Open access peer-reviewed chapter

Perspective Chapter: Drug-Induced Severe Cutaneous Adverse Reactions, Diagnostics and Management

Written By

Miteshkumar Rajaram Maurya, Renuka Munshi, Sachin Bhausaheb Zambare and Sanket Thakur

Submitted: 23 June 2022 Reviewed: 18 October 2022 Published: 12 November 2022

DOI: 10.5772/intechopen.108651

From the Edited Volume

Immunosuppression and Immunomodulation

Edited by Rajeev K. Tyagi, Prakriti Sharma and Praveen Sharma

Chapter metrics overview

591 Chapter Downloads

View Full Metrics

Abstract

Severe cutaneous Adverse Reactions (SCAR) are rare drug hypersensitivity reactions but can be life-threatening if not appropriately and timely managed. Many research studies have shed light on its pathomechanism and triggers that have helped us better understand SCAR. The presence of viral fever and genetics such as HLA genotype with certain drugs have been associated with the occurrence of SCAR. However, the basis of interaction of these causative agents needs further evaluation to understand the predisposition to the reaction occurrence. The different spectrum of SCAR needs to be clinically diagnosed appropriately which includes Drug Reactions with Eosinophilia and Systemic Symptoms (DRESS), Steven Johnson Syndrome/Toxic Epidermal Necrolysis (SJS/TEN), Acute Generalized Exanthematous Pustulosis (AGEP), and generalized bullous fixed drug eruptions (GBFDE). However, due to the rare occurrence of this reaction, there is not sufficient evidence for the best treatment for patients suffering from SCAR. Our review provides detailed information about the disease type, manifestation, pathophysiology, diagnostics, and current treatment aspects of SCAR.

Keywords

  • SCAR
  • adverse drug reaction
  • cutaneous eruptions
  • Steven Johnson syndrome
  • toxic epidermal necrolysis
  • acute generalized exanthematous pustulosis
  • clinical pharmacologists

1. Introduction

Since time immemorial, medications come with some benefits and risks. However the intention of treating patients to cure their ailments remains the utmost priority of all the physicians. Thus stands true, the famous dictum by Hippocrates (460–370 BC) that states “Primum non-nocere” which means first of all be sure you do no harm and benefit come next. The reality however is that at times, risk and benefit cannot be separated out so what we follow today is as long as the benefit outweighs the risk, we are ready to take risks in every walk of life similar to what we do for medicines. Severe Cutaneous Adverse Reactions (SCARs) are drug associated hypersensitivity reactions that involves skin and mucous membranes of various body orifices such as eyes, ears, the inner surface of the nose, buccal mucosa, and lips and may involve damage to internal organs usually mediated by drug specific T lymphocytes [1, 2]. The phrase “Skin is Like an Ocean’s Surface Which Tells Deep Stories If You Watch Carefully” goes very well for SCARs which is a multi-spectrum disease due to its variable manifestations. SCAR has been classified into five main types- Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS)/Drug-Induced Hypersensitivity Syndrome (DIHS), Steven Johnson Syndrome (SJS), Toxic Epidermal Necrolysis (TEN), SJS/TEN overlap syndrome and Acute Generalized Exanthematous Pustulosis (AGEP) [3]. Severe Cutaneous Adverse Reactions (SCAR) terminology was proposed for such conditions, as they were (a) severe, (b) unpredictable, and (c) drug-induced [4]. The pathophysiology remains almost similar in all these five types of severe cutaneous adverse reactions. However, immunological trigger due to viral, drug, and gene interaction is still unknown that requires intensive research. Even the predictability, diagnosis, and treatment remains challenging and uncertain for most dermatologist, immunologists, and clinical pharmacologists.

1.1 Classification of severe cutaneous adverse drug reactions (SCAR) provided by the World Allergy Organization (2014)

  1. Steven Johnson Syndrome

  2. Toxic Epidermal Necrolysis

  3. Steven Johnson Syndrome/Toxic Epidermal Necrolysis Overlap Syndrome [5].

  4. Drug-induced Hypersensitivity Syndrome (DIHS) or Drug reaction with Eosinophilia and Systemic Symptoms (DRESS)

  5. Acute Generalized Exanthematous Pustulosis (AGEP)

1.2 Coombs & Gell’s classification of hypersensitivity skin reactions

Table 1 is summarized below.

Hypersensitivity reactionPredominant inflammatory cellsClinical Conditions
Type IImmediate, IgE-mediated mast cell activationAnaphylactic reaction to bee sting, antibiotic like penicillin, latex allergy.
Type IIAntibodies, cytotoxic, IgG-/IgM-mediated, complementHemolytic reactions
Good pasture Syndrome
Hyper acute graft rejections
Type IIImediated by immune complexes and IgG/IgM, complementHypersensitivity Pneumonitis (HP)
Systemic Lupus Erythematosus (SLE)
Polyarteritis Nodosa (PAN)
Serum sickness
Type IVDelayed-type hypersensitivity reactions mediated by T‑helper and T‑cytotoxic cellsChronic Allograft rejection
Purified Protein Derivative (PPD) test
Latex/Nickel/ Poison ivy allergy
Grading of Type IV hypersensitivity skin reactions by Gell and Coombs
Type IVaMonocytesDrug-induced maculopapular exanthems
Type IVbEosinophilsDrug Reaction with Eosinophilic Systemic Symptoms (DRESS)
Type IVcCytotoxicity of drug specific T cellsSteven Johnson Syndrome/Toxic Epidermal Necrolysis
Type IVdNeutrophilsAcute Generalized Exanthematous Pustulosis (AGEP)

Table 1.

Coombs and Gell classification of hypersensitivity cutaneous reactions.

Advertisement

2. Epidemiology of severe cutaneous adverse reactions (SCARs)

The skin is the most common and easily visible body part that manifest symptom of adverse drug reactions [6]. Adverse Drug Reactions (ADRs) contribute up to 7% of the hospital admissions of which cutaneous ADRs alone contribute to 2–3% of the overall hospitalizations [7, 8]. Few of these cutaneous adverse drug reactions have disabling sequelae or can be life-threatening. The incidence of fatalities among inpatients due to systemic and cutaneous adverse drug reactions reported ranges between 0.1% to 0.3%. However, the mortality rates for SJS/TEN are approximately 5–10%, 30–50% in TEN, 10% in DRESS, and the common leading culprit drugs associated with SCARs, around the world are antibiotics, anti-epileptics, allopurinol, Non-steroidal anti-inflammatory drugs (NSAIDS), and antiretrovirals [9, 10, 11]. Incidence rates of SJS/TEN range from 1.4–12.7/million person-years in different studies [12, 13, 14]. A Nationwide Korean Study of Severe Cutaneous Adverse Reactions by Kang DY et al. based on the Multicenter Registry revealed total of 745 SCAR cases (384 SJS/TEN cases and 361 DRESS cases) due to 149 drugs. The main causative drugs suspected were allopurinol (14.0%), carbamazepine (9.5%), vancomycin (4.7%), and anti-tubercular agents (6.3%). Carbonic anhydrase inhibitors (100%), nonsteroidal anti-inflammatory drugs (84%), and acetaminophen (83%) were common offending drugs for SJS/TEN whereas dapsone (100%), antituberculous agents (81%), and glycopeptide antibacterials (78%) were associated with DRESS. The overall mortality rate reported in this study due to SCARs cases was 6.6% (SJS/TEN-8.9% and DRESS-4.2%) [15]. The phase 1 of regiSCAR project by the European community (last updated on October 25, 2014), includes potential 1889 (69.39%) cases of SJS/TEN/GBFDE/EEMM, 364 (13.37%) cases of AGEP, 469 (17.22%) cases of DIHS/DRESS. Of these suspected cases, the definite or probable cases of SJS/TEN- 1232 (65.21%), EEMM- 251 (13.28%), GBFDE- 5 (0.26%), AGEP- 228 (62.63%) and HSS/DRESS- 281 (59.91%) [16].

Advertisement

3. Steven Johnson syndrome/toxic epidermal necrolysis (SJS-TEN) overlap syndrome

3.1 History of SJS and TEN

Steven Johnson Syndrome was first described in 1922 by American pediatrician named Albert Mason Stevens (1884–1945) and Frank Chambliss Johnson (1894–1934) who reported two cases of boys in New York City, United States, aged 7 years and 8 years presenting with an extraordinary generalized skin eruption with continued fever, inflamed buccal mucosa, and severe purulent conjunctivitis [17]. Both these cases were misdiagnosed by primary care physicians as a case of “hemorrhagic measles” or “black measles”. Similarly, the first description of Toxic Epidermal Necrolysis (TEN) also called Lyell syndrome was given by Scottish dermatologist Dr. Alan Lyell (1917–2007) in 1956 in four patients. Initially, these were considered as the toxic eruption that resembled severe burn or scalding of skin associated with erythematous plaques and widespread areas of epidermal detachment and was referred to as necrolysis due to excessive apoptotic keratinocytes [18]. Skin and mucous membranes were involved but with very little inflammation in the dermis referred to as the phenomenon of “dermal silence” by the dermatologist. This was an acute, rare, life-threatening mucocutaneous disease with an annual incidence of approximately 0.4–1.2 cases per million individuals with a mortality rate of more than 30% [19]. SCAR and EuroSCAR pooled data analysis was performed for children under 15 years of age that revealed that anti-bacterials class of drugs such as sulphonamides, antiepileptics such as phenobarbitol, lamotrigine, and carbamazepine was found to be strongly associated with SJS/TEN in this pediatric population [20]. Following is the case of toxic epidermal necrolysis involving the face and neck (Figure 1), trunk, and all four limbs (Figure 2) in an elderly female on cefixime and paracetamol treatment for dengue fever with no pre-existing comorbidities.

Figure 1.

Epidermal involvement of the face and neck.

Figure 2.

Epidermal involvement of limbs.

Advertisement

4. Criteria for the diagnosis of SJS/TEN based on extent of epidermal detachment

Steven Johnson Syndrome is an immune complex-mediated hypersensitivity reaction that involves mucocutaneous body areas such as oral, nasal, ocular, vaginal, urethral, gastrointestinal, and lower respiratory tract mucous membranes. Moreover, gastrointestinal and lower respiratory tract infections may progress to necrosis. The diagnosis of steven johnson syndrome, Toxic epidermal necrolysis, or overlap syndrome totally depends on the afflicted skin body surface area with epidermal detachment (Figure 3) [21].

  • Steven Johnson syndrome (SJS)- < 10% of the body surface area involvement

  • SJS/TEN overlap syndrome- 10 − 30% of the body surface area involvement

  • Toxic Epidermal Necrolysis (TEN)- >30% of the body surface area involvement

Figure 3.

The extent of epidermal detachment in SJS/TEN. (adapted from fig 21.9 Bolognia and Bastuji-Garin S. et al. arch Derm 129: 92, 1993).

Distinguishing features of different severe bullous skin lesions are tabulated below (Table 2) [22].

ClassificationBullous Erythema multiforme (EM)Steven Johnson Syndrome (SJS)Overlap- Steven Johnson syndromeToxic epidermal necrolysis (TEN) with spotsToxic epidermal necrolysis (TEN) without spots
Epidermal Detachment<10% BSA<10% BSA10–30% BSA>30% BSA>10% BSA
Typical TargetsYesNoNoNoNo
Atypical TargetsRaisedFlatFlatFlatNo
SpotsNoYesYesYesNo

Table 2.

Distinguishing features of different severe bullous skin lesions.

Abbreviations: BSA- Body Surface Area, EM- Erythema Multiforme, SJS- Steven Johnson Syndrome, TEN - toxic epidermal necrolysis. Adapted from Bastuji-Garin S et al. Clinical classification of cases of toxic epidermal necrolysis, Stevens-Johnson syndrome, and erythema multiforme. Arch Dermatol 1993; 129(1): 92–96 (adapted from Bastuji-Garin S et al.).

Advertisement

5. Proposed immunological models of T cell activation in Stevens-Johnson syndrome/toxic epidermal necrolysis

There are three proposed models of T cell activation in Stevens-Johnson syndrome/toxic epidermal necrolysis due to drugs (Figure 4) [23].

  1. Hapten/pro-hapten model: drugs/drug metabolites form complex with carrier proteins and are presented as haptenated peptides in the peptide-binding groove of HLA molecules (covalent).

  2. p-i concept: drugs directly bind to HLA & TCR non-covalently.

  3. Altered peptide model: drugs bind to the peptide-binding groove of HLA, resulting in alteration of HLA-binding peptide repertoire.

Figure 4.

Models of T cell activation in Steven Johnson syndrome and toxic epidermal necrolysis. Abbreviations: APC- antigen presenting cells, HLA- human leukocyte antigen, TCR- T cell receptor, HLA- human leukocyte antigen. Adapted from: Hasegawa a and Abe R. recent advances in managing and understanding Stevens-Johnson syndrome and toxic epidermal necrolysis (version 1; peer review: 2 approved). F1000Research 2020, 9(F1000 faculty rev):612.

Advertisement

6. Pathophysiology of occurrence of SJS/TEN

  1. Drug induced upregulation of Fas-ligand by keratinocytes expressing Fas lead to activation of death receptor-mediated apoptotic pathway [24, 25].

  2. Drug interaction with MHC class I-expressing cells leads to drug-specific CD8 + cytotoxic T cells accumulating within the epidermal blisters releasing perforin & granzyme B that kill keratinocytes [26].

  3. Drug-activated monocytes secrete annexin A1, which induces necroptosis in keratinocytes [11].

  4. Drug triggers activation of CD8 + T cells, NK (Non-Killer) T cells to secrete granulysin leading to keratinocyte death without the need for cell contact [27].

6.1 Histopathologic features

Histopathologic features in SJS/TEN show apoptotic keratinocytes present individually and in clusters within the epidermis. It may also show dendritic cell infiltration, spongiform superficial epidermal pustules, edema of the papillary dermis, and perivascular infiltrates of lymphocytes (Figure 5). Subtle vacuolar changes along the basal layer are accompanied by minimal inflammation with scattered lymphocytes within the epidermis [28].

Figure 5.

Histopathologic features in SJS/TEN show apoptotic keratinocytes present individually and in clusters within the epidermis.

6.2 SCORTEN scale (SCORe of toxic epidermal necrosis) for predicting mortality risk in SJS/TEN

SCORTEN scale is the severity-of-illness scale that measures the severity of certain bullous conditions and can be helpful in systematically determining the disease/reaction prognosis. The table below (Table 3) provides details of seven independent risk factors that need to be evaluated within the first 24 hours of admission or within the first 5 days of reaction that will add to accuracy in predicting mortality. The presence of prognostic factors is given a score of 1 and if not present then assign the score of 0. SCORTEN score is the summation of these individual scores allotted to each of the seven prognostic factors based on their presence or absence that gives the overall prognosis in terms of percentage mortality. More risk factors present, the higher the SCORTEN score, and the higher the mortality rate [29].

Serial numberPrognostic FactorsScoreSCORTEN score% Mortality rate
1.Age > 40 years10–13
2.Tachycardia >120 bpm1212
3.Neoplasia1335
4.Initial detachment >10%1458
5.Serum Urea>10 mmol/l1>490
6.Serum Bicarbonate<20 mmol/l1
7.Blood glucose >14 mmol/l1

Table 3.

Seven independent risk factors for calculating SCORTEN score and predicting mortality risk in SJS/TEN.

Advertisement

7. Algorithm for drug causality for epidermal necrolysis (AlDeN) scale for causality assessment

Table 4 is summarized below.

CriteriaValuesRules to applyValue range
Delay from initial drug component intake to onset of reaction (index day)Suggestive +3From 5 to 28 days-3 to 3
Compatible +2From 29 to 56 days
Likely +1From 1 to 4 days
Unlikely −1>56 days
Excluded −3Drugs started on or after the index day
In case of previous reaction to the same drug only changes for – Suggestive +3: from 1 to 4 days
Likely +1: from 5 to 56 days
Drug present in the body on index dayDefinite 0Drug continued up to index day or stopped at a time point less
than five times the elimination half-life before the index day
-3 to 0
Doubtful −1Drug stopped at a time point prior to the index day by more than
five times the elimination half-lifea but liver or kidney function
alterations or suspected drug interactions are present
Excluded −3Drug stopped at a time point prior to the index day by more
than five times the elimination half-life, without liver or kidney
function alterations or suspected drug interactions
Prechallenge/rechallengePositive specific for
disease and drug: 4
SJS/TEN after use of same drug-2 to 4
Positive specific for
disease or drug: 2
SJS/TEN after use of similar drug or other reaction with
same drug
Positive unspecific: 1Other reaction after use of similar drug
Not done/unknown: 0No known previous exposure to this drug
Negative −2Exposure to this drug without any reaction (before or after
reaction)
DechallengeNeutral 0Drug stopped (or unknown)-2 to 0
Negative −2Drug continued without harm
Type of drug (notoriety)Strongly associated 3Drug of the “high-risk” list according to previous case–control studies-1 to 3
Associated 2Drug with definite but lower risk according to previous
case–control studies
Suspected 1Several previous reports, ambiguous epidemiology
results (drug “under surveillance”)
Unknown 0All other drugs including newly released ones
Not suspected −1No evidence of association from previous epidemiology
study with sufficient number of exposed controls
Intermediate score = total of all previous criteria−11 to 10
Other causePossible −1Rank all drugs from highest to lowest intermediate score−1
If at least one has an intermediate score > 3, subtract 1 point
from the score of each of the other drugs taken by the patient
(another cause is more likely)
Final score − 12 to 10
Causality assessment scale
<0, Very unlikely; 0–1, unlikely; 2–3, possible; 4–5, probable; ≥6, very probable.

Table 4.

Details of AlDeN scale for causality assessment (algorithm for drug causality for epidermal necrolysis).

Advertisement

8. Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS)/Drug-induced Hypersensitivity Syndrome (DIHS)

This DRESS is a life-threatening drug hypersensitivity reaction that can manifest with fever, cutaneous eruptions, and internal organ involvement [30]. Though the disease has a 10% mortality risk, it involves more than 50% of body surface area with skin lesions showing infiltrative papules and plaques with purpuric changes. This may be associated with other clinical features such as facial edema, desquamation during the resolution stage, and mucosal lesion involving the mouth and lips most commonly. Systemic symptoms depend on the organ involved. Eosinophilia (66–95%) is the most common hematological abnormality noted followed by atypical lymphocytosis (27–67%), lymphadenopathy (54%), and the liver is the most common internal organ involved followed by kidney, lung, and heart. The onset of skin reactions is usually observed after 3–8 weeks of the start of suspect drugs. The drugs implicated in such drug reactions are anti-convulsants, anti-infectious agents (anti-tuberculosis, antibiotics, and antiviral agents), sulfonamides, and uric acid-lowering medications. Usually, DRESS lasts for more than 15 days with prolonged courses with flare-ups observed after Human Herpes Virus type 6 reactivation. The immunopathology blamed here is the antiviral and anti-drug immune responses contribute to the disease presentations but the exact interaction is still unclear. Sequelae of this disease could be fulminant type 1 diabetes mellitus, thyroid disorders, autoimmune diseases, or permanent renal dysfunction that needs dialysis [31].

Advertisement

9. Acute generalized exanthematous pustulosis (AGEP)

The clinical characteristics of Acute Generalized Exanthematous Pustulosis (AGEP) are typical with multiple sterile, non-follicular pustules on oedematous erythema mostly on major skin folds. It may be associated with facial edema, blisters, or atypical target lesions. Mucosal lesions are rare and mild. There may be fever and leukocytosis associated with cutaneous eruptions. Systemic involvement is rare (<20%) but may affect the liver followed by the kidney, lung, and bone marrow. The culprit drugs that are strongly associated with these reactions are pristinamycin, ampicillin/amoxicillin, quinilones, hydroxychloroquine, anti-infective sulfonamides, terbinafine, and diltiazem based on multinational case–control EuroSCAR study. The duration between drug intake and occurrence of skin eruption may take a median duration of 1 day in the case of antibiotics and 11 days for other medications. AGEP resolves without any sequelae and has a good prognosis as well [32]. The diagnostic criteria for Acute Generalized Exanthematous Pustulosis (AGEP) are as follows [33, 34] – 1) Acute pustular eruption 2) Fever >38 degrees Celsius 3) Neutrophilia with or without eosinophilia 4) Sub corneal or intradermal pustules on biopsy 5) Spontaneous resolution in less than 15 days.

Advertisement

10. Diagnostic tests for cutaneous drug hypersensitivity

Drug associated cutaneous hypersensitivity reaction requires an adequate clinical and physical examination approach with an evaluation of historical details. In addition to clinical history and examination, the diagnostic test will help to confirm the diagnosis of SCARs. Also, it will help us to determine if the initial reaction is IgE or non-IgE mediated. A diagnostic test can be classified into In Vitro tests and In Vivo tests.

In Vitro Tests: The quantification of various chemokines (histamine, tryptase, leukotrienes) from a different sample such as peripheral blood, nasal or bronchial secretions, urine sample serves as useful means to differentiate between the immediate and delayed types of hypersensitivity reactions (For details, refer Table 5). The Allergen challenge test may be formed to see the difference in the level of chemical mediators from the baseline levels. In case of acute anaphylactic reactions, the test can serially measure serum total tryptase levels, at 1 and 6 hours but is not completely reliable as normal levels may be detected in fatal cases of anaphylaxis. Even plasma histamine levels drop after 1 hour of anaphylactic symptoms making this test not reliable. Moreover, these test kits are expensive.

Function of type of in vitro testTest nameDrug names/Disease ConditionsLimitation of the test/Inferences
Allergen-specific IgE levelsradioallergosorbent tests (RASTs) or radioimmunoassay (RIA) or fluorescent enzyme immunoassay (FEIA) tests (ImmunoCAP®)penicilloyl, amoxicilloyl, ampicilloyl, cefaclor, protamine, insulin, suxamethonium, neuromuscular blocking agents (NMBAs), and chlorhexidinespecific, lack sensitivity compared to clinical history and/or skin tests.
(in clinical practice, a utility for these tests is limited).
CD 63 and CD 203c levels on activated basophils.Flow cytometry-based basophil activation assays (also known as flow cellular antigen stimulation tests [CASTs])beta-lactam, NSAIDs, fluoroquinolones, iodinated contrast media, proton pump inhibitors, NMBAs, and chemotherapeutical agentstechnical concerns, false-positive results, and lack of sensitivity and specificity. (not widely used)
Peripheral blood eosinophilia or elevated total IgE levels- presence or absence.Complete Hemogram profile and Serum IgE levels in the blooddrug hypersensitivity syndromes.not useful in the diagnosis or exclusion of drug allergies.
Positive direct Coomb’s testhematological manifestations of drug hypersensitivity e.g. immune-mediated haemolytic anemia, leukopaenia, thrombocytopaeniano specific diagnostic test or serological test apart from recovery of the cytopaenia following the withdrawal of the putative drug, Drug-induced IgM and IgG have not been found to be clinically useful
Lymphocyte transformation test (LTT)diagnosis of reactions in a wide variety of delayed reactions with a wide variety of drugsT-cell mediated delayed hypersensitivity against a wide variety of drugspositive LTT is useful in confirming the diagnosis, a negative test cannot exclude drug hypersensitivity. Positive LTT is usually drug-specific, and reaction-specific.

Table 5.

The following table provides information about the in-vitro tests.

In Vivo Tests: Many skin tests are available as useful tools in the diagnosis of IgE-mediated allergy. A skin prick test (SPT) is positive if the mean wheal diameter is≥3 mm after 15 to 20 minutes (associated with a flare response) compared to the negative control. Similarly, the intradermal test (IDT) is positive when meaning wheal diameter is ≥3 mm compared to the baseline diameter for the negative control after 15 to 20 minutes of the intradermal administration of allergen (0.02 to 0.05 ml). Though IDT is more sensitive than the skin prick test, there is still a higher risk of irritation, false positive reaction, and IgE-dependent anaphylactic reactions. The observation for immediate hypersensitivity reaction usually appears in 15 to 20 minutes while 24 to 72 hours’ evaluation is required for non-immediate (late) reactions. Patch tests are used for the diagnosis of delayed hypersensitivity drug reactions. In these tests, a patch embedded with the suspected allergen is fixed on the back of the patient for 1 to 2 days and the result is read after 1 day and/or after 2 to 3 days. A photo-patch test is a modified patch test used to diagnose suspected photoallergic or phototoxic reactions. This patch is removed after a day and a skin area of almost 10 J/cm2 is irradiated with ultraviolet A light and the results are read on days 2, 3, and 4. The use of non-irritating skin test concentrations is recommended for SPT, IDT, and patch tests. In very rare cases, a skin biopsy may be helpful in differential diagnoses of skin conditions with typical histological patterns. For instance, connective tissue disease is characterized by interface dermatitis with epidermal atrophy, focal parakeratosis, dermal mucinosis, and fibrinoid deposition in the dermis [35, 36]. Drug provocation (challenge) tests (DPTs) are performed using suspected agents and objectively reproduce the patient’s symptoms and signs of hypersensitivity. However, the positive test does not confirm an immune-mediated reaction. For DPTs, the drug is given using slow, incremental dose escalations at fixed time intervals and observing for the presence or absence of an objective reaction under the strict supervision of trained clinicians/nurses with well-equipped resuscitative tools. The DPT can help to exclude drug hypersensitivity when the history is nonsuggestive or the symptoms nonspecific. To definitively diagnose drug allergy when the clinical history is suggestive but allergological tests are negative, inconclusive, or unavailable. Specific contraindications to DPT include pregnancy; comorbidities in which DPT may provoke the medical situation beyond the ability to control it (e.g., acute infections; uncontrolled asthma; or underlying cardiac, hepatic, or renal diseases); immunobullous drug eruptions; and cases in which the initial reaction was a severe cutaneous and/or systemic reaction (e.g., SJS and TEN). The risks and benefits of any DPT must be explained to the patient and informed consent obtained. Short-acting antihistamines (e.g., chlorpheniramine or hydroxyzine) should be stopped for 3 days and long-acting antihistamines (e.g., cetirizine, loratadine, or fexofenadine) for 7 days before performing any DPT. Patients should also be fasted overnight and carefully observed at all times during the DPT for symptoms or signs of an adverse reaction. Resuscitation equipment should be available at all times, and staff should be trained in the management of acute anaphylaxis [37].

11. Management of Severe Cutaneous Adverse Reactions

11.1 Treatment of SJS/TEN

11.1.1 General management/supportive care

Correct identification of the causative drug and immediate withdrawal of potentially causative drugs. May use the ALDEN algorithm to determine the culprit drug. Supportive management of skin wounds with anti-shear dressings, nutrition status, electrolyte balance, renal and airway function, and adequate pain control, prevent or treat the wound infections. Refer to the specialized unit/burn center for supportive care, silver wraps, apply emollients, air fluidized beds, and prevent infections. Fluid balance is very important to prevent end-organ hypo perfusion with daily monitoring of urine output (maintain 0.5–1.0 ml/kg/hr) or intra-arterial hemodynamic monitoring. Adequate nutrition supplement for protein loss- 20-25 kcal/kg/day in the early phase and 25–30 kcal/kg/day in the recovery phase is recommended either through oral intake or nasogastric feed. Analgesic care with acetaminophen in mild cases or opiod-based analgesics based on the severity of pain [38, 39].

11.1.2 Specific treatment for SJS/TEN

Corticosteroids- Systemic corticosteroids have shown non-inferiority when compared with supportive care in treating patients with SJS. However, in cases of TEN, many studies have shown survival benefits to the patient but some studies have also shown a lack of efficacy and also an increase in mortality. High doses of systemic steroids have shown to be more effective in TEN patients as recommended by Japanese experts. Araki et al have successfully used corticosteroid pulse therapy (methylprednisolone 500 mg/day for 3 days in 5 patients of TEN and all survived also supported by one recent published meta-analysis suggesting systemic corticosteroids as promising immunomodulating therapies for SJS/TEN [40].

Intravenous Immunoglobulin- some studies have shown the survival benefit of Intravenous immunoglobulins (IVIG) with a dose of 2.8 g/kg up to 4 g/kg [41, 42]. Those studies with no survival benefit used doses mostly up to 2 g/kg or lower [43]. Huang et al. performed the first meta-analysis on the efficacy of IVIg for the treatment of TEN showing the benefit of the use of high dose IVIG over low dose IVIG [44]. But recent published reviews and meta-analyses showed no differences in mortality with IVIG compared with only supportive care [45, 46].

Cyclosporine- Inhibits CD8+ cytotoxic T cells with anti-apoptotic effect by inhibition of Fas ligand. Cyclosporine 3 mg/kg for 10 days with gradual tapering over 1 month revealed less skin detachment and a lower mortality rate in one of the pilot studies recruiting 29 patients of SJS/TEN. Chen et al in a meta-analysis found significantly lower mortality than calculated as per SCORTEN score in patients receiving cyclosporine (OR: 0.42, 95% CI- 0.19-0.95) [47, 48]. Zimmermann et al. meta-analysis also showed a better reduction in mortality with the use of cyclosporine when compared with just supportive care. However large-scale randomized controlled studies are required to confirm these findings [49].

Anti-TNF alpha agents: Increase in TNF-alpha in skin specimens, skin blister fluids, and in serum of SJS/TEN patients have been observed and hence the role of anti-TNF alpha agents may prove effective. Infliximab and Etanercept use have shown to be effective as published in many case reports and case series with better survival outcomes. Only one trial by Wolkenstein et al that used Thalidomide for treatment of SJS/TEN was prematurely terminated in view of the increase in mortality observed in patients [50, 51].

Plasmapheresis: Plasmapheresis is known to filter the harmful mediators in blood and have shown dramatic improvement in a patient with SJS/TEN. Narira et al study demonstrated that the use of plasmapheresis in patients refractory to conventional therapy treatment reduced the interleukin levels (IL-6, 8, and TNF-alpha) and is recommended for use by Japanese experts in TEN patients refractory to high dose corticosteroids [52].

11.2 Treatment for drug reaction with eosinophilia and systemic symptoms (DRESS) or drug-induced hypersensitivity syndrome (DIHS)

  • Supportive care is required with hydration maintenance in most of the cases except those with detected elevated levels of auto-antibodies.

  • Systemic corticosteroids such as prednisolone with starting dose of 0.5 to 1 mg/kg/day gradually tapering over 2–3 months suggested by some experts known to decrease flare-up episodes and auto-immune sequelae.

  • However, there is a risk for higher rates of infections, and septicemia and may require intensive care management. Hence suggested use only in those with severe presentations.

  • French group of Dermatology recommends the use of systemic use of corticosteroids for those with a 5-fold increase in serum transaminases level or if there is the involvement of another organ such as kidney, lung, and heart.

  • The results of the use of IVIG are conflicting and its use as monotherapy should be avoided. Several immunosuppressants like cyclosporine, cyclophosphamide, mycophenolate mofetil, and rituximab have been proposed in addition to systemic corticosteroids or IVIG in patients with severe disease and viral reactivation (Human Herpes virus −6 activation has been implicated as a trigger for this reaction) [33].

11.3 Acute generalized Exanthematous Pustulosis (AGEP)

  • Identification and removal of the culprit drugs are sufficient and skin lesions resolve in 6–8 days after suspect drug withdrawal [53].

  • Hospitalization may be required in some patients and may be treated with topical and systemic corticosteroids if required. The beneficial effects of systemic steroids need further evaluation.

11.4 Generalized bullous fixed drug eruptions (GBFDE)

  • Prompt identification and withdrawal of causative suspect drugs remains the mainstay management of this reaction [54].

  • Systemic corticosteroids may be beneficial in severe cases though there is a lack of sufficient evidence to compare with other treatment modalities and supportive care.

12. Future directions using pharmacogenetic tools to identify genetic predisposition to SCARs

The occurrence of SCAR is unpredictable but this uncertainty can be minimized to some extent by performing a pharmacogenetic assessment. If done before initiating patients on potential drugs, the occurrence of SCAR can be prevented as well. Human Leukocyte Antigens (HLA) genes have been evaluated in detail that has been found to have an association with SCAR variations observed in some patients on specific drugs (listed in Table 6). Having knowledge about genetic predisposition will help to detect patients at higher risk of developing SCAR. A study by Esmaeilzadeh H. et al study from Iran has shown Steven Johnson Syndrome (SJS)/Toxic Epidermal Necrolysis (TEN) as the most common drug-induced SCAR presentation and the most common culprit drug as beta-lactam antibiotics followed by carbamazepine. The presence of HLA-A*02:01 and A*51:01 was also shown to increase the risk of SCAR while A*11:01 had a protective role against SCAR. Those with HLA-A*02:01, HLA-A*24:02, and HLA-B*51:01 were an increased SJS as observed in the same study [55].

Predisposing Infectious diseasesDrug-inducedMalignancy relatedHLA allele associated with the risk to specific drugs
AIDS (Acquired Immunodeficiency syndrome)Antiepileptics: Phenytoin, Carbamazepine, Oxcarbazepine, Valproic acid, LamotrigineHematologicalHLA-B*15:02- Carbamazepine
Coxsackie and Dengue virus infectionBarbituratesLung cancerHLA-A*31:01- Carbamazepine
Influenza, Herpes Simplex virusAnti-retrovirals:
NRTI-Nevirapine
NNRTIs - Indinavir
Malignant LymphomaHLA-B*57:01- Allopurinol
Abacavir
Bacterial Infection: Gr A beta-hemolytic streptococci, Diphtheria, Brucellosis, LGV, MycobacteriaNSAIDS: oxicams –meloxicam, piroxicam,
Paracetamol
Urothelial CarcinomaHLA-A*32:01
Vancomycin
Fungal infection: coccidiomycosis, dermatophytosis, trichomoniasisTNF alpha antagonist: Etanercept, Infliximab, AdalimumabHepatocellular Carcinoma

Table 6.

Predisposing factors for Steven Johnson syndrome/toxic epidermal necrolysis.

Abbreviations: AIDS- Acquired immune deficiency syndrome, LGV- Lymphogranuloma venereum, NRTI- Nucleoside reverse transcriptase inhibitors, NNRTIs- Non-nucleoside reverse transcriptase inhibitors, NSAIDs- Non-steroidal anti-inflammatory drugs, TNF alpha- Tumor Necrosis Factor-alpha, HLA- Human leukocyte antigens.

13. Conclusion

Among the various pathophysiology suggested for SCARs, T cells play a major role in the occurrence of delayed hypersensitivity reactions presenting as SCAR variants. There is a need to select structurally different classes of antibiotics in case of antibiotic-induced severe cutaneous adverse reaction (SCAR) to avoid recurrence. Furthermore, there is a need to conduct research and explore the immune mechanism of viral-drug-gene interaction and develop drugs to modulate T cells/other cell lineages/novel therapeutic targets. Animal models for SCAR variants may be an important step toward the development of new drugs but are understudied. Nevertheless, rational use of antibiotics should be promoted as well as implemented into practice by consumers and health care physicians. The important concern with Immunomodulators and targeted therapies is associated with long-term sequelae such as immune reactive inflammatory syndrome and polyglandular autoimmune syndrome III. Validation of more prognostic HLA and other biomarkers for guiding therapy may help to prevent the occurrence of scars. There is a need to develop multicentric, multi-ethnic, and multi-regional registries that will enable us to gather clinical and demographic profiles, along with histopathological, genomic, proteomic, and metabolomic evaluation supplemented with a data mining approach for a better understanding of signals generated. The metabolomic approach may be helpful to detect the drug or its metabolite may be the causative factor for SCAR severity.

Abbreviations

SCARsSevere Cutaneous Adverse Drug Reactions
AGEPAcute Generalized Eaxnthematous Pustulosis
SJSSteven Johnson Syndrome
TENToxic Epidermal Necrolysis
SJS/TEN OverlapSteven Johnson Syndrome/Toxic Epidermal Necrolysis Overlap syndrome
GBFDEGeneralized Bullous Fluid Drug Eruptions
EEMMErythema Exsudativum Multiforme with Mucosal Involvement
EMErythema Multiforme
DRESSDrug Related Eosinophilia with Eosinophilic Systemic Symptom
WAOWorld Allergy Organization
DIHSDrug Induced Hypersensitivity Syndrome
BCBefore Christ
CDClusters of Differentiation
Ig EImmunoglobulin E
Ig GImmunoglobulin G
Ig MImmunoglobulin M
HPHypersensitivity Pneumonitis
PANPolyarthritis Nodosa
SLESystemic Lupus Erythematosus
PPDPurified Protein Derivative
ADRsAdverse Drug Reactions
NSAIDsNon-Steroidal Anti-Inflammatory Drugs
RIARadioimmunoassay
TCRT Cell Receptor
APCAntigen Presenting Cells
FEIAFluorimetric Enzyme-Linked Immunoassay
SPTSkin Prick Test
OROdds Ratio
IDTIntradermal test
IVIGIntravenous Immunoglobulin
TNFalpha- Tumor Necrosis Factor-alpha
HLAHuman Leukocyte Antigen
AIDSAcquired Immunodeficiency Syndrome
LGVLymphogranuloma Venerum
NRTINucleoside Reverse Transcriptase Inhibitors
NNRTINon-Nucleoside Reverse Transcriptase Inhibitors
LTTLymphocyte Transformation Test
DPTDrug Provocation Tests
CASTsCellular Antigen Stimulation Tests

References

  1. 1. Adler NR, Aung AK, Ergen EN, Trubiano J, Goh MSY, Phillips EJ. Recent advances in the understanding of severe cutaneous adverse reactions. The British Journal of Dermatology. 2017;177(5):1234-1247
  2. 2. Bellón T. Mechanisms of severe cutaneous adverse reactions: Recent advances. Drug Safety. 2019;42(8):973-992
  3. 3. Shafiq N, Bhattacharjee S, Malhotra S. Severe cutaneous adverse reaction (SCAR). Indian Dermatology Online Journal. 2022;13(1):10-12
  4. 4. Patel RM, Marfatia YS. Clinical study of cutaneous drug eruptions in 200 patients. Indian Journal of Dermatology, Venereology and Leprology. 2008;74:80
  5. 5. Classification of SCAR. Available from: https://www.worldallergy.org/education-and-programs/education/allergic-disease-resource-center/professionals/drug-allergies. [Accessed: June 22, 2022]
  6. 6. Fierer J, Looney D, Pechère JC. Nature and pathogenicity of micro-organisms. Infectious Diseases. 2017:4-25. e1. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7173513/
  7. 7. Wiffen P, Gill M, Edwards J, Moore A. Adverse drug reactions in hospital patients. A systematic review of the prospective and retrospective studies. Bandolier Extra. 2002;2:1-16
  8. 8. Gruchalla R. Understanding drug allergies. The Journal of Allergy and Clinical Immunology. 2000;105:637-644
  9. 9. Schwartz RA, McDonough PH, Lee BW. Toxic epidermal necrolysis. Journal of the American Academy of Dermatology. 2013;69(2):187.e1-187.e16
  10. 10. Roujeau JC, Bastuji-Garin S. Systematic review of treatments for Stevens-Johnson syndrome and toxic epidermal necrolysis using the SCORTEN score as a tool for evaluating mortality. Ther Adv Drug Saf. 2011;2:87-94
  11. 11. Roujeau JC. Clinical heterogeneity of drug hypersensitivity. Toxicology. 2005;209(2):123-129
  12. 12. Diphoorn J, Cazzaniga S, Gamba C, Schroeder J, Citterio A, Rivolta AL, et al. Incidence, causative factors and mortality rates of Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) in northern Italy: Data from the REACT registry. Pharmacoepidemiology and Drug Safety. 2016;25:196-203
  13. 13. Hsu DY, Brieva J, Silverberg NB, Silverberg JI. Morbidity and mortality of Stevens-Johnson syndrome and toxic epidermal necrolysis in United States adults. The Journal of Investigative Dermatology. 2016;136:1387-1397
  14. 14. Kang DY, Yun J, Lee SY, Koh YI, Sim DW, Kim S, et al. A Nationwide study of severe cutaneous adverse reactions based on the multicenter registry in Korea. The Journal of Allergy and Clinical Immunology. In Practice. 2021;9(2):929-936.e7
  15. 15. Current status of Phase I of the RegiSCAR project. 2014. Available from: http://www.regiscar.org/Status.html. [Accessed: June 22, 2022]
  16. 16. Stevens AM, Johnson FC. A new eruptive fever associated with stomatitis and ophthalmia: Report of two cases in children. American Journal of Diseases of Children. 1922;24:526-533
  17. 17. Callahan SW, Oza VS. Stevens-Johnson syndrome—A look Back. JAMA Dermatology. 2017;153(2):240
  18. 18. Schwartz RA, McDonough PH, Lee BW. Toxic epidermal necrolysis: Part I. introduction, history, classification, clinical features, systemic manifestations, etiology, and immunopathogenesis. Journal of the American Academy of Dermatology. 2013;69(2):173.e1-173.13 quiz 185-6
  19. 19. Levi N, Bastuji-Garin S, Mockenhaupt M, et al. Medications as risk factors of Stevens-Johnson syndrome and toxic epidermal necrolysis in children: A pooled analysis. Pediatrics. 2009;123(2):e297-e304
  20. 20. Bastuji-Garin S, Rzany B, Stern RS, Shear NH, Naldi L, Roujeau JC. Clinical classification of cases of toxic epidermal necrolysis, Stevens-Johnson syndrome, and erythema multiforme. Archives of Dermatology. 1993;129(1):92-96
  21. 21. Harr T, French LE. Toxic epidermal necrolysis and Stevens-Johnson syndrome. Orphanet Journal of Rare Diseases. 2010;5:39
  22. 22. Hasegawa A, Abe R. Recent advances in managing and understanding Stevens-Johnson syndrome and toxic epidermal necrolysis. F1000Res. 2020;9:612
  23. 23. Itoh N, Yonehara S, Ishii A, Yonehara M, Mizushima S, Sameshima M, et al. The polypeptide encoded by the cDNA for human cell surface antigen Fas can mediate apoptosis. Cell. 1991;66:233-243
  24. 24. Iwai K, Miyawaki T, Takizawa T, Konno A, Ohta K, Yachie A, et al. Differential expression of bcl−2 and susceptibility to anti-Fas-mediated cell death in peripheral blood lymphocytes, monocytes, and neutrophils. Blood. 1994;84:1201-1208
  25. 25. Inachi S, Mizutani H, Shimizu M. Epidermal apoptotic cell death in erythema multiforme and Stevens-Johnson syndrome. Contribution of perforin-positive cell infiltration. Archives of Dermatology. 1997;133:845-849
  26. 26. Saito N, Qiao H, Yanagi T, et al. An annexin A1-FPR1 interaction contributes to necroptosis of keratinocytes in severe cutaneous adverse drug reactions. Science Translational Medicine. 2014;6(245):245-295. DOI: 10.1126/scitranslmed.3008227
  27. 27. Rzany B, Hering O, Mockenhaupt M, Schröder W, Goerttler E, Ring J, et al. Histopathological and epidemiological characteristics of patients with erythema exudativum multiforme major, Stevens-Johnson syndrome and toxic epidermal necrolysis. The British Journal of Dermatology. 1996;135(1):6-11
  28. 28. Bastuji-Garin S, Fouchard N, Bertocchi M, Roujeau JC, Revuz J, Wolkenstein P. SCORTEN: A severity-of-illness score for toxic epidermal necrolysis. The Journal of Investigative Dermatology. 2000;115(2):149-153. DOI: 10.1046/j.1523-1747.2000.00061.x
  29. 29. Sassolas B, Haddad C, Mockenhaupt M, Dunant A, Liss Y, Bork K, et al. ALDEN, an algorithm for assessment of drug causality in Stevens-Johnson syndrome and toxic epidermal necrolysis: Comparison with case-control analysis. Clinical Pharmacology and Therapeutics. 2010;88(1):60-68. DOI: 10.1038/clpt.2009.252
  30. 30. Choudhary S, McLeod M, Torchia D, Romanelli P. Drug reaction with eosinophilia and systemic symptoms (DRESS) syndrome. The Journal of Clinical and Aesthetic Dermatology. 2013;6(6):31-37
  31. 31. Feldmeyer L, Heidemeyer K, Yawalkar N. Acute generalized Exanthematous Pustulosis: Pathogenesis, genetic background, clinical variants and therapy. International Journal of Molecular Sciences. 2016;17(8):1214. DOI: 10.3390/ijms17081214
  32. 32. Sidoroff A, Halevy S, Bavinck JN, Vaillant L, Roujeau JC. Acute generalized exanthematous pustulosis (AGEP)--a clinical reaction pattern. Journal of Cutaneous Pathology. 2001;28:113-119
  33. 33. De A, Das S, Sarda A, Pal D, Biswas P. Acute generalised Exanthematous Pustulosis: An update. Indian Journal of Dermatology. 2018;63(1):22-29. DOI: 10.4103/ijd.IJD_581_17
  34. 34. Brockow K, Przybilla B, Aberer W, et al. Guideline for the diagnosis of drug hypersensitivity reactions: S2K-guideline of the German Society for Allergology and Clinical Immunology (DGAKI) and the German dermatological society (DDG) in collaboration with the Association of German Allergologists (AeDA), the German Society for Pediatric Allergology and Environmental Medicine (GPA), the German contact dermatitis research group (DKG), the Swiss Society for Allergy and Immunology (SGAI), the Austrian Society for Allergology and Immunology (ÖGAI), the German academy of Allergology and environmental medicine (DAAU), the German Center for Documentation of severe skin reactions and the German Federal Institute for Drugs and Medical Products (BfArM). Allergo J Int. 2015;24(3):94-105. DOI: 10.1007/s40629-015-0052-6
  35. 35. Bergmann MM, Caubet JC. Role of in vivo and in vitro tests in the diagnosis of severe cutaneous adverse reactions (SCAR) to drug. Current Pharmaceutical Design. 2019;25(36):3872-3880. DOI: 10.2174/1381612825666191107104126
  36. 36. Defrance C, Bousquet PJ, Demoly P. Evaluating the negative predictive value of provocation tests with nonsteroidal anti-inflammatory drugs. Allergy. 2011;66:1410-1414. DOI: 10.1111/j.1398-9995.2011.02671.x
  37. 37. Ushigome Y, Kano Y, Ishida T, Hirahara K, Shiohara T. Short- and long-term outcomes of 34 patients with drug-induced hypersensitivity syndrome in a single institution. Journal of the American Academy of Dermatology. 2013;68(5):721-728
  38. 38. Cho YT, Chu CY. Treatments for severe cutaneous adverse reactions. Journal of Immunology Research. 2017;2017:1503709. DOI: 10.1155/2017/1503709
  39. 39. Auyeung J, Lee M. Successful treatment of Stevens-Johnson syndrome with cyclosporine and corticosteroid. The Canadian Journal of Hospital Pharmacy. 2018;71(4):272-275
  40. 40. Scheuerman O, Nofech-Moses Y, Rachmel A, Ashkenazi S. Successful treatment of antiepileptic drug hypersensitivity syndrome with intravenous immune globulin. Pediatrics. 2001;107(1):e14
  41. 41. Fields KS, Petersen MJ, Chiao E, Tristani-Firouzi P. Case reports: Treatment of nevirapine-associated dress syndrome with intravenous immune globulin (IVIG) journal of drugs. Dermatology. 2005;4(4):510-513
  42. 42. Joly P, Janela B, Tetart F, et al. Poor benefit/risk balance of intravenous immunoglobulins in DRESS. Archives of Dermatology. 2012;148(4):543-544. DOI: 10.1001/archderm.148.4.dlt120002-c
  43. 43. Huang YC, Li YC, Chen TJ. The efficacy of intravenous immunoglobulin for the treatment of toxic epidermal necrolysis: A systematic review and meta-analysis. The British Journal of Dermatology. 2012;167(2):424-432
  44. 44. Bachot N, Revuz J, Roujeau J. Intravenous immunoglobulin treatment for Stevens-Johnson syndrome and toxic epidermal necrolysis: A prospective noncomparative study showing No benefit on mortality or progression. Archives of Dermatology. 2003;139(1):33-36
  45. 45. Ye LP, Zhang C, Zhu QX. The effect of intravenous immunoglobulin combined with corticosteroid on the progression of Stevens-Johnson syndrome and toxic epidermal necrolysis: A meta-analysis. PLoS One. 2016;11(11):e0167120
  46. 46. Chen Y-T, Hsu C-Y, Chien Y-N, Lee W-R, Huang Y-C. Efficacy of cyclosporine for the treatment of Stevens-Johnson syndrome and toxic epidermal necrolysis: Systemic review and meta-analysis. Dermatologica Sinica. 2017;35(3):131-137
  47. 47. Ng QX, De Deyn MLZQ , Venkatanarayanan N, Ho CYX, Yeo WS. A meta-analysis of cyclosporine treatment for Stevens-Johnson syndrome/toxic epidermal necrolysis. Journal of Inflammation Research. 2018;11:135-142
  48. 48. Zimmermann S, Sekula P, Venhoff M, Motschall E, Knaus J, Schumacher M, et al. Systemic Immunomodulating therapies for Stevens-Johnson syndrome and toxic epidermal necrolysis: A systematic review and meta-analysis. JAMA Dermatology. 2017;153(6):514-522. DOI: 10.1001/jamadermatol.2016.5668
  49. 49. Wolkenstein P, Latarjet J, Roujeau JC, Duguet C, Boudeau S, Vaillant L, et al. Randomised comparison of thalidomide versus placebo in toxic epidermal necrolysis. Lancet. 1998;352(9140):1586-1589
  50. 50. Zhang S, Tang S, Li S, Pan Y, Ding Y. Biologic TNF-alpha inhibitors in the treatment of Stevens-Johnson syndrome and toxic epidermal necrolysis: A systemic review. The Journal of Dermatological Treatment. 2020;31(1):66-73
  51. 51. Yamada H, Takamori K. Status of plasmapheresis for the treatment of toxic epidermal necrolysis in Japan. Therapeutic Apheresis and Dialysis. 2008;12(5):355-359
  52. 52. Husain Z, Reddy BY, Schwartz RA. DRESS syndrome: Part II. Management and therapeutics. Journal of the American Academy of Dermatology. 2013;68(5):709.e1-709.e9; quiz 718-20. DOI: 10.1016/j.jaad.2013.01.032
  53. 53. Byrd RC, Mournighan KJ, Baca-Atlas M, Helton MR, Sun NZ, Siegel MB. Generalized bullous fixed-drug eruption secondary to the influenza vaccine. JAAD Case Rep. 2018;4(9):953-955
  54. 54. Esmaeilzadeh H, Farjadian S, Alyasin S, Nemati H, Nabavizadeh H, Esmaeilzadeh E. Epidemiology of severe cutaneous adverse drug reaction and its HLA association among pediatrics. Iran J Pharm Res. 2019;18(1):506-522
  55. 55. Frey N, Jossi J, Bodmer M, Bircher A, Jick SS, Meier CR, et al. The epidemiology of Stevens-Johnson syndrome and toxic epidermal necrolysis in the UK. The Journal of Investigative Dermatology. 2017;137:1240-1247

Written By

Miteshkumar Rajaram Maurya, Renuka Munshi, Sachin Bhausaheb Zambare and Sanket Thakur

Submitted: 23 June 2022 Reviewed: 18 October 2022 Published: 12 November 2022