GET THE APP

Utility of TNF-α as a Biomarker and the Possibility of anti-
Pediatrics & Therapeutics

Pediatrics & Therapeutics
Open Access

ISSN: 2161-0665

+44 1478 350008

Review Article - (2015) Volume 5, Issue 3

Utility of TNF-α as a Biomarker and the Possibility of anti-TNF-α Therapy for Kawasaki Disease

Keiichi Hirono and Fukiko Ichida*
Department of Pediatrics, University of Toyama, Japan
*Corresponding Author: Fukiko Ichida, Assistant Professor, Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan, Tel: +81-76-434-7311, Fax: +81-76-434-5029 Email:

Abstract

Kawasaki disease (KD) is the most common systemic vasculitis syndrome, primarily affecting the coronary arteries. Timely treatment with high-dose intravenous immunoglobulin (IVIG) reduces the duration of fever and incidence of Coronary Artery Lesions (CAL). However, even after IVIG treatment ∼5%-7% of patients develop aneurysms.

TNF-α is a cytokine with multiple biological effects produced primarily by monocytes and macrophages. In the last decade, TNF-α has been the focus of research aimed at uncovering its role during the acute phase of KD. Previous studies reported that TNF-α is responsible for the increase in its soluble receptors and is involved in the pathogenesis of the clinical features and CAL in KD.

Anti-TNF-α therapies, such as infliximab or etanercept, seem to be effective in controlling inflammation in patients with KD who fail to respond to IVIG. Several trials of the usage of infliximab as the first, second, or third line therapy for KD since 2004 showed that infliximab was safe and well tolerated, and patients treated with infliximab had fewer days of fever. Etanercept was recently shown to be safe and well tolerated as adjunctive initial therapy with IVIG in a small study of children with KD.

Keywords: Kawasaki disease, TNF-α

Abbreviations

KD: Kawasaki Disease; CAL: Coronary Artery Lesion; TNF: Tumor Necrosis Factor; IVIG: Intravenous Immunoglobulin; TNFR: Tumor Necrosis Factor Alpha Receptor; sTNFR: Soluble Tumor Necrosis Factor Alpha Receptor

Introduction

Inflammation has often been considered to be a nonspecific response and to play a bridging role in the activation of adaptive immunity. However, it is now accepted that inflammation is the product of an independent innate immune system closely linked to the adaptive immune system. The key mediators of inflammation are inflammatory cytokines, as determined by multiple lines of evidence both in vitro and in vivo. Because of the crucial role of inflammatory cytokines, such as tumor necrosis factor (TNF)-α, in the pathogenesis of autoimmune disorders, anticytokine treatment has been developed as a therapy for rheumatoid arthritis, juvenile idiopathic arthritis, inflammatory bowel diseases and Kawasaki disease (KD).

KD is the most common systemic vasculitis syndrome, primarily affecting small to medium-sized arteries, more particularly the coronary arteries [1]. KD was first described in 1967 and is now identified as the leading cause of acquired heart disease among children in developed countries [2]. The annual incidence of KD in children of Japanese descent is about 218 per 100,000 children less than five years of age, as compared with about 20 per 100,000 in the United States (US) [3,4]. Timely treatment with high-dose intravenous immunoglobulin (IVIG) reduces the duration of fever and incidence of coronary artery lesions (CAL). However, even after IVIG treatment, ∼5%-7% of patients develop aneurysms [5]. To date, the pathogenesis of KD is still not been fully elucidated. Activation of innate and adaptive immune systems is thought to be a central feature of KD. There have been many reports on elevation of plasma levels of multiple inflammatory cytokines, such as TNF-α, interleukin -1b, IL-6, IL-8, and interferon-γ, during the acute phase of KD.

In the last decade, TNF-α has been focused on its role during the acute phase of KD. Here, we review the role of TNF-α, its usefulness as a biomarker, and the effectiveness of anti-TNF-α therapy in KD.

TNF-α

TNF-α is a cytokine with multiple biological effects produced primarily by monocytes and macrophages. TNF-α is a polypeptide cytokine mainly produced by stimulated monocytes, macrophages, and T-lymphocyte subsets. TNF-α plays a key role during the immune response, and it is a potent mediator of inflammatory mechanisms in normal immune surveillance and in pathologic conditions. TNF-α exerts its effects through two cell-surface receptors of 55 and 75 kDa (TNF-α soluble receptors [TNFR] 1 and TNFR 2, respectively) [6]. These cell surface receptors function as transducing elements, providing the intracellular signal for the response to TNF-α. TNFR1 is present on most cells, particularly on those that are susceptible to the cytotoxic action of TNF-α. TNFR2 is also present on many cell types, particularly those of myeloid origin, and it is strongly expressed on stimulated T and B cells.

The histological observation of the association between KD and TNF-α was reported in a 3-month-old female infant with incomplete KD who suddenly died despite IVIG, aspirin, steroid, and heparin treatment [7]. Postmortem examination confirmed the echocardiographically detected giant coronary aneurysms and showed occlusive thrombosis in the giant aneurysm of the left anterior descending coronary artery (LAD). The coronary arteries, including the giant LAD aneurysm, of the KD case showed macrophage infiltration, neoangiogenesis, and immunoreactivity for TNF-α, tissue factor (TF), and thrombopoietin (TPO).

The role of TNF-α in the immune response leading to vascular damage was showed in a murine model of KD using Lactobacillus casei cell wall extract (LCWE) to induce coronary arteritis, where rapid production of TNF-α in the peripheral immune system occurred after disease induction [8]. It was reported that rapid production of TNF-α in the peripheral immune system occurred after disease induction in a murine model of KD. This immune response became site-directed, with migration to the coronary arteries dependent on TNF-α-mediated events. The production of TNF-α in the heart was coincident with the presence of inflammatory infiltrate at the coronary arteries, which persisted during the development of aneurysms. Inflammation and elastin breakdown in the coronary vessels were completely eliminated in the absence of TNF-α effector functions.

Genetic studies also support a role for TNF-α in KD. A systematic review and meta-analysis identified a trend of association between the TNF-α-308 polymorphism and KD [9-13]. The TNF-α-308 polymorphism might be associated with KD via TNF-α production that could induce cell damage [14,15]. This association may be mediated by complex interactions affecting the inflammatory cascade, linkage disequilibrium, or gene interaction, given that the TNF-α-308 polymorphism is linked with nearby polymorphisms, either in the same region, such as TNF-α-238,-857,-863,-1031, or in the major histocompatibility complex (MHC) region, such as the human leukocyte antigen (HLA)-A1, -B8, and -DR3 alleles [13,16,17]

Previous studies reported that the serum or plasma levels of TNF-α before initial treatment in patients with KD were higher than those in patients in the convalescent stage and in healthy controls. TNF-α, soluble TNFR1 (sTNFR1), and soluble TNFR2 (sTNFR2) concentrations are increased in the acute phase of KD and are highest in children who subsequently develop coronary artery aneurysms [18,19]. TNF-α is responsible for the increase in its soluble receptors and is involved in the pathogenesis of the clinical features and CAL in KD [9,18-24]. However, other previous studies reported that the serum or plasma levels of TNF-α had no correlation with clinical and laboratory parameters (Table 1) [25-27]. These discrepancies may come from the stability of TNF-α. Because TNF-α is cleared very rapidly from the circulation, its levels at a given moment may not necessarily reflect local production in the preceding hours [28]. TNF-α is frequently undetectable, and some assays are unable to detect TNF-α bound to sTNFRs; on the other hand, sTNFRs are very stable and can also be determined in stored sera. In addition, serum levels of both sTNFR1 and sTNFR2 correlate well with those of TNF-α [29-31]. Recent studies showed that several immunoassays for TNF-α gave markedly different results even when using international standards [32,33]. Another reason for these discrepancies might be related to the presence of TNF-α/sTNFR complexes and the levels of sTNFR1 or sTNFR2 [34,35]. In addition to sTNFRs in plasma, other factors, including the presence of monomeric or oligomeric forms, and of degradation products of TNF-α, or binding of TNF-α to other molecules, such as immunoglobulins or α2-macroglobulin, may be responsible for these discrepancies by altering TNF-α antigenic properties [36-39]. The difficulties related to TNF-α measurement in plasma make it difficult to compare the results obtained in different laboratories using different methods.

Year Number of patients Serum/plasma TNF-α/sTNFR Concentration level Significance between patients and controls Significance between CAL and nonCAL References
Pre-initial treatment Post-initial treatment 2nd treatment (nonresponder) Convalescent Control Patient with CAL Patient without CAL
2014 40 serum TNF-α 3.4±18.5 pg/mL       0.32±0.67 pg/mL (FC, n=32) 0.59±1.03 pg/mL (AC, n=15) 9.21±31.10 pg/mL (n=12) 0.21±0.22 pg/mL (n=28) no no [25]
2013 143 serum TNF-α 2.5 pg/mL 2.4 pg/mL   2.5 pg/mL (n=33) 3.8 pg/mL (n=80)     no N/A [26]
2012 39 plasma TNF-α 33.2±63.1 pg/mL 18.3±61.8 pg/mL     13.8±61.1 pg/mL (n=15)     yes N/A [18]
2003 24 serum TNF-α 24.1±9.4 pg/mL 11.8±5.8 pg/mL     10.4±4.9 pg/mL (n=12)     yes N/A [9]
2000 30 plasma TNF-α 15.4±11.5 pg/mL     8.8±7.1 pg/mL 7.8±6.9 (children, n=17) 8.2±7.4 (adults, n=10)     no N/A [27]
1990 45 serum TNF-α 18 cases (37.8%) >10 units/mL       <10 units/mL 10/11 (90.1%) >10 units/mL 7/34 (20.6%) >10 units/mL yes yes [19]
1989 39 serum TNF-α           51.1±13.6 pg/mL (n=4) 30.4±15.8 pg/mL (n=35) yes yes [20]
1988 30 serum TNF-α 12 cases (40%) >10 units/mL       <10 units/mL 6/6 (100%) >10 units/mL 6/24 (25%) >10 units/mL yes yes [21]
2009   serum sTNFR1 0.58±0.18 ng/mL (Res, n=18) 0.33 ng/mL (Res, n=18) 0.71±0.16 ng/mL (IFX, n=11) 0.60±0.24 ng/mL (nonRes, n=14) 0.39±0.16 ng/mL (IFX, n=11) 0.47±0.22ng/mL (nonRes, n=14) 0.22±0.08 ng/mL (n=33)     yes N/A [22]
2008 24 serum sTNFR1, 2   sTNFR1 8164.0 pg/mL sTNFR2 4405.5 pg/mL (nonRes, n=11) sTNFR1 12428.0 pg/mL sTNFR2 5784.0 pg/mL (IFX, n=12)         yes N/A [23]
1994 48 serum sTNFR1 6.3±4.2 ng/mL     1.6±0.8 ng/mL 1.5±0.5 ng/mL 13.9±3.5 ng/mL 5.4±3.2 ng/mL yes yes [24]

TNF-α: Tumor Necrosis Factor-α; sTNFR : Soluble Tumor Necrosis Factor-α Receptor; CAL: Coronary Artery Lesions; FC: Febrile Controls; AC: Afebrile Controls; IFX: Infliximab; nonRes, nonresponders; Res, responders

Table 1: Recent studies of TNF-α and TNFR as a biomarker in KD.

Anti-TNF-α Therapy

Infliximab

On the basis of the evidence that serum levels of the proinflammatory cytokine TNF-α are elevated in patients with acute KD, with the highest levels observed in patients who develop coronary artery abnormalities, TNF-α blockade using anti-TNF-α therapy, such as infliximab and etanercept, might be thought to be effective in controlling inflammation in patients with KD who fail to respond to IVIG (Figure 1 and Table 2) [19].

pediatrics-therapeutics-construct-biological-agents

Figure 1: Construct of anti-TNF-α biological agents.

Generic name Trade name Structure Antibody source Mechanism of TNF-α inhibition Half-life Administration pathway
Infliximab Remicade 25% murine, 75% human. Variable region of a mouse monoclonal anti-TNF-α antibody coupled to the construct region of a human IgG1 Human-mouse Chimeric human–mouse IgG1 monoclonal antibody that inhibits binding of TNF-α 8-10 days Intravenous
Adalimumab Humira 100% human peptide sequence and structure.Full-length human IgG1. Phage display technology resulting in human-derived variable regions and human IgG1; k constant regions Human Fully human IgG1 monoclonal antibody that inhibits binding of TNF-α 10-20 days Subcutaneous
Etanercept Enbrel 100% human peptide but artificial construction. Fusion protein made up of two soluble TNF receptor molecules (TNFR2) fused with the Fc fragment from human IgG1 Human + TNF-α receptor Recombinant TNF-α receptor fusion protein that competitively inhibits the binding of TNF-α 3.0-5.5 days Subcutaneous

TNF-α, tumor necrosis factor-α; IgG1, immunoglobulin G1

Table 2: Biological characteristics of anti-TNF-α agents.

Infliximab (Remicade) is a chimeric murine/human immunoglobulin G (IgG)-1 monoclonal antibody that binds specifically to human TNF-α-1 [40] Infliximab is indicated for treating immunemodulated inflammatory disorders, including pediatric Crohn’s disease [41]. The first case report of a child treated with infliximab for KD resistant to several doses of IVIG and methylprednisolone was published in 2004 [42]. This observation led to the trial and successful use of infliximab (5 mg/kg) as the third-line therapy in several case reports and small case series of KD patients with IVIG-resistant disease (Table 3) [42-46]. These reports were followed by a small US multicenter, randomized trial of second IVIG infusion vs. infliximab in 24 children with KD after failure of initial treatment with IVIG. An analysis of the Pediatric Health Information System database from 2001 to 2006 showed that 14 of the 27 participating hospitals had administered infliximab to 48 patients for treatment-resistant KD [47] of these 48 patients, 12 had received infliximab as part of a phase 1 multicenter, randomized, prospective trial of second IVIG vs. infliximab for IVIGresistant KD [23]. This study showed that infliximab was safe and well tolerated. Subsequently, a two-center retrospective review of IVIGresistant patients treated with either a second course of IVIG (n=86) or infliximab (n=20) indicated that patients treated with infliximab had fewer days of fever (median 8 vs. 10 days, p=0.028) and shorter lengths of hospitals stay (median 5.5 vs. 6.0 days, p=0.04) than those given a second IVIG dose [48]. Additional prospective case series from Korea and Japan have also demonstrated the efficacy of infliximab in 13 of 16 (81%) and 18 of 20 (90%) patients with IVIG-resistant KD, respectively [49,50]. Taken together, these studies suggest that a single infusion of 5mg/kg of infliximab is safe and well tolerated and attributed to refractory KD (Table 2).

Anti-TNF-α drug Year Country Illness day of IFX infusion Timing of administration Total Additional therapy CAL
Total IVIG IVMP IFX PE CsA
Infliximab 2008 USA 7.5 2nd (n =12)3rd (n =4) 16 3 (19%) 1 2 0 0 0 N/A (giant CAL 1)
Infliximab 2009 Japan 10.2±1.8 3rd 11 3 (27%) 1 3 0 0 1 4 (36%)
Infliximab 2010 Korea 16.5±6.3 3rd 16 3 (19%) 1 1 1 0 0 5 (31.2%)
Infliximab 2011 USA 7 2nd 20 3 (15%) 15 3 0 0 0 7 (35%)
Infliximab 2014 Japan 8.5±0.14 3rd 76 6 (7.9%) 0 0 0 6 0 3 (3.8%)
Infliximab 2014 Japan 10.5 3rd 17 0 (0%) 0 0 0 0 0 4 (23.5%)
Infliximab 2014 USA 6 1st (with IVIG) 98 11 (11.2%) 2 0 0 0 0 26 (27.1%)
Etanercept 2010 USA <10 2nd 15 0 (0%) 0 0 0 0 0 0 (0%)

TNF-α:Tumor Necrosis Factor-α; KD: Kawasaki Disease; IFX: Infliximab; IVIG: Intravenous Immunoglobulins; IVMP: Intravenous Methylprednisolone Pulse; PE: Plasma Exchange; CsA: Cyclosporin A; CAL: Coronary Artery Lesions.

Table 3: Recent studies of anti-TNF-α therapy in KD.

To further evaluate the role of infliximab in the treatment of KD, a phase 3, randomized, double-blind, placebo-controlled trial was undertaken in two children’s hospitals in the US to assess the addition of infliximab (5mg/ kg) to standard therapy [51]. In total, 196 patients were enrolled and randomized: 98 to the infliximab group and 98 to the placebo. Treatment resistance rate did not differ significantly between the infliximab group and the placebo group. Compared with the placebo group, participants given infliximab had fewer days of fever (median 1 day for infliximab vs. 2 days for placebo; p<0.0001). The infliximab group had a greater mean reduction in C-reactive protein concentration (p=0.0003) and in absolute neutrophil count (p=0.024) at 24h after treatment than did those given placebo, but, by week 2, this difference was not significant. No significant differences were recorded between the two groups in proximal coronary artery Z scores or any other laboratory markers of inflammation measured. They concluded that the addition of infliximab to primary treatment in acute KD did not reduce treatment resistance.

To clarify the role of infliximab in the patients with KD who were resistant to initial and additional therapies and were treated with infliximab, Hirono K et al. investigated the dynamic changes of cytokines during infliximab treatment [22]. They measured serum levels of sTNFR1 and interleukin (IL)-6, as well as vascular endothelial growth factor (VEGF), and the damage-associated molecular pattern (DAMP) molecules myeloid-related protein (MRP) 8/MRP14 and S100A12 sequentially. Although serum levels of proinflammatory cytokines decreased dramatically after infliximab treatment, DAMP molecules, VEGF, and markers of local tissue damage were not suppressed. In IVIG responders, all cytokines decreased markedly after IVIG treatment. Different behaviors of proinflammatory cytokines compared with those of DAMP molecules and VEGF after infliximab treatment suggest that infliximab is effective for the suppression of cytokine-mediated inflammation, but could not completely block local vasculitis.

To investigate the mechanism of action of infliximab therapy in the setting of KD, Ogihara Y et al. used microarray platforms to determine the transcript abundance profiles in whole blood obtained from eight IVIG-resistant KD subjects treated with infliximab therapy and compared them with those in initially IVIG-responsive subjects [52]. The pathway analysis showed a reduced abundance of transcripts in the nucleotide-binding oligomerization domain, matrix metalloproteinase (MMP), and inflammatory cytokine pathways and an increased abundance of transcripts in the T-cell receptor, apoptosis, tumor growth factor (TGF)-β, and IL-2 pathways in the IVIG-resistant, infliximab-treated subjects. The transcript abundance, which was found to be related to signaling pathways of KD inflammation, such as those involving IL-1, IL-6, and TNF-α, changed significantly following the administration of infliximab therapy, suggesting that infliximab therapy regulates important cytokine signaling activities involved in KD inflammation by blocking TNF-α, which may be critical for regulating IVIG resistance factors.

These two observations regarding the dynamic changes of cytokines in KD suggested that infliximab may regulate cytokine signaling relevant to KD inflammation via TNF-α blockade, but could not completely prevent local vasculitis because TNF-α may not have a central role in the development of vasculitis.

Etanercept

Etanercept (Enbrel) is another TNF-α inhibitor that has been studied in a small number of KD patients. Etanercept is a sTNFR and functions as a TNF antagonist with a proposed similar mechanism of action to infliximab in the treatment of KD (Figure 1 and Table 2). Similarly to infliximab, etanercept has been widely used in a broad array of autoimmune and inflammatory diseases. Etanercept was recently shown to be safe and well tolerated as adjunctive initial therapy with IVIG in a small study of children with KD [53]. None of the patients treated in this study-required retreatment. On the basis of these preliminary data, there is a proposed multicenter, doubleblind, randomized, placebo-controlled trial looking at the efficacy of etanercept in addition to IVIG plus aspirin for initial therapy at reducing the rate of IVIG-resistant disease [54].

Recently, three studies reported the role of two TNF-α blockers, infliximab and etanercept, using murine models of KD. Hui-Yuen JS et al. examined the role of TNF-α in the immune response leading to vascular damage in the mouse model of KD that involves the injection of LCWE [55]. Mice treated with the TNF-α-blocking agent etanercept, as well as TNFR1 knockout mice, were resistant to both coronary arteritis development and coronary aneurysm formation. It was concluded that TNF-α was necessary for the development of CAL in an animal model of KD. Oharaseki et al. studied the role of TNFα in a murine model of KD arteritis induced with Candida Albicans Water- Soluble fraction (CAWS) and treated with antiTNFα drugs (etanercept and infliximab) [56]. Their histopathological analysis revealed that the administration of etanercept to the mice reduced not only the incidence of vasculitis but also the scope of lesions and degree of inflammation. Ohashi R et al. reported that etanercept suppresses arteritis in a murine model of KD [51,57]. They compared the efficacy of IVIG, etanercept, methylprednisolone and cyclosporine-A in suppressing CAWSinduced vasculitis in DBA/2 mice. Etanercept was the most effective not only in suppressing inflammation but also in decreasing plasma cytokine levels, and there was a strong correlation between the extent of the vasculitis and the plasma TNF-α levels.

These studies of KD murine models suggest that TNF-α has an important role in the pathogenesis of vasculitis in KD, and etanercept could potentially be a new effective therapy for arteritis in KD. Further clinical trials are required to confirm the efficacy and safety of etanercept therapy and establish its usefulness in the treatment of KD.

Limitations

We review the current knowledge regarding the role of TNF-α and the utilities of infliximab and etanercept. It has been suggest that TNF-α has an important role in the pathogenesis of vasculitis in KD and etanercept could be used as a new effective therapy for arteritis in KD. Further studies will be warranted to confirm the efficacy and safety of etanercept therapy and establish its usefulness in the treatment of KD.

References

  1. Kato H, Sugimura T, Akagi T, Sato N, Hashino K, et al. (1996) Long-term consequences of Kawasaki disease. A 10- to 21-year follow-up study of 594 patients. Circulation 94: 1379-1385.
  2. Wang CL, Wu YT, Liu CA, Kuo HC, Yang KD (2005) Kawasaki disease: infection, immunity and genetics. Pediatr Infect Dis J 24: 998-1004.
  3. Nakamura Y, Yashiro M, Uehara R, Sadakane A, Tsuboi S, et al. (2012) Epidemiologic features of Kawasaki disease in Japan: results of the 2009-2010 nationwide survey. J Epidemiol 22: 216-221.
  4. Holman RC, Belay ED, Christensen KY, Folkema AM, Steiner CA, et al. (2010) Hospitalizations for Kawasaki syndrome among children in the United States, 1997-2007. Pediatr Infect Dis J 29: 483-488.
  5. Ogata S, Tremoulet AH, Sato Y, Ueda K, Shimizu C, et al. (2013) Coronary artery outcomes among children with Kawasaki disease in the United States and Japan. Int J Cardiol 168: 3825-3828.
  6. Hohmann HP, Remy R, Brockhaus M, van Loon AP (1989) Two different cell types have different major receptors for human tumor necrosis factor (TNF alpha). J Biol Chem 264: 14927-14934.
  7. Pucci A, Martino S, Celeste A, Linari A, Tibaldi M, et al. (2008) Angiogenesis, tumor necrosis factor-alpha and procoagulant factors in coronary artery giant aneurysm of a fatal infantile Kawasaki disease. Cardiovasc Pathol 17: 186-189.
  8. Hui-Yuen JS, Duong TT, Yeung RS (2006) TNF-alpha is necessary for induction of coronary artery inflammation and aneurysm formation in an animal model of Kawasaki disease. J Immunol 176: 6294-6301.
  9. Ahn SY, Jang GC, Shin JS, Shin KM, Kim DS (2003) Tumor necrosis factor-alpha levels and promoter polymorphism in patients with Kawasaki disease in Korea. Yonsei Med J 44: 1021-1026.
  10. Cheung YF, Huang GY, Chen SB, Liu XQ, Xi L, et al. (2008) Inflammatory gene polymorphisms and susceptibility to kawasaki disease and its arterial sequelae. Pediatrics 122: e608-614.
  11. Chien YH, Chang KW, Yang YH, Lu MY, Lin YT, et al. (2003) Association between levels of TNF-alpha and TNF-alpha promoter -308 A/A polymorphism in children with Kawasaki disease. J Formos Med Assoc 102: 147-150.
  12. Kamizono S, Yamada A, Higuchi T, Kato H, Itoh K (1999) Analysis of tumor necrosis factor-alpha production and polymorphisms of the tumor necrosis factor-alpha gene in individuals with a history of Kawasaki disease. Pediatr Int 41: 341-345.
  13. Quasney MW, Bronstein DE, Cantor RM, Zhang Q, Stroupe C, et al. (2001) Increased frequency of alleles associated with elevated tumor necrosis factor- alpha levels in children with Kawasaki disease. Pediatr Res 49: 686-690.
  14. Leung DY, Collins T, Lapierre LA, Geha RS, Pober JS (1986) Immunoglobulin M antibodies present in the acute phase of Kawasaki syndrome lyse cultured vascular endothelial cells stimulated by gamma interferon. J Clin Invest 77: 1428-1435.
  15. Leung DY, Geha RS, Newburger JW, Burns JC, Fiers W, et al. (1986) Two monokines, interleukin 1 and tumor necrosis factor, render cultured vascular endothelial cells susceptible to lysis by antibodies circulating during Kawasaki syndrome. J Exp Med 164: 1958-1972.
  16. Kamizono S, Yamada A, Higuchi T, Kato H, Itoh K (1999) Analysis of tumor necrosis factor-alpha production and polymorphisms of the tumor necrosis factor-alpha gene in individuals with a history of Kawasaki disease. Pediatr Int 41: 341-345.
  17. Wilson AG, de Vries N, Pociot F, di Giovine FS, van der Putte LB, et al. (1993) An allelic polymorphism within the human tumor necro- sis factor alpha promoter region is strongly associated with HLA A1, B8, and DR3 alleles. J Exp Med 177: 557-560.
  18. Lin IC, Kuo HC, Lin YJ, Wang FS, Wang L, et al. (2012) Augmented TLR2 expression on monocytes in both human Kawasaki disease and a mouse model of coronary arteritis. PLoS One 7: e38635.
  19. Furukawa S, Matsubara T, Jujoh K, Yone K, Sugawara T, et al. (1988) Peripheral blood monocyte/macrophages and serum tumor necrosis factor in Kawasaki disease. Clin Immunol Immunopathol 48: 247-251.
  20. Maury CP, Salo E, Pelkonen P (1989) Elevated circulating tumor necrosis factor-alpha in patients with Kawasaki disease. J Lab Clin Med 113: 651-654.
  21. Matsubara T, Furukawa S, Yabuta K (1990) Serum levels of tumor necrosis factor, interleukin 2 receptor, and interferon-gamma in Kawasaki disease involved coronary-artery lesions. Clin Immunol Immunopathol 56: 29-36.
  22. Hirono K, Kemmotsu Y, Wittkowski H, Foell D, Saito K, et al. (2009) Infliximab reduces the cytokine-mediated inflammation but does not suppress cellular infiltration of the vessel wall in refractory Kawasaki disease. Pediatr Res 65: 696-701.
  23. Burns JC, Best BM, Mejias A, Mahony L, Fixler DE, et al. (2008) Infliximab treatment of intravenous immunoglobulin-resistant Kawasaki disease. J Pediatr 153: 833-838.
  24. Furukawa S, Matsubara T, Umezawa Y, Okumura K, Yabuta K (1994) Serum levels of p60 soluble tumor necrosis factor receptor during acute Kawasaki disease. J Pediatr 124: 721-725.
  25. Kim HJ, Choi EH, Kil HR (2014) Association between adipokines and coronary artery lesions in children with Kawasaki Disease. J Korean Med Sci 29: 1385-1390.
  26. Wang Y, Wang W, Gong F, Fu S, Zhang Q, et al. (2013) Evaluation of intravenous immunoglobulin resistance and coronary artery lesions in relation to Th1/Th2 cytokine profiles in patients with Kawasaki disease. Arthritis Rheum 65: 805-814.
  27. Takeshita S, Nakatani K, Tsujimoto H, Kawamura Y, Kawase H, et al. (2000) Increased levels of circulating soluble CD14 in Kawasaki disease. Clin Exp Immunol 119: 376-381.
  28. Beutler BA, Milsark IW, Cerami A (1985) Cachectin/tumor necrosis factor: production, distribution, and metabolic fate in vivo. J Immunol 135: 3972-3977.
  29. Engelberts I, Möller A, Schoen GJ, van der Linden CJ, Buurman WA (1991) Evaluation of measurement of human TNF in plasma by ELISA. Lymphokine Cytokine Res 10: 69-76.
  30. Aukrust P, Liabakk NB, Müller F, Lien E, Espevik T, et al. (1994) Serum levels of tumor necrosis factor-a (TNF-a) and soluble TNF receptors in human immunodefi- ciency virus type I infection. Correlations to clinical immu- nologic, and virologic parameters. J Infect Dis: 169: 420-424.
  31. Zangerle R, Gallati H, Sarcletti M, Wachter H, Fuchs D (1994) Tumor necrosis factor alpha and soluble tumor necrosis factor receptors in individuals with human immunodeficiency virus infection. Immunol Lett 41: 229-234.
  32. Ledur A, Fitting C, David B, Hamberger C, Cavaillon JM (1995) Variable estimates of cytokine levels produced by commercial ELISA kits: results using international cytokine standards. J Immunol Methods 186: 171-179.
  33. De Kossodo S, Houba V, Grau GE (1995) Assaying tumor necrosis factor concentrations in human serum. A WHO International Collaborative Study. J Immunol Methods 182: 107-114.
  34. Corti A, Poiesi C, Merli S, Cassani G (1994) Tumor necrosis factor (TNF) alpha quantification by ELISA and bioassay: effects of TNF alpha-soluble TNF receptor (p55) complex dissociation during assay incubations. J Immunol Methods 177: 191-198.
  35. Engelberts I, Stephens S, Francot GJ, van der Linden CJ, Buurman WA (1991) Evidence for different effects of soluble TNF-receptors on various TNF measurements in human biological fluids. Lancet 338: 515-516.
  36. Meager A, Leung H, Woolley J (1989) Assays for tumour necrosis factor and related cytokines. J Immunol Methods 116: 1-17.
  37. Duncombe AS, Brenner MK (1988) Is circulating tumor necrosis factor bioactive? N Engl J Med 319: 1227-1228.
  38. Fomsgaard A, Svenson M, Bendtzen K (1989) Auto-antibodies to tumour necrosis factor alpha in healthy humans and patients with inflammatory diseases and gram-negative bacterial infections. Scand J Immunol 30: 219-223.
  39. James K (1990) Interactions between cytokines and alpha 2-macroglobulin. Immunol Today 11: 163-166.
  40. Knight DM, Trinh H, Le J, Siegel S, Shealy D, et al. (1993) Construction and initial characterization of a mouse-human chimeric anti-TNF antibody. Mol Immunol 30: 1443-1453.
  41. Baldassano R, Braegger CP, Escher JC, DeWoody K, Hendricks DF, et al. (2003) Infliximab (REMICADE) therapy in the treatment of pediatric Crohn's disease. Am J Gastroenterol 98: 833-838.
  42. Weiss JE, Eberhard BA, Chowdhury D, Gottlieb BS (2004) Infliximab as a novel therapy for refractory Kawasaki disease. J Rheumatol 31: 808-810.
  43. Burns JC, Mason WH, Hauger SB, Janai H, Bastian JF, et al. (2005) Infliximab treatment for refractory Kawasaki syndrome. J Pediatr 146: 662-667.
  44. Girish M, Subramaniam G (2008) Infliximab treatment in refractory Kawasaki syndrome. Indian J Pediatr 75: 521-522.
  45. Oishi T, Fujieda M, Shiraishi T, Ono M, Inoue K, et al. (2008) Infliximab treatment for refractory Kawasaki disease with coronary artery aneurysm. Circ J 72: 850-852.
  46. O'connor MJ, Saulsbury FT (2007) Incomplete and atypical Kawasaki disease in a young infant: severe, recalcitrant disease responsive to infliximab. Clin Pediatr (Phila) 46: 345-348.
  47. Son MB , Gauvreau K, Ma L, Baker AL, Sundel RP, et al. (2009) Treatment of Kawasaki disease: analysis of 27 US pediatric hospitals from 2001 to 2006. Pediatrics 124: 1-8.
  48. Son MB, Gauvreau K, Burns JC, Corinaldesi E, Tremoulet AH, et al. (2011) Infliximab for intravenous immunoglobulin resistance in Kawasaki disease: a retrospective study. J Pediatr 158: 644-649.
  49. Song MS, Lee SB, Sohn S, Oh JH, Yoon KL, et al. (2010) Infliximab treatment for refractory kawasaki disease in korean children. Korean Circ J 40: 334-338.
  50. Mori M, Imagawa T, Hara R, Kikuchi M, Hara T, et al. (2012) Efficacy and limitation of infliximab treatment for children with Kawasaki disease intractable to intravenous immunoglobulin therapy: report of an open-label case series. J Rheumatol 39: 864-867.
  51. Tremoulet AH, Jain S, Jaggi P, Jimenez-Fernandez S, Pancheri JM, et al. (2014) Infliximab for intensification of primary therapy for Kawasaki disease: a phase 3 randomised, double-blind, placebo-controlled trial. Lancet 383: 1731-1738.
  52. Ogihara Y, Ogata S, Nomoto K, Ebato T, Sato K, et al. (2014) Transcriptional regulation by infliximab therapy in Kawasaki disease patients with immunoglobulin resistance. Pediatr Res 76: 287-293.
  53. Choueiter NF, Olson AK, Shen DD, Portman MA (2010) Prospective open-label trial of etanercept as adjunctive therapy for kawasaki disease. J Pediatr 157: 960-966.
  54. Portman MA, Olson A, Soriano B, Dahdah N, Williams R, et al. (2011) Etanercept as adjunctive treatment for acute Kawasaki disease: study design and rationale. Am Heart J 161: 494-499.
  55. Hui-Yuen JS, Duong TT, Yeung RS (2006) TNF-alpha is necessary for induction of coronary artery inflammation and aneurysm formation in an animal model of Kawasaki disease. J Immunol 176: 6294-6301.
  56. Oharaseki T, Yokouchi Y, Yamada H, Mamada H, Muto S, et al. (2014) The role of TNF-α in a murine model of Kawasaki disease arteritis induced with a Candida albicans cell wall polysaccharide. Mod Rheumatol 24: 120-128.
  57. Ohashi R, Fukazawa R, Watanabe M, Tajima H, Nagi-Miura N, et al. (2013) Etanercept suppresses arteritis in a murine model of kawasaki disease: a comparative study involving differentbiological agents. Int J Vasc Med 2013:10.
Citation: Hirono K, Ichida F (2015) Utility of TNF-α as a Biomarker and the Possibility of anti-TNF-α Therapy for Kawasaki Disease. Pediat Therapeut 5:257.

Copyright: © 2015 Hirono K, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top