenotypes, may also influence the results. Third, the incidence of KD in Asia is much higher than that in other regions. Thus, the environmental factors or infectious agents between countries should also be considered carefully.

Advertisement

6. Genetic polymorphisms of the ITPKC signaling pathway in Kawasaki disease

A major advancement in the genetic study of KD was made by the discovery of ITPKC in the RIKEN SNP center Japan. In 2008, Onouchi and colleagues first identified a functional polymorphism of ITPKC (rs28493229) that significantly associated with the susceptibility of KD and CAL in both Japanese and US children. [125] By using cell-based functional studies, Onouchi et al. further provided evidence to indicate that the risk C allele of ITPKC can reduce the splicing efficiency of the ITPKC mRNA that, in turn, contributes to the hyperactivation of Ca2+-dependent NFAT pathways in T cells. Thus, in the model of Onouchi et al., ITPKC is a negative regulator of T cells, and it may function as a calcium channel modulator that is involved in controlling immune systems. Interestingly, replication studies in the Taiwanese populations are strikingly controversial. The first replication study was by Chi et al. A total of 385 KD patients and 1158 normal subjects were genotyped. [127] However, no significant association was observed. Lin et al. took similar approaches in another independent medical center in Taipei. Their results indicated that the C allele of rs28493229 is associated with KD susceptibility. [128] Recently, data by meta-analysis support the correlation between rs28493229 of ITPKC and susceptibility of KD in the Taiwanese population. [23]Due to the increase in genetic diversity between cities in the south or north of Taiwan, we attribute the controversial results in the Taiwanese population to population migration.

Figure 10.

Model depicting the cellular pathways of ITPKC/calcium signaling in T cells.

In the non-excitable cells such as T and B cells, calcium entry is mainly through store-operated calcium channels (SOC). The activation of SOC can be controlled by the expression level of IP3, which is the substrate of ITPKC protein. AsITPKCis involved in the Ca2+-dependent NFAT signaling in T cells, genetic association studies between calcium pathways and susceptibility of KD were performed. The calcium-dependent downstream gene CASP3 is a good example. Onouchi et al. reported that a G-to-A substitution in the 5′-untranslated region of CASP3 (rs72689236) is associated with susceptibility to KD in Japanese and in Americans of European descent. [129] In the sample year, CASP3 (rs72689236) was replicated in the KD children in the Taiwanese population. [24] Kuo et al. confirmed that the A allele of rs72689236 is very likely a risk allele in the development of aneurysms in patients with KD. Another 2 important molecules in the SOC are ORAI1 (also known as CRACM1) and STIM1. Feske et al. identified ORAI1 in 2006. Modified linkage analysis completed on data generated by SNP arrays and RNA interference screening led to an important finding. A single missense mutation in ORAI1 was found in patients with severe combined immune deficiency syndrome. In 2011, genetic polymorphisms of ORAI1 were reported to associate with the risk and recurrence of calcium nephrolithiasis [130] and HLA-B27-positive AS [131]. In theKD study, no significant association between OARI1 genotypes ORAI1 andKDclinical parameters (such as CAL formation or IVIG treatment responses) was found. However, a novel genetic polymorphism in the STIM1 gene was detected that associated with CAL formation in KD patients (data not shown). As STIM1 is a key initiator of SOC, DNA sequencing for the STIM1 gene family in a larger population may be helpful to identify novel polymorphisms. Future studies are needed to address the mechanism by which calcium signaling contributes to the development of KD. (Figure 10)

Advertisement

7. Genetic polymorphisms of the TGF- signaling pathway in Kawasaki disease

TGF- is an important molecule that is involved in the regulation of cytokine expression and immune response. It has been shown that TGF--mediated signaling pathways are mainly via transcription factors, Smads, which include at least 3 common proteins: Smad2, Smad3, and Smad4. The binding of TGF- to its receptor results in the phosphorylation of Smad2 or Smad3, which heterodimerizes with Smad4. The formation of the Smad complex further translocates to the nulclus to regulate activation of the target genes. In the cardiovascular system, which is an important target of KD, TGF- signaling is involved in the pathogenesis of multiple cardiovascular diseases via aberrant vascular remodeling. Low expression levels of endogenous TGF- activity in the blood may contribute to the development of atherosclerotic cardiovascular disease. In 2011, a large genetic study revealed a significant association between the polymorphisms in TGF- pathways and KD susceptibility or CAL formation in the European and US populations. In this study, Shimizu et al. [23]were the first to identify 16 SNPs in 6 genes (TGFB2, TGFBR2, SMAD1, ENG, ACVRL1, and SMAD3) associated with the susceptibility to KD. The significance of genetic variation in 3 genes (SMAD3, TGFB2, and TGFBR2) could be replicated in the multiethnic TDT analysis from the independent United States/United Kingdom/Australia subjects.

Kuo et al. [26] performed a replication study of 12 polymorphisms in 950 Taiwanese children. It was confirmed that genetic polymorphisms of SMAD as well as TGFB2 contribute to the susceptibility of KD. These observations, in combination with those of the recent study, support the importance of TGF- pathways for the susceptibility or severity of KD.

Advertisement

8. Genome-wide association study (GWAS) in Kawasaki disease

In 2009, Burgner et al. were the first to perform a genome-wide association study (GWAS) on 119 Caucasian KD cases and 135 matched controls. Forty SNPs and 6 haplotypes were confirmed in an independent cohort of KD families. [132]This insightful work led to the identification of an SNP within the N-acetylated alpha-linked acidic dipeptidase-like 2 gene (NAALADL2; rs17531088], which was significantly associated with the susceptibility to KD. Although the function of NAALADL2 remains unclear, mutations in the gene may be involved in the development of Cornelia de Lange syndrome. In 2010, Kim et al. conducted another GWAS in a Korean population. [133] In total, 786 subjects (186 KD patients and 600 controls) were recruited. A locus in the 1p31 region was identified as a susceptibility locus for KD. Furthermore, the PELI1gene locus in the 2p13.3 region was confirmed to associate with the development of CAL in KD patients. In 2012, two independent research groups by Lee et al. [62] and Onouchi et al. [134] published GWAS data from Taiwanese and Japanese populations, respectively. The results suggested that BLK (encoding B-lymphoid tyrosine kinase) and CD40 are novel susceptibility genes for KD. Consistent with this findings, Kuo et al. conducted a case–control genetic association study and identified another polymorphism in the CD40 gene that associated with susceptibility to KD. Hence, the results from independent groups support a significant role of immune-related genes such as CD40 for KD and CAL formation. [135]

Advertisement

9. Conclusion

Several major advances have been made in understanding the genetic effects of the susceptibility and clinical status of KD over the past decade. Very recently, genome-wide association led 2 groups (Lee et al. and Onouchi et al.) to identify the same novel susceptibility loci as being important for KD in the Asian population. Although the exact functional role of these genes in KD is still unclear, at present, these loci could provide a new direction for future studies. We can expect to see more insightful research beginning to elucidate the genes responsible for KD susceptibility.

Advertisement

Acknowledgement

We thank Siou-Jin Chiu (Kaohsiung Medical University) for help with the figure and the National Science Council (Taiwan) for their research support.

References

  1. 1. KawasakiT.KosakiF.OkawaS.ShigematsuI.YanagawaH. A.newinfantile.acutefebrile.mucocutaneouslymph.nodesyndrome. . M. L. N. S.prevailingin.JapanPediatrics. 1974Sep;5432716
  2. 2. KawasakiT. .Acute febrile mucocutaneous syndrome with lymphoid involvement with specific desquamation of the fingers and toes in children). Arerugi. 1967Mar;163178222
  3. 3. Wang CL, Wu YT, Liu CA, Kuo HC, Yang KD. Kawasaki disease: infection, immunity and genetics.Pediatr Infect Dis J. 2005Nov;24119981004
  4. 4. Burns JC, Glode MP. Kawasaki syndrome. Lancet.2004Aug 7-13;364943353344
  5. 5. Kuo HC, Liang CD, Wang CL, Yu HR, Hwang KP, Yang KD.Serum albumin level predicts initial intravenous immunoglobulin treatment failure in Kawasaki disease. Acta Paediatr. 2010Oct;9910157883
  6. 6. HinksA.KeX.BartonA.EyreS.BowesJ.WorthingtonJ.et al.Association of the IL2RA/CD25 gene with juvenile idiopathic arthritis. Arthritis Rheum. 2009Jan;6012517
  7. 7. CADendrouPlagnol. V.FungE.YangJ. H.DownesK.JDCooperet.alCell-specific protein phenotypes for the autoimmune locus IL2RA using a genotype-selectable human bioresource. Nat Genet. 2009Sep;41910115
  8. 8. QuH. Q.VerlaanD. J.GeB.LuY.LamK. C.GrabsR.etal. A.cis-actingregulatory.variantin.theI. L. R. A.locusJ, Ge B, Lu Y, Lam KC, Grabs R, et al. A cis-acting regulatory variant in the IL2RA locus. J Immunol. 2009Oct 15;1838515862
  9. 9. Huang WC, Huang LM, Chang IS, Chang LY, Chiang BL, Chen PJ, et al.Epidemiologic features of Kawasaki disease in Taiwan, 20032006Pediatrics. 2009Mar;123(3):e401-5.
  10. 10. Park YW, Han JW, Park IS, Kim CH, Cha SH, Ma JS, et al.Kawasaki disease in Korea, 2003-2005. Pediatr Infect Dis J. 2007Sep;2698213
  11. 11. NakamuraY.YashiroM.UeharaR.SadakaneA.ChiharaI.AoyamaY.et al.Epidemiologic features of Kawasaki disease in Japan: results of the 2007-2008 nationwide survey. J Epidemiol. 20102043027
  12. 12. NakamuraY.YashiroM.UeharaR.OkiI.KayabaK.YanagawaH.Increasing incidence of Kawasaki disease in Japan: nationwide survey. Pediatr Int. 2008Jun;50328790
  13. 13. NakamuraY.YashiroM.UeharaR.SadakaneA.TsuboiS.AoyamaY.et al.Epidemiologic Features of Kawasaki Disease in Japan: Results of the 20092010Nationwide Survey. J Epidemiol. 2012Mar 10. doi:10.2188/jea.JE20110126.
  14. 14. TownleyR. G.BarlanI. B.PatinoC.VichyanondP.MinerviniM. C.SimasathienT.et al.The effect of BCG vaccine at birth on the development of atopy or allergic disease in young children. Ann Allergy Asthma Immunol. 2004Mar;9233505
  15. 15. NewburgerJ. W.TakahashiM.MAGerberGewitz. M. H.TaniL. Y.BurnsJ. C.et al.Diagnosistreatment.long-termmanagement.ofKawasaki.diseasea.statementfor.healthprofessionals.fromthe.Committeeon.RheumaticFever.EndocarditisKawasakiDisease.Councilon.CardiovascularDisease.inthe.YoungAmerican.HeartAssociation.Circulation. 2004Oct 26;11017274771
  16. 16. AkagiT.RoseV.BensonL. N.NewmanA.FreedomR. M.Outcome of coronary artery aneurysms after Kawasaki disease. J Pediatr. 1992Nov;121(5 Pt 1):689-94.
  17. 17. ShulmanS. T.De InocencioJ.HirschR.Kawasakidisease.Pediatr Clin North Am. 1995Oct;425120522
  18. 18. YuH. R.KuoH. C.SheenJ. M.WangL.LinI. C.WangC. L.etal. A.uniqueplasma.proteomicprofiling.withimbalanced.fibrinogencascade.inpatients.withKawasaki.diseasePediatr Allergy Immunol. 2009Nov;207699707
  19. 19. Wu MT, Hsieh KS, Lin CC, Yang CF, Pan HB.Images in cardiovascular medicine. Evaluation of coronary artery aneurysms in Kawasaki disease by multislice computed tomographic coronary angiography. Circulation. 2004Oct 5;110(14):e339.
  20. 20. Kuo HC, Yang KD, Juo SH, Liang CD, Chen WC, Wang YS, et al. ITPKC single nucleotide polymorphism associated with the Kawasaki disease in a Taiwanese population.PLoS ONE. 2011e17370.
  21. 21. Kuo HC, Yang KD, Chang WC, Ger LP, Hsieh KS. Kawasaki disease: an update on diagnosis and treatment.Pediatr Neonatol. 2012Feb;531411
  22. 22. NewburgerJ. W.SleeperL. A.Mc CrindleB. W.MinichL. L.GersonyW.VetterV. L.et al.Randomized trial of pulsed corticosteroid therapy for primary treatment of Kawasaki disease. N Engl J Med. 2007Feb 15;356766375
  23. 23. ShimizuC.JainS.DavilaS.HibberdM. L.LinK. O.MolkaraD.et al.Transforming growth factor-beta signaling pathway in patients with Kawasaki disease. Circ Cardiovasc Genet. 2011Feb;411625
  24. 24. Kuo HC, Yu HR,Juo SH, Yang KD, Wang YS, Liang CD, et al. CASP3 gene single-nucleotide polymorphism (rs72689236) and Kawasaki disease in Taiwanese children. J Hum Genet. 2011Feb;5621615
  25. 25. Kuo HC, Lin YJ, Juo SH, Hsu YW, Chen WC, Yang KD, et al.Lack of association between ORAI1/CRACM1 gene polymorphisms and Kawasaki disease in the Taiwanese children. J Clin Immunol. 2011Aug;3146505
  26. 26. KuoH. C.OnouchiY.HsuY. W.ChenW. C.JDHuangHuang. Y. H.et al.Polymorphisms of transforming growth factor-beta signaling pathway and Kawasaki disease in the Taiwanese population. J Hum Genet. 2011Dec;56128405
  27. 27. SabharwalT.ManlhiotC.BenselerS. M.TyrrellP. N.ChahalN.YeungR. S.et al.Comparison of factors associated with coronary artery dilation only versus coronary artery aneurysms in patients with Kawasaki disease. Am J Cardiol. 2009Dec 15;1041217437
  28. 28. KuoH. C.WangC. L.LiangC. D.YuH. R.ChenH. H.WangL.et al.Persistent monocytosis after intravenous immunoglobulin therapy correlated with the development of coronary artery lesions in patients with Kawasaki disease. J Microbiol Immunol Infect. 2007Oct;405395400
  29. 29. KuoH. C.YangK. D.LiangC. D.BongC. N.YuH. R.WangL.et al.The relationship of eosinophilia to intravenous immunoglobulin treatment failure in Kawasaki disease. Pediatr Allergy Immunol. 2007Jun;1843549
  30. 30. Liang CD, Kuo HC, Yang KD, Wang CL, Ko SF.Coronary artery fistula associated with Kawasaki disease. Am Heart J. 2009Mar;15735848
  31. 31. BrosiusC. L.NewburgerJ. W.BurnsJ. C.Hojnowski-DiazP.ZierlerS.LeungD. Y.Increased prevalence of atopic dermatitis in Kawasaki disease. Pediatr Infect Dis J. 1988Dec;7128636
  32. 32. BurnsJ. C.ShimizuC.ShikeH.NewburgerJ. W.SundelR. P.BakerA. L.et al.Family-based association analysis implicates IL-4 in susceptibility to Kawasaki disease. Genes Immun. 2005Aug;6543844
  33. 33. Liew WK, Lim CW, Tan TH, Wong KY, Tai BC, Quek SC, et al.The effect of Kawasaki disease on childhood allergies- a sibling control study. Pediatr Allergy Immunol. Aug;22548893
  34. 34. WebsterR. J.CarterK. W.WarringtonN. M.LohA. M.ZaloumisS.KuijpersT. W.et al.Hospitalisationwith.infectionasthma.allergyin.Kawasakidisease.patientstheirfamilies.genealogicalanalysis.usinglinked.populationdata. P.LoPLoS ONE.6(11):e28004.
  35. 35. KuoH. C.WangC. L.LiangC. D.YuH. R.HuangC. F.WangL.et al.Association of lower eosinophil-related T helper 2 (Th2) cytokines with coronary artery lesions in Kawasaki disease. Pediatr Allergy Immunol. 2009May;20326672
  36. 36. HiraoJ.HibiS.AndohT.IchimuraT.High levels of circulating interleukin-4 and interleukin-10 in Kawasaki disease. Int Arch Allergy Immunol. 1997Feb;11221526
  37. 37. AbeJ.EbataR.JibikiT.YasukawaK.SaitoH.TeraiM.Elevated granulocyte colony-stimulating factor levels predict treatment failure in patients with Kawasaki disease. J Allergy Clin Immunol. 2008Nov;1225100813e8.
  38. 38. MatsubaraT.KatayamaK.MatsuokaT.FujiwaraM.KogaM.FurukawaS.Decreased interferon-gamma (IFN-gamma)-producing T cells in patients with acute Kawasaki disease. Clin Exp Immunol. 1999Jun;11635547
  39. 39. KuoH. C.WangC. L.WangL.YuH. R.YangK. D.Patient characteristics and intravenous immunoglobulin product may affect eosinophils in Kawasaki disease. Pediatr Allergy Immunol. 2008Mar;1921845
  40. 40. Lin LY, Yang TH, Lin YJ, Yu HR, Yang KD, Huang YC, et al.Comparison of the Laboratory Data Between Kawasaki Disease and Enterovirus After Intravenous Immunoglobulin Treatment. Pediatr Cardiol. 2012Mar 25.doi:s00246-012-0293-9.
  41. 41. Liu YC, Hou CP, Kuo CM, Liang CD, Kuo HC. (Atypical kawasaki disease: literature review and clinical nursing).Hu Li Za Zhi. 2010Dec;57610410
  42. 42. AzabS. S.SalamaS. A.Abdel-NaimA. B.KhalifaA. E.El -DemerdashE.Al-HendyA. .Methoxyestradiol and multidrug resistance: can 2-methoxyestradiol chemosensitize resistant breast cancer cells? Breast Cancer Res Treat. 2009Jan;1131919
  43. 43. KobayashiT.SoneK.Effect of dipyridamole on the blood flow in coronary aneurysms resulting from Kawasaki disease. Pediatr Cardiol. 1994Nov-Dec;1562637
  44. 44. UeharaR.IgarashiH.YashiroM.NakamuraY.YanagawaH.Kawasaki disease patients with redness or crust formation at the Bacille Calmette-Guerin inoculation site. Pediatr Infect Dis J. 2010May;2954303
  45. 45. SonobeT.KiyosawaN.TsuchiyaK.AsoS.ImadaY.ImaiY.et al.Prevalence of coronary artery abnormality in incomplete Kawasaki disease. Pediatr Int. 2007Aug;4944216
  46. 46. ChangF. Y.HwangB.ChenS. J.LeeP. C.MengC. C.LuJ. H.Characteristics of Kawasaki disease in infants younger than six months of age. Pediatr Infect Dis J. 2006Mar;2532414
  47. 47. BurnsJ. C.WigginsJ. W.Jr ToewsW. H.NewburgerJ. W.LeungD. Y.WilsonH.et al.Clinical spectrum of Kawasaki disease in infants younger than 6 months of age. J Pediatr. 1986Nov;109575963
  48. 48. HeuclinT.DubosF.HueV.GodartF.FrancartC.VincentP.et al.Increased detection rate of Kawasaki disease using new diagnostic algorithm, including early use of echocardiography. J Pediatr. 2009Nov;15556959e1.
  49. 49. Kuo HC, Liang CD, Yu HR, Wang CL, Lin IC, Liu CA, et al. CTLA-4, position 49 A/G polymorphism associated with coronary artery lesions in Kawasaki disease.J Clin Immunol. 2011Apr;3122404
  50. 50. Hsieh KS, Weng KP, Lin CC, Huang TC, Lee CL, Huang SM.Treatment of acute Kawasaki disease: aspirin’s role in the febrile stage revisited. Pediatrics. 2004Dec;114(6):e68993
  51. 51. Kuo HC, Wang CL, Liang CD, et al.Revisit high dose aspirin in acute stage of Kawasaki disease. Taipei, Taiwan: 6th Asian Society for Pediatric Research; 2010
  52. 52. Lee JH, Hung HY, Huang FY.Kawasaki disease with Reye syndrome: report of one case. Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi. 1992Jan-Feb;3316771
  53. 53. TizardE. J.SuzukiA.LevinM.MJDillonClinical aspects of 100 patients with Kawasaki disease. Arch Dis Child. 1991Feb;6621858
  54. 54. BeutlerE.The molecular biology of G6PD variants and other red cell enzyme defects. Annu Rev Med. 1992434759
  55. 55. YoungsterI.ArcaviL.SchechmasterR.AkayzenY.PopliskiH.ShimonovJ.et al.Medications and glucose-6-phosphate dehydrogenase deficiency: an evidence-based review. Drug Saf. 2010Sep 1;33971326
  56. 56. BeutlerE. G. P. D.deficiencyBlood.1994Dec 1;8411361336
  57. 57. Li JS, Newburger JW.Antiplatelet therapy in pediatric cardiovascular patients. Pediatr Cardiol. 2010May;31445461
  58. 58. CattaneoG.GalvagnoG.MussaF. .Kawasaki disease in a subject with G6PD deficiency). Pediatr Med Chir. 1989Mar-Apr;11221921
  59. 59. Newburger JW, Takahashi M, Beiser AS, Burns JC, Bastian J, Chung KJ, et al. A single intravenous infusion of gamma globulin as compared with four infusions in the treatment of acute Kawasaki syndrome. N Engl J Med. 1991 Jun 6;324(23):1633-9.
  60. 60. Wang CL, Wu YT, Liu CA, Lin MW, Lee CJ, Huang LT, et al. Expression of CD40 ligand on CD4+ T-cells and platelets correlated to the coronary artery lesion and disease progress in Kawasaki disease.Pediatrics. 2003Feb;111(2):E1407
  61. 61. Wang CL, Wu YT, Lee CJ, Liu HC, Huang LT, Yang KD.Decreased nitric oxide production after intravenous immunoglobulin treatment in patients with Kawasaki disease. J Pediatr. 2002Oct;14145605
  62. 62. Lee YC, Kuo HC, Chang JS, Chang LY, Huang LM, Chen MR, et al.Two new susceptibility loci for Kawasaki disease identified through genome-wide association analysis. Nat Genet. 2012Mar 25;4455225
  63. 63. KhorC. C.DavilaS.BreunisW. B.LeeY. C.ShimizuC.WrightV. J.et al.Genome-wide association study identifies FCGR2A as a susceptibility locus for Kawasaki disease. Nat Genet. 2011Dec;431212416
  64. 64. MutaH.IshiiM.EgamiK.FuruiJ.SugaharaY.AkagiT.et al.Early intravenous gamma-globulin treatment for Kawasaki disease: the nationwide surveys in Japan. J Pediatr. 2004Apr;14444969
  65. 65. Fong NC, Hui YW, Li CK, Chiu MC.Evaluation of the efficacy of treatment of Kawasaki disease before day 5 of illness. Pediatr Cardiol. 2004Jan-Feb;251314
  66. 66. ASDajaniTaubert. K. A.MAGerberShulman. S. T.FerrieriP.FreedM.et al.Diagnosis and therapy of Kawasaki disease in children. Circulation. 1993May;875177680
  67. 67. MinichL. L.SleeperL. A.AtzA. M.Mc CrindleB. W.LuM.ColanS. D.et al.Delayed diagnosis of Kawasaki disease: what are the risk factors? Pediatrics. 2007Dec;120(6):e143440
  68. 68. Anderson MS, Todd JK, Glode MP.Delayed diagnosis of Kawasaki syndrome: an analysis of the problem. Pediatrics. 2005Apr;115(4):e42833
  69. 69. MarasiniM.PongiglioneG.GazzoloD.CampelliA.RibaldoneD.CaponnettoS.Late intravenous gamma globulin treatment in infants and children with Kawasaki disease and coronary artery abnormalities. Am J Cardiol. 1991Sep 15;6887967
  70. 70. TremouletA. H.BMBestSong. S.WangS.CorinaldesiE.EichenfieldJ. R.et al.Resistance to intravenous immunoglobulin in children with Kawasaki disease. J Pediatr. 2008Jul;153111721
  71. 71. RiganteD.ValentiniP.RizzoD.LeoA.De RosaG.OnesimoR.et al.Responsiveness to intravenous immunoglobulins and occurrence of coronary artery abnormalities in a single-center cohort of Italian patients with Kawasaki syndrome. Rheumatol Int. 2010Apr;3068416
  72. 72. SleeperL. A.MinichL. L.BMMc CrindleLi. J. S.MasonW.ColanS. D.et al.Evaluation of kawasaki disease risk-scoring systems for intravenous immunoglobulin resistance. J Pediatr. 2011May;15858315e3.
  73. 73. EgamiK.MutaH.IshiiM.SudaK.SugaharaY.IemuraM.et al.Prediction of resistance to intravenous immunoglobulin treatment in patients with Kawasaki disease. J Pediatr. 2006Aug;149223740
  74. 74. Kuo HC, Wu CC, Yang TH, Yu HR, Liang CD, Chen YJ, et al.Non-Langerhans cell histiocytosis in a child with Kawasaki disease. BMJ Case Rep. 2009
  75. 75. WrightD. A.NewburgerJ. W.BakerA.SundelR. P.Treatment of immune globulin-resistant Kawasaki disease with pulsed doses of corticosteroids. J Pediatr. 1996Jan;12811469
  76. 76. SonM. B.GauvreauK.BurnsJ. C.CorinaldesiE.TremouletA. H.WatsonV. E.et al.Infliximab for intravenous immunoglobulin resistance in Kawasaki disease: a retrospective study. J Pediatr. 2011Apr;15846449e1.
  77. 77. Ahn SY, Kim DS.Treatment of intravenous immunoglobulin-resistant Kawasaki disease with methotrexate. Scand J Rheumatol. 2005Mar-Apr;3421369
  78. 78. Pinna GS, Kafetzis DA, Tselkas OI, Skevaki CL. Kawasaki disease: an overview.Curr Opin Infect Dis. 2008Jun;21326370
  79. 79. MoriM.ImagawaT.KatakuraS.MiyamaeT.OkuyamaK.ItoS.et al.Efficacy of plasma exchange therapy for Kawasaki disease intractable to intravenous gamma-globulin. Mod Rheumatol. 2004141437
  80. 80. Chen HH, Liu PM, Bong CN, Wu YT, Yang KD, Wang CL.Methylprednisolone pulse therapy for massive lymphadenopathy in a child with intravenous immunoglobulin-resistant Kawasaki disease. J Microbiol Immunol Infect. 2005Apr;38214952
  81. 81. MiuraM.KohnoK.OhkiH.YoshibaS.SugayaA.SatohM.Effects of methylprednisolone pulse on cytokine levels in Kawasaki disease patients unresponsive to intravenous immunoglobulin. Eur J Pediatr. 2008Oct;16710111923
  82. 82. FurukawaT.KishiroM.AkimotoK.NagataS.ShimizuT.YamashiroY.Effects of steroid pulse therapy on immunoglobulin-resistant Kawasaki disease. Arch Dis Child. 2008Feb;9321426
  83. 83. OgataS.BandoY.KimuraS.AndoH.NakahataY.OgiharaY.et al.The strategy of immune globulin resistant Kawasaki disease: a comparative study of additional immune globulin and steroid pulse therapy. J Cardiol. 2009Feb;531159
  84. 84. HanR. K.SilvermanE. D.NewmanA.Mc CrindleB. W.Management and outcome of persistent or recurrent fever after initial intravenous gamma globulin therapy in acute Kawasaki disease. Arch Pediatr Adolesc Med. 2000Jul;15476949
  85. 85. MiuraM.OhkiH.YoshibaS.UedaH.SugayaA.SatohM.et al.Adverse effects of methylprednisolone pulse therapy in refractory Kawasaki disease. Arch Dis Child. 2005Oct;901010967
  86. 86. OkadaK.HaraJ.MakiI.MikiK.MatsuzakiK.MatsuokaT.et al.Pulse methylprednisolone with gammaglobulin as an initial treatment for acute Kawasaki disease. Eur J Pediatr. 2009Feb;16821815
  87. 87. BALangYeung. R. S.OenK. G.MallesonP. N.HuberA. M.RileyM.et al.Corticosteroid treatment of refractory Kawasaki disease. J Rheumatol. 2006Apr;3348039
  88. 88. OgataS.OgiharaY.NomotoK.AkiyamaK.NakahataY.SatoK.et al.Clinical score and transcript abundance patterns identify Kawasaki disease patients who may benefit from addition of methylprednisolone. Pediatr Res. 2009Nov;66557784
  89. 89. KobayashiT.SajiT.OtaniT.TakeuchiK.NakamuraT.ArakawaH.et al.Efficacy of immunoglobulin plus prednisolone for prevention of coronary artery abnormalities in severe Kawasaki disease (RAISE study): a randomised, open-label, blinded-endpoints trial. Lancet. 2012Mar 7.doi:S0140-6736(11)61930-2.
  90. 90. CPMaurySalo. E.PelkonenP.Elevated circulating tumor necrosis factor-alpha in patients with Kawasaki disease. J Lab Clin Med. 1989May;11356514
  91. 91. BurnsJ. C.BMBestMejias. A.MahonyL.FixlerD. E.JafriH. S.et al.Infliximab treatment of intravenous immunoglobulin-resistant Kawasaki disease. J Pediatr. 2008Dec;15368338
  92. 92. OishiT.FujiedaM.ShiraishiT.OnoM.InoueK.TakahashiA.et al.Infliximab treatment for refractory Kawasaki disease with coronary artery aneurysm. Circ J. 2008May;7258502
  93. 93. ZulianF.ZanonG.MartiniG.MescoliG.MilanesiO.Efficacy of infliximab in long-lasting refractory Kawasaki disease. Clin Exp Rheumatol. 2006Jul-Aug;24(4):453.
  94. 94. Kuo HC, Yu HR, Wu CC, Chang LS, Yang KD.Etanercept treatment for children with refractory juvenile idiopathic arthritis. J Microbiol Immunol Infect. 2011Jan 13;441526
  95. 95. Guo MM, Yang KD, Yu HR, Kuo HC.Hypersensitive joint reaction after etanercept treatment in a patient with juvenile rheumatoid arthritis. J Rheumatol. 2011Mar;3835779
  96. 96. Choueiter NF, Olson AK, Shen DD, Portman MA.Prospective open-label trial of etanercept as adjunctive therapy for kawasaki disease. J Pediatr. 2010Dec;15769606e1.
  97. 97. MAPortmanOlson. A.SorianoB.DahdahN.WilliamsR.KirkpatrickE.Etanercept as adjunctive treatment for acute Kawasaki disease: study design and rationale. Am Heart J. 2011Mar;16134949
  98. 98. SenzakiH.ChenC. H.IshidoH.MasutaniS.MatsunagaT.TaketazuM.et al.Arterial hemodynamics in patients after Kawasaki disease. Circulation. 2005Apr 26;11116211925
  99. 99. BorzutzkyA.GutierrezM.TalesnikE.GodoyI.KrausJ.HoyosR.et al.High sensitivity C-reactive protein and endothelial function in Chilean patients with history of Kawasaki disease. Clin Rheumatol. 2008Jul;27784550
  100. 100. Weng KP, Hsieh KS, Huang SH, Lin CC, Huang DC. Serum HDL level at acute stage of Kawasaki disease.Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi. 1998Jan-Feb;3912832
  101. 101. NewburgerJ. W.BurnsJ. C.ASBeiserLoscalzo. J.Altered lipid profile after Kawasaki syndrome. Circulation. 1991Aug;84262531
  102. 102. RodenburgJ.VissersM. N.WiegmanA.MDTripBakker. H. D.KasteleinJ. J.Familial hypercholesterolemia in children. Curr Opin Lipidol. 2004Aug;15440511
  103. 103. Huang SM, Weng KP, Chang JS, Lee WY, Huang SH, Hsieh KS.Effects of statin therapy in children complicated with coronary arterial abnormality late after Kawasaki disease: a pilot study. Circ J. 2008Oct;721015837
  104. 104. BlankierS.Mc CrindleB. W.ItoS.YeungR. S.The role of atorvastatin in regulating the immune response leading to vascular damage in a model of Kawasaki disease. Clin Exp Immunol. 2011May;1642193201
  105. 105. Williams RV, Wilke VM, Tani LY, Minich LL.Does Abciximab enhance regression of coronary aneurysms resulting from Kawasaki disease? Pediatrics. 2002Jan;109(1):E4.
  106. 106. McCandless RT, Minich LL, Tani LY, Williams RV.Does abciximab promote coronary artery remodeling in patients with Kawasaki disease? Am J Cardiol. 2010Jun 1;1051116258
  107. 107. SuzukiH.TeraiM.HamadaH.HondaT.SuenagaT.TakeuchiT.et al.CyclosporinA.treatmentfor.Kawasakidisease.refractoryto.initialadditionalintravenous.immunoglobulinPediatr Infect Dis J. 2011Oct;30108716
  108. 108. KuijpersT. W.BiezeveldM.AchterhuisA.KuipersI.LamJ.CEHacket.alLongstanding obliterative panarteritis in Kawasaki disease: lack of cyclosporin A effect. Pediatrics. 2003Oct;112498692
  109. 109. SuzukiH.SuenagaT.TakeuchiT.ShibutaS.YoshikawaN.Marker of T-cell activation is elevated in refractory Kawasaki disease. Pediatr Int. 2010Oct;5257859
  110. 110. JiaS.LiC.WangG.YangJ.ZuY.TheT.helpertype.17/regulatoryT.cellimbalance.inpatients.withacute.Kawasakidisease.Clin Exp Immunol. 2010Oct;16211317
  111. 111. FuryW.TremouletA. H.WatsonV. E.BMBestShimizu. C.HamiltonJ.et al.Transcript abundance patterns in Kawasaki disease patients with intravenous immunoglobulin resistance. Hum Immunol. 2010Sep;71986573
  112. 112. DergunM.KaoA.HaugerS. B.NewburgerJ. W.BurnsJ. C.Familial occurrence of Kawasaki syndrome in North America. Arch Pediatr Adolesc Med. 2005Sep;159987681
  113. 113. JibikiT.TeraiM.ShimaM.OgawaA.HamadaH.KanazawaM.et al.Monocyte chemoattractant protein 1 gene regulatory region polymorphism and serum levels of monocyte chemoattractant protein 1 in Japanese patients with Kawasaki disease. Arthritis Rheum. 2001Sep;44922112
  114. 114. Weng KP, Hsieh KS, Hwang YT, Huang SH, Lai TJ, Yuh YS, et al. IL-10 polymorphisms are associated with coronary artery lesions in acute stage of Kawasaki disease.Circ J. 2010May;7459839
  115. 115. Hsieh KS, Lai TJ, Hwang YT, Lin MW, Weng KP, Chiu YT, et al. IL-10 promoter genetic polymorphisms and risk of Kawasaki disease in Taiwan.Dis Markers. 2011301519
  116. 116. HsuehK. C.LinY. J.ChangJ. S.WanL.TsaiY. H.TsaiC. H.et al.Associationof.interleukin-A-5polymorphismC.inTaiwanese.childrenwith.Kawasakidisease.J, Chang JS, Wan L, Tsai YH, Tsai CH, et al. Association of interleukin-10 A-592C polymorphism in Taiwanese children with Kawasaki disease. J Korean Med Sci. 2009Jun;24343842
  117. 117. OnouchiY.OnoueS.TamariM.WakuiK.FukushimaY.YashiroM.etal. C. D.ligandgene.Kawasakidisease.Eur J Hum Genet. 2004Dec;121210628
  118. 118. ChenS. Y.WanL.HuangY. C.SheuJ. J.LanY. C.LaiC. H.et al.Interleukin-gene1.geneticA. C.polymorphismis.associatedwith.thesusceptibility.ofKawasaki.diseaseJJ, Lan YC, Lai CH, et al. Interleukin-18 gene 105A/C genetic polymorphism is associated with the susceptibility of Kawasaki disease. J Clin Lab Anal. 2009232716
  119. 119. Weng KP, Ho TY, Chiao YH, Cheng JT, Hsieh KS, Huang SH, et al.Cytokine genetic polymorphisms and susceptibility to Kawasaki disease in Taiwanese children. Circ J. 2010Nov;7412272633
  120. 120. LinY. J.WanL.WuJ. Y.SheuJ. J.LinC. W.LanY. C.et-Eal. H. L. A.genepolymorphism.associatedwith.susceptibilityto.Kawasakidisease.formationof.coronaryartery.aneurysmsArthritis Rheum. 2009Feb;60260410
  121. 121. BurnsJ. C.ShimizuC.GonzalezE.KulkarniH.PatelS.ShikeH.et al.Genetic variations in the receptor-ligand pair CCR5 and CCL3L1 are important determinants of susceptibility to Kawasaki disease. J Infect Dis. 2005Jul 15;19223449
  122. 122. BreunisW. B.BiezeveldM. H.GeisslerJ.KuipersI. M.LamJ.OttenkampJ.et al.Polymorphisms in chemokine receptor genes and susceptibility to Kawasaki disease. Clin Exp Immunol. 2007Oct;15018390
  123. 123. Jhang WK, Kang MJ, Jin HS, Yu J, Kim BJ, Kim BS, et al. The CCR5 (-2135C/T) polymorphism may be associated with the development of Kawasaki disease in Korean children. J Clin Immunol. 2009 Jan;29(1):22-8.
  124. 124. MamtaniM.MatsubaraT.ShimizuC.FurukawaS.AkagiT.OnouchiY.et al.Associationof. C. C. R.haplotypesC. C. R.copyC. C. L. L.numberwith.KawasakiDisease.coronaryartery.lesionsresponsesI. V. I. G.inJapanese.childrenP.LoPLoS ONE. 2010e11458.
  125. 125. OnouchiY.GunjiT.BurnsJ. C.ShimizuC.NewburgerJ. W.YashiroM.etal. I. T. P. K. C.functionalpolymorphism.associatedwith.Kawasakidisease.susceptibilityformationof.coronaryartery.aneurysmsNat Genet. 2008Jan;4013542
  126. 126. OnouchiY.SuzukiY.SuzukiH.TeraiM.YasukawaK.HamadaH.etal. I. T. P. K. C.polymorphismsC. A. S. P.risksfor. I. V. I. G.unresponsivenesscoronaryartery.lesionformation.inKawasaki.diseasePharmacogenomics J. 2011Oct 11.doi:tpj.2011.45.
  127. 127. ChiH.HuangF. Y.ChenM. R.ChiuN. C.LeeH. C.LinS. P.etal. I. T. P. K. C.geneS. N. P.rs284932Kawasakidisease.inTaiwanese.childrenHum Mol Genet. 2010Mar 15;196114751
  128. 128. Lin MT, Wang JK, Yeh JI, Sun LC, Chen PL, Wu JF, et al.Clinical Implication of the C Allele of the ITPKC Gene SNP rs28493229 in Kawasaki Disease: Association With Disease Susceptibility and BCG Scar Reactivation. Pediatr Infect Dis J. 2011Feb;30214852
  129. 129. OnouchiY.OzakiK.BunsJ. C.ShimizuC.HamadaH.HondaT.et al.Common variants in CASP3 confer susceptibility to Kawasaki disease. Hum Mol Genet. 2010Jul 15;19142898906
  130. 130. ChouY. H.JuoS. H.ChiuY. C.MELiuChen. W. C.ChangC. C.etal. A.polymorphismof.theO. R. A. I.geneis.associatedwith.therisk.recurrenceof.calciumnephrolithiasis.Juo SH, Chiu YC, Liu ME, Chen WC, Chang CC, et al. A polymorphism of the ORAI1 gene is associated with the risk and recurrence of calcium nephrolithiasis. J Urol. 2011May;185517426
  131. 131. Wei JC, Yen JH, Juo SH, Chen WC, Wang YS, Chiu YC, et al. Association of ORAI1 haplotypes with the risk of HLA-B27 positive ankylosing spondylitis.PLoS ONE. 2011e20426.
  132. 132. BurgnerD.DavilaS.BreunisW. B.NgS. B.LiY.BonnardC.etal. A.genome-wideassociation.studyidentifies.novelfunctionallyrelated.susceptibilityLoci.forKawasaki.diseaseP.LoPLoS Genet. 2009Jan;5(1):e1000319.
  133. 133. KimJ. J.HongY. M.SohnS.JangG. Y.HaK. S.YunS. W.etal. A.genome-wideassociation.analysisreveals.1p2p13assusceptibilityloci.forKawasaki.diseaseHum Genet. 2011May;129548795
  134. 134. OnouchiY.OzakiK.BurnsJ. C.ShimizuC.TeraiM.HamadaH.etal. A.genome-wideassociation.studyidentifies.threenew.riskloci.forKawasaki.diseaseNat Genet. 2012 Mar 25.doi:ng.2220. Nat Genet. 2012Mar 25;44551721
  135. 135. Kuo HC CM, Hsu YW, Lin YC, Huang YH, Yu HR, Hou MF, Liang CD, Yang KD, Chang WC andWang CL. CD40 gene polymorphisms associated with susceptibility of Kawasaki disease and coronary artery lesions in the Taiwanese population.The scientific world journal. 2012In press).ScientificWorldJournal. 2012;2012:520865.

Written By

Ho-Chang Kuo and Wei-Chiao Chang

Submitted: 23 November 2011 Published: 18 July 2012