Skip to main content
Chinese Medical Journal logoLink to Chinese Medical Journal
. 2024 Sep 5;137(19):2294–2307. doi: 10.1097/CM9.0000000000003250

Stevens–Johnson syndrome and toxic epidermal necrolysis: Updates in pathophysiology and management

Akito Hasegawa 1,, Riichiro Abe 1,
Editor: Lishao Guo
PMCID: PMC11441865  PMID: 39238098

Abstract

Stevens–Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are life-threatening conditions characterized by extensive detachment of the epidermis and mucous membranes. These severe disorders carry a high mortality rate, and their pathogenesis remains largely unclear. Furthermore, optimal therapeutic strategies for SJS/TEN remain a subject of ongoing debate. Early diagnosis of SJS/TEN is challenging, and reliable biomarkers for diagnosis or severity prediction have not been firmly established. Certain drugs, such as carbamazepine and allopurinol, have shown a strong association with specific human leukocyte antigen (HLA) types. Recently, the potential benefits of HLA screening prior to administering these drugs to reduce the incidence of SJS/TEN have been explored. Epidermal cell death in SJS/TEN lesions is caused by extensive apoptosis, primarily through the Fas–Fas ligand (FasL) and perforin/granzyme pathways. Our findings suggest that necroptosis, a form of programmed necrosis, also contributes to epidermal cell death. Annexin A1, released from monocytes, interacts with the formyl peptide receptor 1 to induce necroptosis. Several biomarkers, such as CC chemokine ligand (CCL)-27, interleukin-15, galectin-7, receptor-interacting protein kinases 3 (RIP3), and lipocalin-2, have been identified for diagnostic and prognostic purposes in SJS/TEN. Supportive care is recommended for treating SJS/TEN, but the efficacy of various therapeutic options–including systemic corticosteroids, intravenous immunoglobulin, cyclosporine, and tumor necrosis factor-α antagonists–remains controversial. Recent studies have investigated the potential benefits of tumor necrosis factor-α antagonists. In this review, we discuss recent advances in the understanding and management of SJS/TEN.

Keywords: Stevens-Johnson syndrome, Toxic epidermal necrolysis, Necroptosis, Apoptosis

Introduction

Stevens–Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are severe, life-threatening mucocutaneous reactions characterized by blisters and extensive skin detachment. Drugs and infections, such as those caused by Mycoplasma or the herpes simplex virus, are the primary triggers.[1]

SJS and TEN are considered part of a disease spectrum differentiated by the extent of skin detachment (SJS: skin detachment area <10%, TEN: >30%, SJS/TEN overlap: 10–30%).[2] Despite the low incidence rates reported in Japan (2.5 cases per million for SJS and 1.0 case per million for TEN), these conditions have high mortality rates (SJS: 4.1%; TEN: 29.9%).[3] Moreover, survivors often suffer from severe sequelae, such as blindness.[1] Therefore, prompt and accurate diagnosis, followed by appropriate treatment, is crucial. However, early diagnosis and severity prediction biomarkers are not yet established, and there is a lack of robust evidence for optimal management strategies for SJS/TEN.

In this review, we describe the clinical features and recent research on the pathomechanisms, diagnostic biomarkers, and management of SJS/TEN.

Clinical Features and Scoring System for Mortality Prediction

The cutaneous manifestations of SJS/TEN include painful erythematous rash, bullae, and erosions predominantly on the face and trunk, spreading to the extremities [Figure 1A, B]. Early lesions appear as round, non-palpable areas with indistinct borders, termed “atypical targets” [Figure 1C]. Skin lesions typically exhibit a positive Nikolsky sign, indicating skin erosion upon gentle pressure. Symptoms such as malaise, fever, and upper respiratory tract issues often precede the rash by a few days. Nearly all patients with SJS/TEN experience mucosal involvement, particularly in the eyes, mouth, and genitalia [Figure 2A, B].[4] Early ocular involvement varies and may include conjunctival hyperemia, corneal and conjunctival erosion, and pseudomembrane formation. Post-recovery, persistent ocular inflammation can lead to sequelae such as dry eyes, reduced visual acuity, conjunctivitis, corneal erosions, and trichiasis, with severe cases resulting in blindness. Dysuria suggests urethral involvement, and erosions may also occur in the lower respiratory tract and intestines.

Figure 1.

Figure 1

Skin manifestations of SJS/TEN. (A,B) Widespread skin detachment in a TEN patient. (C) Atypical targets. Flat erythematous macules with a darker center. SJS: Stevens–Johnson syndrome; TEN: Toxic epidermal necrolysis.

Figure 2.

Figure 2

Mucosal involvement of SJS/TEN. (A) Affected eye with conjunctivitis. (B) Lip involvement. Severe cheilitis with hemorrhagic crust. SJS: Stevens–Johnson syndrome; TEN: Toxic epidermal necrolysis.

SJS/TEN are primarily drug-induced. Onset typically occurs within 1–2 weeks of drug administration. Infections like Mycoplasma or herpes simplex virus are also significant causes. Commonly implicated drugs include antibiotics, allopurinol, non-steroidal anti-inflammatory drugs (NSAIDs), and antiepileptic drugs.[5] The frequent causative drugs are listed in Table 1.[3,5]

Table 1.

Causative drugs of SJS/TEN.

Drug classification Causative drugs
Antibiotics Penicillins, cephems, fluoroquinolones, sulfamethoxazole and trimethoprim combination
Xanthine oxidase inhibitor Allopurinol
Anti-inflammatory drugs Acetaminophen, loxoprofen, celecoxib, ibuprofen
Antiepileptic CBZ, lamotrigine, phenytoin, valproic acid
Peptic ulcer agent Lansoprazole, omeprazole, esomeprazole
Anti-HIV drug Nevirapine

CBZ: Carbamazepine; HIV: Human immunodeficiency virus; SJS: Stevens–Johnson syndrome; TEN: Toxic epidermal necrolysis.

The severity-of-illness score for toxic epidermal necrolysis (SCORTEN) is widely employed to predict mortality in SJS/TEN patients.[6] SCORTEN should be assessed within the first 24 h of admission and re-evaluated on day 3. This scoring system is based on seven independent risk factors: age, malignancy, tachycardia, percentage of epidermal detachment, serum urea, serum glucose, and bicarbonate levels [Table 2]. The presence of more risk factors correlates with higher mortality rates [Table 3].

Table 2.

Risk factors for SCORTEN.

Risk factors Illustration
Age Age >40 years
Tachycardia Heart rate >120 beats/min
Malignancy Presence of cancer or hematologic malignancy
Epidermal detachment Epidermal detachment area involving body surface area >10%
Serum urea Blood urea nitrogen >28 mg/dL (10 mmol/L)
Serum glucose Blood glucose >252 mg/dL (14 mmol/L)
Bicarbonate Bicarbonate <20 mmol/L

SCORTEN: Severity-of-illness score for toxic epidermal necrolysis.

Table 3.

Mortality rate in SCORTEN.

Number of risk factors Mortality rate (%)
0–1 3.2
2 12.1
3 35.3
4 58.3
≥5 90.0

SCORTEN: Severity-of-illness score for toxic epidermal necrolysis.

Recently, our group proposed a novel scoring system, the Clinical Risk Score for TEN (CRISTEN), which predicts early stage mortality using only clinical information, without requiring laboratory data.[7] CRISTEN includes 10 risk factors such as age, percentage of epidermal detachment, use of antibiotics as causative agents, systemic corticosteroid therapy before onset, and mucosal involvement affecting the ocular, buccal, and genital areas. Underlying conditions such as renal impairment, diabetes, cardiovascular disease, malignant neoplasms, and bacterial infections are also considered [Table 4]. CRISTEN has shown statistically comparable results to previous systems.

Table 4.

Risk factors for CRISTEN.

No. Parameter Detailed definition
1 Age Age over 65 years
2 Epidermal detachment Epidermal detachment of >10% of BSA
3 Malignant neoplasm Active phase
4 Diabetes mellitus Under treatment with medication
5 Renal impairment Chronic kidney disease
6 Bacterial infection Pneumonia, sepsis, and urinary-tract infection
7 Cardiac disease Heart failure, valvular disease, arrhythmias, aortic aneurysms, angina, atrial and ventricular septal defects, and hypertension under treatment
8 Drugs Antibiotics in the culprit drugs
9 Mucosal damage Mucosal damage affecting all three of ocular, buccal, and genital mucosa
10 Systemic corticosteroid therapy before the onset of SJS/TEN Regardless of dose or duration of administration

BSA: Body surface area; CRISTEN: Clinical risk score for toxic epidermal necrolysis; SJS: Stevens–Johnson syndrome; TEN: Toxic epidermal necrolysis.

Determine the Causative Drug

To determine the causative drug, drug rechallenge is the most reliable approach. However, in SJS/TEN cases, it is not allowed due to the risk of life-threatening consequences. In delayed-type hypersensitivity reaction such as SJS/TEN mediated by drug-specific T cells, patch testing is a useful adjunct.[8] It is performed by applying predetermined dilutions of the compound on intact skin for different periods and assessing for erythema, edema, or vesiculation, all indicative of specific T-cell reactivity. The positive rate of patch test depends on the type of cutaneous adverse drug reactions (cADRs).[9] Drug-induced hypersensitivity syndrome (DIHS), maculopapular exanthema, and fixed drug eruption (56.3%, 23.9%, and 20.0%, respectively). The positive rate for SJS/TEN is 6.7%. In addition to the disease type, the positive rate also depends on the drug, with contrast media and antiepileptic drugs having high positive rates (41.1% and 41.0%, respectively).

As an in vitro assay, lymphocyte transformation tests (LTT) is used to confirm the causative drug. The LTT measures the proliferation of T cells in response to the drug. Although the sensitivity and specificity of the LTT are limited (sensitivity: 62%, specificity: 91%),[10] it is useful because LTT is an in vitro assay and not invasive for patients. The sensitivity of the test varied with the causative drugs, clinical phenotypes, and the timing of the test.[11] In SJS/TEN, it tends to obtain positive reactions when the test was performed at the acute stage of the disease.[11] Some drugs, such as vancomycin or NSAIDs, may elicit enhanced proliferation even in non-sensitized individuals. Meanwhile, immunosuppressive drugs or cytotoxic chemo-therapy agents interrupt the reaction of LTT.[12] Of these drugs, LTT may not be performed accurately. Interferon (IFN)-γ enzyme-linked immunoSpot (ELISpot) assay is also used as an in vitro assay to identify causative drugs. IFN-γ ELISpot is a method in which the patient’s PBMCs are stimulated with the causative drug and the number of cells that release IFN is evaluated. IFN-γ ELISpot performed within 30 days of cADR onset revealed high sensitivity and specificity than LTT (sensitivity: 75%, specificity: 95%).[13]

Algorithm of Drug causality in Epidermal Necrolysis (ALDEN), an algorithm for assessment of drug causality in epidermal necrolysis, is used to assess drug causality in SJS/TEN.[14] ALDEN is an algorithm for retrospective assessment of drug causality and is not used during the acute phase. ALDEN is based on six independent parameters, and the total score varies from −12 to +10. The scores attributed to each drug were pooled in categories aimed at reflecting causality: “very probable”: score ≥6; “probable”: score 4–5; “possible”: score 2–3; “unlikely”: score 0–1; and “very unlikely”: score <0 [Table 5]. This algorithm proved to be complex because it needed to assess pharmacokinetics and drug notoriety. However, ALDEN provides a better ability to discriminate between “culprit” and “innocent” drugs.

Table 5.

Details of the ALDEN.

Criterion Values Rules to apply Score
Delay from initial drug component intake to onset of reaction (index day) Suggestive +3 From 5 days to 28 days –3 to 3
Compatible +2 From 29 days to 56 days
Likely +1 From 1 days to 4 days
Unlikely –1 >56 days
Excluded –3 Drug started on or after the index day
In case of previous reaction to the same drug, only changes for;Suggestive: +3: from 1 days to 4 daysLikely: +1: from 5 days to 56 days
Drug present in the body on index day Define 0 Drug continued up to index day or stopped at a time point less than five times the elimination half-life* before the index day –3 to 0
Doubtful –1 Drug stopped at a time point prior to the index day by more than five times the elimination half-life* but liver or kidney function alterations or suspected drug interactions are present
Excluded –3 Drug stopped at a time point prior to the index day by more than five times the elimination half-life,* without liver or kidney function alterations or suspected drug interactions
Prechallenge/rechallenge Positive specific for disease and drug: 4 SJS/TEN after use of same drug –2 to 4
Positive specific for disease or drug: 2 SJS/TEN after use of similar drug or other reaction with the same drug
Positive unspecific: 1 Other reaction after use of similar drug
Not done/unknown: 0 No known previous exposure to this drug
Negative –2 Exposure to this drug without any reaction (before or after reaction)
Dechallenge Neutral 0 Drug stopped (or unknown) –2 to 0
Negative –2 Drug continued without harm
Type of drug (notoriety) Strongly associated 3 Drug of the “high risk” list according to previous case-control studies§ –1 to 3
Associated 2 Drug with definite but lower risk according to previous case-control studies§
Suspected 1 Several previous reports, ambiguous epidemiology results (drug “under surveillance”)
Unknown 0 All other drugs including newly released ones
Not suspected –1 No evidence of association from previous epidemiology study§ with sufficient number of exposed controls
Intermediate score = total of all previous criteria –11 to 10
Other cause Possible –1 Rank all drugs from highest to lowest in intermediate score –1
If at least one has an intermediate score >3, subtract 1 point from the score of each of the other drugs taken by the patient (another cause is more likely)
Final score –12 to 10

<0, Very unlikely; 0–1, unlikely; 2–3, possible; 4–5, probable; >6, very probable. *Drug (or active metabolite) elimination half-life from serum and/or tissues (according to pharmacology textbooks), taking into account kidney function for drugs predominantly cleared by kidney and liver function for those with high hepatic clearance. Suspected interaction was considered when more than five drugs were present in a patient’s body at the same time. Similar drug = same ATC code up to the fourth level (chemical subgroups). §Definitions for “high risk”, “lower risk”, and “no evidence of association”. “High risk” drugs include sulfamethoxazole-trimethoprim, sulfonamide anti-infectives, allopurinol, CBZ, phenytoin, phenobarbital, oxicam-NSAIDs. “Lower risk” drugs include acetic acid NSAIDs, macrolides, quinolones, cephalosporins, tetracyclines, aminopenicillins. Drugs with “no evidence of association” with SJS/TEN include beta blockers, angiotensin-converting-enzyme inhibitors, calcium channel blockers, thiazide diuretics, furosemide, propionic acid NSAIDs, sulfonylurea antidiabetics, insulin. ALDEN: Algorithm of Drug causality in Epidermal Necrolysis; ATC: Anatomical therapeutic chemical; CBZ: Carbamazepine; NSAIDs: Non-steroidal anti-inflammatory drugs; SJS: Stevens–Johnson syndrome; TEN: Toxic epidermal necrolysis.

Genetic Factors

A burgeoning body of evidence suggests a substantial genetic predisposition to cADRs. In 2004, Chung et al[15] elucidated a robust correlation between human leukocyte antigen (HLA)-B*15:02 and carbamazepine (CBZ)-induced SJS/TEN in a Han Chinese cohort. HLA alleles, categorized into class I and class II, play a pivotal role in presenting antigenic peptides to T cells, thereby initiating an immune response. In a study comprising 44 individuals with CBZ-induced SJS/TEN, all participants were found to possess the HLA-B*15:02 allele. Subsequent investigations have substantiated the association between CBZ-induced SJS/TEN and the HLA-B*15:02 allele across diverse Asian populations, including those in China, Thailand, Malaysia, and India.[1627] A recent meta-analysis also revealed that the HLA-B*15:02 significantly increased the risk of CBZ-induced SJS/TEN.[28]

The association of HLA-B*15:02 extends to other aromatic antiepileptic drugs such as phenytoin, lamotrigine, and oxcarbazepine, albeit with a lower incidence compared to CBZ.[18,2933] Notably, no such association has been observed in Japanese, Korean, and European populations,[3440] suggesting ethnic variations in HLA-related susceptibilities to SJS/TEN.

Other than HLA-B*15:02, HLA-B*15:11 and HLA-A*31:01 are associated in CBZ-induced cADRs. In the Japanese and Korean population, HLA-B*15:11 is also associated with CBZ-induced SJS/TEN.[35,38] HLA-A*31:01 has been reported to associate with CBZ-induced cADRs among several populations.[26,38,41,42] In a meta-analysis among several populations, HLA-A*31:01 showed a significant association with CBZ-induced DIHS/drug reaction with eosinophilia and systemic symptoms (DRESS) but not for CBZ-induced SJS/TEN.[43] In addition, genome-wide association studies for CBZ-induced cADRs in the Japanese population revealed that HLA-A*31:01 was not associated with SJS/TEN, but associated with DIHS/DRESS, maculopapular exanthema, and erythema multiforme.[44] These studies suggested that the type of cADRs might be determined by genetic variant.

In 2008, the U.S. Food and Drug Administration (FDA) recommended genotyping for HLA-B*15:02 prior to the administration of CBZ.[45] The implementation of HLA-B*15:02 screening has markedly reduced the incidence of CBZ-induced SJS/TEN.[46] Similarly in Japan, the effectiveness of HLA-A*31:01 screening for CBZ-induced SJS/TEN was assessed. The incidence of CBZ-induced cADR was significantly reduced when patients who tested negative for HLA-A*31:01 were prescribed CBZ and those who tested positive for HLA-A*31:01 were prescribed an alternative drug.[47]

Other pharmaceuticals, such as allopurinol and abacavir, also exhibit significant associations with specific HLA alleles. Allopurinol, a primary etiological agent for SJS/TEN, has been linked to HLA-B*58:01 across multiple ethnic groups, including Chinese, Japanese, Korean, Thai, and European populations.[35,36,39,4855] Genotyping for HLA-B*58:01 has proven effective in preventing allopurinol-induced SJS/TEN. Cost-effectiveness analyses suggest that HLA-B*58:01 screening is economically advantageous.[56] In the study from the United States suggested that this screening is cost-effective for Asian and African American populations but not for Caucasians or Hispanics.[57]

Abacavir, used in the treatment of human immunodeficiency virus (HIV) infection, is associated with SJS/TEN in patients carrying HLA-B*57:01.[5862] Since 2008, HLA-B*57:01 screening was recommended in clinical care guidelines to reduce the risk of hypersensitivity reaction from abacavir.[63] It has significantly reduced the incidence of abacavir-induced SJS/TEN.[64] However, many patients have yet to undergo this screening, and expanding its use is anticipated to further decrease the incidence of abacavir-induced SJS/TEN. Regarding anti-HIV drugs, nevirapine has also been shown to have HLA association. In Malawian cohort, HLA-C*04:01 was found to markedly increase the risk for nevirapine-induced SJS/TEN.[65]

It has been shown that HLA is associated not only with the onset of cADR but also with the occurrence of complications. In Thai population, HLA-B*44:03 was revealed to strongly associate with acetaminophen-related SJS/TEN patients who developed severe ocular complications.[66] In addition, HLA-A*02:06 and HLA-B*13:01, HLA-C*14:03 were associated with severe ocular complications in acetaminophen-related SJS/TEN in Japanese patients.[67,68]

Cytochrome P450 (CYP) enzymes, integral to drug metabolism, also play a crucial role in cADRs. The CYP450 gene family comprises 57 isoforms, each exhibiting distinct functional characteristics. Patients with polymorphisms leading to slow drug metabolism face an elevated risk of adverse drug reactions.[69] Chung et al[70] identified significant genetic factors associated with phenytoin-induced SJS/TEN, including 16 notable single nucleotide polymorphisms (SNPs) in CYP2C9. Patients with phenytoin-induced SJS/TEN carrying the CYP2C9*3 allele exhibit delayed phenytoin clearance, thereby exacerbating disease severity.

Pathogenesis and Diagnostic Biomarkers

Immunopathogenesis

SJS/TEN are traditionally regarded as T-cell-mediated disorders. T cells are activated through the binding of drugs to T cell receptors (TCRs) presented by antigen-presenting cells (APCs). Currently, three primary hypotheses elucidate the mechanisms of T cell activation [Figure 3]:[71] hapten/pro-hapten model, pharmacological interaction (p-i) concept, and altered peptide model. Most drugs and their metabolites function as pro-haptens and do not act as haptens independently. They gain immunogenicity by covalently binding to carrier proteins (hapten antigens). These hapten antigens form complexes with HLA molecules in APCs and are recognized by TCRs, triggering drug-specific T cell activation. In this model, antigenic drugs are covalently attached to peptides presented by HLA molecules to TCRs.[7275] However, several drugs, including lamotrigine, celecoxib, CBZ, and sulfamethoxazole, can non-covalently bind directly to HLA and/or TCRs, a mechanism known as the p-i concept.[7680] Generally, HLA polymorphisms depend on the structure of antigen-binding cleft. It has been documented that unmodified abacavir binds to HLA-B*57:01 antigen-binding cleft, altering its shape and chemistry and thereby changing the repertoire of endogenous peptides that can bind to HLA-B*57:01 (altered peptide model).[81,82] The TCR profile is also crucial in the pathogenesis of SJS/TEN. Ko et al[83] reported that 84% of patients with CBZ-associated SJS/TEN had VB-11-ISGSY clonotype, but the clonotype absent in CBZ-tolerant patients.[84] This clonotype specificity has also reported in oxypurinol-induced SJS/TEN.[85] Recently, Pan et al[86] employed next-generation sequencing to explore the TCR repertoire and identified a public αβTCR from the cytotoxic T cells of patients with CBZ-induced SJS/TEN, capable of binding CBZ and mediating an immune response.

Figure 3.

Figure 3

Models of T cell activation in SJS/TEN. (A) Hapten/pro-hapten model. Drugs or drug metabolites form a complex with carrier proteins and are presented as HLA. (B) p-i concept. Drugs directly bind to HLA and TCR non-covalently. (C) Altered peptide model. Drugs bind to the pocket of HLA resulting alteration of HLA-binding peptide repertoire. APC: Antigen-presenting cells; HLA: Human leukocyte antigen; SJS: Stevens–Johnson syndrome; TCR: T cell receptor; TEN: Toxic epidermal necrolysis.

In the initial stages, cytotoxic CD8+ T cells predominantly infiltrate epidermis and blister fluid, while CD4+ T cells primarily infiltrate in the dermis.[87,88] Monocytes are also present in the epidermis of TEN patients, and during the later stages, a decrease in lymphocytes alongside an increase in monocytes is observed. Tohyama et al[89] demonstrated that monocytes play a crucial role in keratinocyte death, likely by activating the cytotoxicity of CD8+ T cells. Elevated levels of soluble IL-2 receptors have been observed in the blister fluid and serum of SJS/TEN patients.[90] Soluble IL-2 receptors serve as markers of T cell activation, underscoring the significance of activated cytotoxic CD8+ T cells in the keratinocyte death of SJS/TEN.

Keratinocyte death

The epidermal damage observed in the skin lesions of SJS/TEN patients is attributed to apoptotic mechanisms.[91] Apoptosis in SJS/TEN is mediated by cytotoxic CD8+ T cells via the Fas-Fas ligand (FasL) pathway, the perforin/granzyme pathway, or through granulysin [Figure 4].

Figure 4.

Figure 4

Apoptosis pathway in SJS/TEN. In SJS/TEN, cytotoxic T cell induces apoptosis through the FasL pathway, perforin/granzyme pathway, or granulysin. FasL: Fas ligand; SJS: Stevens–Johnson syndrome; TEN: Toxic epidermal necrolysis.

FasL produced by cytotoxic CD8+ T cells and natural killer (NK) cells binds to Fas receptors on target cells. This binding activates the caspase cascade, culminating in cell apoptosis.[92] Under normal conditions, Fas is present on the cell surface, while FasL is expressed intracellularly. FasL is translocated to the cell surface when the cell is primed for self-destruction.[93] Viard et al[94] demonstrated that FasL is present on the cell surface of keratinocytes in TEN patients, a feature not observed in keratinocytes from patients with maculopapular drug reactions. Moreover, elevated levels of soluble FasL (sFasL) were detected in the serum of TEN patients, and sFasL also has the capacity to mediate apoptosis.[95]

We demonstrated that serum FasL levels are elevated in TEN patients.[96,97] In this study, it is shown that peripheral blood mononuclear cells (PBMCs) produce sFasL upon exposure to causative drugs. sFasL released from PBMCs binds to Fas expressed on keratinocytes, inducing apoptosis. This finding suggests that increased serum sFasL levels may serve as a valuable diagnostic marker for SJS/TEN. However, a definitive correlation between sFasL levels and disease severity has not yet been established.[97,98]

Nassif et al[99,100] highlighted the significance of the perforin/granzyme pathway. Upon recognizing a target cell, the cytotoxic CD8+ T cells release perforin and granzyme B.[92] Their study reported that mononuclear cells present in the blister fluid of SJS/TEN patients reveled cytotoxic effects when exposed to a causative drug. This cytotoxicity is inhibited by a perforin/granzyme pathway inhibitor, suggesting that this pathway is responsible for the epidermal damage seen in the skin lesions of SJS/TEN.[99,100]

In 2008, Chung et al[101] revealed the apoptosis-inducible effect of granulysin in keratinocyte of SJS/TEN. Granulysin, a pro-apoptotic protein, facilitates cell-mediated cytotoxicity without direct cell-to-cell interaction. Elevated levels of granulysin are detected in SJS/TEN blister fluid. Granulysin is released from blister cells in skin lesions of SJS/TEN, including NK cells and cytotoxic CD8+ T cells. The severity of the skin symptoms correlates with serum granulysin levels. Our research also identified that granulysin could be an early diagnostic marker for SJS/TEN.[102] However, serum granulysin is also increased in patients with DIHS/drug reactions with eosinophilia and systemic symptoms, another category of severe cADRs.[103] Therefore, granulysin is not a specific biomarker for SJS/TEN.

In recent years, the role of exosomes has been attracting attention. Exosomes are small vesicles with a diameter of 50–150 nm containing DNA, miRNA, mRNA, and proteins. Exosomes are secreted by cells and play an important role in cell-to-cell communication including immune response, signal transduction, and antigen presentation. Exosomes are involved not only in the maintenance of homeostasis but also in the pathogenesis of various diseases such as cardiovascular diseases, neurodegenerative diseases, and malignancies.[104] Zhang et al[105] revealed that the expression of miRNA, miR-375-3p, was markedly elevated in the plasma exosomes of SJS/TEN patients, which promotes epidermal apoptosis induction by downregulating the expression of the X-linked inhibitor of apoptosis (XIAP).

In 2014, our research elucidated that necroptosis, induced by the interaction between annexin A1 and formyl peptide receptor 1 (FPR1), associates with keratinocyte death in SJS/TEN.[106] Necroptosis represents a form of programmed cell death that manifests with morphological characteristics akin to necrosis. Unlike apoptosis, necroptotic cells release damage-associated molecular patterns (DAMPs), which include various pro-inflammatory cytokines, thereby inciting inflammation. In contrast, apoptotic cells are rapidly phagocytosed by macrophages and subsequently degraded within phagolysosomes, a process devoid of inflammatory response.[107] Generally, necroptosis is induced by the stimulation of tumor necrosis factor (TNF)-α when apoptosis is inhibited. Upon TNF-α stimulation, receptor-interacting protein kinases 1 (RIP1) and receptor-interacting protein kinases 3 (RIP3) undergo phosphorylation and assemble into a “necrosome” complex. Additionally, the mixed lineage kinase domain-like (MLKL) pseudokinase is recruited to the necrosome and phosphorylated by RIP3. The phosphorylated MLKL (pMLKL) then translocates to the plasma membrane, instigating cell death.[107] Supernatant from PBMCs exposed to the causative drug in SJS/TEN patients has been observed to induce cytotoxicity in SJS/TEN keratinocytes. This cytotoxic effect is inhibited by a specific inhibitor of RIP1, necrostatin-1. Keratinocyte in SJS/TEN skin lesions exhibits significant expression of FPR1. Lipocalin-2 released from CD8+ T cells initiates the formation of neutrophil extracellular traps (NETs). Neutrophils undergoing NETosis release LL-37, which induces FPR1 expression in keratinocytes.[108] The interaction of annexin A1, secreted by monocytes, with FPR1 facilitates necrosome formation [Figure 5]. Inhibition of necroptosis effectively prevents SJS/TEN-like responses in a murine model of SJS/TEN.[106,109] These findings suggest that necroptosis plays an essential role in the pathogenesis of SJS/TEN.

Figure 5.

Figure 5

Necroptosis pathway in SJS/TEN. Drug-stimulated monocytes secrete annexin A1. Annexin A1 binds to FPR1 and RIP1 and RIP3 form necrosome and MLKL is phosphorylated by RIP3. pMLKL is located on plasma membrane and induces cell death. FPR1: Formyl peptide receptor 1; MLKL: Mixed lineage kinase domain-like; pMLKL: Phosphorylated MLKL; RIP1: Receptor-interacting protein kinases 1; RIP3: Receptor-interacting protein kinases 3; SJS: Stevens–Johnson syndrome; TEN: Toxic epidermal necrolysis.

TNF-α also plays an important role in keratinocyte death in SJS/TEN. When TNF-α binds to TNF receptor 1, apoptosis is induced if caspase 8 is activated, while necroptosis is induced if caspase 8 is inactivated.[107] CTLs release TNF-α in addition to granulysin and sFasL. Activated keratinocytes and macrophages also release TNF-α. In fact, high concentrations of TNF-α are found in the serum and blister fluid of SJS/TEN patients.[110,111] TNF-α is involved not only in this direct induction of cell death but also indirectly. TNF-α upregulates granulysin promoter activity and increases granulysin mRNA expression.[112] In SJS/TEN patients, TNF-α and IFN-γ also upregulate FasL expression and induce keratinocyte apoptosis via upregulation of iNOS.[113]

Keratinocyte death in SJS/TEN is a mixture of apoptosis and necroptosis, and it is of interest because it has not been clarified which form of cell death is the key phenomenon in the pathogenesis of SJS/TEN, and how the balance between apoptosis and necroptosis is regulated. In addition, recently, other types of cell death including pyroptosis and ferroptosis have been investigated. It has not been analyzed whether these cell deaths are involved in the pathogenesis of SJS/TEN.

Future research is expected to elucidate the pathogenesis of cell death in SJS/TEN, which may lead to the development of new therapeutic agents.

Diagnostic biomarkers

Although SJS/TEN are severe diseases, the clinical manifestations in early stage are often indistinguishable from those of erythema multiforme and maculopapular exanthema. Although no reliable biomarkers for diagnosis or severity prediction have been established, several biomarkers have been identified promising diagnostic or prognostic biomarkers for SJS/TEN, which are currently in the research phase and have not yet been applied clinically.

Wang et al[114,115] revealed serum CCL-27 levels are increased in SJS/TEN patients, correlating with disease activity. CCL-27 is implicated in cutaneous inflammatory diseases by regulating T cell trafficking to the skin.

In addition, CCL-27 was highly expressed in the skin lesions of SJS/TEN patients.[116] Therefore, it is hypothesized that CCL-27 is produced by keratinocytes in the SJS/TEN skin lesion and released into the circulation.[117]

Su et al[118] identified interleukin-15 (IL-15) as a factor related to mortality and severity of SJS/TEN patients. They further revealed that IL-15 exacerbates TEN severity by enhancing T cell- and NK-cell-mediated responses. IL-15 induces the production of TNF-α and downstream cytokines/chemokines,[119] suggesting that the elevation of numerous cytokines and chemokines in SJS/TEN might be secondary to IL-15 activity.

Through proteomics analysis, we identified galectin-7 as a diagnostic biomarker for SJS/TEN.[120] We hypothesized that certain soluble factors might be secreted exclusively by drug-specific lymphocytes in SJS/TEN patients, differentiating them from those with non-severe cADRs. These soluble factors could serve as biomarkers for SJS/TEN. PBMCs from SJS/TEN patients were cultured with the causative drugs, and the supernatant was collected for proteomic analysis of the elevated proteins.[121] Hama et al[120] concluded that this approach facilitated the identification of novel SJS/TEN-specific biomarkers not associated with the pathogenesis.

Focusing on the mechanisms of epidermal necroptosis, we identified serum RIP3, a key mediator of necroptosis, as a diagnostic and severity marker.[122] It has been reported that RIP3 expression increases in necroptotic cells.[123] We demonstrated that RIP3 expression also increases in necroptotic keratinocytes, and serum RIP3 levels are elevated in the acute phase of SJS/TEN patients, potentially correlating with disease activity.

Lipocalin-2 is also a biomarker in relation to the mechanism of necroptosis.[108] Lipocalin-2 is associated with early stage of necroptosis in SJS/TEN. It is released from CD8+ T cells and induces NETs formation, resulting in FPR1 expression. The serum levels of lipocalin-2 were elevated in early stage of SJS/TEN, but not in other types of cADRs.

Management

In patients with SJS/TEN, the epidermis and mucosal membranes are mainly affected. However, SJS/TEN can also induce various organ complications, such as the kidneys, liver, and respiratory tract. Consequently, a multidisciplinary evaluation and prompt intervention within a specialized hospital setting including intensive care unit or burn care unit are pivotal in enhancing survival outcomes. Initially, immediate cessation of the suspected causative drugs is imperative for SJS/TEN. Furthermore, supportive care, encompassing supplemental oxygen, nutritional assessment, fluid replacement, pain management, and supplemental oxygen is essential. Given that infection resulting from skin detachment is a common complication in SJS/TEN patients, which impairs re-epithelialization and has potential progression to sepsis, meticulous daily skin care is necessary. The blister fluid should be aspirated, and the detached skin retained to cover the dermis as a biologic dressing. However, it is necessary to remove necrotic epidermis. In the areas of skin erosion, non-adherent dressings should be applied. There is no evidence to suggest which dressing is superior, but antibiotic ointments and petrolatum are commonly used.[124] Antibiotic agent should be administrated if cutaneous infection is clinically suspected.[125]

The optimal therapeutic strategy for SJS/TEN remains a subject of debate.[126] In 2016, UK guidelines emphasized the prioritization of culprit drug withdrawal and comprehensive supportive care over systemic treatment due to the lack of evidence supporting the benefits.[127] However, some studies have reported benefits with the treatment of systemic corticosteroids, intravenous immunoglobulins (IVIGs), cyclosporine, TNF-α antagonists (infliximab and etanercept), and plasmapheresis (PP).[124,128]

The effectiveness of systemic therapy may be contingent upon the disease phase. For instance, during the acute phase, immunosuppressive treatments may be appropriate due to the occurrence of a robust inflammatory response resembling a “cytokine storm”. Conversely, during the peak period characterized by extensive skin detachment, strong immunosuppressive treatment might impede re-epithelialization and elevate the risk of infection. Previous studies have not adequately considered this aspect, often including results from all phases, thereby leading to inconsistent conclusions. We will introduce each treatment modality in detail below.

Systemic corticosteroids

Previous studies have demonstrated that corticosteroid treatment in SJS/TEN patients is associated with an increased risk of infection and overall complications, including higher mortality rates.[129131] Comprehensive analyses and systematic reviews have not shown a survival benefit with the use of systemic corticosteroids.[132134]

However, recent investigations suggested a beneficial effect of corticosteroid therapy. A European multicenter retrospective study, along with recent meta-analysis of observational studies, indicated positive effects of corticosteroids.[128,135] A recent survey in Japan showed that the ratio of expected to observed mortality, calculated using SCORTEN score, was lowest with high-dose of corticosteroid therapy, followed by steroid pulse therapy.[3] Given that cutaneous infection is a critical concern in the administration of corticosteroids for SJS/TEN patients, short-term corticosteroid use, enhanced infection control, and meticulous wound management are essential to reduce mortality rates.

Steroid pulse therapy is used in patients with severe cases of SJS/TEN that progress rapidly.[136] An observational study revealed that the early administration of steroids pulse therapy reduced mortality without increasing infection risk.[137] Notably, it is reported steroid pulse therapy at disease onset could prevent ocular complications.[138]

IVIG

IVIG has been widely administrated to patients with SJS/TEN. Although the precise mechanisms of IVIG therapy remain unclear, it has been suggested that IVIG inhibits apoptosis by blocking Fas receptor with anti-Fas antibodies.[139] Although some case reports have concluded that IVIG does not confer a beneficial effect in reducing mortality,[140142] other reports have shown the beneficial effects of IVIG for SJS/TEN patients.[143147] The largest retrospective study, the European Study of Severe Cutaneous Adverse Reactions (EuroSCAR), found that IVIG did not improve mortality compared with supportive care alone.[135] However, recent meta-analyses have shown that high-dose IVIG (<2 g/kg) has a beneficial effect in decreasing the mortality of SJS/TEN.[148] Recently, there is increasing evidence that combination of IVIG and corticosteroid therapy, rather than IVIG monotherapy, improves the prognosis of SJS/TEN.[148,149] A retrospective study from China revealed combination therapy was prone to bring a better prognosis for SJS/TEN patients compared with those treated by corticosteroids alone. In addition, the incidence of infections was significantly lower in the IVIG and steroid combined therapy group compared to patients treated with corticosteroids alone.[150]

Cyclosporine

Cyclosporine, a calcineurin inhibitor, has demonstrated therapeutic efficacy in the treatment of SJS/TEN. This immunosuppressive agent modulates T-lymphocyte-mediated cytotoxicity and inhibits key molecules such as FasL, nuclear factor-kB, and TNF-α.[151] Several case reports and meta-analyses have indicated that cyclosporine treatment enhances survival rates in patients with SJS/TEN.[152159] It is reported that cyclosporine not only improves mortality outcomes but also exhibits minimal adverse effects in the treatment of SJS/TEN.[160] These findings suggest a promising role for cyclosporine in managing SJS/TEN. Nevertheless, the limited number of patients included in these studies necessitates further research to validate cyclosporine’s efficacy.

Plasmapheresis

Several case series have demonstrated the efficacy of PP in treating SJS/TEN.[161166] The primary objective of PP is to eliminate pathogenic factors, including drugs, drug metabolites, and disease-induced cytokines/chemokines, from the patient’s bloodstream. PP sessions are typically conducted every other day or daily. This procedure is generally considered safe, with few adverse side effects. Despite one observational study concluding that PP is ineffective, the overall survival rate in this study was 87.5%.[167] Narita et al[168] found that PP was effective in TEN patients who did not respond to supportive or systemic corticosteroid therapies and observed a reduction in serum cytokine levels following PP.

A study suggested that combining PP with IVIG therapy may provide additional benefits.[169] However, another study reported unfavorable results for the combined therapy, while treatment with PP alone yielded positive outcomes.[170] Consequently, randomized controlled trials are essential to further elucidate the efficacy of PP in SJS/TEN treatment.

Tumor necrosis factor inhibitors

As previously described, TNF-α is associated with keratinocyte death in SJS/TEN. TNF-α inhibitors have been used and, in some cases, beneficial effects have been suggested. In 2002, Fischer et al[171] reported a TEN case recovered with a single dose of infliximab. Since then, many cases have been reported in which infliximab was effective in SJS/TEN,[172174] and some reports suggest that infliximab in combination with IVIG may be useful.[175,176] Infliximab has been reported mainly in case reports, and no randomized controlled trials have been conducted. Etanercept is also expected to be a promising treatment. Etanercept improved clinical outcomes and mortality in patients with SJS/TEN in a randomized trial comparing the effects of etanercept and corticosteroids.[177] Although there was no statistical significance, the actual mortality rate in the etanercept group was lower (8.3%) than that predicted by SCORTEN (17.7%). A recent multicenter observational study showed the efficacy of combination therapy with etanercept and systemic corticosteroids.[178] Patients who received combination therapy with etanercept and corticosteroids had lower mortality than those with corticosteroid monotherapy and those with IVIG combined with corticosteroids.

Conclusions

This review encapsulates the recent advancements in understanding the pathophysiology, diagnosis, and treatment of SJS/TEN. SJS/TEN is a formidable disease with a high mortality rate, and its diagnostic methods and therapeutic algorithms have not been fully established. However, recent studies have increasingly focused on therapeutic approaches. In particular, TNF-α inhibitors are expected to be a promising treatment. In addition, research on the pathogenesis of SJS/TEN is gradually progressing. As the pathology is elucidated, it is hoped that a novel therapeutic target will be developed.

Funding

This work was supported by Grant-in-Aid for Scientific Research (KAKENHI) (No. 23K15264).

Conflicts of interest

None.

Footnotes

How to cite this article: Hasegawa A, Abe R. Stevens–Johnson syndrome and toxic epidermal necrolysis: Updates in pathophysiology and management. Chin Med J 2024;137:2294–2307. doi: 10.1097/CM9.0000000000003250

References

  • 1.Hasegawa A, Abe R. Recent advances in managing and understanding Stevens-Johnson syndrome and toxic epidermal necrolysis. F1000Res 2020;9:F1000FacultyRev–612. doi: 10.12688/f1000research.24748.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bastuji-Garin S, Rzany B, Stern RS, Shear NH, Naldi L, Roujeau JC. Clinical classification of cases of toxic epidermal necrolysis, Stevens-Johnson syndrome, and erythema multiforme. Arch Dermatol 1993;129:92–96. doi: 10.1001/archderm.1993.01680220104023. [PubMed] [Google Scholar]
  • 3.Sunaga Y Kurosawa M Ochiai H Watanabe H Sueki H Azukizawa H, et al. The nationwide epidemiological survey of Stevens-Johnson syndrome and toxic epidermal necrolysis in Japan, 2016-2018. J Dermatol Sci 2020;100:175–182. doi: 10.1016/j.jdermsci.2020.09.009. [DOI] [PubMed] [Google Scholar]
  • 4.Revuz J Penso D Roujeau JC Guillaume JC Payne CR Wechsler J, et al. Toxic epidermal necrolysis. Clinical findings and prognosis factors in 87 patients. Arch Dermatol 1987;123:1160–1165. doi: 10.1001/archderm.123.9.1160. [DOI] [PubMed] [Google Scholar]
  • 5.Mockenhaupt M Viboud C Dunant A Naldi L Halevy S Bouwes Bavinck JN, et al. Stevens-Johnson syndrome and toxic epidermal necrolysis: Assessment of medication risks with emphasis on recently marketed drugs. The EuroSCAR-study. J Invest Dermatol 2008;128:35–44. doi: 10.1038/sj.jid.5701033. [DOI] [PubMed] [Google Scholar]
  • 6.Bastuji-Garin S, Fouchard N, Bertocchi M, Roujeau JC, Revuz J, Wolkenstein P. SCORTEN: A severity-of-illness score for toxic epidermal necrolysis. J Invest Dermatol 2000;115:149–153. doi: 10.1046/j.1523-1747.2000.00061.x. [DOI] [PubMed] [Google Scholar]
  • 7.Hama N Sunaga Y Ochiai H Kokaze A Watanabe H Kurosawa M, et al. Development and validation of a novel score to predict mortality in Stevens-Johnson syndrome and toxic epidermal necrolysis: CRISTEN. J Allergy Clin Immunol Pract 2023;11:3161–3168.e2. doi: 10.1016/j.jaip.2023.07.001. [DOI] [PubMed] [Google Scholar]
  • 8.Thaiwat S, Rojanapanthu P. Cutaneous adverse drug eruption: The role of drug patch testing. Int J Dermatol 2023;62:108–114. doi: 10.1111/ijd.16398. [DOI] [PubMed] [Google Scholar]
  • 9.Ohtoshi S, Kitami Y, Sueki H, Nakada T. Utility of patch testing for patients with drug eruption. Clin Exp Dermatol 2014;39:279–283. doi: 10.1111/ced.12239. [DOI] [PubMed] [Google Scholar]
  • 10.Glässner A, Dubrall D, Weinhold L, Schmid M, Sachs B. Lymphocyte transformation test for drug allergy detection: When does it work? Ann Allergy Asthma Immunol 2022;129:497–506.e3. doi: 10.1016/j.anai.2022.06.014. [DOI] [PubMed] [Google Scholar]
  • 11.Kano Y, Hirahara K, Mitsuyama Y, Takahashi R, Shiohara T. Utility of the lymphocyte transformation test in the diagnosis of drug sensitivity: Dependence on its timing and the type of drug eruption. Allergy 2007;62:1439–1444. doi: 10.1111/j.1398-9995.2007.01553.x. [DOI] [PubMed] [Google Scholar]
  • 12.Pichler WJ, Tilch J. The lymphocyte transformation test in the diagnosis of drug hypersensitivity. Allergy 2004;59:809–820. doi: 10.1111/j.1398-9995.2004.00547.x. [DOI] [PubMed] [Google Scholar]
  • 13.Porebski G, Piotrowicz-Wojcik K, Spiewak R. ELISpot assay as a diagnostic tool in drug hypersensitivity reactions. J Immunol Methods 2021;495:113062. doi: 10.1016/j.jim.2021.113062. [DOI] [PubMed] [Google Scholar]
  • 14.Sassolas B Haddad C Mockenhaupt M Dunant A Liss Y Bork K, et al. ALDEN, an algorithm for assessment of drug causality in Stevens-Johnson syndrome and toxic epidermal necrolysis: Comparison with case-control analysis. Clin Pharmacol Ther 2010;88:60–68. doi: 10.1038/clpt.2009.252. [DOI] [PubMed] [Google Scholar]
  • 15.Chung WH Hung SI Hong HS Hsih MS Yang LC Ho HC, et al. Medical genetics: A marker for Stevens-Johnson syndrome. Nature 2004;428:486. doi: 10.1038/428486a. [DOI] [PubMed] [Google Scholar]
  • 16.Hung SI Chung WH Jee SH Chen WC Chang YT Lee WR, et al. Genetic susceptibility to carbamazepine-induced cutaneous adverse drug reactions. Pharmacogenet Genomics 2006;16:297–306. doi: 10.1097/01.fpc.0000199500.46842.4a. [DOI] [PubMed] [Google Scholar]
  • 17.Man CB Kwan P Baum L Yu E Lau KM Cheng AS, et al. Association between HLA-B*1502 allele and antiepileptic drug-induced cutaneous reactions in Han Chinese. Epilepsia 2007;48:1015–1018. doi: 10.1111/j.1528-1167.2007.01022.x. [DOI] [PubMed] [Google Scholar]
  • 18.Locharernkul C Loplumlert J Limotai C Korkij W Desudchit T Tongkobpetch S, et al. Carbamazepine and phenytoin induced Stevens-Johnson syndrome is associated with HLA-B*1502 allele in Thai population. Epilepsia 2008;49:2087–2091. doi: 10.1111/j.1528-1167.2008.01719.x. [DOI] [PubMed] [Google Scholar]
  • 19.Mehta TY Prajapati LM Mittal B Joshi CG Sheth JJ Patel DB, et al. Association of HLA-B*1502 allele and carbamazepine-induced Stevens-Johnson syndrome among Indians. Indian J Dermatol Venereol Leprol 2009;75:579–582. doi: 10.4103/0378-6323.57718. [DOI] [PubMed] [Google Scholar]
  • 20.Tassaneeyakul W Tiamkao S Jantararoungtong T Chen P Lin SY Chen WH, et al. Association between HLA-B*1502 and carbamazepine-induced severe cutaneous adverse drug reactions in a Thai population. Epilepsia 2010;51:926–930. doi: 10.1111/j.1528-1167.2010.02533.x. [DOI] [PubMed] [Google Scholar]
  • 21.Wu XT Hu FY An DM Yan B Jiang X Kwan P, et al. Association between carbamazepine-induced cutaneous adverse drug reactions and the HLA-B*1502 allele among patients in central China. Epilepsy Behav 2010;19:405–408. doi: 10.1016/j.yebeh.2010.08.007. [DOI] [PubMed] [Google Scholar]
  • 22.Chang CC, Too CL, Murad S, Hussein SH. Association of HLA-B*1502 allele with carbamazepine-induced toxic epidermal necrolysis and Stevens-Johnson syndrome in the multi-ethnic Malaysian population. Int J Dermatol 2011;50:221–224. doi: 10.1111/j.1365-4632.2010.04745.x. [DOI] [PubMed] [Google Scholar]
  • 23.Then SM, Rani ZZ, Raymond AA, Ratnaningrum S, Jamal R. Frequency of the HLA-B*1502 allele contributing to carbamazepine-induced hypersensitivity reactions in a cohort of Malaysian epilepsy patients. Asian Pac J Allergy Immunol 2011;29:290–293. [PubMed] [Google Scholar]
  • 24.Wang Q Zhou JQ Zhou LM Chen ZY Fang ZY Chen SD, et al. Association between HLA-B*1502 allele and carbamazepine-induced severe cutaneous adverse reactions in Han people of southern China mainland. Seizure 2011;20:446–448. doi: 10.1016/j.seizure.2011.02.003. [DOI] [PubMed] [Google Scholar]
  • 25.Zhang Y Wang J Zhao LM Peng W Shen GQ Xue L, et al. Strong association between HLA-B*1502 and carbamazepine-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in mainland Han Chinese patients. Eur J Clin Pharmacol 2011;67:885–887. doi: 10.1007/s00228-011-1009-4. [DOI] [PubMed] [Google Scholar]
  • 26.Khor AH Lim KS Tan CT Kwan Z Tan WC Wu DB, et al. HLA-A*31: 01 and HLA-B*15:02 association with Stevens-Johnson syndrome and toxic epidermal necrolysis to carbamazepine in a multiethnic Malaysian population. Pharmacogenet Genomics 2017;27:275–278. doi: 10.1097/fpc.0000000000000287. [DOI] [PubMed] [Google Scholar]
  • 27.Ou GJ Wang J Ji X Yu H Jiang L Li L, et al. A study of HLA-B*15:02 in 9 different Chinese ethnics: Implications for carbamazepine related SJS/TEN. HLA 2017;89:225–229. doi: 10.1111/tan.12970. [DOI] [PubMed] [Google Scholar]
  • 28.Biswas M, Ershadian M, Shobana J, Nguyen AH, Sukasem C. Associations of HLA genetic variants with carbamazepine-induced cutaneous adverse drug reactions: An updated meta-analysis. Clin Transl Sci 2022;15:1887–1905. doi: 10.1111/cts.13291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hung SI Chung WH Liu ZS Chen CH Hsih MS Hui RC, et al. Common risk allele in aromatic antiepileptic-drug induced Stevens-Johnson syndrome and toxic epidermal necrolysis in Han Chinese. Pharmacogenomics 2010;11:349–356. doi: 10.2217/pgs.09.162. [DOI] [PubMed] [Google Scholar]
  • 30.Cheung YK, Cheng SH, Chan EJ, Lo SV, Ng MH, Kwan P. HLA-B alleles associated with severe cutaneous reactions to antiepileptic drugs in Han Chinese. Epilepsia 2013;54:1307–1314. doi: 10.1111/epi.12217. [DOI] [PubMed] [Google Scholar]
  • 31.Chang CC Ng CC Too CL Choon SE Lee CK Chung WH, et al. Association of HLA-B*15:13 and HLA-B*15:02 with phenytoin-induced severe cutaneous adverse reactions in a Malay population. Pharmacogenomics J 2017;17:170–173. doi: 10.1038/tpj.2016.10. [DOI] [PubMed] [Google Scholar]
  • 32.Chen CB Hsiao YH Wu T Hsih MS Tassaneeyakul W Jorns TP, et al. Risk and association of HLA with oxcarbazepine-induced cutaneous adverse reactions in Asians. Neurology 2017;88:78–86. doi: 10.1212/wnl.0000000000003453. [DOI] [PubMed] [Google Scholar]
  • 33.Shi YW Min FL Zhou D Qin B Wang J Hu FY, et al. HLA-A*24:02 as a common risk factor for antiepileptic drug-induced cutaneous adverse reactions. Neurology 2017;88:2183–2191. doi: 10.1212/wnl.0000000000004008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Alfirevic A, Jorgensen AL, Williamson PR, Chadwick DW, Park BK, Pirmohamed M. HLA-B locus in Caucasian patients with carbamazepine hypersensitivity. Pharmacogenomics 2006;7:813–818. doi: 10.2217/14622416.7.6.813. [DOI] [PubMed] [Google Scholar]
  • 35.Kaniwa N Saito Y Aihara M Matsunaga K Tohkin M Kurose K, et al. HLA-B locus in Japanese patients with anti-epileptics and allopurinol-related Stevens-Johnson syndrome and toxic epidermal necrolysis. Pharmacogenomics 2008;9:1617–1622. doi: 10.2217/14622416.9.11.1617. [DOI] [PubMed] [Google Scholar]
  • 36.Lonjou C Borot N Sekula P Ledger N Thomas L Halevy S, et al. A European study of HLA-B in Stevens-Johnson syndrome and toxic epidermal necrolysis related to five high-risk drugs. Pharmacogenet Genomics 2008;18:99–107. doi: 10.1097/FPC.0b013e3282f3ef9c. [DOI] [PubMed] [Google Scholar]
  • 37.Kaniwa N Saito Y Aihara M Matsunaga K Tohkin M Kurose K, et al. HLA-B*1511 is a risk factor for carbamazepine-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in Japanese patients. Epilepsia 2010;51:2461–2465. doi: 10.1111/j.1528-1167.2010.02766.x. [DOI] [PubMed] [Google Scholar]
  • 38.Kim SH Lee KW Song WJ Kim SH Jee YK Lee SM, et al. Carbamazepine-induced severe cutaneous adverse reactions and HLA genotypes in Koreans. Epilepsy Res 2011;97:190–197. doi: 10.1016/j.eplepsyres.2011.08.010. [DOI] [PubMed] [Google Scholar]
  • 39.Park HJ Kim YJ Kim DH Kim J Park KH Park JW, et al. HLA allele frequencies in 5802 Koreans: Varied allele types associated with SJS/TEN according to culprit drugs. Yonsei Med J 2016;57:118–126. doi: 10.3349/ymj.2016.57.1.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ramírez E Bellón T Tong HY Borobia AM de Abajo FJ Lerma V, et al. Significant HLA class I type associations with aromatic antiepileptic drug (AED)-induced SJS/TEN are different from those found for the same AED-induced DRESS in the Spanish population. Pharmacol Res 2017;115:168–178. doi: 10.1016/j.phrs.2016.11.027. [DOI] [PubMed] [Google Scholar]
  • 41.McCormack M Alfirevic A Bourgeois S Farrell JJ Kasperavičiūtė D Carrington M, et al. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N Engl J Med 2011;364:1134–1143. doi: 10.1056/NEJMoa1013297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ozeki T Mushiroda T Yowang A Takahashi A Kubo M Shirakata Y, et al. Genome-wide association study identifies HLA-A*3101 allele as a genetic risk factor for carbamazepine-induced cutaneous adverse drug reactions in Japanese population. Hum Mol Genet 2011;20:1034–1041. doi: 10.1093/hmg/ddq537. [DOI] [PubMed] [Google Scholar]
  • 43.Genin E Chen DP Hung SI Sekula P Schumacher M Chang PY, et al. HLA-A*31:01 and different types of carbamazepine-induced severe cutaneous adverse reactions: An international study and meta-analysis. Pharmacogenomics J 2014;14:281–288. doi: 10.1038/tpj.2013.40. [DOI] [PubMed] [Google Scholar]
  • 44.Fukunaga K Tsukagoshi E Kurata M Mizukawa Y Niihara H Morita E, et al. Differential effects of HLA-B*15:11 and HLA-A*31:01 on carbamazepine-induced cutaneous adverse reactions. J Invest Dermatol 2024;144:908–911.e7. doi: 10.1016/j.jid.2023.09.282. [DOI] [PubMed] [Google Scholar]
  • 45.Ferrell PB Jr., McLeod HL. Carbamazepine, HLA-B*1502 and risk of Stevens-Johnson syndrome and toxic epidermal necrolysis: US FDA recommendations. Pharmacogenomics 2008;9:1543–1546. doi: 10.2217/14622416.9.10.1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chen P Lin JJ Lu CS Ong CT Hsieh PF Yang CC, et al. Carbamazepine-induced toxic effects and HLA-B*1502 screening in Taiwan. N Engl J Med 2011;364:1126–1133. doi: 10.1056/NEJMoa1009717. [DOI] [PubMed] [Google Scholar]
  • 47.Mushiroda T Takahashi Y Onuma T Yamamoto Y Kamei T Hoshida T, et al. Association of HLA-A*31:01 screening with the incidence of carbamazepine-induced cutaneous adverse reactions in a Japanese population. JAMA Neurol 2018;75:842–849. doi: 10.1001/jamaneurol.2018.0278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hung SI Chung WH Liou LB Chu CC Lin M Huang HP, et al. HLA-B*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol. Proc Natl Acad Sci U S A 2005;102:4134–4139. doi: 10.1073/pnas.0409500102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tassaneeyakul W Jantararoungtong T Chen P Lin PY Tiamkao S Khunarkornsiri U, et al. Strong association between HLA-B*5801 and allopurinol-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in a Thai population. Pharmacogenet Genomics 2009;19:704–709. doi: 10.1097/FPC.0b013e328330a3b8. [DOI] [PubMed] [Google Scholar]
  • 50.Gonçalo M Coutinho I Teixeira V Gameiro AR Brites MM Nunes R, et al. HLA-B*58:01 is a risk factor for allopurinol-induced DRESS and Stevens-Johnson syndrome/toxic epidermal necrolysis in a Portuguese population. Br J Dermatol 2013;169:660–665. doi: 10.1111/bjd.12389. [DOI] [PubMed] [Google Scholar]
  • 51.Niihara H Kaneko S Ito T Sugamori T Takahashi N Kohno K, et al. HLA-B*58:01 strongly associates with allopurinol-induced adverse drug reactions in a Japanese sample population. J Dermatol Sci 2013;71:150–152. doi: 10.1016/j.jdermsci.2013.04.013. [DOI] [PubMed] [Google Scholar]
  • 52.Cheng L Xiong Y Qin CZ Zhang W Chen XP Li J, et al. HLA-B*58:01 is strongly associated with allopurinol-induced severe cutaneous adverse reactions in Han Chinese patients: A multicentre retrospective case-control clinical study. Br J Dermatol 2015;173:555–558. doi: 10.1111/bjd.13688. [DOI] [PubMed] [Google Scholar]
  • 53.Sukasem C Jantararoungtong T Kuntawong P Puangpetch A Koomdee N Satapornpong P, et al. HLA-B (*) 58:01 for allopurinol-induced cutaneous adverse drug reactions: Implication for clinical interpretation in Thailand. Front Pharmacol 2016;7:186. doi: 10.3389/fphar.2016.00186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wu R Cheng YJ Zhu LL Yu L Zhao XK Jia M, et al. Impact of HLA-B*58:01 allele and allopurinol-induced cutaneous adverse drug reactions: Evidence from 21 pharmacogenetic studies. Oncotarget 2016;7:81870–81879. doi: 10.18632/oncotarget.13250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yu KH, Yu CY, Fang YF. Diagnostic utility of HLA-B*5801 screening in severe allopurinol hypersensitivity syndrome: An updated systematic review and meta-analysis. Int J Rheum Dis 2017;20:1057–1071. doi: 10.1111/1756-185x.13143. [DOI] [PubMed] [Google Scholar]
  • 56.Ke CH Chung WH Wen YH Huang YB Chuang HY Tain YL, et al. Cost-effectiveness analysis for genotyping before allopurinol treatment to prevent severe cutaneous adverse drug reactions. J Rheumatol 2017;44:835–843. doi: 10.3899/jrheum.151476. [DOI] [PubMed] [Google Scholar]
  • 57.Jutkowitz E, Dubreuil M, Lu N, Kuntz KM, Choi HK. The cost-effectiveness of HLA-B*5801 screening to guide initial urate-lowering therapy for gout in the United States. Semin Arthritis Rheum 2017;46:594–600. doi: 10.1016/j.semarthrit.2016.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hetherington S Hughes AR Mosteller M Shortino D Baker KL Spreen W, et al. Genetic variations in HLA-B region and hypersensitivity reactions to abacavir. Lancet 2002;359:1121–1122. doi: 10.1016/s0140-6736(02)08158-8. [DOI] [PubMed] [Google Scholar]
  • 59.Mallal S Nolan D Witt C Masel G Martin AM Moore C, et al. Association between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet 2002;359:727–732. doi: 10.1016/s0140-6736(02)07873-x. [DOI] [PubMed] [Google Scholar]
  • 60.Hughes AR Mosteller M Bansal AT Davies K Haneline SA Lai EH, et al. Association of genetic variations in HLA-B region with hypersensitivity to abacavir in some, but not all, populations. Pharmacogenomics 2004;5:203–211. doi: 10.1517/phgs.5.2.203.27481. [DOI] [PubMed] [Google Scholar]
  • 61.Martin AM Nolan D Gaudieri S Almeida CA Nolan R James I, et al. Predisposition to abacavir hypersensitivity conferred by HLA-B*5701 and a haplotypic Hsp70-Hom variant. Proc Natl Acad Sci U S A 2004;101:4180–4185. doi: 10.1073/pnas.0307067101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Saag M Balu R Phillips E Brachman P Martorell C Burman W, et al. High sensitivity of human leukocyte antigen-b*5701 as a marker for immunologically confirmed abacavir hypersensitivity in white and black patients. Clin Infect Dis 2008;46:1111–1118. doi: 10.1086/529382. [DOI] [PubMed] [Google Scholar]
  • 63.Mallal S Phillips E Carosi G Molina JM Workman C Tomazic J, et al. HLA-B*5701 screening for hypersensitivity to abacavir. N Engl J Med 2008;358:568–579. doi: 10.1056/NEJMoa0706135. [DOI] [PubMed] [Google Scholar]
  • 64.Mounzer K Hsu R Fusco JS Brunet L Henegar CE Vannappagari V, et al. HLA-B*57:01 screening and hypersensitivity reaction to abacavir between 1999 and 2016 in the OPERA(®) observational database: A cohort study. AIDS Res Ther 2019;16:1. doi: 10.1186/s12981-019-0217-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Carr DF Chaponda M Jorgensen AL Castro EC van Oosterhout JJ Khoo SH, et al. Association of human leukocyte antigen alleles and nevirapine hypersensitivity in a Malawian HIV-infected population. Clin Infect Dis 2013;56:1330–1339. doi: 10.1093/cid/cit021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Jongkhajornpong P Ueta M Lekhanont K Puangsricharern V Prabhasawat P Chantaren P, et al. Association of HLA polymorphisms and acetaminophen-related Steven-Johnson syndrome with severe ocular complications in Thai population. Br J Ophthalmol 2022;106:884–888. doi: 10.1136/bjophthalmol-2020-317315. [DOI] [PubMed] [Google Scholar]
  • 67.Ueta M. Findings by an international collaboration on SJS/TEN with severe ocular complications. Front Med (Lausanne) 2021;8:649661. doi: 10.3389/fmed.2021.649661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ueta M Nakamura R Saito Y Tokunaga K Sotozono C Yabe T, et al. Association of HLA class I and II gene polymorphisms with acetaminophen-related Stevens-Johnson syndrome with severe ocular complications in Japanese individuals. Hum Genome Var 2019;6:50. doi: 10.1038/s41439-019-0082-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wilkinson GR. Drug metabolism and variability among patients in drug response. N Engl J Med 2005;352:2211–2221. doi: 10.1056/NEJMra032424. [DOI] [PubMed] [Google Scholar]
  • 70.Chung WH Chang WC Lee YS Wu YY Yang CH Ho HC, et al. Genetic variants associated with phenytoin-related severe cutaneous adverse reactions. JAMA 2014;312:525–534. doi: 10.1001/jama.2014.7859. [DOI] [PubMed] [Google Scholar]
  • 71.Abe R. Immunological response in Stevens-Johnson syndrome and toxic epidermal necrolysis. J Dermatol 2015;42:42–48. doi: 10.1111/1346-8138.12674. [DOI] [PubMed] [Google Scholar]
  • 72.Weltzien HU, Padovan E. Molecular features of penicillin allergy. J Invest Dermatol 1998;110:203–206. doi: 10.1046/j.1523-1747.1998.00122.x. [DOI] [PubMed] [Google Scholar]
  • 73.Naisbitt DJ, Hough SJ, Gill HJ, Pirmohamed M, Kitteringham NR, Park BK. Cellular disposition of sulphamethoxazole and its metabolites: Implications for hypersensitivity. Br J Pharmacol 1999;126:1393–1407. doi: 10.1038/sj.bjp.0702453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Friedmann PS, Lee MS, Friedmann AC, Barnetson RS. Mechanisms in cutaneous drug hypersensitivity reactions. Clin Exp Allergy 2003;33:861–872. doi: 10.1046/j.1365-2222.2003.01718.x. [DOI] [PubMed] [Google Scholar]
  • 75.Callan HE Jenkins RE Maggs JL Lavergne SN Clarke SE Naisbitt DJ, et al. Multiple adduction reactions of nitroso sulfamethoxazole with cysteinyl residues of peptides and proteins: Implications for hapten formation. Chem Res Toxicol 2009;22:937–948. doi: 10.1021/tx900034r. [DOI] [PubMed] [Google Scholar]
  • 76.Pichler WJ. Pharmacological interaction of drugs with antigen-specific immune receptors: The p-i concept. Curr Opin Allergy Clin Immunol 2002;2:301–305. doi: 10.1097/00130832-200208000-00003. [DOI] [PubMed] [Google Scholar]
  • 77.Pichler WJ. Delayed drug hypersensitivity reactions. Ann Intern Med 2003;139:683–693. doi: 10.7326/0003-4819-139-8-200310210-00012. [DOI] [PubMed] [Google Scholar]
  • 78.Pichler WJ Beeler A Keller M Lerch M Posadas S Schmid D, et al. Pharmacological interaction of drugs with immune receptors: The p-i concept. Allergol Int 2006;55:17–25. doi: 10.2332/allergolint.55.17. [DOI] [PubMed] [Google Scholar]
  • 79.Yang CW Hung SI Juo CG Lin YP Fang WH Lu IH, et al. HLA-B*1502-bound peptides: Implications for the pathogenesis of carbamazepine-induced Stevens-Johnson syndrome. J Allergy Clin Immunol 2007;120:870–877. doi: 10.1016/j.jaci.2007.06.017. [DOI] [PubMed] [Google Scholar]
  • 80.Yun J Marcaida MJ Eriksson KK Jamin H Fontana S Pichler WJ, et al. Oxypurinol directly and immediately activates the drug-specific T cells via the preferential use of HLA-B*58:01. J Immunol 2014;192:2984–2993. doi: 10.4049/jimmunol.1302306. [DOI] [PubMed] [Google Scholar]
  • 81.Illing PT Vivian JP Dudek NL Kostenko L Chen Z Bharadwaj M, et al. Immune self-reactivity triggered by drug-modified HLA-peptide repertoire. Nature 2012;486:554–558. doi: 10.1038/nature11147. [DOI] [PubMed] [Google Scholar]
  • 82.Ostrov DA Grant BJ Pompeu YA Sidney J Harndahl M Southwood S, et al. Drug hypersensitivity caused by alteration of the MHC-presented self-peptide repertoire. Proc Natl Acad Sci U S A 2012;109:9959–9964. doi: 10.1073/pnas.1207934109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ko TM Chung WH Wei CY Shih HY Chen JK Lin CH, et al. Shared and restricted T-cell receptor use is crucial for carbamazepine-induced Stevens-Johnson syndrome. J Allergy Clin Immunol 2011;128:1266–1276.e11. doi: 10.1016/j.jaci.2011.08.013. [DOI] [PubMed] [Google Scholar]
  • 84.Wei CY, Chung WH, Huang HW, Chen YT, Hung SI. Direct interaction between HLA-B and carbamazepine activates T cells in patients with Stevens-Johnson syndrome. J Allergy Clin Immunol 2012;129:1562–1569.e5. doi: 10.1016/j.jaci.2011.12.990. [DOI] [PubMed] [Google Scholar]
  • 85.Chung WH Pan RY Chu MT Chin SW Huang YL Wang WC, et al. Oxypurinol-specific T cells possess preferential TCR clonotypes and express granulysin in allopurinol-induced severe cutaneous adverse reactions. J Invest Dermatol 2015;135:2237–2248. doi: 10.1038/jid.2015.165. [DOI] [PubMed] [Google Scholar]
  • 86.Pan RY Chu MT Wang CW Lee YS Lemonnier F Michels AW, et al. Identification of drug-specific public TCR driving severe cutaneous adverse reactions. Nat Commun 2019;10:3569. doi: 10.1038/s41467-019-11396-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Correia O, Delgado L, Ramos JP, Resende C, Torrinha JA. Cutaneous T-cell recruitment in toxic epidermal necrolysis. Further evidence of CD8+ lymphocyte involvement. Arch Dermatol 1993;129:466–468. doi: 10.1001/archderm.1993.01680250078010. [PubMed] [Google Scholar]
  • 88.Le Cleach L Delaire S Boumsell L Bagot M Bourgault-Villada I Bensussan A, et al. Blister fluid T lymphocytes during toxic epidermal necrolysis are functional cytotoxic cells which express human natural killer (NK) inhibitory receptors. Clin Exp Immunol 2000;119:225–230. doi: 10.1046/j.1365-2249.2000.01119.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Tohyama M Watanabe H Murakami S Shirakata Y Sayama K Iijima M, et al. Possible involvement of CD14+ CD16+ monocyte lineage cells in the epidermal damage of Stevens-Johnson syndrome and toxic epidermal necrolysis. Br J Dermatol 2012;166:322–330. doi: 10.1111/j.1365-2133.2011.10649.x. [DOI] [PubMed] [Google Scholar]
  • 90.Correia O, Delgado L, Roujeau JC, Le Cleach L, Fleming-Torrinha JA. Soluble interleukin 2 receptor and interleukin 1alpha in toxic epidermal necrolysis: A comparative analysis of serum and blister fluid samples. Arch Dermatol 2002;138:29–32. doi: 10.1001/archderm.138.1.29. [DOI] [PubMed] [Google Scholar]
  • 91.Paul C Wolkenstein P Adle H Wechsler J Garchon HJ Revuz J, et al. Apoptosis as a mechanism of keratinocyte death in toxic epidermal necrolysis. Br J Dermatol 1996;134:710–714. doi: 10.1111/j.1365-2133.1996.tb06976.x. [DOI] [PubMed] [Google Scholar]
  • 92.Elmore S. Apoptosis: A review of programmed cell death. Toxicol Pathol 2007;35:495–516. doi: 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Viard-Leveugle I Bullani RR Meda P Micheau O Limat A Saurat JH, et al. Intracellular localization of keratinocyte Fas ligand explains lack of cytolytic activity under physiological conditions. J Biol Chem 2003;278:16183–16188. doi: 10.1074/jbc.M212188200. [DOI] [PubMed] [Google Scholar]
  • 94.Viard I Wehrli P Bullani R Schneider P Holler N Salomon D, et al. Inhibition of toxic epidermal necrolysis by blockade of CD95 with human intravenous immunoglobulin. Science 1998;282:490–493. doi: 10.1126/science.282.5388.490. [DOI] [PubMed] [Google Scholar]
  • 95.Tanaka M Suda T Haze K Nakamura N Sato K Kimura F, et al. Fas ligand in human serum. Nat Med 1996;2:317–322. doi: 10.1038/nm0396-317. [DOI] [PubMed] [Google Scholar]
  • 96.Abe R, Shimizu T, Shibaki A, Nakamura H, Watanabe H, Shimizu H. Toxic epidermal necrolysis and Stevens-Johnson syndrome are induced by soluble Fas ligand. Am J Pathol 2003;162:1515–1520. doi: 10.1016/s0002-9440(10)64284-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Abe R. Toxic epidermal necrolysis and Stevens-Johnson syndrome: Soluble Fas ligand involvement in the pathomechanisms of these diseases. J Dermatol Sci 2008;52:151–159. doi: 10.1016/j.jdermsci.2008.06.003. [DOI] [PubMed] [Google Scholar]
  • 98.Stur K, Karlhofer FM, Stingl G. Soluble FAS ligand: A discriminating feature between drug-induced skin eruptions and viral exanthemas. J Invest Dermatol 2007;127:802–807. doi: 10.1038/sj.jid.5700648. [DOI] [PubMed] [Google Scholar]
  • 99.Nassif A Bensussan A Dorothée G Mami-Chouaib F Bachot N Bagot M, et al. Drug specific cytotoxic T-cells in the skin lesions of a patient with toxic epidermal necrolysis. J Invest Dermatol 2002;118:728–733. doi: 10.1046/j.1523-1747.2002.01622.x. [DOI] [PubMed] [Google Scholar]
  • 100.Nassif A Bensussan A Boumsell L Deniaud A Moslehi H Wolkenstein P, et al. Toxic epidermal necrolysis: Effector cells are drug-specific cytotoxic T cells. J Allergy Clin Immunol 2004;114:1209–1215. doi: 10.1016/j.jaci.2004.07.047. [DOI] [PubMed] [Google Scholar]
  • 101.Chung WH Hung SI Yang JY Su SC Huang SP Wei CY, et al. Granulysin is a key mediator for disseminated keratinocyte death in Stevens-Johnson syndrome and toxic epidermal necrolysis. Nat Med 2008;14:1343–1350. doi: 10.1038/nm.1884. [DOI] [PubMed] [Google Scholar]
  • 102.Abe R, Yoshioka N, Murata J, Fujita Y, Shimizu H. Granulysin as a marker for early diagnosis of the Stevens-Johnson syndrome. Ann Intern Med 2009;151:514–515. doi: 10.7326/0003-4819-151-7-200910060-00016. [DOI] [PubMed] [Google Scholar]
  • 103.Saito N, Abe R, Yoshioka N, Murata J, Fujita Y, Shimizu H. Prolonged elevation of serum granulysin in drug-induced hypersensitivity syndrome. Br J Dermatol 2012;167:452–453. doi: 10.1111/j.1365-2133.2012.10921.x. [DOI] [PubMed] [Google Scholar]
  • 104.Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci 2019;9:19. doi: 10.1186/s13578-019-0282-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhang C Zhu Z Gao J Yang L Dang E Fang H, et al. Plasma exosomal miR-375-3p regulates mitochondria-dependent keratinocyte apoptosis by targeting XIAP in severe drug-induced skin reactions. Sci Transl Med 2020;12:eaaw6142. doi: 10.1126/scitranslmed.aaw6142. [DOI] [PubMed] [Google Scholar]
  • 106.Saito N Qiao H Yanagi T Shinkuma S Nishimura K Suto A, et al. An annexin A1-FPR1 interaction contributes to necroptosis of keratinocytes in severe cutaneous adverse drug reactions. Sci Transl Med 2014;6:245ra295. doi: 10.1126/scitranslmed.3008227. [DOI] [PubMed] [Google Scholar]
  • 107.Linkermann A, Green DR. Necroptosis. N Engl J Med 2014;370:455–465. doi: 10.1056/NEJMra1310050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kinoshita M Ogawa Y Hama N Ujiie I Hasegawa A Nakajima S, et al. Neutrophils initiate and exacerbate Stevens-Johnson syndrome and toxic epidermal necrolysis. Sci Transl Med 2021;13:eaax2398. doi: 10.1126/scitranslmed.aax2398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Saito N Yoshioka N Abe R Qiao H Fujita Y Hoshina D, et al. Stevens-Johnson syndrome/toxic epidermal necrolysis mouse model generated by using PBMCs and the skin of patients. J Allergy Clin Immunol 2013;131:434–441.e1–9. doi: 10.1016/j.jaci.2012.09.014. [DOI] [PubMed] [Google Scholar]
  • 110.Nassif A Moslehi H Le Gouvello S Bagot M Lyonnet L Michel L, et al. Evaluation of the potential role of cytokines in toxic epidermal necrolysis. J Invest Dermatol 2004;123:850–855. doi: 10.1111/j.0022-202X.2004.23439.x. [DOI] [PubMed] [Google Scholar]
  • 111.Paquet P, Nikkels A, Arrese JE, Vanderkelen A, Piérard GE. Macrophages and tumor necrosis factor alpha in toxic epidermal necrolysis. Arch Dermatol 1994;130:605–608. doi: 10.1001/archderm.130.5.605. [PubMed] [Google Scholar]
  • 112.Kida Y, Kuwano K, Zhang Y, Arai S. Acholeplasma laidlawii up-regulates granulysin gene expression via transcription factor activator protein-1 in a human monocytic cell line, THP-1. Immunology 2001;104:324–332. doi: 10.1046/j.1365-2567.2001.01310.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Viard-Leveugle I Gaide O Jankovic D Feldmeyer L Kerl K Pickard C, et al. TNF-α and IFN-γ are potential inducers of Fas-mediated keratinocyte apoptosis through activation of inducible nitric oxide synthase in toxic epidermal necrolysis. J Invest Dermatol 2013;133:489–498. doi: 10.1038/jid.2012.330. [DOI] [PubMed] [Google Scholar]
  • 114.Wang F, He D, Tang X, Zhang X. Chemokine expression in diverse nonimmediate drug hypersensitivity reactions: Focus on thymus activation-regulated chemokine, cutaneous T-cell-attracting chemokine, and interleukin-10. Ann Allergy Asthma Immunol 2014;113:204–208. doi: 10.1016/j.anai.2014.05.006. [DOI] [PubMed] [Google Scholar]
  • 115.Wang F, Ye Y, Luo ZY, Gao Q, Luo DQ, Zhang X. Diverse expression of TNF-α and CCL27 in serum and blister of Stevens-Johnson syndrome/toxic epidermal necrolysis. Clin Transl Allergy 2018;8:12. doi: 10.1186/s13601-018-0199-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.McCully ML, Moser B. The human cutaneous chemokine system. Front Immunol 2011;2:33. doi: 10.3389/fimmu.2011.00033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Tapia B Padial A Sánchez-Sabaté E Alvarez-Ferreira J Morel E Blanca M, et al. Involvement of CCL27-CCR10 interactions in drug-induced cutaneous reactions. J Allergy Clin Immunol 2004;114:335–340. doi: 10.1016/j.jaci.2004.04.034. [DOI] [PubMed] [Google Scholar]
  • 118.Su SC Mockenhaupt M Wolkenstein P Dunant A Le Gouvello S Chen CB, et al. Interleukin-15 is associated with severity and mortality in Stevens-Johnson syndrome/toxic epidermal necrolysis. J Invest Dermatol 2017;137:1065–1073. doi: 10.1016/j.jid.2016.11.034. [DOI] [PubMed] [Google Scholar]
  • 119.McInnes IB, Leung BP, Sturrock RD, Field M, Liew FY. Interleukin-15 mediates T cell-dependent regulation of tumor necrosis factor-alpha production in rheumatoid arthritis. Nat Med 1997;3:189–195. doi: 10.1038/nm0297-189. [DOI] [PubMed] [Google Scholar]
  • 120.Hama N Nishimura K Hasegawa A Yuki A Kume H Adachi J, et al. Galectin-7 as a potential biomarker of Stevens-Johnson syndrome/toxic epidermal necrolysis: Identification by targeted proteomics using causative drug-exposed peripheral blood cells. J Allergy Clin Immunol Pract 2019;7:2894–2897.e7. doi: 10.1016/j.jaip.2019.05.002. [DOI] [PubMed] [Google Scholar]
  • 121.Vidova V, Spacil Z. A review on mass spectrometry-based quantitative proteomics: Targeted and data independent acquisition. Anal Chim Acta 2017;964:7–23. doi: 10.1016/j.aca.2017.01.059. [DOI] [PubMed] [Google Scholar]
  • 122.Hasegawa A Shinkuma S Hayashi R Hama N Watanabe H Kinoshita M, et al. RIP3 as a diagnostic and severity marker for Stevens-Johnson syndrome and toxic epidermal necrolysis. J Allergy Clin Immunol Pract 2020;8:1768–1771.e7. doi: 10.1016/j.jaip.2020.01.006. [DOI] [PubMed] [Google Scholar]
  • 123.Qing DY Conegliano D Shashaty MG Seo J Reilly JP Worthen GS, et al. Red blood cells induce necroptosis of lung endothelial cells and increase susceptibility to lung inflammation. Am J Respir Crit Care Med 2014;190:1243–1254. doi: 10.1164/rccm.201406-1095OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Schneider JA, Cohen PR. Stevens-Johnson syndrome and toxic epidermal necrolysis: A concise review with a comprehensive summary of therapeutic interventions emphasizing supportive measures. Adv Ther 2017;34:1235–1244. doi: 10.1007/s12325-017-0530-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.de Prost N Ingen-Housz-Oro S Duong TA Valeyrie-Allanore L Legrand P Wolkenstein P, et al. Bacteremia in Stevens-Johnson syndrome and toxic epidermal necrolysis: Epidemiology, risk factors, and predictive value of skin cultures. Medicine (Baltimore) 2010;89:28–36. doi: 10.1097/MD.0b013e3181ca4290. [DOI] [PubMed] [Google Scholar]
  • 126.White KD Abe R Ardern-Jones M Beachkofsky T Bouchard C Carleton B, et al. SJS/TEN 2017: Building multidisciplinary networks to drive science and translation. J Allergy Clin Immunol Pract 2018;6:38–69. doi: 10.1016/j.jaip.2017.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Creamer D Walsh SA Dziewulski P Exton LS Lee HY Dart JK, et al. U.K. guidelines for the management of Stevens-Johnson syndrome/toxic epidermal necrolysis in adults 2016. Br J Dermatol 2016;174:1194–1227. doi: 10.1111/bjd.14530. [DOI] [PubMed] [Google Scholar]
  • 128.Zimmermann S Sekula P Venhoff M Motschall E Knaus J Schumacher M, et al. Systemic immunomodulating therapies for Stevens-Johnson syndrome and toxic epidermal necrolysis: A systematic review and meta-analysis. JAMA Dermatol 2017;153:514–522. doi: 10.1001/jamadermatol.2016.5668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Ginsburg CM. Stevens-Johnson syndrome in children. Pediatr Infect Dis 1982;1:155–158. doi: 10.1097/00006454-198205000-00005. [DOI] [PubMed] [Google Scholar]
  • 130.Halebian PH, Corder VJ, Madden MR, Finklestein JL, Shires GT. Improved burn center survival of patients with toxic epidermal necrolysis managed without corticosteroids. Ann Surg 1986;204:503–512. doi: 10.1097/00000658-198611000-00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Kelemen JJ, 3rd, Cioffi WG, McManus WF, Mason AD Jr., Pruitt BA Jr. Burn center care for patients with toxic epidermal necrolysis. J Am Coll Surg 1995;180:273–278. [PubMed] [Google Scholar]
  • 132.Finkelstein Y Soon GS Acuna P George M Pope E Ito S, et al. Recurrence and outcomes of Stevens-Johnson syndrome and toxic epidermal necrolysis in children. Pediatrics 2011;128:723–728. doi: 10.1542/peds.2010-3322. [DOI] [PubMed] [Google Scholar]
  • 133.Roujeau JC, Bastuji-Garin S. Systematic review of treatments for Stevens-Johnson syndrome and toxic epidermal necrolysis using the SCORTEN score as a tool for evaluating mortality. Ther Adv Drug Saf 2011;2:87–94. doi: 10.1177/2042098611404094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Sekula P Dunant A Mockenhaupt M Naldi L Bouwes Bavinck JN Halevy S, et al. Comprehensive survival analysis of a cohort of patients with Stevens-Johnson syndrome and toxic epidermal necrolysis. J Invest Dermatol 2013;133:1197–1204. doi: 10.1038/jid.2012.510. [DOI] [PubMed] [Google Scholar]
  • 135.Schneck J, Fagot JP, Sekula P, Sassolas B, Roujeau JC, Mockenhaupt M. Effects of treatments on the mortality of Stevens-Johnson syndrome and toxic epidermal necrolysis: A retrospective study on patients included in the prospective EuroSCAR Study. J Am Acad Dermatol 2008;58:33–40. doi: 10.1016/j.jaad.2007.08.039. [DOI] [PubMed] [Google Scholar]
  • 136.Hirahara K Kano Y Sato Y Horie C Okazaki A Ishida T, et al. Methylprednisolone pulse therapy for Stevens-Johnson syndrome/toxic epidermal necrolysis: Clinical evaluation and analysis of biomarkers. J Am Acad Dermatol 2013;69:496–498. doi: 10.1016/j.jaad.2013.04.007. [DOI] [PubMed] [Google Scholar]
  • 137.Kardaun SH, Jonkman MF. Dexamethasone pulse therapy for Stevens-Johnson syndrome/toxic epidermal necrolysis. Acta Derm Venereol 2007;87:144–148. doi: 10.2340/00015555-0214. [DOI] [PubMed] [Google Scholar]
  • 138.Araki Y Sotozono C Inatomi T Ueta M Yokoi N Ueda E, et al. Successful treatment of Stevens-Johnson syndrome with steroid pulse therapy at disease onset. Am J Ophthalmol 2009;147:1004–1011, 1011.e1. doi: 10.1016/j.ajo.2008.12.040. [DOI] [PubMed] [Google Scholar]
  • 139.Ballow M. The IgG molecule as a biological immune response modifier: Mechanisms of action of intravenous immune serum globulin in autoimmune and inflammatory disorders. J Allergy Clin Immunol 2011;127:315–323; quiz 324–325. doi: 10.1016/j.jaci.2010.10.030. [DOI] [PubMed] [Google Scholar]
  • 140.Bachot N, Revuz J, Roujeau JC. Intravenous immunoglobulin treatment for Stevens-Johnson syndrome and toxic epidermal necrolysis: A prospective noncomparative study showing no benefit on mortality or progression. Arch Dermatol 2003;139:33–36. doi: 10.1001/archderm.139.1.33. [DOI] [PubMed] [Google Scholar]
  • 141.Brown KM, Silver GM, Halerz M, Walaszek P, Sandroni A, Gamelli RL. Toxic epidermal necrolysis: Does immunoglobulin make a difference? J Burn Care Rehabil 2004;25:81–88. doi: 10.1097/01.Bcr.0000105096.93526.27. [DOI] [PubMed] [Google Scholar]
  • 142.Lee HY, Lim YL, Thirumoorthy T, Pang SM. The role of intravenous immunoglobulin in toxic epidermal necrolysis: A retrospective analysis of 64 patients managed in a specialized centre. Br J Dermatol 2013;169:1304–1309. doi: 10.1111/bjd.12607. [DOI] [PubMed] [Google Scholar]
  • 143.Morici MV Galen WK Shetty AK Lebouef RP Gouri TP Cowan GS, et al. Intravenous immunoglobulin therapy for children with Stevens-Johnson syndrome. J Rheumatol 2000;27:2494–2497. [PubMed] [Google Scholar]
  • 144.Metry DW, Jung P, Levy ML. Use of intravenous immunoglobulin in children with Stevens-Johnson syndrome and toxic epidermal necrolysis: Seven cases and review of the literature. Pediatrics 2003;112(6 Pt 1):1430–1436. doi: 10.1542/peds.112.6.1430. [DOI] [PubMed] [Google Scholar]
  • 145.Prins C Vittorio C Padilla RS Hunziker T Itin P Förster J, et al. Effect of high-dose intravenous immunoglobulin therapy in Stevens-Johnson syndrome: A retrospective, multicenter study. Dermatology 2003;207:96–99. doi: 10.1159/000070957. [DOI] [PubMed] [Google Scholar]
  • 146.Faye O, Roujeau JC. Treatment of epidermal necrolysis with high-dose intravenous immunoglobulins (IV Ig): Clinical experience to date. Drugs 2005;65:2085–2090. doi: 10.2165/00003495-200565150-00002. [DOI] [PubMed] [Google Scholar]
  • 147.Huang YC, Li YC, Chen TJ. The efficacy of intravenous immunoglobulin for the treatment of toxic epidermal necrolysis: A systematic review and meta-analysis. Br J Dermatol 2012;167:424–432. doi: 10.1111/j.1365-2133.2012.10965.x. [DOI] [PubMed] [Google Scholar]
  • 148.Barron SJ, Del Vecchio MT, Aronoff SC. Intravenous immunoglobulin in the treatment of Stevens-Johnson syndrome and toxic epidermal necrolysis: A meta-analysis with meta-regression of observational studies. Int J Dermatol 2015;54:108–115. doi: 10.1111/ijd.12423. [DOI] [PubMed] [Google Scholar]
  • 149.Tsai TY Huang IH Chao YC Li H Hsieh TS Wang HH, et al. Treating toxic epidermal necrolysis with systemic immunomodulating therapies: A systematic review and network meta-analysis. J Am Acad Dermatol 2021;84:390–397. doi: 10.1016/j.jaad.2020.08.122. [DOI] [PubMed] [Google Scholar]
  • 150.Yang L, Shou YH, Li F, Zhu XH, Yang YS, Xu JH. Intravenous immunoglobulin combined with corticosteroids for the treatment of Stevens-Johnson syndrome/toxic epidermal necrolysis: A propensity-matched retrospective study in China. Front Pharmacol 2021;12:750173. doi: 10.3389/fphar.2021.750173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Chung WH, Wang CW, Dao RL. Severe cutaneous adverse drug reactions. J Dermatol 2016;43:758–766. doi: 10.1111/1346-8138.13430. [DOI] [PubMed] [Google Scholar]
  • 152.Valeyrie-Allanore L Wolkenstein P Brochard L Ortonne N Maître B Revuz J, et al. Open trial of ciclosporin treatment for Stevens-Johnson syndrome and toxic epidermal necrolysis. Br J Dermatol 2010;163:847–853. doi: 10.1111/j.1365-2133.2010.09863.x. [DOI] [PubMed] [Google Scholar]
  • 153.Kirchhof MG, Miliszewski MA, Sikora S, Papp A, Dutz JP. Retrospective review of Stevens-Johnson syndrome/toxic epidermal necrolysis treatment comparing intravenous immunoglobulin with cyclosporine. J Am Acad Dermatol 2014;71:941–947. doi: 10.1016/j.jaad.2014.07.016. [DOI] [PubMed] [Google Scholar]
  • 154.Kumar P, Kanti Das N. Cyclosporine in toxic epidermal necrolysis: A brief review of the emerging therapeutic modality. Dermatol Online J 2016;22:13030/qt3m82s074. doi: 10.5070/D32210032890. [PubMed] [Google Scholar]
  • 155.González-Herrada C Rodríguez-Martín S Cachafeiro L Lerma V González O Lorente JA, et al. Cyclosporine use in epidermal necrolysis is associated with an important mortality reduction: Evidence from three different approaches. J Invest Dermatol 2017;137:2092–2100. doi: 10.1016/j.jid.2017.05.022. [DOI] [PubMed] [Google Scholar]
  • 156.Lee HY, Fook-Chong S, Koh HY, Thirumoorthy T, Pang SM. Cyclosporine treatment for Stevens-Johnson syndrome/toxic epidermal necrolysis: Retrospective analysis of a cohort treated in a specialized referral center. J Am Acad Dermatol 2017;76:106–113. doi: 10.1016/j.jaad.2016.07.048. [DOI] [PubMed] [Google Scholar]
  • 157.Roujeau JC, Mockenhaupt M, Guillaume JC, Revuz J. New evidence supporting cyclosporine efficacy in epidermal necrolysis. J Invest Dermatol 2017;137:2047–2049. doi: 10.1016/j.jid.2017.07.828. [DOI] [PubMed] [Google Scholar]
  • 158.Ng QX, De Deyn M, Venkatanarayanan N, Ho CYX, Yeo WS. A meta-analysis of cyclosporine treatment for Stevens-Johnson syndrome/toxic epidermal necrolysis. J Inflamm Res 2018;11:135–142. doi: 10.2147/jir.S160964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Shah R, Chen ST, Kroshinsky D. Use of cyclosporine for the treatment of Stevens-Johnson syndrome/toxic epidermal necrolysis. J Am Acad Dermatol 2021;85:512–513. doi: 10.1016/j.jaad.2018.09.063. [DOI] [PubMed] [Google Scholar]
  • 160.Gilbert M, Scherrer LA. Efficacy and safety of cyclosporine in Stevens-Johnson syndrome and toxic epidermal necrolysis. Dermatol Ther 2019;32:e12758. doi: 10.1111/dth.12758. [DOI] [PubMed] [Google Scholar]
  • 161.Kamanabroo D, Schmitz-Landgraf W, Czarnetzki BM. Plasmapheresis in severe drug-induced toxic epidermal necrolysis. Arch Dermatol 1985;121:1548–1549. doi: 10.1001/archderm.1985.01660120074023. [PubMed] [Google Scholar]
  • 162.Sakellariou G Koukoudis P Karpouzas J Alexopoulos E Papadopoulou D Chrisomalis F, et al. Plasma exchange (PE) treatment in drug-induced toxic epidermal necrolysis (TEN). Int J Artif Organs 1991;14:634–638. doi: 10.1177/039139889101401006. [PubMed] [Google Scholar]
  • 163.Chaidemenos GC, Chrysomallis F, Sombolos K, Mourellou O, Ioannides D, Papakonstantinou M. Plasmapheresis in toxic epidermal necrolysis. Int J Dermatol 1997;36:218–221. doi: 10.1046/j.1365-4362.1997.00192.x. [DOI] [PubMed] [Google Scholar]
  • 164.Yamada H, Takamori K, Yaguchi H, Ogawa H. A study of the efficacy of plasmapheresis for the treatment of drug induced toxic epidermal necrolysis. Ther Apher 1998;2:153–156. doi: 10.1111/j.1744-9987.1998.tb00094.x. [DOI] [PubMed] [Google Scholar]
  • 165.Egan CA, Grant WJ, Morris SE, Saffle JR, Zone JJ. Plasmapheresis as an adjunct treatment in toxic epidermal necrolysis. J Am Acad Dermatol 1999;40:458–461. doi: 10.1016/s0190-9622(99)70497-4. [DOI] [PubMed] [Google Scholar]
  • 166.Bamichas G Natse T Christidou F Stangou M Karagianni A Koukourikos S, et al. Plasma exchange in patients with toxic epidermal necrolysis. Ther Apher 2002;6:225–228. doi: 10.1046/j.1526-0968.2002.00409.x. [DOI] [PubMed] [Google Scholar]
  • 167.Furubacke A, Berlin G, Anderson C, Sjöberg F. Lack of significant treatment effect of plasma exchange in the treatment of drug-induced toxic epidermal necrolysis? Intensive Care Med 1999;25:1307–1310. doi: 10.1007/s001340051063. [DOI] [PubMed] [Google Scholar]
  • 168.Narita YM, Hirahara K, Mizukawa Y, Kano Y, Shiohara T. Efficacy of plasmapheresis for the treatment of severe toxic epidermal necrolysis: Is cytokine expression analysis useful in predicting its therapeutic efficacy? J Dermatol 2011;38:236–245. doi: 10.1111/j.1346-8138.2010.01154.x. [DOI] [PubMed] [Google Scholar]
  • 169.Lissia M, Figus A, Rubino C. Intravenous immunoglobulins and plasmapheresis combined treatment in patients with severe toxic epidermal necrolysis: Preliminary report. Br J Plast Surg 2005;58:504–510. doi: 10.1016/j.bjps.2004.12.007. [DOI] [PubMed] [Google Scholar]
  • 170.Han F Zhang J Guo Q Feng Y Gao Y Guo L, et al. Successful treatment of toxic epidermal necrolysis using plasmapheresis: A prospective observational study. J Crit Care 2017;42:65–68. doi: 10.1016/j.jcrc.2017.07.002. [DOI] [PubMed] [Google Scholar]
  • 171.Fischer M, Fiedler E, Marsch WC, Wohlrab J. Antitumour necrosis factor-alpha antibodies (infliximab) in the treatment of a patient with toxic epidermal necrolysis. Br J Dermatol 2002;146:707–709. doi: 10.1046/j.1365-2133.2002.46833.x. [DOI] [PubMed] [Google Scholar]
  • 172.Scott-Lang V, Tidman M, McKay D. Toxic epidermal necrolysis in a child successfully treated with infliximab. Pediatr Dermatol 2014;31:532–534. doi: 10.1111/pde.12029. [DOI] [PubMed] [Google Scholar]
  • 173.Wojtkiewicz A, Wysocki M, Fortuna J, Chrupek M, Matczuk M, Koltan A. Beneficial and rapid effect of infliximab on the course of toxic epidermal necrolysis. Acta Derm Venereol 2008;88:420–421. doi: 10.2340/00015555-0462. [DOI] [PubMed] [Google Scholar]
  • 174.Zárate-Correa LC, Carrillo-Gómez DC, Ramírez-Escobar AF, Serrano-Reyes C. Toxic epidermal necrolysis successfully treated with infliximab. J Investig Allergol Clin Immunol 2013;23:61–63. [PubMed] [Google Scholar]
  • 175.Gaitanis G Spyridonos P Patmanidis K Koulouras V Nakos G Tzaphlidou M, et al. Treatment of toxic epidermal necrolysis with the combination of infliximab and high-dose intravenous immunoglobulin. Dermatology 2012;224:134–139. doi: 10.1159/000338202. [DOI] [PubMed] [Google Scholar]
  • 176.Patmanidis K Sidiras A Dolianitis K Simelidis D Solomonidis C Gaitanis G, et al. Combination of infliximab and high-dose intravenous immunoglobulin for toxic epidermal necrolysis: Successful treatment of an elderly patient. Case Rep Dermatol Med 2012;2012:915314. doi: 10.1155/2012/915314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Wang CW Yang LY Chen CB Ho HC Hung SI Yang CH, et al. Randomized, controlled trial of TNF-α antagonist in CTL-mediated severe cutaneous adverse reactions. J Clin Invest 2018;128:985–996. doi: 10.1172/jci93349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Zhang J Lu CW Chen CB Wang CW Chen WT Cheng B, et al. Evaluation of combination therapy with etanercept and systemic corticosteroids for Stevens-Johnson syndrome and toxic epidermal necrolysis: A multicenter observational study. J Allergy Clin Immunol Pract 2022;10:1295–1304.e6. doi: 10.1016/j.jaip.2022.01.038. [DOI] [PubMed] [Google Scholar]

Articles from Chinese Medical Journal are provided here courtesy of Wolters Kluwer Health

RESOURCES