Review Article


A Literature Review on Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis and its Complications

Authors: Marium Ali, Ibram Zaki, Shamail Zia, Muhammad Haseeb, Rahul Pawate, Meeran Asher Syed , Muhammad Naveed Uz Zafar, Vardah Zia, Syed Jawwad Ali, Fazail Zia, Farozaan Shamail
DOI: https://doi.org/10.37184/jlnh.2959-1805.1.2
Year: 2023
Volume: 1



Abstract

Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are delayed-type hypersensitivity reactions to medications. Early signs of identification and proper management are critical for patients’ survival. The distinct features initially resemble influenza-like conditions, malaise, and fever with gradual pain and lesions in the oral, ocular, and genital areas along with other systemic symptoms. SJS/TEN can be differentiated: SJS by the degree of skin involvement and TEN by the degree of exfoliation. In SJS, skin involvement is less than 10%, whereas in TEN, skin involvement is gretater than 30%; overlapping among the two develops with 10–30% skin involvement. For the identification of culprit drugs, oral provocation tests, patch testing, and in vitro assay tests are used. The estimation of mortality rates for SJS/TEN has been performed by using SCORTEN- severity-of-illness score for toxic epidermal necrolysis. Sulfonamides, anti-epileptics, NSAIDs (oxicams), and allopurinol are some of the medications with a high risk of SJS/TEN development. Drug-mediated SJS/TEN is facilitated by genetic involvement particularly HLA antigens along with CYP450 genetic variants. Most frequent clinical features include mucosal involvement of nasopharyngeal, esophageal, and genital regions accompanying blisters and erosions. A multidisciplinary approach is required for the handling and treatment of SJS/TEN. It is mandatory to withdraw the contributing medicine followed by supportive therapy. Clinical management involves the use of corticosteroids, cyclosporine, and immunoglobulins.

Keywords:
Stevens-Johnson syndrome, skin involvement, toxic epidermal necrolysis, delayed hypersensitivity.

INTRODUCTION

Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), described by blisters and skin exfoliation, are fatal, cutaneous, and mucosal membrane reactions [1]. The main causative sources include some drugs and infectious species like Mycoplasma [2] and the herpes simplex virus. SJS/TEN share similarities amid the disease spectrum with variations in severity and are mainly classified based on skin detachment throughout the body [3, 4]. Around 1.58 -2.26/million cases were reported in the USA which is contradictory to its high mortality rate being 4.8% (SJS) and 14.8% (TEN) [5]. Besides, sequelae are also reported after recovery like blindness, etc. Therefore, very precise and early diagnosis and appropriate therapeutic approach are required to initiate the management as soon as possible. Prediction of severity can be assessed with the early identification of biomarkers [3].

SJS/TEN presents purpuric macules, blisters with pain, and lesions that involve skin and mucosa. The appearance of lesions starts 4 to 28 days after exposure to the causative medication. The development of skin lesions is led by fever, weakness, and respiratory tract indications. Nearly every patient shows the involvement of mucosa in the eyes, genitalia, and mouth. Additionally, systemic involvements include the complications of cardiopulmnary, urinary tract system, and gastrointestinal tract. Sequelae and complications due to multiple organ involvement that lead to mortality were also reported and include sepsis, cutaneous infection, hepatitis, and pneumonia [4]. The recovery phase of SJS/TEN also carries events of sequelae including mucosal and dermal issues. According to one study, the prevalence of sequelae accounts for 84.3% of skin complications, 59.5% of ocular complications, and 50.8% of oral mucosal complications [5]. Neurological complications include depression associated with anxiety, along with post-traumatic stress disorder. The physical and psychological sequelae reported by a study showed that 28.2% of patients lost the ability to work whereas 30% – 68.1% of the patients were either scared of or non-compliant with medication administration [5].

SJS/TEN is a stressful illness and no standard treatment has been reported so far. The paucity of SJS/TEN is the obstacle in conducting double-blind controlled research of high-quality to interpret the effectiveness of drugs.

Mostly, data is gathered through retrospective studies and case reports but without any obvious agreement on the usefulness of multiple therapeutic agents. This review focuses on the provision of evidence-based data about using different treatment modalities for SJS/TEN [6].

METHODOLOGY

This review article emphasizes the knowledge overview and updates on the development and management of SJS/TEN. PubMed and Google Scholar were searched from the year 2000 to 2022, using the keywords Stevens-Johnson syndrome and toxic epidermal necrolysis. The exploration was confined to articles delivering generalized data on SJS/TEN in human adults (≥18 years old) and to articles written in English. Further articles were explored after hand-searching for appropriate ones, as well as assessing the references of major articles.

We conducted a systematic review of all English language publications. We included all interventional, observational, meta-analysis, and retrospective studies and case reports. MeSH terms (medical subject headings) included Stevens-Johnson syndrome and toxic epidermal necrolysis. The article selection methodology is displayed in Fig. (1).

RESULTS

After applying the inclusion criteria, 12,700 articles were recognized in the search, of which 119 were incorporated in the final review

DISCUSSION

Genetic Contribution to the Development of SJS/TEN

An incline in the evidence of genetic incidence of cutaneous adverse reactions has been described in various studies. Chung and co-investigators (2004) studied a Han Chinese populace and reported a robust association between human leukocyte antigen (HLA)-B*15:02 and carbamazepine (CBZ)-prompted SJS/TEN. There is a division of HLA alleles (class I and class II), which are specific for presenting antigenic peptides to T cells, which then activate the immune reaction. In this research, 44 cases of CBZ-prompted SJS/TEN were incorporatedand there was an HLA-B*15:02 allele in 100% of cases. More studies described the relationship between CBZ-prompted SJS/TEN and the HLA-B*15:02 allele in populaces comprising China, Thailand, Malaysia, and India [4, 7-9].

Other than CBZ, the link between SJS/TEN and HLA-B*15:02 was established for antiepileptic medications also. Even though the occurrence was lesser than that noted with CBZ, a sturdy relationship with phenytoin-, lamotrigine- and oxcarbazepine-prompted SJS/TEN was exhibited by HLA-*15:02 [10]. On the other hand, no relationship was established amid CBZ-prompted SJS/TEN and HLA-B*15:02 in Japan, Korea, and Europe [11, 12]. The link between HLA-A*31:01 and CBZ-prompted SJS/TEN was revealed by Ozeki et al. [13]. HLA-A*31:01 exhibited a link with CBZ-prompted SJS/TEN in Korean, European, and Japanese populaces [14, 15] while in Asian populaces, most of CBZ-prompted SJS/TEN is linked with HLA-B*15:02; the relationship with HLA-A*31:01 is revealed in populaces of multiple ethnicities [4]. Before the administration of CBZ, the US FDA, in 2008, recommended executing HLA-B*15:02 genotyping [4]. HLA-B*15:02 screening is stated to have a sturdy relationship with a decline in CBZ-prompted SJS/TEN incidences in Taiwan [16].

Apart from antiepileptics, several other medications like allopurinol and abacavir have HLA relations. Allopurinol, which is an anti-hyperuricemic medication, is rendered as a key reason for SJS/TEN. Several ethnicities have demonstrated a link between HLA-B*58:01 and allopurinol-prompted SJS/TEN together with Korean, Thai, Taiwanese, European, and Japanese people. So, such data proposed that HLA-B*58:01 genotyping could perhaps be beneficial in the prevention of allopurinol-prompted SJS/TEN [17-20].

Cytochrome Ps (CYPs) are genetically significant elements that are involved in medication metabolism. There are 57 variants of CYP450 genes and each one displays practical variances. Cases with slow drug metabolism due to CYP450 variants have a great risk to develop ADRs (adverse drug reactions). Particular genetic factors related to phenytoin-induced SJS/TEN were identified by Chung et al. [21]. This study, in CYP2C9, identified 16 important single nucleotide polymorphisms. Cases with phenytoin-prompted SJS/TEN having CYP2C9*3 presented late phenytoin clearance, leading to more severe illness [4].

Clinical Presentation

Except for BSA (body surface area) involved, the clinical presentation is comparable to the spectrum of disease. In most cases, the cutaneous connection is led by a prodromal symptoms stage i.e. fever, sore throat, malaise, and cough [22-24]. Subsequent cutaneous and mucosal contributions are general and characteristically look like erythematous macules or uncharacteristic trunk lesions that then develop as merging erythematous regions, with shadowy midpoints, saggy blisters with a Nikolsky sign and stripped epidermal sheets [25, 26]. There is mucosal involvement in the majority of cases; 80% of cases have involvement of two or more mucosal surfaces. Dermal hyperpigmentation, scarring, alopecia, and nail dystrophy are commonly observed, too. Ocular problems comprise foreign body feelings, dry eyes, chronic conjunctivitis, trichiasis, and also loss of sight [5].

There is common involvement of the oral cavity in up to 100% of cases, with mucositis and ulceration [27]. Ocular involvement is also frequent with hyperemia of the conjunctiva and total epidermal sloughing at the surface. To avert long-term ocular sequelae, early consultation is necessary [23, 27-29]. Gynecologic involvement is noticed in about 77% of female cases but is variable in severity [27, 30].

Pathophysiology

Although medications mostly trigger SJS/TEN, however, infections (most commonly Mycoplasma pneumonia) have also been related [2, 22, 31-38]. In about 15–30% of cases, no offending cause was recognized [39]. Though the triggers of these ailments have been well-reported, their pathophysiology still requires elucidation. These are thought to be type IV hypersensitivity reactions, intervened by the T-cells. Several hypotheses exist about how medications produce an immunological reaction causing SJS/TEN [4, 40]. The foremost is the hapten theory stating that molecular medications will make a covalent complex with the serum proteins that are identified by specific HLA molecules and offered to T-cells, generating an immune reaction. Another theory is the concept of pharmacological interaction (p-i), stating that chemically inert medications and those that cannot bind covalently with serum proteins, attach directly with HLA molecules, resulting in the activation of the T cells. The last theory is the concept of altered peptide, stating that medications attach inside HLA binding sections in a manner that modifies self-protein presentation to T cells, so that they are not identified as self, resulting in the immune reaction [41, 42]. Regardless of doubt about the particular mechanism, the conclusion is T-cells activation responds to a medication or infection and subsequential epidermal necrosis. Initial theories hypothesized that keratinocyte expiry was interceded by interactions of soluble Fas ligand (sFasL) with the Fas receptor on the keratinocyte surface. Consequent research established granulysin as a noteworthy facilitator of apoptosis [30]. More studies established granulysin's role as a foremost driver of the illness and exhibited that granulysin levels in the sore fluid were linked with the disease seriousness [43, 44]. Granulysin does not act alone though it appears to be the chief propeller of epidermal necrosis [45]. There was a study of 28 diverse cytokines and chemokines regarding their serum levels by Su et al. [45] and it was found that several were upregulated in SJS/TEN cases, of which granulysin and IL-15 were linked with disease seriousness. Furthermore, the character of necroptosis or programmed necrosis was studied and its contribution to keratinocyte death was revealed, that possibly has significant diagnostic inferences [46-48].

Management

Owing to the greater SJS/TEN-related morbidity and mortality, multidisciplinary attention in a dedicated burn division is suggested for such cases. The recommendations of the UK advise transfer of cases to a Burn unit with TEN and confirmation of these signs: clinical decline, further detachment of epidermis, localized sepsis, lesion conversion, and/or late curing. As per UK guidelines for SJS/TEN management, removal of the offending medication and integrative supportive care have to be lined up above systemic therapy due to the paucity of confirmation of the management effectiveness [6, 49]. Yet, Japanese SJS/TEN recommendations suggest the first-line treatment of prompt systemic corticosteroids (alone or combined with cyclosporine) [50].

Offending Drugs: their Identification and Withdrawal

Recognition and removal of the offending medication is the key element for treatment. Offending drugs were recognized in 85% of SJS/TEN patients in a study. In some patients, offending drug identification is challenging, particularly in cases of taking many drugs concomitantly. The ALDEN (Algorithm of Drug causality for Epidermal Necrolysis) algorithm is commonly employed retrospectively (not in the acute stage) for assessing medication causality [39]. Pharmacovigilance statistics have an imperative part in recognizing medications that have a much sturdy link with SJS/TEN [6].

Diagnostic Tests for the Identification of Offending Drugs

Numerous tests are available to identify the offending medication; for the offending medication of cutaneous reactions like acute generalized exanthematous pustulosis (AGEP), medication rashes with eosinophilia and systemic symptoms (DRESS), or maculopapular exanthema, patch testing has been used. Yet, no standard formulations of a testing agent are available. For AGEP,Wolkenstein and co-investigators described 50% affirmative rates, however, a positive test for only 2 cases among the 22 SJS/TEN patients was reported by them [6]. Similar results were reported with the offending medication by a multi-center study, recognized in 64% (46/72) cases with DRESS, 58% (26/45) cases with AGEP, and only 24% (4/17) cases with SJS/TEN [51]. On the contrary, Lin and co-investigators stated confirmatory patch test in 62.5% (n=10 out of 16) cases with CBZ-prompted SJS/TEN, with negative relapse of hypersensitivity response. There had been reports of cross-sensitivity to structure-associated anti-epileptics (i.e. oxcarbazepine, phenytoin, and lamotrigine) in CBZ-prompted SJS/TEN cases. It was proposed that those aromatic anti-epileptic agents should be evaded in such patients. It was concluded that patch testing had extremely variable sensitivity and specificity for various medications [52].

The T cell proliferation responding to medication, in vitro, is measured by LTT (lymphocyte transformation test) and was reported as positive in 21–56% of SJS/TEN cases [53-55]. Researchers altered the LTT to increase specificity by the addition of IL-2, IL-7/IL-15, antigen-presenting cells, or by removing regulatory T cells (CD3+ CD25+) [56]. Cytokine assays are used to evaluate cytokines levels or other arbitrators, developed by lymphocytes secondary to medication provocation. Several cytokines i.e. IFN- γ, IL-2, IL-4, and IL-5 are expressed and then liberated during the delayed hypersensitivity test of medication. Some new researchers have reported that IFN-γ assays and IL-4 assays could ascertain the offending medication in 78% and 50% of SJS/TEN cases, correspondingly [54]. In separate research, the offending medication was recognized in 55%, 43%, and 38% of patients with IFN-γ, IL-5, and IL-2 assays respectively. This research also proposed a combination of different assays as a more practicable method to recognize the offending medication in SJS/TEN cases. At present, cytokine assays are not usually employed in clinical practice to detect the offending medication in SJS/TEN cases [55].

SCORTEN: Severity-of-Illness Score for TEN

This score (SCORTEN) is generally employed for the evaluation and prediction of death owing to SJS/TEN [57]. SCORTEN comprises of 7 individual risk factors, e.g. age (> 40 years), tachycardia (> 120 beats/minute), malignancy, > 10% skin detachment, glucose (> 14 mmol/L), serum urea (> 10 mmol/L) and serum bicarbonate (less than 20 mmol/L). SCORTEN has to be evaluated after admission of the patient within 24 hours and on the 3rd day [6].

Supportive Therapy

The supportive therapy is akin to the therapy of a serious burn case for SJS/TEN cases. It includes protection and restoration of the skin's barrier function, sustaining fluid balance, protection of the respiratory tract, and infection treatment. Fluid and electrolyte observation and replacement are vital along with nutritional support, owing to the high catabolic rate. Also, thermoregulation and sufficient analgesia are generally desirable [27, 58]. Clinical recommendations for the care of skin in SJS/TEN cases are unavailable. Epidermis detachment is proposed as a natural bandage for hastening re-epithelialization [59].

Integrated timely care is necessitated encompassing experts from various disciplines like gynecology, urology, ENT, ophthalmology, etc. to avert the SJS/TEN consequences. Referring to the optometric physician is important as most cases have an ophthalmic association. Management with combative lubrication, topical corticosteroids, topical antibiotics, and breakdown of adhesions for eyes is essential. Lately, amniotic membrane transplants displayed efficacy in the preservation of visual acuity and an intact ocular surface [27]. Since SJS/TEN may have psychological effects, proper knowledge, and emotional support are necessary [60].

In conclusion, timely diagnosis, removal of offending medication, and integrated and supportive care are the most important elements of SJS/TEN therapy. Quite a lot of immunosuppressive and immunomodulating treatments have been suggested, together with cyclosporine, corticosteroids, IVIg, and TNF-α antagonists, based on the existing information about SJS/TEN pathophysiology [6].

Systemic Corticosteroids

Corticosteroids are broadly employed as anti-inflammatory and anti-hypersensitivity drugs. Initially documented therapy for SJS/TEN was systemic corticosteroid and its effectiveness to treat SJS/TEN has long been argued. Earlier research instituted that corticosteroids to treat SJS/TEN cases may elevate infection risk, general complications, and death rate [6]. Multi-center research conducted retrospectively with n = 281 cases in Europe could not establish adequate confirmation of the benefits of corticosteroids [61]. Several meta-analyses could not prove the advantageous effects of systemic corticosteroids to reduce mortality. Yet, meta-analyses performed by Zimmermann and coworkers (2017) and Houschyar and coworkers (2021) proposed that steroids may enhance existence rates [1, 62].

In retrospective research performed by Kardaun and co-investigators in the Netherlands (2007), 12 patients with SJS/TEN were given high-dose of dexamethasone, in the short term, which reduced the mortality rate [63]. The useful effects of pulse methylprednisolone treatment for survival and deterrence of ocular problems have been established by 2 Japanese studies [64, 65]. In SJS/TEN cases, the serum levels of IFN-γ, TNF-α, IL-6, and IL-10 were reduced as reported by Hirahara et al. following 4 days of methylprednisolone pulse treatment, though, a statistically substantial drop was detected only in levels of IFN-γ and IL-6 [65]. In Indian research conducted prospectively, 18 TEN cases were managed with dexamethasone (1 mg/kg/day) intramuscular injection, and all cases were revived [66]. Mieno et al. in a retrospective research of 85 cases, instituted that administration within 4 days from the onset of pulse corticosteroids, may decrease serious ocular consequences [67]. One more retrospective research with 70 SJS/TEN cases also discovered an advantageous outcome of corticosteroids irrespective of the therapeutic plan, whether low-dose (≤2 mg/kg/day) or high-dose (>2 mg/kg/day) [68]. While the advantageous effects of systemic corticosteroids were generally established on outcomes from retrospective/ single-arm research, Japanese recommendations suggest pulse corticosteroid treatment as among the first-line management for SJS/TEN with proper infection control [50]. Systemic corticosteroids can be regarded as life-savers and a cost-effective treatment in resource-restricted circumstances [66].

Intravenous Immunoglobulin (IVIg)

Apart from antibodies having anti-infectious actions, a variety of naturally existing autoantibodies in IVIg may control vital immune activities [69].

In a retrospective study with 12 SJS cases, IVIg treatment for an average of 4 days and a mean daily dose of 0.6 g/kg, prohibited the development of epidermal necrolysis and decreased the period to total mucocutaneous cure [70]. The useful effects of IVIg have been demonstrated by 2 more single-arm and non-randomized research [71]. Yet, in 2008, a retrospective study of 281 cases revealed no noteworthy difference in deaths when corresponding with cases administered with median total dosage (1.9 g/kg IVIg) and those who got supportive management [61]. Several retrospective cohort and case series researches also described that IVIg is ineffective in decreasing deaths or the development of skin sloughing [72-75].

A complete dose of more than 2 g/ kg of body weight of IVIg is regarded as a high-dose plan for SJS/TEN, generally. Retrospective research on 48 TEN cases found that the survival rate was 88% for cases administered with high-dose IVIg [76]. A noteworthy decline in mortality rate by 17%, in a cohort of 16 TEN cases, was also reported by Trent et al., by using high-dose IVIg [77]. Increasing doses of IVIg displayed an inverse correlation with mortality even though a meta-analysis by Barron and coworkers established no useful outcome with a total dose > 2 g/kg of IVIg, in reducing the deaths related to SJS/TEN [78]. On the other hand, two meta-analyses conducted by Huang and co-investigators revealed insignificant survival gain of low-dose/ high-dose IVIg in TEN cases [79]. The varying outcomes could be due to target cells’ sensitivity to Fas (FasL or CD95L or CD178), the IVIg concentration employed, and the relative proportions of agonistic and antagonistic anti-Fas autoantibodies in IVIg formulations [80].

Systemic Corticosteroids and IVIg Combination

In Chinese research done retrospectively, 20 patients administered with a combination of 0.4 g/kg/day IVIg and systemic steroid for 5 days exhibited an insignificant decline in deaths or the period to taper corticosteroids [81]. In one more Chinese retrospective research, 24 cases treated with >2 g/kg IVIg (high-dose), combined with systemic steroids, presented an insignificant decrease in standardized death ratio in comparison to those administered with corticosteroids only [81]. Both of these researches displayed insignificant benefits of combined treatment in decreasing mortality rate. Contrariwise, an Indian, prospective, open-label research with 36 TEN cases employed low-dose (0.2–0.5 g/kg) IVIg and systemic corticosteroids combined, resulting in a considerably lesser standardized death ratio in comparison to the sole corticosteroids [82]. Management with both steroids and IVIg was revealed to decrease deaths in multi-center retrospective research [83].

Ye et al. in 2016 performed a meta-analysis with 26 researchers and revealed that the combined IVIg and corticosteroid noticeably decreased the recovery period but not death. Subgroup investigation among Asian cases also discovered a larger outcome [84]. Moreover, Torres-Navarro et al. in meta-regression analysis revealed that IVIg, combined with corticosteroids, was linked with reduced mortality than anticipated by SCORTEN [85]. The greater significance of combination treatment with IVIg and corticosteroid was also ranked by 2 new network meta-analyses, for decreasing mortality among the existing systemic treatments [86, 87].

Cyclosporine A (CsA)

CsA also has anti-apoptotic actions so, it may hypothetically have an advantage for SJS/TEN cases [88]. No mortality was detected in a non-blinded, prospective, phase II clinical study, among the 29 SJS/TEN cases who were administered with 3 mg/kg/day CsA, but some cases (n = 3) necessitated termination of treatment or (n = 2) dose-tapering owing to some ADRs [89]. In further studies, the favorable effect of CsA was also seen with a first dose of 3–5 mg/kg/day following tapering [90-95].

Several meta-analyses propose advantageous effects of CsA [96, 97]. Some researchers criticized for an element of selection bias that cases with renal insufficiency, uncontrolled diabetes mellitus, arterial hypertension, severe infection, immunodeficiency or malignancy, not included in certain series. Also, the outcomes of a meta-analysis are expected to be influenced by publication biasness [98]. There are alarms relating to the renal and hepatic toxicity of CsA too, as CsA ought to be evaded or employed with attention in cases of already existent renal insufficiency, having creatinine clearance = less than 60 mL/min or uncontrolled levels of diabetes [88].

TNF-Alpha (TNF-α) Inhibitors

TNFα was presented to increase HLA class I expression on keratinocytes, presenting them to be more susceptible to T cell-prompted cytotoxicity [99]. The augmented TNF-α levels have steered the suggestions to use TNF-α inhibitors in SJS/TEN cases [100]. Certain case reports and case series have defined advantageous activities of biological TNF-α inhibitors like etanercept, which is a soluble fusion protein, and infliximab, which is an anti-TNF-α monoclonal antibody [101-103]. Etanercept considerably reduced TNF-α and granulysin levels in sore fluids and plasma and augmented the regulatory T cell populace in the peripheral blood [104].

Combined Biologic Anti-TNF-α and Corticosteroids

Some case reports have defined using anti-TNF-α and corticosteroids in combination [105-110]. Sachdeva, in 2021, performed a systematic review and discovered that biological monotherapy and combination treatment both were linked with enhanced consequences in SJS/TEN [111]. AO and coworkers (2022) employed 25 SJS/TEN cases and reported that combination treatment with etanercept and corticosteroids considerably reduced acute phase period, hospitalization, and skin re-epithelialization as compared to sole corticosteroid therapy. Both therapies declined the serum levels of IL-15 significantly but combined treatment reduced the serum levels of IL-6 and IL-18, too [112]. Zhang and co-investigators conducted a study from Taiwan and China on SJS/TEN cases (n= 242) retrospectively and established that cases that were administered with combined therapy of etanercept and corticosteroids had lesser death rates as compared to corticosteroid only or IVIg combined with corticosteroids [113].

Combined Biologic Anti-TNF-α with Further Therapies

Combination treatment with anti-TNF-α and IVIg has been defined by a few cases reports [114, 115]. Adding etanercept to IVIg along with supportive care was described by Pham & coworkers in a case series of 13 cases, which improved results in comparison to IVIg with supportive care only [116].

Plasmapheresis

Through plasmapheresis, medication, its metabolites, and cytokines are removed from the sufferer. The 3 first-line treatments of choice are followed as per the Japanese recommendations: IVIg, systematic steroids, and plasmapheresis [50]. A prospective observational research enrolling 28 TEN SJS/TEN overlap cases was performed by Han et al. who revealed that plasmapheresis was much better than conventional treatments like IVIg or corticosteroids to reduce the deaths and the length of hospitalization [117].

Giudice et al. (2017), at the burn division, stated the safety and efficacy of plasmapheresis and CsA in combination with TEN therapy with 12 TEN cases [118]. The improved results with a combination of plasmapheresis and IVIg were also reported by Krajewski & coworkers and Lissia & coworkers [119].

CONCLUSION

This review article updates the knowledge about SJS/TEN by discussing genetic contribution, pathophysiology, clinical features, diagnosis and identification of culprit drugs, and pharmacotherapeutic options of SJS/TEN. Apart from genetic modulation, the adverse drug reaction mediated SJS/TEN is also discussed in detail. Evidence showed that antiepileptic drugs, allopurinol, and CYP450 can initiate this cutaneous complication with genetic involvement. However, despite the greater mortality rate, its management process is still not significantly determined. Among the therapeutic options, a dispute still exists for using systemic corticosteroids and IVIg. Nevertheless, positive treatment outcomes have been reported while using biologic anti-TNF-α and cyclosporine. Collation of further data is required to achieve a revolution for developing novel therapeutic agents.

CONFLICT OF INTEREST

There is no conflict of interest.

REFERENCES

  1. Houschyar KS, Tapking C, Borrelli MR, Puladi B, Ooms M, Wallner C, et al. Stevens-Johnson syndrome and toxic epidermal necrolysis: a systematic review and meta-analysis. J Wound Care 2021; 30(12): 1012-9.

    DOI: https://doi.org/10.12968/jowc.2021.30.12.1012 PMID: 34881995

  2. Alajmi A, Jfri A, Gomolin A, Jafarian F. A pediatric case of Stevens-Johnson syndrome/toxic epidermal necrolysis with rapid response to intravenous cyclosporine. JAAD Case Rep 2020; 6(6): 555-7.

    DOI: https://doi.org/10.1016%2Fj.jdcr.2020.04.003 PMID: 32509946

  3. Lerch M, Mainetti C, Terziroli Beretta-Piccoli B, Harr T. Current perspectives on Stevens-Johnson syndrome and toxic epidermal necrolysis. Clin Rev Allergy Immunol 2018; 54(1): 147-76.

    DOI: https://doi.org/10.1007/s12016-017-8654-z PMID: 29188475

  4. Hasegawa A, Abe R. Recent advances in managing and understanding Stevens-Johnson syndrome and toxic epidermal necrolysis. Version 1. F1000Res 2020; 9: F1000 Faculty Rev-612.

    DOI: https://doi.org/10.12688%2Ff1000research.24748.1 PMID: 32595945

  5. Hsu DY, Brieva J, Silverberg NB, Silverberg JI. Morbidity and mortality of Stevens-Johnson syndrome and toxic epidermal necrolysis in United States adults. J Invest Dermatol 2016; 136(7): 1387-97.

    DOI: https://doi.org/10.1016/j.jid.2016.03.023 PMID: 27039263

  6. Chang HC, Wang TJ, Lin MH, Chen TJ. A review of the systemic treatment of Stevens-Johnson syndrome and toxic epidermal necrolysis. Biomedicines 2022; 10(9): 2105.

    DOI: https://doi.org/10.3390/biomedicines10092105 PMID: 36140207

  7. Khor AHP, Lim KS, Tan CT, Kwan Z, Tan WC, Wu DBC, 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(7): 275-8.

    DOI: https://doi.org/10.1097/fpc.0000000000000287 PMID: 28570299

  8. 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(9): 885-7.

    DOI: https://doi.org/10.1007/s00228-011-1009-4 PMID: 21424386

  9. Wang Q, Zhou JQ, Zhou LM, Chen ZY, Fang ZY, Yang LB, 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(6): 446-8.

    DOI: https://doi.org/10.1016/j.seizure.2011.02.003 PMID: 21397523

  10. 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(1): 78-86.

    DOI: https://doi.org/10.1212/wnl.0000000000003453 PMID: 27913699

  11. 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(1): 118-26.

    DOI: https://doi.org/10.3349%2Fymj.2016.57.1.118 PMID: 26632391

  12. 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: 168-78.

    DOI: https://doi.org/10.1016/j.phrs.2016.11.027 PMID: 27888155

  13. 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(5): 1034-41.

    DOI: https://doi.org/10.1093/hmg/ddq537 PMID: 21149285

  14. 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(1-2): 190-7.

    DOI: https://doi.org/10.1016/j.eplepsyres.2011.08.010 PMID: 21917426

  15. Amstutz U, Shear NH, Rieder MJ, Hwang S, Fung V, Nakamura H, et al. Recommendations for HLA‐B* 15: 02 and HLA‐A* 31: 01 genetic testing to reduce the risk of carbamazepine‐induced hypersensitivity reactions. Epilepsia 2014; 55(4): 496-506.

    DOI: https://doi.org/10.1111/epi.12564 PMID: 24597466

  16. Chen P, Lin JJ, Lu C-S, 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(12): 1126-33.

    DOI: https://doi.org/10.1056/nejmoa1009717 PMID: 21428768

  17. Cheng L, Xiong Y, Qin C, Zhang W, Chen X, 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(2): 555-8.

    DOI: https://doi.org/10.1111/bjd.13688 PMID: 26104483

  18. 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: https://doi.org/10.3389/fphar.2016.00186 PMID: 27486401

  19. 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(9): 1057-71.

    DOI: https://doi.org/10.1111/1756-185x.13143 PMID: 28857441

  20. Wu R, Cheng YJ, Zhu LI, 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(49): 81870-9.

    DOI: https://doi.org/10.18632%2Foncotarget.13250 PMID: 27835909

  21. 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(5): 525-34.

    DOI: https://doi.org/10.1001/jama.2014.7859 PMID: 25096692

  22. Charlton OA, Harris V, Phan K, Mewton E, Jackson C, Cooper A. Toxic epidermal necrolysis and Steven-Johnson syndrome: a comprehensive review. Adv Wound Care (New Rochelle) 2020; 9(7): 426-39.

    DOI: https://doi.org/10.1089/wound.2019.0977 PMID: 32520664

  23. Guvenir H, Arikoglu T, Vezir E, Misirlioglu ED. Clinical phenotypes of severe cutaneous drug hypersensitivity reactions. Curr Pharm Des 2019; 25(36): 3840-54.

    DOI: https://doi.org/10.2174/1381612825666191107162921 PMID: 31696807

  24. Paulmann M, Mockenhaupt M. Fever in Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis in Pediatric Cases: Laboratory Work-up and Antibiotic Therapy. Pediatr Infect Dis J 2017; 36(5): 513-5.

    DOI: https://doi.org/10.1097/inf.0000000000001571 PMID: 28403057

  25. Alerhand S, Cassella C, Koyfman A. Stevens-Johnson syndrome and toxic epidermal necrolysis in the pediatric population: a review. Pediatr Emerg Care 2016; 32(7): 472-6.

    DOI: https://doi.org/10.1097/pec.0000000000000840 PMID: 27380605

  26. Grünwald P, Mockenhaupt M, Panzer R, Emmert S. Erythema multiforme, Stevens‐Johnson syndrome/toxic epidermal necrolysis–diagnosis and treatment. J Dtsch Dermatol Ges 2020; 18(6): 547-53.

    DOI: https://doi.org/10.1111/ddg.14118 PMID: 32469468

  27. Shanbhag SS, Chodosh J, Fathy C, Goverman J, Mitchell C, Saeed HN. Multidisciplinary care in Stevens-Johnson syndrome. Therapeutic Advances in Chronic Disease 2020; 11: 2040622319894469.

    DOI:https://doi.org/10.1177%2F2040622319894469 PMID: 32523661

  28. Basu S, Shanbhag SS, Gokani A, Kedar R, Bahuguna C, Sangwan VS. Chronic ocular sequelae of Stevens-Johnson syndrome in children: long-term impact of appropriate therapy on natural history of disease. Am J Ophthalmol 2018; 189: 17-28.

    DOI: https://doi.org/10.1016/j.ajo.2018.01.028 PMID: 29421293

  29. Gregory DG. New grading system and treatment guidelines for the acute ocular manifestations of Stevens-Johnson syndrome. Ophthalmology 2016; 123(8): 1653-8.

    DOI: https://doi.org/10.1016/j.ophtha.2016.04.041 PMID: 27297404

  30. Frantz R, Huang S, Are A, Motaparthi K. Stevens–Johnson Syndrome and Toxic Epidermal Necrolysis: A Review of Diagnosis and Management. Medicina (Kaunas) 2021; 57(9): 895.

    DOI: https://doi.org/10.3390/medicina57090895 PMID: 34577817

  31. Aghdam MK, Ahmadiafshar A, Eftekhari K. Toxic epidermal necrolysis syndrome following single-dose diclofenac suppository, a case report. J Comprehen Pediatr 2020; 11(2): e100496.

    DOI: http://dx.doi.org/10.5812/compreped.100496

  32. Maloney NJ, Ravi V, Cheng K, Bach DQ, Worswick S. Stevens‐Johnson syndrome and toxic epidermal necrolysis‐like reactions to checkpoint inhibitors: a systematic review. Int J Dermatol 2020; 59(6): e183-8.

    DOI: https://doi.org/10.1111/ijd.14811 PMID: 32052409

  33. Simonsen AB, Kaae J, Ellebaek E, Svane IM, Zachariae C. Cutaneous adverse reactions to anti-PD-1 treatment-A systematic review. J Am Acad Dermatol 2020; 83(5): 1415-24.

    DOI: https://doi.org/10.1016/j.jaad.2020.04.058 PMID: 32320766

  34. Madabhavi I, Revannasiddaiah S, Patel A, Anand A. Toxic epidermal necrolysis with the use of tamoxifen. BMJ Case Rep 2015; 2015: bcr2014209102.

    DOI: https://doi.org/10.1136/bcr-2014-209102 PMID: 25878231

  35. Mani R, Monteleone C, Schalock PC, Truong T, Zhang XB, Wagner ML. Rashes and other hypersensitivity reactions associated with antiepileptic drugs: a review of current literature. Seizure 2019; 71: 270-8.

    DOI: https://doi.org/10.1016/j.seizure.2019.07.015 PMID: 31491658

  36. Rashid M, Kashyap A, Undela K. Valproic acid and Stevens‐Johnson syndrome: a systematic review of descriptive studies. Int J Dermatol 2019; 58(9): 1014-22.

    DOI: https://doi.org/10.1111/ijd.14411 PMID: 30809807

  37. Gupta SS, Sabharwal N, Patti R, Kupfer Y. Allopurinol-Induced Stevens - Johnson syndrome. Am J Med Sci 2019; 357(4): 348-51.

    DOI: https://doi.org/10.1016/j.amjms.2018.11.018 PMID: 30638600

  38. Sibbald C, Putterman E, Micheletti R, Treat J, Castelo‐Soccio L. Retrospective review of drug‐induced Stevens‐Johnson syndrome and toxic epidermal necrolysis cases at a pediatric tertiary care institution. Pediatric Dermatol 2020; 37(3): 461-6.

    DOI: https://doi.org/10.1111/pde.14118 PMID: 32058621

  39. 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(1): 60-8.

    DOI: https://doi.org/10.1038/clpt.2009.252 PMID: 20375998

  40. Abe R. Immunological response in Stevens–Johnson syndrome and toxic epidermal necrolysis. J Dermatol 2015; 42(1): 42-8.

    DOI: https://doi.org/10.1111/1346-8138.12674 PMID: 25355273

  41. Adam J, Pichler WJ, Yerly D. Delayed drug hypersensitivity: models of T‐cell stimulation. Br J Clin Pharmacol 2011; 71(5): 701-7.

    DOI: https://doi.org/10.1111%2Fj.1365-2125.2010.03764.x PMID: 21480949

  42. Pichler WJ. Modes of presentation of chemical neoantigens to the immune system. Toxicology 2002; 181-182: 49-54.

    DOI: https://doi.org/10.1016/s0300-483x(02)00254-8 PMID: 12505284

  43. Chen CB, Kuo KL, Wang CW, Lu CW, Hui RCY, Lu KL, et al. Detecting Lesional Granulysin Levels for Rapid Diagnosis of Cytotoxic T lymphocyte–Mediated Bullous Skin Disorders. J Allergy Clin Immunol Pract 2021; 9(3): 1327-37.e3.

    DOI: https://doi.org/10.1016/j.jaip.2020.09.048 PMID: 33039642

  44. 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(2): 452-3.

    DOI: https://doi.org/10.1111/j.1365-2133.2012.10921.x PMID: 22384988

  45. 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(5): 1065-73.

    DOI: https://doi.org/10.1016/j.jid.2016.11.034 PMID: 28011147

  46. Hasegawa A, Shinkuma S, Hayashi R, Hama N, Watanabe H, Kinoshita M, et al. 019 Serum RIP3 level as a severity-predictive marker for Stevens-Johnson syndrome and toxic epidermal necrolysis. J Invest Dermatol 2019; 139(5): S4.

    DOI: http://dx.doi.org/10.1016/j.jid.2019.03.095

  47. 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(5): 1768-71.e7.

    DOI: https://doi.org/10.1016/j.jaip.2020.01.006 PMID: 31954192

  48. 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(245): 245ra95.

    DOI: https://doi.org/10.1126/scitranslmed.3008227 PMID: 25031270

  49. Creamer D, Walsh SA, Dziewulski P, Exton LS, Lee HY, Dart JKG, et al. UK guidelines for the management of Stevens-Johnson syndrome/toxic epidermal necrolysis in adults 2016. Br J Dermatol 2016; 174(6): 1194-227.

    DOI: https://doi.org/10.1111/bjd.14530 PMID: 27317286

  50. Aihara M, Kano Y, IIJIMA M, IKEZAWA Z, SHIOHARA T, MORITA E, et al. Guidelines for the management of Stevens-Johnson syndrome and toxic epidermal necrolysis 2009, established by the Japanese research committee on Severe adverse reaction (J-SCAR) supported by the Ministry of Health, Labour and Welfare of Japan. Japanese J Dermatol 2009; 119(11): 2157-63.https://doi.org/10.14924/dermatol.119.2157
  51. Barbaud A, Collet E, Milpied B, Assier H, Staumont D, Avenel‐Audran M, et al. A multicentre study to determine the value and safety of drug patch tests for the three main classes of severe cutaneous adverse drug reactions. Br J Dermatol 2013; 168(3): 555-62.

    DOI: https://doi.org/10.1111/bjd.12125 PMID: 23136927

  52. Lin YT, Chang YC, Hui RY, Yang CH, Ho HC, Hung SI, et al. A patch testing and cross‐sensitivity study of carbamazepine‐induced severe cutaneous adverse drug reactions. J Eur Acad Dermatol Venereol 2013; 27(3): 356-64.

    DOI: https://doi.org/10.1111/j.1468-3083.2011.04418.x PMID: 22211830

  53. Tang Y, Mockenhaupt M, Henry A, Bounoua M, Naldi L, Le Gouvello S, et al. Poor relevance of a lymphocyte proliferation assay in lamotrigine‐induced Stevens–Johnson syndrome or toxic epidermal necrolysis. Clin Exp Allergy 2012; 42(2): 248-54.

    DOI: https://doi.org/10.1111/j.1365-2222.2011.03875.x PMID: 22092454

  54. Polak M, Belgi G, McGuire C, Pickard C, Healy E, Friedmann P, et al. In vitro diagnostic assays are effective during the acute phase of delayed‐type drug hypersensitivity reactions. Br J Dermatol 2013; 168(3): 539-49.

    DOI: https://doi.org/10.1111/bjd.12109 PMID: 23106791

  55. Porebski G, Pecaric‐Petkovic T, Groux‐Keller M, Bosak M, Kawabata T, Pichler WJ. In vitro drug causality assessment in Stevens-Johnson syndrome-alternatives for lymphocyte transformation test. Clin Exp Allergy 2013; 43(9): 1027-37.

    DOI: https://doi.org/10.1111/cea.12145 PMID: 23957338

  56. Fatangare A, Glässner A, Sachs B, Sickmann A. Future perspectives on in-vitro diagnosis of drug allergy by the lymphocyte transformation test. J Immunol Methods 2021; 495: 113072.

    DOI: https://doi.org/10.1016/j.jim.2021.113072 PMID: 34000289

  57. 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(2): 149-53.

    DOI: https://doi.org/10.1046/j.1523-1747.2000.00061.x PMID: 10951229

  58. Valeyrie-Allanore L, Ingen-Housz-Oro S, Chosidow O, Wolkenstein P. French referral center management of Stevens–Johnson syndrome/toxic epidermal necrolysis. Dermatologica Sinica 2013; 31(4): 191-5.

    DOI: https://doi.org/10.1016/j.dsi.2013.09.008

  59. Mockenhaupt M. The current understanding of Stevens–Johnson syndrome and toxic epidermal necrolysis. Expert Rev Clin Immunol 2011; 7(6): 803-15.

    DOI: https://doi.org/10.1586/eci.11.66 PMID: 22014021

  60. Hoffman M, Chansky PB, Bashyam AR, Boettler MA, Challa N, Dominguez A, et al. Long-term physical and psychological outcomes of Stevens-Johnson syndrome/toxic epidermal necrolysis. JAMA Dermatol 2021; 157(6): 712-5.

    DOI: https://doi.org/10.1001/jamadermatol.2021.1136 PMID: 33950191

  61. Schneck J, Fagot J-P, 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(1): 33-40.

    DOI: https://doi.org/10.1016/j.jaad.2007.08.039 PMID: 17919775

  62. 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(6): 514-22.

    DOI: https://doi.org/10.1001/jamadermatol.2016.5668 PMID: 28329382

  63. Kardaun SH, Jonkman MF. Dexamethasone pulse therapy for Stevens-Johnson syndrome/toxic epidermal necrolysis. Acta Derm Venereol 2007; 87(2): 144-8.

    DOI: https://doi.org/10.2340/00015555-0214 PMID: 17340021

  64. 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(6): 1004-11, 1011.e1.

    DOI: https://doi.org/10.1016/j.ajo.2008.12.040 PMID: 19285657

  65. 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(3): 496-8.

    DOI: https://doi.org/10.1016/j.jaad.2013.04.007 PMID: 23957982

  66. Das S, Roy AK, Biswas I. A six-month prospective study to find out the treatment outcome, prognosis and offending drugs in toxic epidermal necrolysis from an urban institution in Kolkata. Indian J Dermatol 2013; 58(3): 191-3.

    DOI: https://doi.org/10.4103/0019-5154.110826 PMID: 23723468

  67. Mieno H, Ueta M, Kinoshita F, Teramukai S, Kinoshita S, Sotozono C. Corticosteroid pulse therapy for Stevens-Johnson syndrome and toxic epidermal necrolysis patients with acute ocular involvement. Am J Ophthalmol 2021; 231: 194-9.

    DOI: https://doi.org/10.1016/j.ajo.2021.06.015 PMID: 34214456

  68. Liu W, Nie X, Zhang L. A retrospective analysis of Stevens–Johnson syndrome/toxic epidermal necrolysis treated with corticosteroids. Int J Dermatol 2016; 55(12): 1408-13.

    DOI: https://doi.org/10.1111/ijd.13379 PMID: 27652522

  69. Jarczak D, Kluge S, Nierhaus A. Use of intravenous immunoglobulins in sepsis therapy-a clinical view. Int J Mol Sci 2020; 21(15): 5543.

    DOI: https://doi.org/10.3390%2Fijms21155543 PMID: 32756325

  70. 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(1): 96-9.

    DOI: https://doi.org/10.1159/000070957 PMID: 12835566

  71. Al‐Mutairi N, Arun J, Osama NE, Amr Z, Mazen AS, Ibtesam EA, et al. Prospective, noncomparative open study from Kuwait of the role of intravenous immunoglobulin in the treatment of toxic epidermal necrolysis. Int J Dermatol 2004; 43(11): 847-51.

    DOI: https://doi.org/10.1111/j.1365-4632.2004.02048.x PMID: 15533072

  72. Firoz BF, Henning JS, Zarzabal LA, Pollock BH. Toxic epidermal necrolysis: five years of treatment experience from a burn unit. J Am Acad Dermatol 2012; 67(4): 630-5.

    DOI: https://doi.org/10.1016/j.jaad.2011.12.014 PMID: 22285617

  73. Bachot N, Revuz J, Roujeau J-C. 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(1): 33-6.

    DOI: https://doi.org/10.1001/archderm.139.1.33 PMID: 12533161

  74. Wolf R, Davidovici B. Severe cutaneous adverse drug reactions: who should treat, where and how? Facts and controversies. Clin Dermatol 2010; 28(3): 344-8.

    DOI: https://doi.org/10.1016/j.clindermatol.2009.06.020 PMID: 20541690

  75. Mittmann N, Chan B, Knowles S, Cosentino L, Shear N. Intravenous immunoglobulin use in patients with toxic epidermal necrolysis and Stevens-Johnson syndrome. Am J Clin Dermatol 2006; 7(6): 359-68.

    DOI: https://doi.org/10.2165/00128071-200607060-00004 PMID: 17173470

  76. Prins C, Kerdel FA, Padilla RS, Hunziker T, Chimenti S, Viard I, et al. Treatment of toxic epidermal necrolysis with high-dose intravenous immunoglobulins: multicenter retrospective analysis of 48 consecutive cases. Arch Dermatol 2003; 139(1): 26-32.

    DOI: https://doi.org/10.1001/archderm.139.1.26 PMID: 12533160

  77. Trent JT, Kirsner RS, Romanelli P, Kerdel FA. Analysis of intravenous immunoglobulin for the treatment of toxic epidermal necrolysis using SCORTEN: The University of Miami Experience. Arch Dermatol 2003; 139(1): 39-43.

    DOI: https://doi.org/10.1001/archderm.139.1.39 PMID: 12533162

  78. 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(1): 108-15.

    DOI: https://doi.org/10.1111/ijd.12423 PMID: 24697283

  79. 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(2): 424-32.

    DOI: https://doi.org/10.1111/j.1365-2133.2012.10965.x PMID: 22458671

  80. Reipert B, Stellamor M, Poell M, Ilas J, Sasgary M, Reipert S, et al. Variation of anti‐Fas antibodies in different lots of intravenous immunoglobulin. Vox Sang 2008; 94(4): 334-41.

    DOI: https://doi.org/10.1111/j.1423-0410.2008.001036.x PMID: 18266779

  81. Yang Y, Xu J, Li F, Zhu X. Combination therapy of intravenous immunoglobulin and corticosteroid in the treatment of toxic epidermal necrolysis and Stevens–Johnson syndrome: a retrospective comparative study in China. Int J Dermatol 2009; 48(10): 1122-8.

    DOI: https://doi.org/10.1111/j.1365-4632.2009.04166.x PMID: 19775409

  82. Jagadeesan S, Sobhanakumari K, Sadanandan S, Ravindran S, Divakaran M, Skaria L, et al. Low dose intravenous immunoglobulins and steroids in toxic epidermal necrolysis: a prospective comparative open-labelled study of 36 cases. Indian J Dermatol Venereol Leprol 2013; 79(4): 506-11.

    DOI: https://doi.org/10.4103/0378-6323.113080 PMID: 23760320

  83. Micheletti RG, Chiesa-Fuxench Z, Noe MH, Stephen S, Aleshin M, Agarwal A, et al. Stevens-Johnson syndrome/toxic epidermal necrolysis: a multicenter retrospective study of 377 adult patients from the United States. J Invest Dermatol 2018; 138(11): 2315-21.

    DOI: https://doi.org/10.1016/j.jid.2018.04.027 PMID: 29758282

  84. Ye LP, Zhang C, Zhu QX. The effect of intravenous immunoglobulin combined with corticosteroid on the progression of Stevens-Johnson syndrome and toxic epidermal necrolysis: a meta-analysis. PloS one 2016; 11(11): e0167120.

    DOI: https://doi.org/10.1371%2Fjournal.pone.0167120 PMID: 27902746

  85. Torres‐Navarro I, Briz‐Redón Á, Botella‐Estrada R. Systemic therapies for Stevens–Johnson Syndrome and Toxic Epidermal Necrolysis: a SCORTEN‐based systematic review and meta‐analysis. J Eur Acad Dermatol Venereol 2021; 35(1): 159-71.

    DOI: https://doi.org/10.1111/jdv.16685 PMID: 32946187

  86. Patel TK, Patel PB, Thakkar S. Comparison of effectiveness of interventions in reducing mortality in patients of toxic epidermal necrolysis: A network meta-analysis. Indian J Dermatol Venereol Leprol 2021; 87(5): 628-44.

    DOI: https://doi.org/10.25259/ijdvl_605_19 PMID: 33871208

  87. Tsai T-Y, Huang I-H, Chao Y-C, Li H, Hsieh T-S, Wang H-H, et al. Treating toxic epidermal necrolysis with systemic immunomodulating therapies: a systematic review and network meta-analysis. J Am Acad Dermatol 2021; 84(2): 390-7.

    DOI: https://doi.org/10.1016/j.jaad.2020.08.122 PMID: 32898587

  88. Gilbert M, Scherrer LA. Efficacy and safety of cyclosporine in Stevens–Johnson syndrome and toxic epidermal necrolysis. Dermatol Ther 2019; 32(1): e12758.

    DOI: https://doi.org/10.1111/dth.12758 PMID: 30285308

  89. 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(4): 847-53.

    DOI: https://doi.org/10.1111/j.1365-2133.2010.09863.x PMID: 20500799

  90. Singh G, Chatterjee M, Verma R. Cyclosporine in Stevens-Johnson syndrome and toxic epidermal necrolysis and retrospective comparison with systemic corticosteroid. Indian J Dermatol Venereol Leprol 2013; 79(5): 686-92.

    DOI: https://doi.org/10.4103/0378-6323.116738 PMID: 23974585

  91. 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(5): 941-7.

    DOI: https://doi.org/10.1016/j.jaad.2014.07.016 PMID: 25087214

  92. McKenzie E, Owen C, Callen J, editors. The use of cyclosporine for Stevens-Johnson syndrome and toxic epidermal necrolysis: The University of Louisville experience. J Am Acad Dermatol 2016; 74(5) suppl 1: AB175.

    DOI: https://doi.org/10.1016/j.jaad.2016.02.688

  93. Mohanty S, Das A, Ghosh A, Sil A, Gharami R, Bandyopadhyay D, et al. Effectiveness, safety and tolerability of cyclosporine versus supportive treatment in Stevens-Johnson syndrome/toxic epidermal necrolysis: a record-based study. Indian J Dermatol Venereol Leprol 2017; 83(3): 312-6.

    DOI: https://doi.org/10.4103/ijdvl.ijdvl_201_16 PMID: 28366923

  94. 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(1): 106-13.

    DOI: https://doi.org/10.1016/j.jaad.2016.07.048 PMID: 27717620

  95. Gonzalez-Herrada C, Rodriguez-Martin S, Cachafeiro L, Lerma V, Gonzalez 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(10): 2092-100.

    DOI: https://doi.org/10.1016/j.jid.2017.05.022 PMID: 28634032

  96. Ng QX, De Deyn MLZQ, Venkatanarayanan N, Ho CYX, Yeo WS. A meta-analysis of cyclosporine treatment for Stevens–Johnson syndrome/toxic epidermal necrolysis. Journal of inflammation research 2018; 11: 135-42.

    DOI: https://doi.org/10.2147/jir.s160964 PMID: 29636627

  97. Krajewski A, Maciejewska-Markiewicz D, Jakubczyk K, Markowska M, Strużyna J, Mądry R, et al. Impact of multiple medical interventions on mortality, length of hospital stay and reepithelialization time in toxic epidermal necrolysis, Steven-Johnsons syndrome, and TEN/SJS overlap-metanalysis and metaregression of observational studies. Burns 2021; 48(2): 263-80.

    DOI: https://doi.org/10.1016/j.burns.2021.11.004 PMID: 34903405

  98. Noe MH, Mostaghimi A, Rosenbach M, Shinkai K, Micheletti RG. Selective use of cyclosporine for Stevens-Johnson syndrome/toxic epidermal necrolysis may exclude patients with poor prognostic factors. J Invest Dermatol 2018; 138(9): 2068-72.

    DOI: https://doi.org/10.1016/j.jid.2018.03.1496 PMID: 29559345

  99. Kuijper E, French L, Tensen C, Vermeer M, Bouwes Bavinck J. Clinical and pathogenic aspects of the severe cutaneous adverse reaction epidermal necrolysis (EN). J Eur Acad Dermatol Venereol 2020; 34(9): 1957-71.

    DOI: https://doi.org/10.1111%2Fjdv.16339 PMID: 32415695

  100. Caproni M, Torchia D, Schincaglia E, Volpi W, Frezzolini A, Schena D, et al. Expression of cytokines and chemokine receptors in the cutaneous lesions of erythema multiforme and Stevens–Johnson syndrome/toxic epidermal necrolysis. Br J Dermatol 2006; 155(4): 722-8.

    DOI: https://doi.org/10.1111/j.1365-2133.2006.07398.x PMID: 16965421

  101. 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: https://doi.org/10.1155/2012/915314 PMID: 23259092

  102. 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(1): 61-3.PMID: 23653980
  103. Scott‐Lang V, Tidman M, McKay D. Toxic epidermal necrolysis in a child successfully treated with infliximab. Pediatr Dermatol 2014; 31(4): 532-4.

    DOI: https://doi.org/10.1111/pde.12029 PMID: 23072342

  104. 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(3): 985-96.

    DOI: https://doi.org/10.1172/jci93349 PMID: 29400697

  105. Chong I, Chao A. Stevens-Johnson syndrome/toxic epidermal necrolysis and treatment with a biologic: a case report. Perm J 2017; 21: 16-060.

    DOI: https://doi.org/10.7812/tpp/16-060 PMID: 28488978

  106. Vivar KL, Deschaine M, Messina J, Divine JM, Rabionet A, Patel N, et al. Epidermal programmed cell death‐ligand 1 expression in TEN associated with nivolumab therapy. J Cutan Pathol 2017; 44(4): 381-4.

    DOI: https://doi.org/10.1111/cup.12876 PMID: 28000240

  107. Jiang YY, Nguyen GH, Jin HZ, Zeng YP. Methazolamide‐induced toxic epidermal necrolysis in a man carrying HLA‐B* 59: 01: successful treatment with infliximab and glucocorticoid. Int J Dermatol 2018; 57(4): 494-6.

    DOI: https://doi.org/10.1111/ijd.13924 PMID: 29359334

  108. Wang F, Gao X, Chen X, Tang X, Chen H, Han J. Successful treatment of interstitial lung disease related to Stevens–Johnson syndrome/toxic epidermal necrolysis overlap with etanercept: A case report and published work review. J Dermatol 2019; 46(11): 1035-8.

    DOI: https://doi.org/10.1111/1346-8138.15058 PMID: 31436331

  109. Shen MH, Liu MT, Chung WH, Lu CW. Toxic epidermal necrolysis induced by human herpesvirus 7 treated with a tumor necrosis factor‐α inhibitor. J Dermatol 2020; 47(10): 1179-81.

    DOI: https://doi.org/10.1111/1346-8138.15493 PMID: 32686137

  110. Estébanez A, Sáez‐Martín LC, Muñoz JI, Silva E, Monrabal A, Monteagudo C, et al. Levetiracetam‐induced pediatric toxic epidermal necrolysis successfully treated with etanercept. Pediatr Dermatol 2020; 37(4): 701-5.

    DOI: https://doi.org/10.1111/pde.14179 PMID: 32319121

  111. Sachdeva M, Maliyar K, Ponzo MG. A Systematic Review of Efficacy and Safety of Monotherapy and Combination Therapy with Biologic for Stevens - Johnson syndrome and Toxic Epidermal Necrolysis. J Cutan Med Surg 2021; 25(6): 598-615.

    DOI: https://doi.org/10.1177/1203475421993779 PMID: 33631950

  112. Ao S, Gao X, Zhan J, Ai L, Li M, Su H, et al. Inhibition of tumor necrosis factor improves conventional steroid therapy for Stevens-Johnson syndrome/toxic epidermal necrolysis in a cohort of patients. J Am Acad Dermatol 2022; 86(6): 1236-45.

    DOI: https://doi.org/10.1016/j.jaad.2022.01.039 PMID: 35121075

  113. 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(5): 1295-304.e6.

    DOI: https://doi.org/10.1016/j.jaip.2022.01.038 PMID: 35131514

  114. Wang R, Zhong S, Tu P, Li R, Wang M. Rapid remission of Stevens-Johnson syndrome by combination therapy using etanercept and intravenous immunoglobulin and a review of the literature. Dermatol Ther 2019; 32(4): e12832.

    DOI: https://doi.org/10.1111/dth.12832 PMID: 30659711

  115. Kreft B, Lieser U, Haase R, Marsch WC, Wohlrab J. Extensive hypertrophic scarring after toxic epidermal necrolysis in a child. Pediatr Dermatol 2014; 31(4): 527-8.

    DOI: https://doi.org/10.1111/j.1525-1470.2012.01863.x PMID: 23025572

  116. Pham CH, Gillenwater TJ, Nagengast E, McCullough MC, Peng DH, Garner WL. Combination therapy: Etanercept and intravenous immunoglobulin for the acute treatment of Stevens-Johnson syndrome/toxic epidermal necrolysis. Burns 2019; 45(7): 1634-8.

    DOI: https://doi.org/10.1016/j.burns.2018.12.018 PMID: 31466921

  117. 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: 65-8.

    DOI: https://doi.org/10.1016/j.jcrc.2017.07.002 PMID: 28688239

  118. Giudice G, Maggio G, Bufano L, Memeo G, Vestita M. Management of toxic epidermal necrolysis with plasmapheresis and cyclosporine A: our 10 years’ experience. Plast Reconstr Surg Glob Open 2017; 5(2): e1221.

    DOI: https://doi.org/10.1097%2FGOX.0000000000001221 PMID: 28280663

  119. Krajewski A, Mazurek MJ, Mlynska-Krajewska E, Piorun K, Knakiewicz M, Markowska M. Toxic epidermal necrolysis therapy with TPE and IVIG-10 years of experience of the burns treatment center. J Burn Care Res 2019; 40(5): 652-7.

    DOI: https://doi.org/10.1093/jbcr/irz073 PMID: 31032516