ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Stevens-Johnson syndrome and toxic epidermal necrolysis: Management, prognosis, and long-term sequelae

Stevens-Johnson syndrome and toxic epidermal necrolysis: Management, prognosis, and long-term sequelae
Literature review current through: Jan 2024.
This topic last updated: Nov 14, 2023.

INTRODUCTION — Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are severe, mucocutaneous, adverse reactions most commonly induced by medications. Clinical features include fever, prodromal symptoms, mucositis, and extensive epidermal detachment. SJS and TEN are considered a disease continuum and are collectively known as epidermal necrolysis.

The classification of the disease is based on the total body surface area (BSA) involved with skin detachment [1]:

SJS is defined as skin detachment affecting <10 percent of the BSA (picture 1A-C).

TEN is defined as skin detachment affecting >30 percent of the BSA (picture 2A-D).

SJS/TEN overlap is defined as skin detachment involving 10 to 30 percent of the BSA.

We will use the term "Stevens-Johnson syndrome/toxic epidermal necrolysis" to refer collectively to SJS, TEN, and SJS/TEN overlap.

The management, prognosis, and long-term sequelae of SJS/TEN are discussed in this topic. The pathogenesis, clinical manifestations, and diagnosis of SJS/TEN are discussed separately. (See "Stevens-Johnson syndrome and toxic epidermal necrolysis: Pathogenesis, clinical manifestations, and diagnosis".)

GENERAL PRINCIPLES — The management of SJS/TEN can be broadly classified into two phases [2]:

Acute phase – During the acute phase, progressive worsening of the cutaneous detachment and mucositis occur for five to seven days, followed by disease arrest and re-epithelization. Due to extensive skin detachment during this phase, the patient is prone to fluid and electrolyte imbalances, sepsis, organ decompensation, and death, with mortality rates of 10 to 50 percent depending on the extent and severity of disease and the patient's age and comorbidities [3,4]. Management during this phase is focused on supportive care and prevention of short- and long-term complications.

Chronic phase – This phase occurs during the convalescent and recovery stage of SJS/TEN. Physical and psychologic sequelae may occur [5]. Management principles include the screening and treatment of these complications in order to maintain life quality.

INITIAL IN-HOSPITAL EVALUATION — Patients suspected of having SJS/TEN require immediate in-hospital evaluation. As soon as the diagnosis of SJS/TEN has been established, the severity and prognosis of the disease should be rapidly determined to define the appropriate medical setting for management [2,6]. (See "Stevens-Johnson syndrome and toxic epidermal necrolysis: Pathogenesis, clinical manifestations, and diagnosis", section on 'Diagnosis'.)

Prompt identification and withdrawal of the causative drug — Prompt identification and withdrawal of the offending agent may improve the prognosis. In a 10-year, observational study of 113 patients with TEN or SJS, early withdrawal of the causative drugs with short half-lives reduced the risk of death by 30 percent for each day before the development of blisters and erosions (odds ratio [OR] 0.69, 95% CI 0.53-0.89) [7]. However, in drugs with long half-lives, there was an increased risk of death, independent of early or late withdrawal (OR 4.9, 95% CI 1.3-18.9). This observed difference between short and long half-life medications may be due to the persistence of the causative drug or substrate despite withdrawal. (See "Stevens-Johnson syndrome and toxic epidermal necrolysis: Pathogenesis, clinical manifestations, and diagnosis", section on 'Drug causality assessment'.)

Causality attribution is generally determined by two principles:

Temporal sequence – Most causative drugs are started 5 to 28 days (occasionally up to two months) prior to the onset of symptoms [8].

Drug notoriety – The majority of SJS/TEN cases are caused by a few high-risk medications (table 1) [9-11]. An algorithm for determining drug causality has been formulated (algorithm of drug causality for epidermal necrolysis [ALDEN]) and is useful for the determination of the culprit drug (table 2) [8].

Assessment of the extent of skin detachment — The Lund-Browder chart (figure 1) is commonly used for a rapid estimation of the percentage of the total body surface area (BSA) involved in adults and children. In adult patients, the "rule of nines" can be alternatively used:

The head represents 9 percent total BSA.

Each arm represents 9 percent total BSA.

Each leg represents 18 percent total BSA.

The anterior and posterior trunk each represent 18 percent total BSA.

Assessment of prognosis — The prognosis of individual patients can be rapidly evaluated on admission by applying prognostic scoring systems. Two scores have been proposed:

SCORTEN score – The severity of illness score of toxic epidermal necrolysis (SCORTEN) is based upon seven independent clinical and laboratory variables (table 3) [12]. The extent of the cutaneous score has been validated for use on days 1 and 3 of hospitalization [13]. The survival curves based upon the SCORTEN score at admission (figure 2) may be helpful when discussing a patient's prognosis [13]. However, due to progress in the management of patients with SJS/TEN over time, particularly in specialized centers, the SCORTEN score may overestimate the mortality risk in these settings [14,15].

ABCD-10 – A prognostic scoring system consisting of five variables (age, serum bicarbonate level, cancer, dialysis, involvement of >10 percent BSA [ABCD-10]) has been proposed based on data from a multicenter, United States cohort (n = 370) of adult patients with SJS/TEN admitted between 2000 and 2015 [16]. One point is given each for age 50 years and older, epidermal detachment >10 percent BSA, and serum bicarbonate <20 mmol/L. Two points are given for presence of active cancer, and three points are given for dialysis prior to admission. This translated to a mortality risk of 2.3, 5.4, 12.3, 25.5, 45.7, 67.4, and 83.6 percent for scores of 0, 1, 2, 3, 4, 5, and ≥6, respectively.

Comparisons between the two prognostic scores suggest that the performance of SCORTEN remains superior and should be considered as the default prognostic tool [15,17].

A study evaluating the association between inflammatory markers and in-hospital mortality in 192 patients with SJS/TEN showed that red cell distribution width to hemoglobin ratio (RDW/Hb) had a similar predictive accuracy as SCORTEN [18]. Furthermore, when incorporated into SCORTEN (revision of SCORTEN [Re-SCORTEN]), the prognostic ability was further augmented. In the Re-SCORTEN prognostic score, an additional two points are added to SCORTEN if the RDW/Hb is 1.2 or higher. This translates to a mortality risk of 1.9, 4.3, 8.7, 14, 23, 35.3, 54.5, 73.2, and 86.1 percent for scores of 0, 1, 2, 3, 4, 5, 6, 7, and ≥8, respectively.

Transfer to a referral center — Patients with BSA detachment of ≥10 percent, rapidly progressive disease, or worsening biochemical/organ function should be transferred and managed in specialized centers, such as specialized dermatology units, burn centers, or intensive care units [2,6]. Several studies indicate that prognosis is better for patients promptly transferred to a reference center or burn care unit. It has also been shown that survival is increased for patients treated in reference centers with a high volume of admissions for SJS/TEN.

In a retrospective, multicenter review of 199 patients with TEN treated at burn care centers in the United States, the overall mortality was 32 percent compared with 51 percent among patients initially cared for in other settings and transferred to a burn center more than one week after disease onset [19].

In an analysis of the French nationwide hospital discharge database that identified 991 patients with SJS/TEN, the OR of mortality was 0.5 (95% CI 0.3-1.0) for one supplemental admission to centers with experience in treating TEN compared with centers without experience, even after adjusting for individual risk factors [20].

In a retrospective cohort study, late admission (>5 days after skin detachment) was associated with increased risk of death (OR 2.9, 95% CI 1.0-8.3), bacteremia (OR 3.7, 95% CI 1.2-11.2), and intensive care unit admission (OR 3.3, 95% CI 1.1-9.8) compared with early admission (within four days of skin detachment) [21].

ACUTE MANAGEMENT — The management of SJS/TEN is multidisciplinary and should be driven by a team of clinicians experienced in treating this disease [2,6,22-24].

Supportive care — Supportive care is the cornerstone of management and includes wound care, fluid and electrolyte management, nutritional support, temperature management, pain control, infection prevention and management, ocular care, and organ support, if needed [2,6,23-25]. Mucosal surfaces of the eyes, mouth, and urogenital tract are frequently affected, and multidisciplinary care is recommended [2,6].

Wound care — The ideal wound management strategy in SJS/TEN has not been determined. Wound care for patients with SJS/TEN varies among centers and can be broadly divided into surgical and conservative approaches [26-29]:

Surgical approach – In the surgical approach, the devitalized epidermis is removed via operative debridement, manual scrubbing, or the use of whirlpools [30]. In some centers, biologic membranes (eg, allografts, xenografts, biosynthetic dressings) are applied to the dermis following debridement [29].

Conservative approach – In the conservative approach or "antishear" approach [31], the detached/detachable epidermis is left in situ as a biologic dressing, and additional nonadhesive dressings may be used to promote healing. Gentle cleansing of the skin with sterile water or diluted chlorhexidine should be performed at each wound change. Large bullae may be ruptured with a sterile needle and drained without deroofing the bullae. Petrolatum-impregnated gauze is traditionally used as the primary dressing in many centers. Modern dressings (eg, nonadherent nanocrystalline gauze materials containing silver and biosynthetic dressings) are increasingly used. They may be left in situ for up to seven days, decreasing the frequency of painful dressing changes. However, the use of modern dressings does not seem to impact the healing time [32-34]. Air-fluidized beds are also useful to reduce pressure on the skin and prevent shearing of the skin [2].

Surgical and conservative approaches have not been evaluated in randomized trials. Data from observational studies suggest that they result in comparable outcomes (eg, survival, time to re-epithelization) [31,35].

Fluid and temperature management — Fluid and electrolyte imbalances occur secondarily to increased water loss from the denuded dermis and decreased oral intake in patients with extensive oral mucosal involvement. Strict intake/output monitoring should be instituted. Fluid replacement is essential to prevent end-organ hypoperfusion and shock. Replacement volumes are approximately one-third lower than those needed for burn victims [2,36]. In the first 24 hours, fluid requirements are estimated to be 2 mL/kg/body surface area (BSA) percent affected. Subsequently, fluid should be titrated according to the patient's response and urine output, aiming for urine volume of 0.5 to 1 mL/kg/hour [37].

Room temperature should be maintained between 82.4 to 89.6°F (28 to 32°C) to prevent excessive caloric expenditures due to epidermal loss and to prevent hypothermia [2,23,38]. Heated-air body warmers may also be used.

Nutrition — Nutritional supplementation should be initiated early to support metabolic disturbances and promote healing. The estimated caloric requirement is 20 to 25 kcal/kg per day during the early, catabolic phase of SJS/TEN and 25 to 30 kcal/kg per day during the anabolic, recovery phase of the disease [6].

Oral feeding may be difficult if the oral mucosal involvement is extensive. If required, nasogastric feeding should be initiated as soon as possible and continued throughout the acute phase of SJS/TEN [2,6,23,24]. The passage of a nasogastric tube should be carefully performed to minimize damage to affected mucous membranes.

Pain control — SJS/TEN is an extremely painful condition, and the pain is exacerbated by mobilization and wound care procedures. Every effort should be taken to optimize pain control. (See "Pain control in the critically ill adult patient" and "Paradigm-based treatment approaches for management of burn pain".)

The intensity of pain can be graded by asking the patient to describe the level of pain on a numerical scale of increasing severity (ranging from 0 to 10 (figure 3)). This evaluation may be repeated every four hours during the acute phase. The choice of analgesia is dependent on the level of pain, and the aim of treatment is to keep the level of pain to ≤2 [39]. Additional doses of analgesia may be required during patient mobilization or wound care procedures.

Mild pain (intensity <4) – Nonopioid analgesics (eg, acetaminophen [paracetamol], ibuprofen).

Moderate/severe pain (intensity ≥4) – Opioids are the drug of choice due to their potency, rapid onset of action, and dose-dependent degree of sedation. These can be delivered enterally, subcutaneously, via patient-controlled analgesia, or via intravenous infusions. For severe pain, the optimal route of administration is intravenous, which provides faster pain relief and can be titrated to meet the individual needs of the patient [2,39].

Prevention and treatment of infections — Patients with SJS/TEN are at high risk of infection. Up to 30 to 50 percent of patients with SJS/TEN develop bacteremia, and sepsis is the most common cause of death [40-42]. BSA involvement of ≥10 percent, hemoglobin levels ≤10 g/dL, and existing cardiovascular disease are associated with increased risk of developing bacteremia [41].

Prevention, active clinical surveillance, and prompt treatment of infections are key components of SJS/TEN management. Prophylactic systemic antibiotics are not recommended [2,6,41]. (See "Nosocomial infections in the intensive care unit: Epidemiology and prevention".)

Prevention – Prevention of infection involves the following:

Sterile handling – Sterile handling is essential. In specialized centers, patients are nursed in reverse isolation procedure rooms [2,6].

Skin antisepsis – Antiseptic solutions containing octenidine, polyhexanide, or chlorhexidine or silver nitrate preparations may be used for disinfection. Silver sulfadiazine should be avoided if SJS/TEN is suspected to be caused by sulfonamides, but silver nitrate and silver-imbued nanocrystalline gauze materials may be used safely. (See 'Wound care' above.)

Repeated bacterial cultures – Repeated cultures of the skin, as well as blood, catheters, gastric, and urinary tubes, should be obtained at regular intervals (eg, every 48 hours) to allow early detection of infections [41,42]. Positive skin cultures may predict blood culture positivity from the same organisms. In a French cohort of 98 patients with SJS/TEN involving ≥10 percent of the BSA, the positive and negative predictive values of skin cultures growing Staphylococcus aureus or Pseudomonas aeruginosa were 58 and 89 percent and 50 and 81 percent, respectively [43].

Surveillance for clinical signs of sepsis – Signs of bacteremia include hypothermia, confusion, hypotension, and reduced urinary output [41,42]. Hypothermia and elevated procalcitonin ≥1 mcg at the time of blood culture have been shown to be predictive of the blood culture positivity [41]. Traditional markers of sepsis (eg, fever, elevated white cell count, erythrocyte sedimentation rates, and C-reactive proteins) are not useful in SJS/TEN [41,42].

Treatment – Antibiotic choice should be directed by culture results whenever possible [41]. Commonly isolated pathogens include S. aureus and P. aeruginosa [42]. However, other bacteria species, including Enterobacteriaceae, have also been isolated, suggesting that bacteremia occurs both through the skin as well as gut translocation [42]. In patients treated in burn centers and intensive care units, local nosocomial microbiota may additionally influence culture results [41]. (See "Clinical approach to Staphylococcus aureus bacteremia in adults" and "Principles of antimicrobial therapy of Pseudomonas aeruginosa infections" and "Gram-negative bacillary bacteremia in adults".)

Management of acute respiratory involvement — Up to 40 percent of patients with SJS/TEN may develop acute respiratory complications [44]. These include specific bronchial erosions as well as nonspecific pulmonary manifestations (eg, pneumonia, pulmonary edema, atelectasis). Twenty-five to 38 percent of patients may develop acute respiratory failure requiring ventilatory support [44-46]. Among intubated patients, the mortality rate increases up to over 50 percent [45]. Risk factors for mechanical ventilation include serum bicarbonate <20 mmol/L, serum urea >10 mmol/L, baseline detached BSA >10 percent, white blood cell count >12,000/mm3, hemoglobin <8 g/dL, bacteremia, and shock/organ failure on admission [45-47].

All patients need to be closely monitored for respiratory involvement during the acute phase and for referral to a specialized intensive care unit. Baseline evaluation should include chest radiographs and arterial blood gases. Features suggestive of pulmonary involvement include cough, dyspnea, hypoxemia, or radiologic abnormalities [44].

Bronchoscopy may be warranted to confirm bronchial involvement, evaluate for the presence of pulmonary infection, and prevent atelectasis and airway obstruction through the mechanical removal of sloughed bronchial epithelium [44].

Noninvasive ventilation is contraindicated because of the skin lesions and the risk of obstruction due to laryngeal involvement. (See "Noninvasive ventilation in adults with acute respiratory failure: Benefits and contraindications".)

Oral intubation may be challenging due to the oral/pharyngeal mucosal involvement. Patients needing mechanical ventilation should be referred to an intensive care unit [2].

Management of urogenital involvement — Urogenital involvement occurs in up to 70 percent of patients with SJS/TEN [48]. It may present acutely as erosions of the scrotum/labia and penis/vulva and may result in dysuria and urinary retention. Early urogenital/gynecologic examination should be part of a baseline assessment [49].

Nonadhesive dressings should be applied to urogenital erosions to reduce pain and prevent adhesions. Moderate-potency topical corticosteroids (group 4 (table 4)) can be applied daily until resolution of the acute phase of illness [49,50]. Estrogen cream and barrier creams (eg, petrolatum, zinc oxide) may also promote re-epithelization [49-51]. The use of viscous lidocaine may also reduce mucosal pain [51].

In female patients, additional local vulvar care includes gentle water washes or sitz baths. If vaginal involvement is suspected, an intravaginal potent corticosteroid ointment should be applied, and silicone vaginal dilators can be used to prevent strictures and adhesions [49]. These should be placed prophylactically during the acute phase of the illness and used regularly until complete healing of the erosions has occurred. In severe vaginal involvement, a vagina estrogen ring may be helpful, as it separates the anterior and posterior vaginal walls while providing low-dose estrogen to aid in epithelial healing. Menstrual suppression during the acute phase of illness in female patients of reproductive age is recommended to reduce the risk of vaginal adenosis and endometriosis [49,50]. To prevent periurethral stricture, patients with vulvovaginal involvement should receive a urinary catheter.

In uncircumcised male patients, daily retraction of the foreskin can prevent preputial adhesions [2].

Management of ocular involvement — Acute ocular involvement affects 70 to 90 percent of patients [52-54]. The severity of the acute ocular involvement does not correlate with the score of toxic epidermal necrolysis (SCORTEN) or BSA involved. The development of chronic ocular sequelae is unpredictable. In a French cohort of 159 patients, the severity of the acute ocular involvement was the strongest predictor for chronic ocular sequelae [52]. Thus, effective management of the acute ocular involvement may mitigate and prevent long-term ocular sequelae.

Evaluation — Repeated ophthalmic evaluation of all patients with suspected SJS/TEN should be performed even if ocular involvement is not visible at the onset of the disease, as it may follow cutaneous involvement. Examination should be performed daily until the ocular condition stabilizes [24].

The entire ocular surface should be carefully examined. Examination should include fluorescein staining to document the presence of membranes and loss of surface epithelium. The severity of ocular lesions can be graded using a simple score [55]:

0 (none) – No ocular involvement

1 (mild) – Conjunctival hyperemia

2 (severe) – Either ocular surface epithelial defect or pseudomembrane formation

3 (very severe) – Both ocular surface epithelial defect and pseudomembrane formation

Ophthalmic therapy — The principles of ophthalmic care are to reduce destructive inflammation at the ocular surface and lid margin; prevention of conjunctival adhesions; infection prophylaxis; and prompt identification and management of blinding complications of corneal exposure, ulceration, and infection [6].

Saline rinsesSaline rinses can be used to clean the eyes and eyelids as well as to remove mucous and inflammatory debris. Local ocular therapy should be started based upon the severity of eye involvement [56].

Lubricants – For patients with no apparent eye involvement (grade 0), multiple daily lubrication with preservative-free eye drops (artificial tears) or ointments is indicated.

Topical corticosteroids/antibiotics – For patients with conjunctival hyperemia (grade 1), ophthalmic preparations containing topical corticosteroids and broad-spectrum antibiotics (eg, moxifloxacin hydrochloride 0.5%) should be applied four to six times per day along with lubricants.

Adhesion separation – Conjunctival adhesions should be separated daily. Forceps, squint hooks, or scissors can be used.

Amniotic membrane transplantation – In addition to topical antibiotics, corticosteroids, and lubricants, amniotic membrane transplantation (AMT) should be considered for patients with extensive sloughing of the bulbar conjunctiva and/or pseudomembrane formation (grades 2 and 3) [57]. Amniotic membrane sheets can be applied to the ocular surface without sutures using a symblepharon ring, a technique that avoids general anesthesia [58]. Cryopreserved amniotic membrane is commercially available in the United States and Canada. In the United Kingdom, it can be obtained from the National Health Service (NHS) Blood and Transplant tissue services.

The use of AMT for the treatment of ocular involvement is supported by several case series and one randomized trial [57,59-61]. In this trial including 25 patients (50 eyes), the eyes were randomized to receive AMT or standard medical therapy [59]. The main endpoints were the maintenance of the best corrected visual acuity (BCVA) and stable ocular surface (eg, absence of corneal haze, limbal stem cell deficiency, symblepharon, ankyloblepharon, or lid-related complications). After six months, the eyes treated with AMT had better BCVA, tear film break-up time, and Schirmer test results than eyes treated with medical therapy. Conjunctival hyperemia persisted in 4 percent of the eyes treated with AMT versus 44 percent of those treated with medical therapy. Corneal haze, corneal vascularization and conjunctivalization, and symblepharon occurred in 44, 24, and 16 percent, respectively, of the eyes treated with medical therapy but in none of the eyes treated with AMT.

Role of systemic therapy — There is scarce and conflicting evidence on the role of systemic adjunctive therapies in halting the ocular damage and improving the visual outcome.

In a Korean, retrospective study, 43 patients with SJS/TEN were allocated into five treatment groups: systemic corticosteroids, intravenous immune globulin (IVIG), systemic steroids and immunoglobulins, pulse corticosteroids, and supportive care only. There were no benefits of systemic immunomodulatory treatments in ocular outcomes measured, such as BCVA and chronic ocular surface complications [62].

In a retrospective, historically controlled study, 10 patients treated with IVIG (2 g/kg) were compared with 18 historical controls (5 received prednisolone). Outcomes of ocular complications and visual acuity six weeks post-IVIG were similar in both groups [63].

In another study, five patients with SJS or TEN with ocular complications during the acute stage were treated within the first four days from disease onset with intravenous pulses of methylprednisolone 500 or 1000 mg per day for three to four days along with 0.1% betamethasone solution, 0.1% betamethasone eye ointment, or both five to eight times daily [64]. At one year, the BCVA was 20/20 or better in all eyes. Epithelial defect, loss of limbal stem cells, conjunctivalization, neovascularization, opacification, or keratinization were absent in all eyes. Only mild, superficial, punctate keratopathy was noted in five eyes.

A retrospective study of 85 patients with SJS/TEN and severe ocular involvement found that corticosteroid pulse therapy given within four days of disease onset (n = 36) was more effective in preventing long-term ocular sequelae than corticosteroid pulse therapy given after five days of disease onset, corticosteroids given as nonpulse therapy, or no corticosteroids (n = 49) [65]. A BCVA in the worse eye of ≥1 was found in 52.8 percent of the eyes in the corticosteroid pulse therapy group compared with 14.3 percent in the other group. In addition, there was a reduction in chronic sequelae affecting the cornea, conjunctiva, and eyelid.

In a small study, seven patients who received systemic cyclosporine at a median dose of 2.5 mg/kg/day over 12±8 days were compared with a control group of seven patients who did not receive cyclosporine. Visual outcomes of BCVA and a chronic ocular surface complication score were not significantly different between the eyes of patients who received cyclosporine versus those who did not receive cyclosporine [66].

Additional supportive measures — Similar to other patients in intensive care units, patients with SJS/TEN are at risk of additional complications, including stress-related peptic ulcer disease; disseminated intravascular coagulation, particularly in those with sepsis; venous thromboembolism in those who are immobile; hyper- or hypoglycemia; and neutropenia.

Stress ulcer prophylaxis – Stress ulcer prophylaxis with proton pump inhibitor may be necessary in those who are unable to feed enterally [6]. (See "Stress ulcers in the intensive care unit: Diagnosis, management, and prevention".)

Disseminated intravascular coagulation – In patients with disseminated intravascular coagulation and active bleeding, blood component replacement (eg, cryoprecipitate, fresh frozen plasma, platelets, red blood cells) may be necessary [67,68]. (See "Evaluation and management of disseminated intravascular coagulation (DIC) in adults".)

Prevention of deep vein thrombosis – Deep vein thrombosis prevention is encouraged with compression stockings, calf pumps, and anticoagulation, if there are no contraindications [6]. (See "Overview of the treatment of proximal and distal lower extremity deep vein thrombosis (DVT)".)

Hyperglycemia – Glycemic levels should be monitored frequently and managed according to intensive care unit principles. (See "Glycemic control in critically ill adult and pediatric patients".)

Neutropenia – Neutropenia is known to occur during the acute phase of the disease. The use of granulocyte colony-stimulating factor (G-CSF) has been advocated by some, as it may mitigate the risk of infection as well as aid in wound re-epithelization [69]. (See "Management of the adult with non-chemotherapy-induced neutropenia".)

ADJUNCTIVE PHARMACOLOGIC THERAPIES

General considerations — There is no established pharmacologic treatment for SJS/TEN. A variety of systemic immunosuppressive or immunomodulating agents, such as systemic corticosteroids, intravenous immune globulin (IVIG) [70], cyclosporine [71], plasmapheresis [72], and anti-tumor necrosis factor (TNF) agents [73], have been utilized for the treatment of SJS/TEN.

The evidence of benefit for such interventions is limited, and none of them can be conclusively recommended. However, the results of several meta-analyses suggest a potential benefit for (see 'Evidence overview' below):

Cyclosporine

Etanercept

Systemic corticosteroids

Combination of IVIG and systemic corticosteroids

When considering the use of any of these agents, it is important to remember that the progression of SJS/TEN in the acute phase is self-limiting, lasting approximately eight days from the onset of symptoms to maximal skin detachment [3]. Thus, the initiation of any immunomodulatory agent outside this window is unlikely to modify disease progression.

Evidence overview — Only two randomized trials have evaluated treatments for SJS/TEN [73,74].

A 1998 trial comparing thalidomide with supportive care was prematurely terminated due to excess death in the thalidomide group [74]. Ten of 12 patients who received thalidomide 400 mg daily for five days died compared with 3 of 10 patients in the placebo group. (See 'Harmful agents' below.)

A 2018 randomized trial that included 93 patients compared etanercept 25 or 50 mg twice a week with prednisolone 1 to 1.5 mg/kg per day [73]. There was no statistically significant difference in mortality between the corticosteroid group and the etanercept group (16.3 versus 8.3 percent), although mortality in both groups was lower than that predicted by the score of toxic epidermal necrolysis (SCORTEN; 20.3 and 17.7 percent for the corticosteroid and etanercept groups, respectively). (See 'Tumor necrosis factor inhibitors' below.)

Evidence on the efficacy of immunomodulatory treatments is largely derived from observational studies and has been summarized in several meta-analyses [75-79].

A 2017 meta-analysis of 27 studies (1209 patients) found that systemic corticosteroids and cyclosporine, in addition to supportive treatment, were associated with a decreased risk of death compared with supportive treatment alone (odds ratio [OR] 0.67, 95% CI 0.46-0.97 and OR 0.1, 95% CI 0.0-0.4, respectively). Treatment with IVIG did not improve survival [78].

A 2021 meta-analysis of data from 38 studies published between 2000 and 2019 evaluating the efficacy of systemic corticosteroids, supportive care, IVIG plus systemic corticosteroids, cyclosporine, and etanercept in reducing mortality as predicted by SCORTEN found that only cyclosporine (standardized mortality rate [SMR] -0.88, 95% CI -1.47 to 0.29), IVIG plus corticosteroids (SMR -0.56, 95% CI -0.94 to -0.19), and etanercept (SMR -0.95, 95% CI -1.82 to -0.07) were found to have survival benefits [75].

In a 2021 network meta-analysis of 67 studies (2079 patients), no treatment was superior to supportive care in reducing the mortality rate. However, when considering the mortality rates predicted by SCORTEN, combination therapy with IVIG and corticosteroids was the only treatment associated with survival benefits (SCORTEN-based SMR 0.53, 95% CI 0.31-0.93) [76].

In another 2021 meta-analysis (24 studies, 979 patients), direct pairwise comparison showed that cyclosporine was associated with a lower mortality risk compared with supportive care (OR 0.32, 95% CI 0.13-0.82) and IVIG (OR 0.08, 95% CI 0.01-0.54). Systemic corticosteroids plus IVIG was also associated with a reduction in mortality compared with IVIG alone (OR 0.16, 95% CI 0.04-0.61). On network analysis, the risk of death was reduced for cyclosporine compared with supportive care (OR 0.19, 95% CI 0.05-0.59). Hierarchy ranking of treatments showed that cyclosporine was the most effective treatment, followed by corticosteroids plus IVIG and etanercept [77].

A 2022 Cochrane review that included nine studies (308 patients) concluded that treatment with etanercept may result in a reduction in mortality compared with systemic corticosteroids, although evidence was of low certainty. Evidence of benefit was very uncertain for corticosteroids versus no corticosteroids, IVIG versus no IVIG, and cyclosporine versus IVIG [79].

It should be noted that the quality of the studies included in these meta-analyses was generally poor and that nearly all were retrospective cohort studies. The studies were also heterogeneous, with high variability in population, type of treatment and dosing, timing and duration of treatment, and site of care.

Most studies utilized SCORTEN as a comparator or internal control to determine the observed/expected death ratio in the patient cohort. However, it has been shown that SCORTEN may overestimate the expected mortality, resulting in a positively biased estimate of the efficacy of any intervention [15]. In addition, the "center effect" (ie, better outcomes seen in reference centers/specialized centers due to improvements in supportive care and treatment protocols) cannot be excluded in many studies [20].

Systemic corticosteroids — Varying regimens of systemic corticosteroids have been used for the treatment of SJS/TEN. These include prednisolone 60 to 250 mg/day for 2 to 12 days, intravenous dexamethasone 1.5 mg/kg/day for three days, and intravenous methylprednisolone 250 to 1000 mg/day for three days [80].

However, the efficacy of systemic corticosteroids has not been proven. Thus, routine use of systemic corticosteroids cannot be recommended.

Early, observational studies indicated a higher frequency of complications and increased mortality for patients with TEN treated with corticosteroids in burn units [81,82].

An initial analysis of French and German patients recruited in the EuroSCAR study (n = 281) comparing 159 patients who received variable doses of corticosteroids (ranging from 60 to 250 mg per day of prednisone equivalents), 75 patients who received IVIG (40 in association with corticosteroids), and 87 patients who received supportive care alone showed that the mortality rate was 18 percent in the corticosteroid group, 25 percent in the IVIG group, and 25 percent in the supportive care group [83]. The OR of death for patients treated with corticosteroids compared with patients treated with supportive care alone was 0.6 (95% CI 0.3-1.0), suggesting a potential benefit.

However, in a subsequent, updated review of patients recruited in the RegiSCAR study for whom treatment details were available (n = 442), there was no observed benefit or harm with corticosteroids (hazard ratio [HR] 1.3, 95% CI 0.8-1.9) [4].

A 2017 meta-analysis using individual data from 1209 patients, of whom 367 received systemic corticosteroids in addition to supportive treatment, found that corticosteroid treatment was associated with a modest decreased risk of death compared with supportive treatment alone (OR 0.67, 95% CI 0.46-0.97) [78].

A 2018 randomized trial compared the use of etanercept 25 or 50 mg two times per week (48 patients) versus intravenous prednisolone 1 to 1.5 mg/kg/day (43 patients). The mortality rate in the etanercept group was lower than that in the corticosteroid group (8.3 versus 16.3 percent, respectively), although the difference was not statistically significant [73]. The number of observed deaths in both the corticosteroid and etanercept arms was lower than that predicted by SCORTEN.

In a systematic review of 31 pediatric case series with 128 patients, no deaths were reported among the 20 patients who received either prednisolone/prednisone (1 mg/kg/day) or methylprednisolone (4 mg/kg/day) for five to seven days [84]. Complications occurred in five patients (mild skin infections in three children and bronchiolitis in two children).

In a retrospective study including 898 pediatric patients hospitalized with a primary diagnosis of SJS or TEN and treated with glucocorticoids only (18.6 percent), IVIG only (25.5 percent), IVIG plus glucocorticoids (17 percent), or supportive care only, there was no association between the use of systemic steroids, IVIG, or IVIG and steroids during the first two days of hospitalization and decreased length of stay or need for mechanical ventilation [85]. The overall hospital mortality was 0.56 percent (0.13 percent for SJS and 3.2 percent for TEN).

Cyclosporine — There is increasing evidence from multiple case series and meta-analyses suggesting that the cyclosporine administered during the acute phase may be associated with reduced mortality [71,76,78,86-90]. The use of cyclosporine early in the course of the disease has consequently been advocated by some experts [91]. We suggest using cyclosporine 3 to 5 mg/kg/day as adjunctive therapy within 24 to 48 hours of symptom onset.

The mechanism of action of cyclosporine in SJS/TEN involves the inhibition of T cell activation, therefore preventing the production and release of cytokines, which play a critical role in the pathogenesis and propagation of SJS/TEN by cytotoxic T cells and natural killer cells. (See "Stevens-Johnson syndrome and toxic epidermal necrolysis: Pathogenesis, clinical manifestations, and diagnosis", section on 'Pathogenesis'.)

An open-label trial conducted at a French center for SJS/TEN showed that the use of cyclosporine at 3 mg/kg/day tapered over a month may be associated with survival benefits. Utilizing SCORTEN, four deaths were predicted, although none were observed [71].

In a study of 71 patients in Spain with SJS/TEN, of whom 49 were treated with cyclosporine and 22 were treated with other therapies, the observed mortality rates were 10 and 32 percent, respectively [89]. The expected mortality rates according to the SCORTEN at admission were 24 percent in the cyclosporine group (observed-to-expected mortality rate ratio [MRR] 0.42, 95% CI 0.14-0.99) and 29 percent in the other therapies group (observed-to-expected MRR 1.09, 95% CI 0.44-2.25).

Similar findings have been demonstrated in Canada, Singapore, and India [88,90,92].

In a 2017 meta-analysis including five observational studies with a total of 134 patients with SJS/TEN who were treated with cyclosporine, the observed-to-expected MRR was 0.41 (95% CI 0.21-0.80) [89].

A 2018 meta-analysis of nine observational studies including 255 patients with TEN found that treatment with cyclosporine was associated with an approximately 70 percent reduction in mortality risk (pooled SMR 0.32, 95% CI 0.12-0.52) compared with supportive care and other treatments, including cyclophosphamide, corticosteroids, IVIG, and plasmapheresis [93].

However, in a French retrospective cohort of 174 patients with SJS/TEN hospitalized between 2005 and 2016, whereby 95 patients received cyclosporine and 79 patients received supportive care, there was no difference in primary outcome (progression of skin detachment on day 5) and secondary outcomes (proportion of patients with cutaneous re-epithelialization starting on day 5, overall in-hospital mortality) when analyzed via traditional exposed/unexposed or propensity-matched methodology [94].

Intravenous immune globulin — Intravenous immune globulin (IVIG; given at a dose of 2 to 4 g/kg over two to five days) was initially proposed as a treatment for SJS/TEN based on the hypothesis that Fas ligand (FasL) was the main mediator of widespread keratinocyte apoptosis in TEN and on the finding that high-dose IVIG was able to antagonize FasL effects [95]. However, it is now widely accepted that granulysin, a cytotoxic protein found in cytotoxic T cells, is the most important mediator [96]. (See "Stevens-Johnson syndrome and toxic epidermal necrolysis: Pathogenesis, clinical manifestations, and diagnosis".)

There is little evidence:

A 2012 systematic review and meta-analysis of 17 observational studies including 113 patients treated with IVIG and 130 patients treated with supportive care only found no difference in the risk of death between the two groups (OR 1.00, 95% CI 0.58-1.75) [97]. However, a subgroup analysis showed a statistically nonsignificant reduction in the mortality risk for patients treated with high-dose IVIG (total dose ≥2 g/kg) compared with those treated with <2 g/kg (OR 0.49, 95% CI 0.11-2.30).

In a subsequent, single-institution study of 64 cases of TEN and SJS/TEN overlap (mean SCORTEN 2.6) treated with IVIG, the overall mortality rate was 31 percent (SCORTEN-predicted mortality 28 percent, SMR 1.1, 95% CI 0.6-1.6) [98]. Mortality was similar among patients treated with <3 g/kg IVIG and those treated with ≥3 g/kg IVIG (31 and 26 percent, respectively).

A 2015 meta-analysis of 13 studies of patients who received IVIG for SJS or TEN, the severity of which was determined using SCORTEN, found an overall SMR of 0.81 (95% CI 0.62-1.08), with a nonsignificant difference in SMR between patients treated with IVIG and those who did not receive IVIG (SMR difference 0.32, 95% CI -0.77 to 0.12) [99]. However, a meta-regression performed to determine the effect of IVIG doses on SMR found significant reductions in SMR with increasing IVIG doses (≥2 g/kg).

A 2017 meta-analysis using individual data from 1209 patients, of whom 215 received IVIG in addition to supportive treatment, found that IVIG treatment was not associated with a decreased risk of death compared with supportive treatment alone (OR 0.99, 95% CI 0.64-1.54) [78].

Adverse effects of IVIG include renal, hematologic, and thrombotic complications. The risk for serious complications is increased in patients receiving high-dose IVIG, in older patients, and in those with pre-existing renal or cardiovascular disorders. (See "Intravenous immune globulin: Adverse effects".)

Intravenous immune globulin plus systemic corticosteroids — Systemic corticosteroids and intravenous immune globulin (IVIG) have been administered in combination to patients with SJS/TEN, but the data remain limited.

In a United States, retrospective, multicenter study including 377 hospitalized patients with SJS/TEN, the SMR among patients receiving both systemic corticosteroids (mean daily dose 148 mg prednisone equivalent) and IVIG (mean dose 1 g/kg per day for three days) was lower (SMR 0.52, 95% CI 0.21-0.79) than among patients receiving corticosteroids only (SMR 0.72, 95% CI 0.48-0.89), IVIG only (SMR 0.79, 95% CI 0.55-0.92), or supportive care alone (SMR 0.70, 95% CI 0.47-0.87) [100].

In an observational, propensity-matched, retrospective study from China, 93 patients who received combination therapy were compared with 52 patients on corticosteroid monotherapy [101]. There was no difference in the mortality rate between the two groups. However, compared with corticosteroid monotherapy, combination therapy was associated with a shorter mean hospitalization time (-3.37 days) and a lower rate of skin infections.

In a prospective, nonrandomized study, 36 patients with TEN received low-dose IVIG (0.2 to 0.5 g/kg) plus intravenous dexamethasone (0.1 to 0.3 mg/kg per day tapered in one to two weeks) or dexamethasone alone [102]. One death occurred in the combined therapy group, and three deaths occurred in the corticosteroid-only group. The SCORTEN-predicted deaths were five in both groups.

In the EuroSCAR cohort study, 35 patients were treated with IVIG alone, and 40 patients were treated with IVIG plus systemic corticosteroids [83]. The dose of IVIG ranged from 0.7 to 2.3 g/kg and was given in one to seven days. The mortality rate was 34 percent in the group treated with IVIG alone and 18 percent in the group treated with IVIG and corticosteroids.

In a retrospective study of 55 patients with TEN, 39 patients were treated with IVIG 0.4 g/kg per day for five days plus methylprednisolone 1.5 mg/kg per day for three to five days, and 22 patients were treated with methylprednisolone alone [103]. The mortality rate was 13 percent among patients treated with combination therapy (5 of 39) and 23 percent among those treated with corticosteroids alone (5 of 22).

A retrospective study compared 65 patients over a 14-year period. Forty-five patients were treated with corticosteroids (methylprednisolone 1 to 1.5 mg/kg/day), and 20 patients were treated with IVIG (2 g/kg over five days) and corticosteroids (methylprednisolone 1 to 1.5 mg/kg/day). There was no statistical difference in mortality between the two groups. The SMR was 1.16 (95% CI 0.56-2.13) in the corticosteroid group and 0.85 (95% CI 0.18-2.5) in the combination group [104].

Another similar, retrospective, Chinese study compared 82 patients who were treated with corticosteroids (n = 58) or IVIG and corticosteroids (n = 24). The SMR was 0.48 (95% CI 0.08-1.92) in the corticosteroids group and 0.57 (95% CI 0.32-1.91) in the combination group. There was no significant difference in SMR between the two groups [105].

Tumor necrosis factor inhibitors — In a few case reports, a single infusion of 5 mg/kg of the tumor necrosis factor (TNF)-alpha inhibitor infliximab halted the progression of skin detachment and induced a rapid re-epithelization of the denuded skin [106-109]. Etanercept, given in a single 50 mg subcutaneous injection, was used successfully in a small number of patients [110-112].

The efficacy of etanercept for the treatment of SJS/TEN was subsequently evaluated in a randomized, unblinded trial including 91 patients with a diagnosis of SJS/TEN [73]. Patients received etanercept 25 or 50 mg subcutaneously twice weekly or intravenous prednisolone 1 to 1.5 mg/kg per day. Seventy-one patients completed the study and were included in the analysis. The mortality rates were 8.3 and 16.3 percent in the etanercept and glucocorticoid groups, respectively. Although the difference was not statistically significant, the observed mortality rates in both groups were lower than those predicted by SCORTEN at admission (17.7 and 20.3 percent for the etanercept and steroid groups, respectively). The median time to complete re-epithelization in patients with ≥10 percent body surface area (BSA) detachment was shorter in the etanercept group than in the corticosteroid group (14 versus 19 days, respectively). Serious adverse events (including sepsis, respiratory failure, and upper gastrointestinal bleeding) occurred in 13 percent of patients in the etanercept group and in 27 percent of patients in the corticosteroid group but, except for gastrointestinal bleeding, were deemed to be unrelated to treatments.

Although this study adds further evidence to support the use of etanercept for SJS/TEN, additional studies are needed to confirm these findings and determine the optimal dose and duration of treatment.

Tumor necrosis factor inhibitors plus corticosteroids — Following initial experience with anti-tumor necrosis factor (TNF) inhibitors, there have been two further studies evaluating combinations of etanercept and systemic corticosteroids in SJS/TEN.

In a retrospective study of 242 patients with SJS/TEN from Taiwan and China, 196 patients (81 percent) received methylprednisolone monotherapy (average dose of 2 mg/kg/day), 25 patients received etanercept at a dose of 25 or 50 mg twice a week combined with systemic corticosteroids (1.2 mg/kg/day), and 21 patients were treated with IVIG and systemic corticosteroids (1.5 mg/kg/day) [113]. At baseline, those treated with combination therapy had higher mean SCORTEN values, with predicted mortality rates based on SCORTEN of 9.3, 16, and 14.4 percent in the methylprednisolone, methylprednisolone plus IVIG, and methylprednisolone plus etanercept groups, respectively. The observed mortality rate was 6.6 percent (SMR 0.7, 95% CI -0.8-2.6) in the methylprednisolone group, 4.8 percent (SMR 0.3, 95% CI -0.7 to 6.2) in the methylprednisolone plus IVIG group, and 0 percent (SMR 0, 95% CI -1.8 to 3.59) in the methylprednisolone plus etanercept group. Combination of methylprednisolone and etanercept also showed a reduced mean skin healing time of 12 days versus 13 days (methylprednisolone) and 13.5 days (methylprednisolone plus IVIG).

In a cohort of 25 patients with SJS/TEN (17 with SJS, 1 with SJS/TEN, and 7 with TEN), 10 patients received methylprednisolone, and 15 patients received etanercept plus methylprednisolone [114]. At baseline, there were no significant differences in terms of age, SCORTEN, and disease classification. There were no deaths in both groups. The combination therapy shortened skin healing time (median 12 days) compared with the corticosteroid monotherapy group (median 16 days).

Plasmapheresis — The use of plasmapheresis alone or in combination with other treatments has been reported in several small case series of TEN with variable results [115-118]. Removal of a toxin, such as a drug, drug metabolite, or other cytotoxic mediator, has been proposed as a mechanism of action. (See "Therapeutic apheresis (plasma exchange or cytapheresis): Indications and technology".)

In a Japanese retrospective study comparing the outcomes of patients with SJS/TEN who did not respond to pulse corticosteroid therapy and were subsequently treated with plasmapheresis (n = 53) or IVIG (n = 213), the mortality rates were similar in the two groups (18.3 and 19.5 percent, respectively) [119]. However, the length of hospital stays and costs were greater for patients undergoing plasmapheresis.

Harmful agents — Thalidomide, a potent inhibitor of TNF-alpha, should not be used for the treatment of SJS/TEN. A randomized, placebo-controlled trial of thalidomide in patients with TEN was prematurely stopped because of excess mortality among patients given thalidomide [74].

PROGNOSIS

Mortality — The overall mortality rate among patients with SJS and TEN is approximately 25 percent, ranging from 10 percent for SJS to >30 percent for TEN [4]. In the United States, the mean adjusted mortality rates between 2009 and 2012 were 4.8 percent for SJS, 19.4 percent for SJS/TEN, and 14.8 percent for TEN [120]. Mortality is considerably lower in children, ranging from 0 to 9.5 percent among centers [121-123], although a very high mortality rate (39 percent) has been reported in infants with TEN [124].

Sepsis, acute respiratory distress syndrome, disseminated intravascular coagulation, and multiple organ failure are the most common causes of in-hospital death [40,41,45,67,125]. Patients who develop acute renal failure requiring renal replacement therapy also have an increased mortality risk [126]. Older age (>70 years) and presence of comorbidities (eg, liver cirrhosis, metastatic cancer) are also associated with an increased risk of death [127].

Disease severity (ie, the extent of cutaneous involvement) is the main risk factor for mortality within 90 days of disease onset [4]. However, the risk of death remains high beyond 90 days and up to one year after the onset of the reaction and is mainly influenced by the patient's age and presence of comorbidities. Among 460 patients with SJS/TEN included in the European RegiSCAR study between 2003 and 2007, death rates of 23 and 34 percent were reported at six weeks and one year after discharge, respectively [4].

Recurrence — The likelihood of recurrence of SJS/TEN is unknown. Recurrence is generally rare in adults unless there is re-exposure to the same drug/structurally similar drug [128,129]. In two studies based on the International Classification of Diseases (ICD) coding and hospital records, recurrence of up to 18 percent occurred in children [130], and recurrence of up to 7 percent has been reported in adults (16 recurrent episodes per 1000 person-years) [131]. However, in a cohort of 106 validated cases of SJS/TEN followed up in a reference center, there were two recurrences (one from Mycoplasma infection and another due to re-exposure to the causative drug; 4.2 recurrent episodes per 1000 person-years) [128]. These differences in recurrence rates may be due to the methodology of utilizing electronic disease coding (eg, ICD coding), which may allow for the inclusion of cases of erythema multiforme and bullous fixed drug eruptions, which recur more frequently.

In children, up to 50 percent of SJS/TEN cases do not have a drug trigger and are believed to be due to infections (eg, Mycoplasma) or idiopathic [130]. The high recurrence in children may be due to the higher incidence of infections as a trigger.

Long-term sequelae — Long-term sequelae include cutaneous, mucosal, ocular, visceral, and psychologic complications, which are increasingly reported as survival improves [5,132,133].

Skin, hair, and nails — Cutaneous sequelae are common, affecting up to 80 percent of survivors, and include postinflammatory hypo- or hyperpigmentation, scarring, eruptive nevi, telogen effluvium, and chronic pruritus [5,133]. Scarring may occur when superinfection complicates re-epithelialization or as a consequence of surgical debridement of necrotic skin or skin grafting. Nail changes occur in up to 70 percent of survivors and include Beau lines and onychomadesis [134]. (See "Overview of nail disorders", section on 'Transverse grooves (Beau lines)' and "Overview of nail disorders", section on 'Onychomadesis'.)

Eyes — Ocular sequelae develop in approximately 50 to 90 percent of patients with acute ocular involvement [130]. These sequelae include dry eye, photophobia, ingrown eyelashes (trichiasis), neovascularization of the cornea, lid margin changes (eg, posterior migration of the mucocutaneous junction and keratinization), keratitis, symblepharon, and corneal scarring leading to visual impairment and, rarely, blindness [52-56,135].

Ocular sequelae are mainly due to functional alteration of the conjunctival epithelium, decreased lacrimal film, ocular surface failure due to loss of corneal stem cells and recurrent episodic inflammation, scleritis, and progressive conjunctival cicatrization [6].

The onset of ocular involvement may be delayed. In a study of 54 patients with SJS or TEN, only 11 percent of patients had severe ocular involvement detected during the acute phase, but nearly 90 percent developed chronic ocular sequelae six months or later after the initial presentation [136]. Late complications were severe in 17 percent of the patients, including trichiasis, mucocutaneous junction abnormalities, and extensive corneal opacities.

Management of long-term ocular sequelae involves the use of artificial tears and scleral lenses [137]. Systemic immunosuppression has been used to suppress conjunctival inflammation with variable results. Surgical interventions to restore ocular surface include limbal stem cell transplantation, amniotic membrane transplantation (AMT), cultivated oral mucosal epithelial transplantation, and corneal transplant.

Oral and dental — Oral and dental sequelae have been observed in survivors of SJS/TEN. These include mouth discomfort, xerostomia, gingival inflammation and synechiae, caries, and periodontal disease [138]. Severe dental growth abnormalities (eg, dental agenesia, root dysmorphia, root-building abortion, microdontia) may occur in survivors who suffered from SJS/TEN in childhood [138].

Urogenital — Long-term consequences include adhesion formation, scarring, urethral stenosis, and strictures. In females, specific urogenital sequelae affect up to 28 percent of survivors. These include introital stenosis, vaginal dryness, dyspareunia, urinary retention, hematocolpos, and vaginal adenosis [48-51,139-141]. In males, strictures and adhesions can lead to phimosis.

Pulmonary — Long-term pulmonary complications include chronic bronchitis/bronchiolitis with obstructive changes (including bronchiolitis obliterans and organizing pneumonia [formerly called bronchiolitis obliterans organizing pneumonia]), bronchiectasis, and obstructive disorders [5,142,143]. Pulmonary sequelae may also occur in asymptomatic patients. A series of 32 patients with SJS/TEN underwent pulmonary function tests (PFTs) at a median time of three months after the acute episode. Although only two patients had dyspnea, abnormal PFTs were demonstrated in more than one-half of the cases [144]. The most frequent PFT abnormality was a reduced carbon monoxide (CO) diffusion, which persisted at 12 months in 8 of 10 patients. (See "Overview of bronchiolar disorders in adults" and "Clinical manifestations and diagnosis of bronchiectasis in adults".)

Chronic pain — Chronic pain at mucocutaneous sites of involvement has been reported in approximately one-third of patients [145].

Psychiatric — Long-term psychosocial complications and psychiatric disorders (eg, anxiety, depression) and decreased health-related quality of life have been reported in survivors of SJS/TEN [133,146,147]. Post-traumatic stress disorder affects 20 percent of survivors six months after the initial episode. Depression and anxiety may affect up to 30 to 50 percent and 40 percent of survivors, respectively. More than 60 percent of respondents were fearful of taking newly prescribed medications, and 30 percent of patients have avoided taking prescribed medications for their diagnosed medical conditions. Close to 30 percent of survivors were unable to work following their acute episode [133].

FUTURE DRUG AVOIDANCE

Patient education — Patients who survive SJS/TEN attributed to a medication must be educated about future avoidance and understand that re-exposure to the culprit drug may be fatal. Relevant information should be inscribed on a medical information bracelet or necklace and worn at all times. Electronic medical records, "allergy cards," and alerts should also be updated accordingly. Patients should learn all the various names for the causative medication, be able to report that they have a history of SJS or TEN, and be able to recognize closely related medications.

Future use of related medications — Recurrent SJS/TEN with readministration of the causative drug or closely, chemically related agents has been widely documented in multiple case reports [128,148,149]. The risk of recurrent SJS/TEN with structurally distinct agents (within the same therapeutic class of drugs) is unknown but probably very low. As an example, patients with past SJS/TEN due to sulfamethoxazole should avoid other anti-infectious sulfonamides (eg, sulfadiazine, sulfapyridine) but can use thiazide diuretics or sulfonylurea-derived oral antidiabetics [150].

Patients with SJS/TEN caused by "aromatic" anticonvulsants (eg, phenytoin, carbamazepine, phenobarbital) should be informed that other agents in the same class may cause similar reactions in both the patient and their family members (table 1) [151,152]. Although there are no data about the safety of nonaromatic anticonvulsants (eg, valproate, succinimides, benzodiazepines, gabapentin) for patients with a history of SJS/TEN induced by aromatic anticonvulsants, the risk of SJS/TEN recurrence after exposure to a different class of anticonvulsants is probably extremely low and certainly lower than the risk related to the absence of antiepileptic treatment [10]. The pharmacology of antiepileptic agents is reviewed in detail elsewhere. (See "Antiseizure medications: Mechanism of action, pharmacology, and adverse effects".)

Because human leukocyte antigen groups are associated with carbamazepine-, phenytoin-, and allopurinol-induced SJS/TEN, family members of patients should be alerted against the use of the same medication. (See "Stevens-Johnson syndrome and toxic epidermal necrolysis: Pathogenesis, clinical manifestations, and diagnosis".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Drug allergy and hypersensitivity".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Stevens-Johnson syndrome and toxic epidermal necrolysis (The Basics)")

SUMMARY AND RECOMMENDATIONS

Initial in-hospital evaluation – Patients suspected to have Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) require immediate in-hospital evaluation.

Identification and withdrawal of the offending drug – Prompt withdrawal of the offending drug (table 1) may improve the prognosis. The algorithm of drug causality for epidermal necrolysis (ALDEN (table 2)) can help identify the offending agent. (See 'Prompt identification and withdrawal of the causative drug' above.)

Assessment of severity and prognosis – The severity and prognosis of the disease should be rapidly determined by using the score of toxic epidermal necrolysis (SCORTEN) to define the appropriate medical setting for management. (See 'Assessment of the extent of skin detachment' above and 'Assessment of prognosis' above.)

Transfer to a referral center – Patients with body surface area (BSA) detachment of ≥10 percent, rapidly progressive disease, or worsening biochemical/organ function should be transferred and managed in specialized centers, such as specialized dermatology units, burn centers, or intensive care units. (See 'Transfer to a referral center' above.)

Supportive care – Supportive care is the mainstay of treatment and includes wound care, fluid and electrolyte management, nutritional support, temperature management, pain control, prevention and treatment of infection, ocular care, and organ support if needed. (See 'Supportive care' above.)

Wound care – The optimal approach to wound care has not been determined. Success has been reported with both repeated debridement of exfoliating skin and "antishear" wound care, in which the necrotic skin is left in place to act as a biologic dressing. (See 'Wound care' above.)

Ocular care – Ocular lesions require immediate attention and care. Specific interventions aimed at preventing long-term ocular sequelae in severe cases with extensive sloughing of the bulbar conjunctiva include the use of preservative-free corticosteroid eye drops, coverage of the eye surface with cryopreserved amniotic membrane, and use of scleral spacers. (See 'Management of ocular involvement' above.)

Infection prevention – Because sepsis is the major cause of death, key infection control measures include sterile handling, topical antiseptic agents, and surveillance cultures of possible sites of superinfection. Prophylactic systemic antibiotics are not utilized by most burn centers, although antimicrobials should be administered at the first sign of infection, and choice of agent should be guided by specific culture data. (See 'Prevention and treatment of infections' above.)

Monitoring of respiratory involvement – All patients need close monitoring for respiratory involvement and referral to an intensive care unit if they develop features suggestive of pulmonary involvement (eg, cough, dyspnea, hypoxemia, radiologic abnormalities). (See 'Management of acute respiratory involvement' above.)

Adjunctive pharmacologic therapies

Beyond supportive care, we suggest cyclosporine rather than other systemic adjunctive therapies for patients with SJS/TEN. Oral cyclosporine at the dose of 3 to 5 mg/kg/day should be given early in the course of the disease (ie, within 24 to 48 hours of symptom onset) (Grade 2C). For patients in whom the use of cyclosporine is contraindicated, a single dose of the tumor necrosis factor (TNF) inhibitor etanercept is an alternative option. (See 'Cyclosporine' above and 'Tumor necrosis factor inhibitors' above.)

Although frequently used, the role of systemic corticosteroids in the management of SJS/TEN remains uncertain, and treatment modality (eg, oral versus intravenous "pulse" administration), dose, timing, and duration have not been determined. Thus, routine use of systemic corticosteroids cannot be recommended. (See 'Systemic corticosteroids' above.)

We suggest not using intravenous immune globulin (IVIG) for SJS/TEN as monotherapy (Grade 2C). In several meta-analyses, no clear survival advantage has been found for patients with SJS/TEN treated with IVIG. The role of IVIG in combination with systemic steroids needs to be further investigated. (See 'Intravenous immune globulin' above.)

Prognosis – The overall mortality rate among patients with SJS/TEN is approximately 25 percent, ranging from approximately 10 percent for SJS to >30 percent for TEN. Sepsis, acute respiratory distress syndrome, and multiple organ failure are the most common causes of in-hospital death. Long-term sequelae involving the skin and eyes are common among survivors. (See 'Mortality' above and 'Long-term sequelae' above.)

Future drug avoidance – Patients who survive SJS/TEN attributed to a medication must be educated about future avoidance of the causative drug and other chemically related agents and understand that re-exposure may be fatal. Relevant information should be inscribed on a medical information bracelet or necklace and worn at all times. Electronic medical records, "allergy cards," and alerts should also be updated accordingly.

  1. Bastuji-Garin S, Rzany B, Stern RS, et al. Clinical classification of cases of toxic epidermal necrolysis, Stevens-Johnson syndrome, and erythema multiforme. Arch Dermatol 1993; 129:92.
  2. Ingen-Housz-Oro S, Duong TA, Bensaid B, et al. Epidermal necrolysis French national diagnosis and care protocol (PNDS; protocole national de diagnostic et de soins). Orphanet J Rare Dis 2018; 13:56.
  3. Heng YK, Lee HY, Roujeau JC. Epidermal necrolysis: 60 years of errors and advances. Br J Dermatol 2015; 173:1250.
  4. Sekula P, Dunant A, Mockenhaupt M, et al. Comprehensive survival analysis of a cohort of patients with Stevens-Johnson syndrome and toxic epidermal necrolysis. J Invest Dermatol 2013; 133:1197.
  5. Lee HY, Walsh SA, Creamer D. Long-term complications of Stevens-Johnson syndrome/toxic epidermal necrolysis (SJS/TEN): the spectrum of chronic problems in patients who survive an episode of SJS/TEN necessitates multidisciplinary follow-up. Br J Dermatol 2017; 177:924.
  6. Creamer D, Walsh SA, Dziewulski P, et al. U.K. guidelines for the management of Stevens-Johnson syndrome/toxic epidermal necrolysis in adults 2016. Br J Dermatol 2016; 174:1194.
  7. Garcia-Doval I, LeCleach L, Bocquet H, et al. Toxic epidermal necrolysis and Stevens-Johnson syndrome: does early withdrawal of causative drugs decrease the risk of death? Arch Dermatol 2000; 136:323.
  8. Sassolas B, Haddad C, Mockenhaupt M, 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.
  9. Roujeau JC, Kelly JP, Naldi L, et al. Medication use and the risk of Stevens-Johnson syndrome or toxic epidermal necrolysis. N Engl J Med 1995; 333:1600.
  10. Mockenhaupt M, Viboud C, Dunant A, 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.
  11. Wang YH, Chen CB, Tassaneeyakul W, et al. The Medication Risk of Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis in Asians: The Major Drug Causality and Comparison With the US FDA Label. Clin Pharmacol Ther 2019; 105:112.
  12. Bastuji-Garin S, Fouchard N, Bertocchi M, et al. SCORTEN: a severity-of-illness score for toxic epidermal necrolysis. J Invest Dermatol 2000; 115:149.
  13. Guégan S, Bastuji-Garin S, Poszepczynska-Guigné E, et al. Performance of the SCORTEN during the first five days of hospitalization to predict the prognosis of epidermal necrolysis. J Invest Dermatol 2006; 126:272.
  14. Torres-Navarro I, Briz-Redón Á, Botella-Estrada R. Accuracy of SCORTEN to predict the prognosis of Stevens-Johnson syndrome/toxic epidermal necrolysis: a systematic review and meta-analysis. J Eur Acad Dermatol Venereol 2020; 34:2066.
  15. Koh HK, Fook-Chong S, Lee HY. Assessment and Comparison of Performance of ABCD-10 and SCORTEN in Prognostication of Epidermal Necrolysis. JAMA Dermatol 2020; 156:1294.
  16. Noe MH, Rosenbach M, Hubbard RA, et al. Development and Validation of a Risk Prediction Model for In-Hospital Mortality Among Patients With Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis-ABCD-10. JAMA Dermatol 2019; 155:448.
  17. Duplisea MJ, Roberson ML, Chrisco L, et al. Performance of ABCD-10 and SCORTEN mortality prediction models in a cohort of patients with Stevens-Johnson syndrome/toxic epidermal necrolysis. J Am Acad Dermatol 2021; 85:873.
  18. Koh HK, Fook-Chong SMC, Lee HY. Improvement of Mortality Prognostication in Patients With Epidermal Necrolysis: The Role of Novel Inflammatory Markers and Proposed Revision of SCORTEN (Re-SCORTEN). JAMA Dermatol 2022; 158:160.
  19. Palmieri TL, Greenhalgh DG, Saffle JR, et al. A multicenter review of toxic epidermal necrolysis treated in U.S. burn centers at the end of the twentieth century. J Burn Care Rehabil 2002; 23:87.
  20. Traikia C, Hua C, Le Cleach L, et al. Individual- and hospital-level factors associated with epidermal necrolysis mortality: a nationwide multilevel study, France, 2012-2016. Br J Dermatol 2020; 182:900.
  21. Clark AE, Fook-Chong S, Choo K, et al. Delayed admission to a specialist referral center for Stevens-Johnson syndrome and toxic epidermal necrolysis is associated with increased mortality: A retrospective cohort study. JAAD Int 2021; 4:10.
  22. McPherson T, Exton LS, Biswas S, et al. British Association of Dermatologists' guidelines for the management of Stevens-Johnson syndrome/toxic epidermal necrolysis in children and young people, 2018. Br J Dermatol 2019; 181:37.
  23. Brüggen MC, Le ST, Walsh S, et al. Supportive care in the acute phase of Stevens-Johnson syndrome and toxic epidermal necrolysis: an international, multidisciplinary Delphi-based consensus. Br J Dermatol 2021; 185:616.
  24. Seminario-Vidal L, Kroshinsky D, Malachowski SJ, et al. Society of Dermatology Hospitalists supportive care guidelines for the management of Stevens-Johnson syndrome/toxic epidermal necrolysis in adults. J Am Acad Dermatol 2020; 82:1553.
  25. Roujeau JC, Chosidow O, Saiag P, Guillaume JC. Toxic epidermal necrolysis (Lyell syndrome). J Am Acad Dermatol 1990; 23:1039.
  26. Lee HY. Wound management strategies in Stevens-Johnson syndrome/toxic epidermal necrolysis: An unmet need. J Am Acad Dermatol 2018; 79:e87.
  27. Ying S, Ho W, Chan HH. Toxic epidermal necrolysis: 10 years experience of a burns centre in Hong Kong. Burns 2001; 27:372.
  28. Atiyeh BS, Dham R, Yassin MF, El-Musa KA. Treatment of toxic epidermal necrolysis with moisture-retentive ointment: a case report and review of the literature. Dermatol Surg 2003; 29:185.
  29. Boorboor P, Vogt PM, Bechara FG, et al. Toxic epidermal necrolysis: use of Biobrane or skin coverage reduces pain, improves mobilisation and decreases infection in elderly patients. Burns 2008; 34:487.
  30. Spies M, Sanford AP, Aili Low JF, et al. Treatment of extensive toxic epidermal necrolysis in children. Pediatrics 2001; 108:1162.
  31. Dorafshar AH, Dickie SR, Cohn AB, et al. Antishear therapy for toxic epidermal necrolysis: an alternative treatment approach. Plast Reconstr Surg 2008; 122:154.
  32. Castillo B, Vera N, Ortega-Loayza AG, Seminario-Vidal L. Wound care for Stevens-Johnson syndrome and toxic epidermal necrolysis. J Am Acad Dermatol 2018; 79:764.
  33. Dunn K, Edwards-Jones V. The role of Acticoat with nanocrystalline silver in the management of burns. Burns 2004; 30 Suppl 1:S1.
  34. Huang SH, Yang PS, Wu SH, et al. Aquacel Ag with Vaseline gauze in the management of toxic epidermal necrolysis (TEN). Burns 2010; 36:121.
  35. Haravu PN, Gottlieb LJ, Vrouwe SQ. Antishear Therapy for Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis: A Follow-up Study. J Burn Care Res 2021; 42:1152.
  36. Pruitt BA Jr. Burn treatment for the unburned. JAMA 1987; 257:2207.
  37. Shiga S, Cartotto R. What are the fluid requirements in toxic epidermal necrolysis? J Burn Care Res 2010; 31:100.
  38. Kelemen JJ 3rd, Cioffi WG Jr, Mason AD Jr, et al. Effect of ambient temperature on metabolic rate after thermal injury. Ann Surg 1996; 223:406.
  39. Valeyrie-Allanore L, Ingen-Housz-Oro S, Colin A, et al. [Pain management in Stevens-Johnson syndrome, toxic epidermal necrolysis and other blistering diseases]. Ann Dermatol Venereol 2011; 138:694.
  40. Revuz J, Penso D, Roujeau JC, et al. Toxic epidermal necrolysis. Clinical findings and prognosis factors in 87 patients. Arch Dermatol 1987; 123:1160.
  41. Koh HK, Chai ZT, Tay HW, et al. Risk factors and diagnostic markers of bacteremia in Stevens-Johnson syndrome and toxic epidermal necrolysis: A cohort study of 176 patients. J Am Acad Dermatol 2019; 81:686.
  42. de Prost N, Ingen-Housz-Oro S, Duong Ta, 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.
  43. Lecadet A, Woerther PL, Hua C, et al. Incidence of bloodstream infections and predictive value of qualitative and quantitative skin cultures of patients with overlap syndrome or toxic epidermal necrolysis: A retrospective observational cohort study of 98 cases. J Am Acad Dermatol 2019; 81:342.
  44. Lebargy F, Wolkenstein P, Gisselbrecht M, et al. Pulmonary complications in toxic epidermal necrolysis: a prospective clinical study. Intensive Care Med 1997; 23:1237.
  45. de Prost N, Mekontso-Dessap A, Valeyrie-Allanore L, et al. Acute respiratory failure in patients with toxic epidermal necrolysis: clinical features and factors associated with mechanical ventilation. Crit Care Med 2014; 42:118.
  46. Beck A, Cooney R, Gamelli RL, Mosier MJ. Predicting Mechanical Ventilation and Mortality: Early and Late Indicators in Steven-Johnson Syndrome and Toxic Epidermal Necrolysis. J Burn Care Res 2016; 37:e47.
  47. Phan K, Oh LJ, Issler-Fisher A, et al. Ventilatory support in Stevens-Johnson syndrome and toxic epidermal necrolysis: a systematic review and meta-analysis. J Dermatolog Treat 2022; 33:525.
  48. Meneux E, Wolkenstein P, Haddad B, et al. Vulvovaginal involvement in toxic epidermal necrolysis: a retrospective study of 40 cases. Obstet Gynecol 1998; 91:283.
  49. Kaser DJ, Reichman DE, Laufer MR. Prevention of vulvovaginal sequelae in stevens-johnson syndrome and toxic epidermal necrolysis. Rev Obstet Gynecol 2011; 4:81.
  50. Crowder CA, Jeney SES, Kraus CN, et al. Vulvovaginal involvement in Stevens-Johnson syndrome and toxic epidermal necrolysis: management and techniques used to reduce gynecologic sequelae. Int J Dermatol 2022; 61:158.
  51. O'Brien KF, Bradley SE, Mitchell CM, et al. Vulvovaginal manifestations in Stevens-Johnson syndrome and toxic epidermal necrolysis: Prevention and treatment. J Am Acad Dermatol 2021; 85:523.
  52. Gueudry J, Roujeau JC, Binaghi M, et al. Risk factors for the development of ocular complications of Stevens-Johnson syndrome and toxic epidermal necrolysis. Arch Dermatol 2009; 145:157.
  53. Chang YS, Huang FC, Tseng SH, et al. Erythema multiforme, Stevens-Johnson syndrome, and toxic epidermal necrolysis: acute ocular manifestations, causes, and management. Cornea 2007; 26:123.
  54. Yip LW, Thong BY, Lim J, et al. Ocular manifestations and complications of Stevens-Johnson syndrome and toxic epidermal necrolysis: an Asian series. Allergy 2007; 62:527.
  55. Sotozono C, Ueta M, Nakatani E, et al. Predictive Factors Associated With Acute Ocular Involvement in Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis. Am J Ophthalmol 2015; 160:228.
  56. Kohanim S, Palioura S, Saeed HN, et al. Acute and Chronic Ophthalmic Involvement in Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis - A Comprehensive Review and Guide to Therapy. II. Ophthalmic Disease. Ocul Surf 2016; 14:168.
  57. Gregory DG. Treatment of acute Stevens-Johnson syndrome and toxic epidermal necrolysis using amniotic membrane: a review of 10 consecutive cases. Ophthalmology 2011; 118:908.
  58. Shay E, Khadem JJ, Tseng SC. Efficacy and limitation of sutureless amniotic membrane transplantation for acute toxic epidermal necrolysis. Cornea 2010; 29:359.
  59. Sharma N, Thenarasun SA, Kaur M, et al. Adjuvant Role of Amniotic Membrane Transplantation in Acute Ocular Stevens-Johnson Syndrome: A Randomized Control Trial. Ophthalmology 2016; 123:484.
  60. Shammas MC, Lai EC, Sarkar JS, et al. Management of acute Stevens-Johnson syndrome and toxic epidermal necrolysis utilizing amniotic membrane and topical corticosteroids. Am J Ophthalmol 2010; 149:203.
  61. Shanbhag SS, Hall L, Chodosh J, Saeed HN. Long-term outcomes of amniotic membrane treatment in acute Stevens-Johnson syndrome/toxic epidermal necrolysis. Ocul Surf 2020; 18:517.
  62. Kim DH, Yoon KC, Seo KY, et al. The role of systemic immunomodulatory treatment and prognostic factors on chronic ocular complications in Stevens-Johnson syndrome. Ophthalmology 2015; 122:254.
  63. Yip LW, Thong BY, Tan AW, et al. High-dose intravenous immunoglobulin in the treatment of toxic epidermal necrolysis: a study of ocular benefits. Eye (Lond) 2005; 19:846.
  64. Araki Y, Sotozono C, Inatomi T, et al. Successful treatment of Stevens-Johnson syndrome with steroid pulse therapy at disease onset. Am J Ophthalmol 2009; 147:1004.
  65. Mieno H, Ueta M, Kinoshita F, et al. Corticosteroid Pulse Therapy for Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis Patients With Acute Ocular Involvement. Am J Ophthalmol 2021; 231:194.
  66. Hall LN, Shanbhag SS, Rashad R, et al. The effects of systemic cyclosporine in acute Stevens-Johnson syndrome/toxic epidermal necrolysis on ocular disease. Ocul Surf 2021; 19:128.
  67. Chen CB, Hsu TH, Chung-Yee Hui R, et al. Disseminated intravascular coagulation in Stevens-Johnson syndrome and toxic epidermal necrolysis. J Am Acad Dermatol 2021; 84:1782.
  68. El-Azhary RA, Nowsheen S, Gibson LE, DiCaudo DJ. Disseminated intravascular coagulopathy: a complication of Stevens-Johnson syndrome/toxic epidermal necrolysis. Int J Dermatol 2021; 60:185.
  69. de Sica-Chapman A, Williams G, Soni N, Bunker CB. Granulocyte colony-stimulating factor in toxic epidermal necrolysis (TEN) and Chelsea & Westminster TEN management protocol [corrected]. Br J Dermatol 2010; 162:860.
  70. 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.
  71. Valeyrie-Allanore L, Wolkenstein P, Brochard L, et al. Open trial of ciclosporin treatment for Stevens-Johnson syndrome and toxic epidermal necrolysis. Br J Dermatol 2010; 163:847.
  72. Chaidemenos GC, Chrysomallis F, Sombolos K, et al. Plasmapheresis in toxic epidermal necrolysis. Int J Dermatol 1997; 36:218.
  73. Wang CW, Yang LY, Chen CB, et al. Randomized, controlled trial of TNF-α antagonist in CTL-mediated severe cutaneous adverse reactions. J Clin Invest 2018; 128:985.
  74. Wolkenstein P, Latarjet J, Roujeau JC, et al. Randomised comparison of thalidomide versus placebo in toxic epidermal necrolysis. Lancet 1998; 352:1586.
  75. 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; 35:159.
  76. Tsai TY, Huang IH, Chao YC, et al. Treating toxic epidermal necrolysis with systemic immunomodulating therapies: A systematic review and network meta-analysis. J Am Acad Dermatol 2021; 84:390.
  77. 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; 87:628.
  78. Zimmermann S, Sekula P, Venhoff 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.
  79. Jacobsen A, Olabi B, Langley A, et al. Systemic interventions for treatment of Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), and SJS/TEN overlap syndrome. Cochrane Database Syst Rev 2022; 3:CD013130.
  80. Kim KJ, Lee DP, Suh HS, et al. Toxic epidermal necrolysis: analysis of clinical course and SCORTEN-based comparison of mortality rate and treatment modalities in Korean patients. Acta Derm Venereol 2005; 85:497.
  81. Halebian PH, Corder VJ, Madden MR, et al. Improved burn center survival of patients with toxic epidermal necrolysis managed without corticosteroids. Ann Surg 1986; 204:503.
  82. Kelemen JJ 3rd, Cioffi WG, McManus WF, et al. Burn center care for patients with toxic epidermal necrolysis. J Am Coll Surg 1995; 180:273.
  83. Schneck J, Fagot JP, Sekula P, et al. 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.
  84. Del Pozzo-Magana BR, Lazo-Langner A, Carleton B, et al. A systematic review of treatment of drug-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in children. J Popul Ther Clin Pharmacol 2011; 18:e121.
  85. Antoon JW, Goldman JL, Shah SS, Lee B. A Retrospective Cohort Study of the Management and Outcomes of Children Hospitalized with Stevens-Johnson Syndrome or Toxic Epidermal Necrolysis. J Allergy Clin Immunol Pract 2019; 7:244.
  86. Arévalo JM, Lorente JA, González-Herrada C, Jiménez-Reyes J. Treatment of toxic epidermal necrolysis with cyclosporin A. J Trauma 2000; 48:473.
  87. Reese D, Henning JS, Rockers K, et al. Cyclosporine for SJS/TEN: a case series and review of the literature. Cutis 2011; 87:24.
  88. Lee HY, Fook-Chong S, Koh HY, et al. 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.
  89. González-Herrada C, Rodríguez-Martín S, Cachafeiro L, 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.
  90. Kirchhof MG, Miliszewski MA, Sikora S, et al. Retrospective review of Stevens-Johnson syndrome/toxic epidermal necrolysis treatment comparing intravenous immunoglobulin with cyclosporine. J Am Acad Dermatol 2014; 71:941.
  91. Roujeau JC, Mockenhaupt M, Guillaume JC, Revuz J. New Evidence Supporting Cyclosporine Efficacy in Epidermal Necrolysis. J Invest Dermatol 2017; 137:2047.
  92. Singh GK, 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; 79:686.
  93. Ng QX, De Deyn MLZQ, Venkatanarayanan N, et al. A meta-analysis of cyclosporine treatment for Stevens-Johnson syndrome/toxic epidermal necrolysis. J Inflamm Res 2018; 11:135.
  94. Poizeau F, Gaudin O, Le Cleach L, et al. Cyclosporine for Epidermal Necrolysis: Absence of Beneficial Effect in a Retrospective Cohort of 174 Patients-Exposed/Unexposed and Propensity Score-Matched Analyses. J Invest Dermatol 2018; 138:1293.
  95. Viard I, Wehrli P, Bullani R, et al. Inhibition of toxic epidermal necrolysis by blockade of CD95 with human intravenous immunoglobulin. Science 1998; 282:490.
  96. Chung WH, Hung SI, Yang JY, et al. Granulysin is a key mediator for disseminated keratinocyte death in Stevens-Johnson syndrome and toxic epidermal necrolysis. Nat Med 2008; 14:1343.
  97. 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.
  98. 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.
  99. 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.
  100. Micheletti RG, Chiesa-Fuxench Z, Noe MH, et al. Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis: A Multicenter Retrospective Study of 377 Adult Patients from the United States. J Invest Dermatol 2018; 138:2315.
  101. Yang L, Shou YH, Li F, et al. 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.
  102. Jagadeesan S, Sobhanakumari K, Sadanandan SM, 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; 79:506.
  103. Zhu QY, Ma L, Luo XQ, Huang HY. Toxic epidermal necrolysis: performance of SCORTEN and the score-based comparison of the efficacy of corticosteroid therapy and intravenous immunoglobulin combined therapy in China. J Burn Care Res 2012; 33:e295.
  104. 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; 48:1122.
  105. Chen J, Wang B, Zeng Y, Xu H. High-dose intravenous immunoglobulins in the treatment of Stevens-Johnson syndrome and toxic epidermal necrolysis in Chinese patients: a retrospective study of 82 cases. Eur J Dermatol 2010; 20:743.
  106. 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.
  107. Patmanidis K, Sidiras A, Dolianitis K, 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.
  108. Scott-Lang V, Tidman M, McKay D. Toxic epidermal necrolysis in a child successfully treated with infliximab. Pediatr Dermatol 2014; 31:532.
  109. Wojtkiewicz A, Wysocki M, Fortuna J, et al. Beneficial and rapid effect of infliximab on the course of toxic epidermal necrolysis. Acta Derm Venereol 2008; 88:420.
  110. Gubinelli E, Canzona F, Tonanzi T, et al. Toxic epidermal necrolysis successfully treated with etanercept. J Dermatol 2009; 36:150.
  111. Famularo G, Di Dona B, Canzona F, et al. Etanercept for toxic epidermal necrolysis. Ann Pharmacother 2007; 41:1083.
  112. Paradisi A, Abeni D, Bergamo F, et al. Etanercept therapy for toxic epidermal necrolysis. J Am Acad Dermatol 2014; 71:278.
  113. Zhang J, Lu CW, Chen CB, 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.
  114. Ao S, Gao X, Zhan J, 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; 86:1236.
  115. Bamichas G, Natse T, Christidou F, et al. Plasma exchange in patients with toxic epidermal necrolysis. Ther Apher 2002; 6:225.
  116. Giudice G, Maggio G, Bufano L, et al. Management of Toxic Epidermal Necrolysis with Plasmapheresis and Cyclosporine A: Our 10 Years' Experience. Plast Reconstr Surg Glob Open 2017; 5:e1221.
  117. 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.
  118. Krajewski A, Mazurek MJ, Mlynska-Krajewska E, et al. Toxic Epidermal Necrolysis Therapy with TPE and IVIG-10 Years of Experience of the Burns Treatment Center. J Burn Care Res 2019; 40:652.
  119. Miyamoto Y, Ohbe H, Kumazawa R, et al. Evaluation of Plasmapheresis vs Immunoglobulin as First Treatment After Ineffective Systemic Corticosteroid Therapy for Patients With Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis. JAMA Dermatol 2023; 159:481.
  120. 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; 136:1387.
  121. Quirke KP, Beck A, Gamelli RL, Mosier MJ. A 15-year review of pediatric toxic epidermal necrolysis. J Burn Care Res 2015; 36:130.
  122. Rizzo JA, Johnson R, Cartie RJ. Pediatric Toxic Epidermal Necrolysis: Experience of a Tertiary Burn Center. Pediatr Dermatol 2015; 32:704.
  123. Techasatian L, Panombualert S, Uppala R, Jetsrisuparb C. Drug-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in children: 20 years study in a tertiary care hospital. World J Pediatr 2017; 13:255.
  124. Iriarte C, Karim SA, Nassim JS, et al. Infantile Stevens Johnson syndrome and toxic epidermal necrolysis: A systematic review of clinical features and outcomes in children ages 12 months and under. Pediatr Dermatol 2022; 39:876.
  125. Wasuwanich P, So JM, Chakrala TS, et al. Epidemiology of Stevens-Johnson syndrome and toxic epidermal necrolysis in the United States and factors predictive of outcome. JAAD Int 2023; 13:17.
  126. Papo M, Valeyrie-Allanore L, Razazi K, et al. Renal replacement therapy during Stevens-Johnson syndrome and toxic epidermal necrolysis: a retrospective observational study of 238 patients. Br J Dermatol 2017; 176:1370.
  127. Ezaldein H, Totonchy M, Chow C, et al. The effect of comorbidities on overall mortality in Stevens- Johnson Syndrome: an analysis of the Nationwide Inpatient Sample. Dermatol Online J 2017; 23.
  128. Lee HY, Tharmotharampillai T, Pang SM. Recurrence of Stevens-Johnson syndrome and toxic epidermal necrolysis in adults. Int J Dermatol 2017; 56:e78.
  129. Guillaume JC, Roujeau JC, Revuz J, et al. The culprit drugs in 87 cases of toxic epidermal necrolysis (Lyell's syndrome). Arch Dermatol 1987; 123:1166.
  130. Finkelstein Y, Soon GS, Acuna P, et al. Recurrence and outcomes of Stevens-Johnson syndrome and toxic epidermal necrolysis in children. Pediatrics 2011; 128:723.
  131. Finkelstein Y, Macdonald EM, Li P, et al. Recurrence and mortality following severe cutaneous adverse reactions. JAMA 2014; 311:2231.
  132. Sheridan RL, Schulz JT, Ryan CM, et al. Long-term consequences of toxic epidermal necrolysis in children. Pediatrics 2002; 109:74.
  133. Hoffman M, Chansky PB, Bashyam AR, et al. Long-term Physical and Psychological Outcomes of Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis. JAMA Dermatol 2021; 157:712.
  134. Lian SB, Oh CC, Yeo YW, Lee HY. Spectrum of Nail Sequelae in Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis. JAMA Dermatol 2021; 157:117.
  135. De Rojas MV, Dart JK, Saw VP. The natural history of Stevens Johnson syndrome: patterns of chronic ocular disease and the role of systemic immunosuppressive therapy. Br J Ophthalmol 2007; 91:1048.
  136. Van Zyl L, Carrara H, Lecuona K. Prevalence of chronic ocular complications in Stevens-Johnson syndrome and toxic epidermal necrolysis. Middle East Afr J Ophthalmol 2014; 21:332.
  137. Tougeron-Brousseau B, Delcampe A, Gueudry J, et al. Vision-related function after scleral lens fitting in ocular complications of Stevens-Johnson syndrome and toxic epidermal necrolysis. Am J Ophthalmol 2009; 148:852.
  138. Gaultier F, Rochefort J, Landru MM, et al. Severe and unrecognized dental abnormalities after drug-induced epidermal necrolysis. Arch Dermatol 2009; 145:1332.
  139. Wilson EE, Malinak LR. Vulvovaginal sequelae of Stevens-Johnson syndrome and their management. Obstet Gynecol 1988; 71:478.
  140. Emberger M, Lanschuetzer CM, Laimer M, et al. Vaginal adenosis induced by Stevens-Johnson syndrome. J Eur Acad Dermatol Venereol 2006; 20:896.
  141. Marquette GP, Su B, Woodruff JD. Introital adenosis associated with Stevens-Johnson syndrome. Obstet Gynecol 1985; 66:143.
  142. Kamada N, Kinoshita K, Togawa Y, et al. Chronic pulmonary complications associated with toxic epidermal necrolysis: report of a severe case with anti-Ro/SS-A and a review of the published work. J Dermatol 2006; 33:616.
  143. Park H, Ko YB, Kwon HS, Lim CM. Bronchiolitis obliterans associated with Stevens-Johnson syndrome: a case report. Yonsei Med J 2015; 56:578.
  144. Duong TA, de Prost N, Ingen-Housz-Oro S, et al. Stevens-Johnson syndrome and toxic epidermal necrolysis: follow-up of pulmonary function after remission. Br J Dermatol 2015; 172:400.
  145. Lefaucheur JP, Valeyrie-Allanore L, Ng Wing Tin S, et al. Chronic pain: a long-term sequela of epidermal necrolysis (Stevens-Johnson syndrome/toxic epidermal necrolysis) - prevalence, clinical characteristics and risk factors. J Eur Acad Dermatol Venereol 2021; 35:188.
  146. Dodiuk-Gad RP, Olteanu C, Feinstein A, et al. Major psychological complications and decreased health-related quality of life among survivors of Stevens-Johnson syndrome and toxic epidermal necrolysis. Br J Dermatol 2016; 175:422.
  147. Hefez L, Zaghbib K, Sbidian E, et al. Post-traumatic stress disorder in Stevens-Johnson syndrome and toxic epidermal necrolysis: prevalence and risk factors. A prospective study of 31 patients. Br J Dermatol 2019; 180:1206.
  148. Roujeau JC. Immune mechanisms in drug allergy. Allergol Int 2006; 55:27.
  149. Chaby G, Valeyrie-Allanore L, Duong TA, et al. Severe cutaneous adverse reactions due to inappropriate medication use. Br J Dermatol 2018; 179:329.
  150. Strom BL, Schinnar R, Apter AJ, et al. Absence of cross-reactivity between sulfonamide antibiotics and sulfonamide nonantibiotics. N Engl J Med 2003; 349:1628.
  151. Hung SI, Chung WH, Liu ZS, et al. Common risk allele in aromatic antiepileptic-drug induced Stevens-Johnson syndrome and toxic epidermal necrolysis in Han Chinese. Pharmacogenomics 2010; 11:349.
  152. Tangamornsuksan W, Chaiyakunapruk N, Somkrua R, et al. Relationship between the HLA-B*1502 allele and carbamazepine-induced Stevens-Johnson syndrome and toxic epidermal necrolysis: a systematic review and meta-analysis. JAMA Dermatol 2013; 149:1025.
Topic 2092 Version 47.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟