Journal List > Allergy Asthma Immunol Res > v.2(2) > 1052196

Thong: Update on the Management of Antibiotic Allergy

Abstract

Drug allergy to antibiotics may occur in the form of immediate or non-immediate (delayed) hypersensitivity reactions. Immediate reactions are usually IgE-mediated whereas non-immediate hypersensitivity reactions are usually non-IgE or T-cell mediated. The clinical manifestations of antibiotic allergy may be cutaneous, organ-specific (e.g., blood dyscracias, hepatitis, interstitial nephritis), systemic (e.g., anaphylaxis, drug induced hypersensitivity syndrome) or various combinations of these. Severe cutaneous adverse reactions manifesting as Stevens Johnson syndrome or toxic epidermal necrolysis (TEN) may be potentially life-threatening. The management of antibiotic allergy begins with the identification of the putative antibiotic from a detailed and accurate drug history, complemented by validated in-vivo and in-vitro allergological tests. This will facilitate avoidance of the putative antibiotic through patient education, use of drug alert cards, and electronic medical records with in-built drug allergy/adverse drug reaction prescription and dispensing checks. Knowledge of the evidence for specific antibiotic cross-reactivities is also important in patient education. Apart from withdrawal of the putative antibiotic, immunomodulatory agents like high-dose intravenous immunoglobulins may have a role in TEN. Drug desensitization where the benefits outweigh the risks, and where no alternative antibiotics can be used for various reasons, may be considered in certain situations. Allergological issues pertaining to electronic drug allergy alerts, computerized physician prescriptions and decision support systems, and antibiotic de-escalation in antimicrobial stewardship programmes are also discussed.

INTRODUCTION

Antibiotics are one of the most common causes of drug allergy in most epidemiological studies, both among adults and children.1-6 Among the various classes of antibiotics, beta-lactam antibiotics (penicillins and cephalosporins), cotrimoxazole and quinolones are some of the most common causes of antibiotic allergy.
Antibiotic allergy may occur in the form of immediate or non-immediate (delayed) hypersensitivity reactions. Immediate reactions are usually IgE-mediated whereas non-immediate hypersensitivity reactions are usually non-IgE or T-cell mediated.7 The clinical manifestations of antibiotic allergy may be cutaneous, organ-specific (e.g., blood dyscracias, hepatitis, interstitial nephritis), systemic (e.g., anaphylaxis, drug induced hypersensitivity syndrome) or various combinations of these. Severe cutaneous adverse reactions (SCAR) manifesting as Stevens Johnson syndrome (SJS) or toxic epidermal necrolysis (TEN) may be potentially life-threatening.8

DIAGNOSIS OF ANTIBIOTIC ALLERGY

The management of antibiotic allergy begins with the identification of the putative antibiotic from a detailed and accurate drug history.9 Not infrequently, the drug history may need to be obtained from a combination of sources other than the patient, including care-givers, records from other prescribing physicians and both non-electronic and electronic medical records.10 With the use of digital photography, instructing patients to take digital photographs of the initial rash may become increasingly important in helping the allergist to diagnose a drug eruption, especially when the rash is likely to have resolved by the time the patient sees the allergist.11-13
In the diagnosis of immediate allergic reactions to antibiotics, the in-vivo tests available are skin prick tests (SPT) and intradermal tests (IDT).14,15 However, these have been well validated mainly for beta-lactam antibiotics and less so for other classes of antibiotics. For in-vitro tests, commercially available assays include fluorescent enzyme immunoassays (FEIA) (ImmunoCAP®, Phadia) which are less sensitive and specific compared to skin tests. Again, these tests are available mainly for penicillins and cephalosporins. Radioimmunoassays previously used mainly for the diagnosis of penicillin allergy (including the radioallergosorbent test, RAST) have over the years been replaced with the FEIA assays.16,17 Flow-cytometric based basophil activation tests (BAT) (flow assay stimulation test, FAST/FlowCAST®, Buhlmann Laboratories) which measure CD69 or CD203c on drug-specific activated basophils may have a role in the diagnosis of antibiotic allergy, with studies so far mainly focused on beta-lactam allergy.18
For non-immediate reactions, delayed readings of IDT are done at 24 hours and 72 hours.19 Delayed reactions are considered positive when there is an infiltrated erythematous reaction. Patch tests are often done in Europe to assist in the diagnosis of non-immediate reactions to various antibiotics. The tests are read on day 2, day 4, and day 7 (if negative on days 2 and 4), and the vehicle used is usually petrolatum.20 The patch test allergens can be prepared in-house or using commercially available products (Chemotechnique Diagnostics®, Sweden). However, the sensitivity of the test is usually drug- and reaction-specific. Patch tests have been described in the diagnosis of non-immediate reactions to amoxicillin, cefcapene pivoxil, clindamycin, ciprofloxacin, clarithromycin, cotrimoxazole, doxycycline, erythromycin, fluoroquinolones, isoniazid, metronidazole, minocycline, pristinamycin, rifampicin, spiramycin, teicoplanin and vancomycin. Patch tests are generally useful in maculopapular exanthema (MPE), eczema, acute generalized exanthematous pustulosis (AGEP), fixed drug eruptions (FDE) (when done on the lesional skin), symmetric drug-related intertriginous and flexural exanthema (SDRIFE, Baboon's syndrome); but have not been shown to be very useful in SJS/TEN and vasculitis.19
In-vivo tests available for non-immediate reactions include the lymphocyte transformation test (LTT) which is a proliferation assay which detects drug-specific T-cells.21 This test can be technically difficult to carry out and are thus often done in specialized centres, mostly in Europe. Like the patch test, the LTT is usually positive in a drug- and reaction-specific manner. Antibiotics which have been found to often test positive in LTT are beta-lactams, quinolones, macrolides, sulfonamides, tetracycline, isoniazid and rifampicin. Similar to patch tests, LTT are often positive in MPE, bullous exanthema, AGEP, and drug rash with eosinophilia and systemic symptoms (DRESS). It is occasionally positive in hepatitis and nephritis, but rarely positive in TEN, cytopaenias and vasculitis.21,22 Novel in-vitro tests evaluating cytokine secretion, up-regulation of cell surface activation markers (e.g., CD69), and analysis of cytotoxic potential (granzyme B, CD107) remain as research tools.22
In view of the limited number of in-vivo and in-vitro tests commercially available for most antibiotics, and also because non-immediate reactions are generally more common than immediate reactions in clinical practice, drug provocation tests (DPT) often have to used in the diagnostic evaluation of drug allergy.23-27 The indications for DPT are as follows:23
  • to exclude hypersensitivity in non-suggestive history of drug hypersensitivity and in patients with non-specific symptoms, e.g. vagal symptoms following the use of an antibiotic

  • to provide safe pharmacologically and/or structurally non-related drugs in proven hypersensitivity e.g. other antibiotics in beta lactam-allergic patients, anxious people who would refuse to take the recommended drug without proof of tolerance

  • to exclude cross-reactivity of related drugs in proven hypersensitivity, e.g. a cephalosporin in a penicillin-allergic subject

  • to establish a firm diagnosis in suggestive history of drug hypersensitivity with negative, non-conclusive or non-available allergologic tests, e.g. MPE during aminopenicillin treatment with negative allergological tests.

DPT can generally be carried out safely with careful patient selection.28 Blinded (single- or double-blind placebo-control) challenges may sometimes be needed in patients with non-suggestive history and non-specific symptoms.

TREATMENT OF ANTIBIOTIC ALLERGY

Definitive treatment involves cessation of the suspected antibiotic. In certain instances where the antibiotic is required because there are no better alternatives (e.g., infection with multiresistant organisms, or when alternative drugs are more expensive), drug desensitization can be carried out. Desensitization is a method of reintroducing antibiotics into highly sensitized patients to induce tolerance. However such individuals are still considered as being allergic to the antibiotic. Recent studies of in vitro rapid antigen desensitizations implicate mast cells and basophils as cellular targets, as well as syk, a signal transducing molecule, and signal transducer and activator of transcription 6 (STAT6), which is responsible for the transcription of interleukin (IL)-4 and IL-13.29 Rapid desensitization results in patients achieving the target total dose of the drug through rapidly escalating doses usually within 24 hours, slow desensitization results in patients achieving the total target dose within a few days to weeks. Desensitization should be avoided should the initial reaction be potentially life-threatening reactions like immunobullous eruptions and SJS/TEN, with the exception of anaphylaxis. Various desensitization protocols are available for penicillin (benzylpenicillin, ampicillin), cephalosporins (ceftazidime, cefotaxime), cotrimoxazole, ethambutol, imipenam, isoniazid, meropenam, metronidazole, rifamipicin, streptomycin, vancomycin and fluoroquinolones.

BETA-LACTAM ALLERGY

Penicillin allergy

Allergic reactions to beta-lactam antibiotics are the most common cause of drug allergies in most epidemiological studies on adverse drug reactions. SPT and IDT using commercially available penicilloyl polylysine (PPL), minor determinant mix (MDM) and benzylpenicillin G or amoxicillin have been validated in various studies and shown to be useful in the evaluation of suspected immediate reactions to penicillin.30,31 In 2004, Allergopharma and Hollister-Stier announced their decision to stop the commercial production of penicillin reagents (Allergopen® and PrePen® respectively). A Spanish product (Diater®) was subsequently found to be a reliable and consistent alternative32,33 and is presently used in many countries worldwide. In September 2009, Pre-Pen® was approved for marketing by the Food and Drug Administration (FDA) through ALK-Abello and Allerquest LLC. In countries where commercial PPL and MDM are not available, skin testing with benzylpenicillin may be used in lieu.34 However, this may miss patients who may have tested positive to PPL or MDM, and thus could result in potentially positive drug provocation tests being done.
In-vitro tests are often less sensitive and more expensive when compared to skin tests, with the FEIA currently being the most widely commercially available test. The determinants used in FEIA are benzylpenicilloyl and amoxicilloyl. However, the sensitivity (42-74%) and specificity (85-100%) reported varied among studies,35,36 depending on when the sample was taken from the time of the initial clinical reaction, and the outcomes of skin tests to PPL, MDM and/or amoxicillin in the respective studies.
The flow cytometric BAT assay, when used in the diagnosis of beta-lactam allergy, has a sensitivity of 50%, and specificity of 93%.37,38 However, the test is unable to differentiate between selective reactors and cross-reactors, and tests become negative the longer the duration from the initial reaction.39 Using a combination of skin tests, specific IgE assays, followed by cellular tests in negative patients, can facilitate confirmation of beta-lactam allergy, avoiding DPT in up to two-thirds of patients.40 Using an alternative marker like CD203c may increase the sensitivity of these tests.41
Patch tests when used, should be carried out with benzylpenicillin, amoxicillin, ampicillin, and any suspect penicillins and/or cephalosporins. LTT for beta lactam allergy has a low sensitivity of 60-70%, hence a positive test is useful in confirming beta lactam allergy but a negative test does not rule it out. The LTT is often positive in AGEP and DRESS, but rarely positive (<10%) in blood dyscracias and TEN associated with drug allergy.42

Cephalosporin allergy

The reported cross-reactivity for IgE-mediated hypersensitivity between cephalosporins and penicillins in patients with Ig-E mediated penicillin allergy of 5-10%, were based on early studies from the 1970s on patients with a history of penicillin allergy who developed allergic reactions to cephalexin, cephalothin and cephaloridine.43,44 In addition, early cephalosporin antibiotics contained traces of penicillin.45 Although the practice parameters of the AAAAI in 1999 did not advocate the use of cephalosporin skin testing,27 this is recommended by the British Society of Allergy and Clinical Immunology (BSACI)46 and the European Academy of Allergy and Clinical Immunology (EAACI).14 The R1 side chain rather than the betalactam structure, shared by penicillins and cephalosporins, seems to play a dominant role in determining the specificity of immunologic reactions to cephalosporins.47 Thus, penicillin can be administered safely to patients allergic to cephalosporins and with a negative skin test result to penicillin determinants.48 Similarly, this may the reason why the penicillin allergic individuals appear to be able to tolerate most third and fourth generation cephalosporins.
The flow cytometric BAT assay appears to be a promising invitro test in the diagnosis of cephalosporin allergy as well as penicillin allergy.37,38

Carbapenem allergy

Earlier studies from the late 1980s showed that cross-reactivity between penicillin and imipenem allergy was 50% based on 10 of 20 patients with penicillin allergy being skin test positive to one or more penicillin or imipenem determinants.49 Recent prospective studies in adults and children with penicillin (predominantly amoxicillin) IgE-mediated allergy have shown that the cross-reactivity based on positive skin tests to imipenamcilastatin50 and meropenam51,52 was 0.9%, and that patients who were SPT/IDT negative to imipenam-cilastatin and meropenam were able to tolerate a graded, challenge dose of intravenous imipenam-cilastatin and meropenam respectively. For delayed reactions to carbapenams, the cross-reactivity with penicillins was 5.5% based on patients with cell-mediated allergy to penicillins showing positive patch tests to at least one penicillin reagent and imipenem-cilastatin. All patients with negative patch test and delayed IDT reading to imipenamcilastatin tolerated an intramuscular provocation test.53

COTRIMOXAZOLE ALLERGY

Cotrimoxazole is an immunogenic drug which may cause both immediate and non-immediate reactions. Non-immediate reactions range from mild MPE and FDE to serious SJS and TEN,54,55 and are more common than immediate reactions. This is especially prevalent in HIV-infected individuals where cotrimoxazole is used for the treatment and prophylaxis for Pneumocystis jiroveci infection and toxoplasmosis.56 Slow acetylator phenotype and genotype,57,58 and major histocompatibility complex (MHC) polymorphisms59 have not been shown to be major predisposing risk factors for cotrimoxazole hypersensitivity in HIV-infected individuals. Rapid and slow desensitization to cotrimoxazole especially in the setting of HIV infection, has been shown to be effective and safe.60

FLUOROQUINOLONE ALLERGY

Fluoroquinolone allergy may present in the form of immediate and non-immediate reactions. The immediate reactions may be IgE mediated or non IgE mediated, with non-IgE mediated reactions occurring after the first dose with no previous history of sensitization.61,62 Although previous studies had shown that skin tests to quinolones lack sensitivity and specificity,63 a negative skin test could predict a negative challenge test in 94% of the challenged cases.64 Cross-reactivity has been demonstrated for immediate reactions through positive skin tests to a range of quinolones,62 and delayed reactions through generation and analysis (flow cytometry and proliferation assays) of quinolone-specific T cell clones respectively.65 Thus, patients with allergy to a fluoroquinolone should avoid other fluoroquinolones.

MACROLIDE ALLERGY

Macrolides are classified according to the number of carbon atoms in the chemical structure: 14 membered (erythromicin, roxithromycin, dirithromycin, clarithromycin), 15 membered (azithromycin) and 16 membered (spiramycin, josamycin, midecamycin) macrolides. Allergic reactions to macrolide antibiotics appear to be relatively uncommon (0.4% to 3% of treatments).66 Cases of immediate reactions in the form of anaphylaxis,67 and non-immediate reactions like fixed drug eruptions, toxic epidermal necrolysis and leukocytoclastic vasculitis have been reported, in children and adults, for clarithromycin and azithromycin. Successful desensitization has also been reported.68

TETRACYCLINE ALLERGY

Minocycline can cause serious adverse reactions including drug hypersensitivity syndrome, serum sickness and drug-induced lupus. These occur on average within 4 weeks of therapy, whereas minocycline-induced lupus occurs on average 2 years after the initiation of therapy.69 Apart from photodermatoses and photo-onycholysis which are usually phototoxic in nature, adverse drug reactions, in particular drug allergies to doxycycline and tetracycline are relatively rare.70

CLINDAMYCIN ALLERGY

Clindamycin may be associated with both immediate and non-immediate allergic reactions.71 However, the prevalence of such reactions is rare.72 Apart from exanthematous eruptions, cases reported in the literature include contact dermatitis, AGEP73 and TEN.74 The use of a combination of skin prick tests, patch tests and oral challenges if skin tests are negative, appear to be more useful compared to SPT and IDT alone as negative skin tests may still result in positive challenges.75,76 Clindamycin desensitization has been reported in the literature in particular in HIV-infected individuals.77,78

VANCOMYCIN AND TEICOPLANIN ALLERGY

Vancomycin, a glycopeptide, has rarely been reported to be associated with allergic drug reactions including exfoliative dermatitis and maculopapular rash. This is in contrast to vancomycin red man syndrome, which is commonly associated with too rapid an infusion of vancomycin resulting in direct mast cell histamine release.79
Anaphylaxis from vancomycin may be through IgE mediated allergic mechanisms or non-IgE mediated non-allergic mechanisms. Various effective desensitization regimes have been described in the treatment of vancomycin anaphylaxis.80-83
Linear IgA bullous dermatosis (LABD) is an autoimmune, subepidermal, vesiculobullous disease that has been commonly associated with the use of vancomycin.84,85 Lesions typically appear during vancomycin therapy, 24 hours to 15 days after the first dose. Histopathologic examination and immunofluorescence studies are diagnostic, showing linear IgA and C3 deposits at the basement membrane zone on direct immunofluorescence. Withdrawal of vancomycin is all that is required.
Teicoplanin, another glycopeptide, has fewer side effects compared to vancomycin.79 Red man syndrome is very unusual with teicoplanin because this compound does not cause histamine release even at faster infusion rates than those of vancomycin. Immediate reactions [anaphylaxis86,87] and non-immediate reactions [rash,88 AGEP89 and DHS90] are infrequent. Although there have been reports of cross-reactivity between individuals with vancomycin and teicoplanin allergy,91-95 there have also been reports of patients with teicoplanin who tolerated vancomycin.96,97
Pre-operative allergy clinic assessment together with penicillin skin testing has been shown to be an effective intervention in reducing unnecessary use of prophylactic vancomycin perioperatively.98,99 This would be helpful in the long-term in reducing the spread of vancomycin resistant infections in hospitals and within the community, and the need for potentially expensive antibiotics like linezolid and tigecycline.

TUBERCULOUS DRUG ALLERGY

Mycobacterium tuberculosis (MTC) infection remains endemic in certain parts of Asia. Treatment of MTC infections involves combinations of anti-tuberculous drugs including isoniazid, rifampicin, ethambutol and pyrazinamide. Non-immediate reactions are much more common than immediate reactions to anti-tuberculous drugs. Drug eruptions100 in the form of MPE and lichenoid drug eruptions,101 haematological reactions, hepatitis, DHS, SJS/TEN102,103 have all been reported in the literature. Diagnosis using LTT have not been useful to date.104-106 Patch tests are also not consistently useful as they are dependent on the type of cutaneous drug eruption.19,20 In practice, it is often not clinically feasible to leave MTC infection untreated for 6 weeks pending evaluation using LTT or patch tests, which in the end may not be helpful. As such, rapid oral desensitization regimes have been described for isoniazid, rifampicin and ethambutol.107-111 These regimes often involve reintroducing the anti-tuberculous drugs as soon as the allergic reaction has settled. In addition, more than one drug often needs to be reintroduced, with at most a 3-5 day interval apart, because leaving patients on anti-tuberculous monotherapy would increase the risk of emergence of drug-resistant tuberculosis. If the initial allergic reaction was SJS/TEN, desensitization would need to be considered very carefully in consultation with the attending infectious diseases physician or pulmonologist. The risks of desensitization need to be explained carefully to the patient provided combinations of second-line anti-tuberculous drugs (e.g., quinolones, dapsone, cycloserine) are not an option.

SEVERE CUTANEOUS ADVERSE REACTIONS (SCAR)

Severe cutaneous adverse reactions (SCAR) include SJS, TEN and DHS or DRESS).8 In the study of Roujeau et al.,54 sulfonamides were the most strongly associated with TEN, followed by antibiotic drugs (in descending order of frequency: cephalosporins, quinolones, aminopenicillins, tetracyclines, macrolides), imidazole antifungals, anticonvulsants (phenobarbital, phenytoin, valproic acid, carbamazepine, and lamotrigine), then nonsteriodal anti-inflammatory drugs (especially oxicam) and allopurinol. HLA B*38 showed only a weak association with sulfamethoxazole induced SJS/TEN54 in contrast to antiepileptic drugs and allopurinol where HLA associations are stronger and ethnically related.112
In DHS, systemic corticosteroids (0.5 to 1 mg/kg/day) tapered over 6-8 weeks rapidly improves symptoms and laboratory measurements, but its impact on the long term disease course is not known. Controlled clinical trials are lacking on the use of systemic corticosteroids in DHS. Relapses of rash and hepatitis may occur as corticosteroids are tapered.113 Sequential reactivation of herpes viruses (e.g., human herpes virus 6, Ebstein Barr virus, cytomegalovirus)114 and subsequent triggering of autoimmunity115 may explain these relapses, and hence the effectiveness of systemic corticosteroids.
In SJS, the use of systemic corticosteroids has been supported by case reports and series (prospective and retrospective) which showed positive outcomes with the early use of corticosteroids (prednisolone 1 mg/kg/day or methylprednisolone 1-2 mg/kg/day) within 72 hours was beneficial in arresting the progression of SJS.116-120 However, there were also other studies which showed harm or no benefit.121
TEN is defined as the detachment of the epidermis affecting more than 30% body surface area of skin involvement. In early TEN, between 10-30% of epidermal detachment occurs which can sometimes be diagnosed clinically from a positive Nikolsky's sign or histological evidence of epidermal necrolysis. Apart from prompt withdrawal of the suspected drug, supportive measures including specialized nursing, early referral to a specialized unit, nutritional and respiratory care and support, skin care including the use of Biobrane dressings, are standard of care for which there are no controlled trials.122 Systemic corticosteroids should not been used as most series have suggested that the risks outweigh the benefits. The use of oral and intravenous cyclosporine 3-5 mg/kg/day, of duration of up to 3 weeks in case series of patients with severe TEN suggest that the risks of infection outweighed the benefits.123-125 The only double-blind placebo-controlled trial to date in the management of TEN, using thalidomide was stopped because there was excessive mortality in the thalidomide group.126 Other therapies like cyclophosphamide127 and plasmapharesis128 have not been shown to be useful.
In the last decade, several case series129-133 have described the use of high dose intravenous immunoglobulins (IVIg) from 0.8-3 g/day in the treatment of TEN. The rationale for the use of IVIg is based on the inhibition of Fas-mediated keratinocyte apoptosis in TEN by naturally occurring Fas-blocking antibodies within the IVIg. Although there were wide variation in patients and treatment protocols, different brands of IVIg used with different dosing regimens, the overall mortality rate was around 20% with earlier re-epithelialization demonstrated in some of the studies.
The prevalence of acute ocular complications ranges from 6% to 100%, and long-term sequelae from 1% to 50%. The most common long-term sequelae is sicca syndrome. Others include corneal ulceration, corneal epithelial defect, symblepharon and fornix foreshortening. Treatment modalities for ocular complications include topical antibiotics, topical corticosteroids, lubricants, and fornix sweeping. High-dose IVIg did not appear to reduce the severity of visually significant ocular complications.134 Early intervention with cryopreserved amniotic membrane transplantation was shown in a recent study to suppress inflammation and promote epithelial healing at the acute stage.135 Significant dry eye problems and photophobia may also be avoided with this intervention.
A recent retrospective study from China suggested that combination therapy with corticosteroid and high dose IVIG exhibited a tendency to reduce the mortality rate in comparison with administration of corticosteroid alone. The decrease in the mortality rate, however, was not statistically significant. Combination therapy also arrested progression earlier and decreased the hospitalization time, meaning that the total dose of corticosteroid may be reduced. Combination therapy, however, did not lead to earlier tapering of corticosteroid.136

DRUG-INDUCED LUPUS

Drug-induced lupus erythematosus (DILE) is defined as a lupus-like syndrome temporally related to continuous drug exposure which resolves after discontinuation of the offending drug. There are currently no standard diagnostic criteria for DILE and the pathomechanisms are still unclear. Among the antibiotics, minocycline and isoniazid are most often associated with DILE. Systemic DILE is characterized by typical lupus-like symptoms including skin signs, usually mild systemic involvement and a typical laboratory profile with positive antinuclear and anti-histone antibodies. In most cases of classic DILE, visceral involvement, low serum complement levels as well as anti-extractable nuclear antigen antibodies and anti-dsDNA antibodies are rarely present. In contrast, these are present in half the cases of anti-tumour necrosis factor (TNF) alpha inhibitor induced DILE. The diagnosis of DILE is based on a temporal association (months to years) of use of the putative drug with characteristic lupus-like symptoms, and resolution of symptoms upon withdrawal of the drug. Systemic corticosteroids and immunosuppressive drugs are only needed in refractory cases.137

ANTIBIOTIC ALLERGY AND ANTIMICROBIAL STEWARDSHIP PROGRAMMES

Antimicrobial stewardship programs in hospitals seek to optimize antimicrobial prescribing in order to improve individual patient care, reduce hospital costs and slow the spread of antibiotic resistant organisms. Such programs are often administered by multidisciplinary teams comprising infectious diseases physicians, clinical pharmacists, clinical microbiologists, and infection control practitioners. Strategies for changing antimicrobial prescribing behaviour include education of prescribers regarding proper antimicrobial usage, creation of an antimicrobial formulary with restricted prescribing of targeted agents, and review of antimicrobial prescribing with feedback to prescribers. De-escalation from broad-spectrum empirical antibiotics to narrow-spectrum, culture and sensitivity specific antibiotic is a supplemental strategy used in such programmes to reduce antibiotic resistance from the use of broad-spectrum antibiotics.138 However, de-escalation in a patient with unconfirmed antibiotic allergy should be exercised with caution as drug provocation tests in the presence of negative skin tests, should be avoided in the presence of on-going sepsis unless no other alternative antibiotics are available.23 Similarly, in the patient with a high probability of allergy to a narrow spectrum antibiotic (e.g., penicillin G) who has been tolerating a broad-spectrum antibiotic (e.g., meropenam), it would be prudent to continue the broad-spectrum antibiotic rather than to consider skin testing and desensitization to penicillin G in the presence of on-going sepsis where alternative antibiotic choices remain available.

ANTIBIOTIC ALLERGY ALERTS AND DECISION SUPPORT FOR COMPUTERIZED PHYSICIAN ORDERS

Antibiotic stewardship programmes may also be complemented by electronic computerized physician prescriptions with decision support systems139 utilizing drug/antibiotic allergy checks.140 However, the data from electronic drug allergy physician reporting systems are often inaccurate or incomplete. Thus, using such electronic alerts in any type of electronic medication record system as a decision support tool to facilitate antibiotic prescribing has to be done very cautiously.

CONCLUSIONS

Antibiotics may cause various types of allergic drug reactions ranging from mild to serious cutaneous reactions, organ-specific or systemic reactions. A high index of clinical suspicion and immediate withdrawal of the suspected drug/drugs are the most important steps in the management of antibiotic allergy. Systemic immunomodulatory drugs may be required to suppress severe cutaneous/systemic reactions. Drug desensitization may be considered in cases where the risks of retrying the drug outweigh the benefits, in particular where no alternative medications are available or are as effective.

Notes

There are no financial or other issues that might lead to conflict of interest.

References

1. Cars O, Molstad S, Melander A. Variation in antibiotic use in the European Union. Lancet. 2001. 357:1851–1853.
2. McCaig LF, Hughes JM. Trends in antimicrobial drug prescribing among office-based physicians in the United States. JAMA. 1995. 273:214–219.
3. Bigby M, Jick S, Jick H, Arndt K. Drug-induced cutaneous reactions. A report from the Boston Collaborative Drug Surveillance Program on 15,438 consecutive inpatients, 1975 to 1982. JAMA. 1986. 256:3358–3363.
4. Rademaker M, Oakley A, Duffill MB. Cutaneous adverse drug reactions in a hospital setting. N Z Med J. 1995. 108:165–166.
5. Hunziker T, Kunzi UP, Braunschweig S, Zehnder D, Hoigne R. Comprehensive hospital drug monitoring (CHDM): adverse skin reactions, a 20-year survey. Allergy. 1997. 52:388–393.
6. Thong BY, Leong KP, Tang CY, Chng HH. Drug allergy in a general hospital: Results of a novel prospective inpatient reporting system. Ann Allergy Asthma Immunol. 2003. 90:342–347.
7. Johansson SG, Bieber T, Dahl R, Friedmann PS, Lanier BQ, Lockey RF, Motala C, Ortega Martell JA, Platts-Mills TA, Ring J, Thien F, Van Cauwenberge P, Williams HC. Revised nomenclature for allergy for global use: Report of the Nomenclature Review Committee of the World Allergy Organization, October 2003. J Allergy Clin Immunol. 2004. 113:832–836.
8. Bastuji-Garin S, Rzany B, Stern RS, Shear NH, Naldi L, Roujeau JC. Clinical classification of cases of toxic epidermal necrolysis, Stevens-Johnson syndrome, and erythema multiforme. Arch Dermatol. 1993. 129:92–96.
9. Demoly P, Kropf R, Bircher A, Pichler WJ. Drug hypersensitivity: questionnaire. EAACI interest group on drug hypersensitivity. Allergy. 1999. 54:999–1003.
10. Park CS, Kim TB, Kim SL, Kim JY, Yang KA, Bae YJ, Cho YS, Moon HB. The use of an electronic medical record system for mandatory reporting of drug hypersensitivity reactions has been shown to improve the management of patients in the university hospital in Korea. Pharmacoepidemiol Drug Saf. 2008. 17:919–925.
11. Mann T, Colven R. A picture is worth more than a thousand words: enhancement of a pre-exam telephone consultation in dermatology with digital images. Acad Med. 2002. 77:742–743.
12. Leggett P, Gilliland AE, Cupples ME, McGlade K, Corbett R, Stevenson M, O'Reilly D, Steele K. A randomized controlled trial using instant photography to diagnose and manage dermatology referrals. Fam Pract. 2004. 21:54–56.
13. Eminovic N, de Keizer NF, Wyatt JC, ter Riet G, Peek N, van Weert HC, Bruijnzeel-Koomen CA, Bindels PJ. Teledermatologic consultation and reduction in referrals to dermatologists: a cluster randomized controlled trial. Arch Dermatol. 2009. 145:558–564.
14. Position paper: Allergen standardization and skin tests. The European Academy of Allergology and Clinical Immunology. Allergy. 1993. 48:48–82.
15. Brockow K, Romano A, Blanca M, Ring J, Pichler W, Demoly P. General considerations for skin test procedures in the diagnosis of drug hypersensitivity. Allergy. 2002. 57:45–51.
16. Edwards RG, Spackman DA, Dewdney JM. Development and use of three new radioallergosorbent tests in the diagnosis of penicillin allergy. Int Arch Allergy Appl Immunol. 1982. 68:352–357.
17. Garcia JJ, Blanca M, Moreno F, Vega JM, Mayorga C, Fernandez J, Juarez C, Romano A, de Ramon E. Determination of IgE antibodies to the benzylpenicilloyl determinant: a comparison of the sensitivity and specificity of three radio allergo sorbent test methods. J Clin Lab Anal. 1997. 11:251–257.
18. Hausmann OV, Gentinetta T, Bridts CH, Ebo DG. The basophil activation test in immediate-type drug allergy. Immunol Allergy Clin North Am. 2009. 29:555–566.
19. Barbaud A. Skin testing in delayed reactions to drugs. Immunol Allergy Clin North Am. 2009. 29:517–535.
20. Barbaud A, Goncalo M, Bruynzeel D, Bircher A. Guidelines for performing skin tests with drugs in the investigation of cutaneous adverse drug reactions. Contact Dermatitis. 2001. 45:321–328.
21. Pichler WJ, Tilch J. The lymphocyte transformation test in the diagnosis of drug hypersensitivity. Allergy. 2004. 59:809–820.
22. Lochmatter P, Zawodniak A, Pichler WJ. In vitro tests in drug hypersensitivity diagnosis. Immunol Allergy Clin North Am. 2009. 29:537–554.
23. Aberer W, Bircher A, Romano A, Blanca M, Campi P, Fernandez J, Brockow K, Pichler WJ, Demoly P. Drug provocation testing in the diagnosis of drug hypersensitivity reactions: general considerations. Allergy. 2003. 58:854–863.
24. Nizankowska-Mogilnicka E, Bochenek G, Mastalerz L, Swierczynska M, Picado C, Scadding G, Kowalski ML, Setkowicz M, Ring J, Brockow K, Bachert C, Wohrl S, Dahlen B, Szczeklik A. EAACI/GA2LEN guideline: aspirin provocation tests for diagnosis of aspirin hypersensitivity. Allergy. 2007. 62:1111–1118.
25. Patterson R, DeSwarte RD, Greenberger PA, Grammer LC, Brown JE, Choy AC. Drug allergy and protocols for management of drug allergies. Allergy Proc. 1994. 15:239–264.
26. Patterson R, DeSwarte RD, Greenberger PA, Grammer LC. Drug allergy and protocols for management of drug allergies. N Engl Reg Allergy Proc. 1986. 7:325–342.
27. Executive summary of disease management of drug hypersensitivity: a practice parameter. Joint Task Force on Practice Parameters, the American Academy of Allergy, Asthma and Immunology, the American Academy of Allergy, Asthma and Immunology, and the Joint Council of Allergy, Asthma and Immunology. Ann Allergy Asthma Immunol. 1999. 83:665–700.
28. Messaad D, Sahla H, Benahmed S, Godard P, Bousquet J, Demoly P. Drug provocation tests in patients with a history suggesting an immediate drug hypersensitivity reaction. Ann Intern Med. 2004. 140:1001–1006.
29. Castells M. Rapid desensitization for hypersensitivity reactions to medications. Immunol Allergy Clin North Am. 2009. 29:585–606.
30. Torres MJ, Blanca M, Fernandez J, Romano A, Weck A, Aberer W, Brockow K, Pichler WJ, Demoly P. Diagnosis of immediate allergic reactions to beta-lactam antibiotics. Allergy. 2003. 58:961–972.
31. Bousquet PJ, Co-Minh HB, Arnoux B, Daures JP, Demoly P. Importanceof mixture of minor determinants and benzylpenicilloyl poly-L-lysine skin testing in the diagnosis of beta-lactam allergy. J Allergy Clin Immunol. 2005. 115:1314–1316.
32. Romano A, Viola M, Bousquet PJ, Gaeta F, Valluzzi R, Caruso C, Demoly P. A comparison of the performance of two penicillin reagent kits in the diagnosis of beta-lactam hypersensitivity. Allergy. 2007. 62:53–58.
33. Matheu V, Perez E, Gonzalez R, Poza P, de la Torre F, Sanchez-Machin I, Garcia-Robaina JC. Assessment of a new brand of determinants for skin testing in a large group of patients with suspected beta-lactam allergy. J Investig Allergol Clin Immunol. 2007. 17:257–260.
34. Romano A, Bousquet-Rouanet L, Viola M, Gaeta F, Demoly P, Bousquet PJ. Benzylpenicillin skin testing is still important in diagnosing immediate hypersensitivity reactions to penicillins. Allergy. 2009. 64:249–253.
35. Blanca M, Mayorga C, Torres MJ, Reche M, Moya MC, Rodriguez JL, Romano A, Juarez C. Clinical evaluation of Pharmacia CAP System RAST FEIA amoxicilloyl and benzylpenicilloyl in patients with penicillin allergy. Allergy. 2001. 56:862–870.
36. Blanca M, Mayorga C, Perez E, Suau R, Juarez C, Vega JM, Carmona MJ, Perez-Estrada M, Garcia J. Determination of IgE antibodies to the benzyl penicilloyl determinant. A comparison between poly-L-lysine and human serum albumin as carriers. J Immunol Methods. 1992. 153:99–105.
37. Sanz ML, Gamboa PM, Antepara I, Uasuf C, Vila L, Garcia-Aviles C, Chazot M, De Weck AL. Flow cytometric basophil activation test by detection of CD63 expression in patients with immediate-type reactions to betalactam antibiotics. Clin Exp Allergy. 2002. 32:277–286.
38. Torres MJ, Padial A, Mayorga C, Fernandez T, Sanchez-Sabate E, Cornejo-Garcia JA, Antunez C, Blanca M. The diagnostic interpretation of basophil activation test in immediate allergic reactions to betalactams. Clin Exp Allergy. 2004. 34:1768–1775.
39. Fernandez TD, Torres MJ, Blanca-Lopez N, Rodriguez-Bada JL, Gomez E, Canto G, Mayorga C, Blanca M. Negativization rates of IgE radioimmunoassay and basophil activation test in immediate reactions to penicillins. Allergy. 2009. 64:242–248.
40. De Week AL, Sanz ML, Gamboa PM, Aberer W, Sturm G, Bilo MB, Montroni M, Blanca M, Torres MJ, Mayorga L, Campi P, Manfredi M, Drouet M, Sainte-Laudy J, Romano A, Merk H, Weber JM, Jermann TM. Diagnosis of immediate-type beta-lactam allergy in vitro by flow-cytometric basophil activation test and sulfidoleukotriene production: a multicenter study. J Investig Allergol Clin Immunol. 2009. 19:91–109.
41. Abuaf N, Rostane H, Rajoely B, Gaouar H, Autegarden JE, Leynadier F, Girot R. Comparison of two basophil activation markers CD63 and CD203c in the diagnosis of amoxicillin allergy. Clin Exp Allergy. 2008. 38:921–928.
42. Romano A, Blanca M, Torres MJ, Bircher A, Aberer W, Brockow K, Pichler WJ, Demoly P. Diagnosis of nonimmediate reactions to beta-lactam antibiotics. Allergy. 2004. 59:1153–1160.
43. Dash CH. Penicillin allergy and the cephalosporins. J Antimicrob Chemother. 1975. 1:107–118.
44. Saxon A, Beall GN, Rohr AS, Adelman DC. Immediate hypersensitivity reactions to beta-lactam antibiotics. Ann Intern Med. 1987. 107:204–215.
45. Kelkar PS, Li JT. Cephalosporin allergy. N Engl J Med. 2001. 345:804–809.
46. Mirakian R, Ewan PW, Durham SR, Youlten LJ, Dugue P, Friedmann PS, English JS, Huber PA, Nasser SM. BSACI guidelines for the management of drug allergy. Clin Exp Allergy. 2009. 39:43–61.
47. Perez-Inestrosa E, Suau R, Montanez MI, Rodriguez R, Mayorga C, Torres MJ, Blanca M. Cephalosporin chemical reactivity and its immunological implications. Curr Opin Allergy Clin Immunol. 2005. 5:323–330.
48. Antunez C, Blanca-Lopez N, Torres MJ, Mayorga C, Perez-Inestrosa E, Montanez MI, Fernandez T, Blanca M. Immediate allergic reactions to cephalosporins: evaluation of cross-reactivity with a panel of penicillins and cephalosporins. J Allergy Clin Immunol. 2006. 117:404–410.
49. Saxon A, Adelman DC, Patel A, Hajdu R, Calandra GB. Imipenem cross-reactivity with penicillin in humans. J Allergy Clin Immunol. 1988. 82:213–217.
50. Romano A, Viola M, Gueant-Rodriguez RM, Gaeta F, Pettinato R, Gueant JL. Imipenem in patients with immediate hypersensitivity to penicillins. N Engl J Med. 2006. 354:2835–2837.
51. Romano A, Viola M, Gueant-Rodriguez RM, Gaeta F, Valluzzi R, Gueant JL. Brief communication: tolerability of meropenem in patients with IgE-mediated hypersensitivity to penicillins. Ann Intern Med. 2007. 146:266–269.
52. Atanaskovic-Markovic M, Gaeta F, Medjo B, Viola M, Nestorovic B, Romano A. Tolerability of meropenem in children with IgE-mediated hypersensitivity to penicillins. Allergy. 2008. 63:237–240.
53. Schiavino D, Nucera E, Lombardo C, Decinti M, Pascolini L, Altomonte G, Buonomo A, Patriarca G. Cross-reactivity and tolerability of imipenem in patients with delayed-type, cell-mediated hypersensitivity to beta-lactams. Allergy. 2009. 64:1644–1648.
54. Mockenhaupt M, Viboud C, Dunant A, Naldi L, Halevy S, Bouwes Bavinck JN, Sidoroff A, Schneck J, Roujeau JC, Flahault A. Stevens-Johnson syndrome and toxic epidermal necrolysis: assessment of medication risks with emphasis on recently marketed drugs. The EuroSCAR-study. J Invest Dermatol. 2008. 128:35–44.
55. Levi N, Bastuji-Garin S, Mockenhaupt M, Roujeau JC, Flahault A, Kelly JP, Martin E, Kaufman DW, Maison P. Medications as risk factors of Stevens-Johnson syndrome and toxic epidermal necrolysis in children: a pooled analysis. Pediatrics. 2009. 123:e297–e304.
56. Ryan C, Madalon M, Wortham DW, Graziano FM. Sulfa hypersensitivity in patients with HIV infection: onset, treatment, critical review of the literature. WMJ. 1998. 97:23–27.
57. O'Neil WM, MacArthur RD, Farrough MJ, Doll MA, Fretland AJ, Hein DW, Crane LR, Svensson CK. Acetylator phenotype and genotype in HIV-infected patients with and without sulfonamide hypersensitivity. J Clin Pharmacol. 2002. 42:613–619.
58. Pirmohamed M, Alfirevic A, Vilar J, Stalford A, Wilkins EG, Sim E, Park BK. Association analysis of drug metabolizing enzyme gene polymorphisms in HIV-positive patients with co-trimoxazole hypersensitivity. Pharmacogenetics. 2000. 10:705–713.
59. Alfirevic A, Vilar FJ, Alsbou M, Jawaid A, Thomson W, Ollier WE, Bowman CE, Delrieu O, Park BK, Pirmohamed M. TNF, LTA, HSPA1L and HLA-DR gene polymorphisms in HIV-positive patients with hypersensitivity to cotrimoxazole. Pharmacogenomics. 2009. 10:531–540.
60. Lin D, Li WK, Rieder MJ. Cotrimoxazole for prophylaxis or treatment of opportunistic infections of HIV/AIDS in patients with previous history of hypersensitivity to cotrimoxazole. Cochrane Database Syst Rev. 2007. CD005646.
61. Schmid DA, Campi P, Pichler WJ. Hypersensitivity reactions to quinolones. Curr Pharm Des. 2006. 12:3313–3326.
62. Gonzalez I, Lobera T, Blasco A, del Pozo MD. Immediate hypersensitivity to quinolones: moxifloxacin cross-reactivity. J Investig Allergol Clin Immunol. 2005. 15:146–149.
63. Seitz CS, Brocker EB, Trautmann A. Diagnostic testing in suspected fluoroquinolone hypersensitivity. Clin Exp Allergy. 2009. 39:1738–1745.
64. Venturini Diaz M, Lobera Labairu T, del Pozo Gil MD, Blasco Sarramian A, Gonzalez Mahave I. In vivo diagnostic tests in adverse reactions to quinolones. J Investig Allergol Clin Immunol. 2007. 17:393–398.
65. Schmid DA, Depta JP, Pichler WJ. T cell-mediated hypersensitivity to quinolones: mechanisms and cross-reactivity. Clin Exp Allergy. 2006. 36:59–69.
66. Araujo L, Demoly P. Macrolides allergy. Curr Pharm Des. 2008. 14:2840–2862.
67. Pascual C, Crespo JF, Quiralte J, Lopez C, Wheeler G, Martin-Esteban M. In vitro detection of specific IgE antibodies to erythromycin. J Allergy Clin Immunol. 1995. 95:668–671.
68. Holmes NE, Hodgkinson M, Dendle C, Korman TM. Report of oral clarithromycin desensitization. Br J Clin Pharmacol. 2008. 66:323–324.
69. Shapiro LE, Knowles SR, Shear NH. Comparative safety of tetracycline, minocycline, and doxycycline. Arch Dermatol. 1997. 133:1224–1230.
70. Smith K, Leyden JJ. Safety of doxycycline and minocycline: a systematic review. Clin Ther. 2005. 27:1329–1342.
71. Lammintausta K, Tokola R, Kalimo K. Cutaneous adverse reactions to clindamycin: results of skin tests and oral exposure. Br J Dermatol. 2002. 146:643–648.
72. Mazur N, Greenberger PA, Regalado J. Clindamycin hypersensitivity appears to be rare. Ann Allergy Asthma Immunol. 1999. 82:443–445.
73. Sulewski RJ Jr, Blyumin M, Kerdel FA. Acute generalized exanthematous pustulosis due to clindamycin. Dermatol Online J. 2008. 14:14.
74. Paquet P, Schaaf-Lafontaine N, Pierard GE. Toxic epidermal necrolysis following clindamycin treatment. Br J Dermatol. 1995. 132:665–666.
75. Notman MJ, Phillips EJ, Knowles SR, Weber EA, Shear NH. Clindamycin skin testing has limited diagnostic potential. Contact Dermatitis. 2005. 53:335–338.
76. Seitz CS, Brocker EB, Trautmann A. Allergy diagnostic testing in clindamycin-induced skin reactions. Int Arch Allergy Immunol. 2009. 149:246–250.
77. Marcos C, Sopena B, Luna I, Gonzalez R, de la Fuente J, Martinez-Vazquez C. Clindamycin desensitization in an AIDS patient. AIDS. 1995. 9:1201–1202.
78. Martin JA, Alonso MD, Navas E, Antela A. [Clindamycin desensitization in a patient with the acquired immunodeficiency syndrome]. Med Clin (Barc). 1992. 98:478–479.
79. Svetitsky S, Leibovici L, Paul M. Comparative efficacy and safety of vancomycin versus teicoplanin: systematic review and meta-analysis. Antimicrob Agents Chemother. 2009. 53:4069–4079.
80. Chopra N, Oppenheimer J, Derimanov GS, Fine PL. Vancomycin anaphylaxis and successful desensitization in a patient with end stage renal disease on hemodialysis by maintaining steady antibiotic levels. Ann Allergy Asthma Immunol. 2000. 84:633–635.
81. Anne S, Middleton E Jr, Reisman RE. Vancomycin anaphylaxis and successful desensitization. Ann Allergy. 1994. 73:402–404.
82. Wazny LD, Daghigh B. Desensitization protocols for vancomycin hypersensitivity. Ann Pharmacother. 2001. 35:1458–1464.
83. Kitazawa T, Ota Y, Kada N, Morisawa Y, Yoshida A, Koike K, Kimura S. Successful vancomycin desensitization with a combination of rapid and slow infusion methods. Intern Med. 2006. 45:317–321.
84. Neughebauer BI, Negron G, Pelton S, Plunkett RW, Beutner EH, Magnussen R. Bullous skin disease: an unusual allergic reaction to vancomycin. Am J Med Sci. 2002. 323:273–278.
85. Waldman MA, Black DR, Callen JP. Vancomycin-induced linear IgA bullous disease presenting as toxic epidermal necrolysis. Clin Exp Dermatol. 2004. 29:633–636.
86. Polk RE. Anaphylactoid reactions to glycopeptide antibiotics. J Antimicrob Chemother. 1991. 27:Suppl B. 17–29.
87. Asero R. Teicoplanin-induced anaphylaxis. Allergy. 2006. 61:1370.
88. Unal S, Ikizoglu G, Demirkan F, Kaya TI. Teicoplanin-induced skin eruption. Int J Dermatol. 2002. 41:948–949.
89. Chu CY, Wu J, Jean SS, Sun CC. Acute generalized exanthematous pustulosis due to teicoplanin. Dermatology. 2001. 202:141–142.
90. Perrett CM, McBride SR. Teicoplanin induced drug hypersensitivity syndrome. BMJ. 2004. 328:1292.
91. Knudsen JD, Pedersen M. IgE-mediated reaction to vancomycin and teicoplanin after treatment with vancomycin. Scand J Infect Dis. 1992. 24:395–396.
92. McElrath MJ, Goldberg D, Neu HC. Allergic cross-reactivity of teicoplanin and vancomycin. Lancet. 1986. 1:47.
93. Marshall C, Street A, Galbraith K. Glycopeptide-induced vasculitis -cross-reactivity between vancomycin and teicoplanin. J Infect. 1998. 37:82–83.
94. Grek V, Andrien F, Collignon J, Fillet G. Allergic cross-reaction of teicoplanin and vancomycin. J Antimicrob Chemother. 1991. 28:476–477.
95. Kwon HS, Chang YS, Jeong YY, Lee SM, Song WJ, Kim HB, Kim YK, Cho SH, Kim YY, Min KU. A case of hypersensitivity syndrome to both vancomycin and teicoplanin. J Korean Med Sci. 2006. 21:1108–1110.
96. de Vries E, van Weel-Sipman MH, Vossen JM. A four-year-old child with teicoplanin allergy but no evidence of cross-reaction with vancomycin. Pediatr Infect Dis J. 1994. 13:167.
97. Macias E, Moreno E, Davila I, Laffond E, Ruiz A, Batista JC, Lorente F. Reaction to teicoplanin with tolerance to vancomycin. J Investig Allergol Clin Immunol. 2008. 18:71–72.
98. Park M, Markus P, Matesic D, Li JT. Safety and effectiveness of a preoperative allergy clinic in decreasing vancomycin use in patients with a history of penicillin allergy. Ann Allergy Asthma Immunol. 2006. 97:681–687.
99. Frigas E, Park MA, Narr BJ, Volcheck GW, Danielson DR, Markus PJ, Olson KE, Schroeder DR, Kita H. Preoperative evaluation of patients with history of allergy to penicillin: comparison of 2 models of practice. Mayo Clin Proc. 2008. 83:651–662.
100. Forget EJ, Menzies D. Adverse reactions to first-line antituberculosis drugs. Expert Opin Drug Saf. 2006. 5:231–249.
101. Lee AY, Jung SY. Two patients with isoniazid-induced photosensitive lichenoid eruptions confirmed by photopatch test. Photodermatol Photoimmunol Photomed. 1998. 14:77–78.
102. Leenutaphong V, Sivayathorn A, Suthipinittharm P, Sunthonpalin P. Stevens-Johnson syndrome and toxic epidermal necrolysis in Thailand. Int J Dermatol. 1993. 32:428–431.
103. Kuaban C, Bercion R, Koulla-Shiro S. HIV seroprevalence rate and incidence of adverse skin reactions in adults with pulmonary tuberculosis receiving thiacetazone free anti-tuberculosis treatment in Yaounde, Cameroon. East Afr Med J. 1997. 74:474–477.
104. Schreiber J, Zissel G, Greinert U, Schlaak M, Muller-Quernheim J. Lymphocyte transformation test for the evaluation of adverse effects of antituberculous drugs. Eur J Med Res. 1999. 4:67–71.
105. Yano S, Kobayashi K, Kato K, Tatsukawa T, Shishido S. [The usefulness of lymphocyte stimulation test (LST) in side effects of antituberculosis drugs]. Kekkaku. 2004. 79:7–10.
106. Suzuki Y, Miwa S, Shirai M, Ohba H, Murakami M, Fujita K, Suda T, Nakamura H, Hayakawa H, Chida K. Drug lymphocyte stimulation test in the diagnosis of adverse reactions to antituberculosis drugs. Chest. 2008. 134:1027–1032.
107. Kobashi Y, Okimoto N, Matsushima T, Abe T, Nishimura K, Shishido S, Kawahara S, Shigeto E, Takeyama H, Kuraoka T. [Desensitization therapy for allergic reactions of antituberculous drugs--evaluation of desensitization therapy according to the guideline of the Japanese Society for Tuberculosis]. Kekkaku. 2000. 75:699–704.
108. Holland CL, Malasky C, Ogunkoya A, Bielory L. Rapid oral desensitization to isoniazid and rifampin. Chest. 1990. 98:1518–1519.
109. Matz J, Borish LC, Routes JM, Rosenwasser LJ. Oral desensitization to rifampin and ethambutol in mycobacterial disease. Am J Respir Crit Care Med. 1994. 149:815–817.
110. Kim JH, Kim HB, Kim BS, Hong SJ. Rapid oral desensitization to isoniazid, rifampin, and ethambutol. Allergy. 2003. 58:540–541.
111. Buergin S, Scherer K, Hausermann P, Bircher AJ. Immediate hypersensitivity to rifampicin in 3 patients: diagnostic procedures and induction of clinical tolerance. Int Arch Allergy Immunol. 2006. 140:20–26.
112. Chung WH, Hung SI, Chen YT. Human leukocyte antigens and drug hypersensitivity. Curr Opin Allergy Clin Immunol. 2007. 7:317–323.
113. Shiohara T, Inaoka M, Kano Y. Drug-induced hypersensitivity syndrome (DIHS): a reaction induced by a complex interplay among herpesviruses and antiviral and antidrug immune responses. Allergol Int. 2006. 55:1–8.
114. Asano Y, Kagawa H, Kano Y, Shiohara T. Cytomegalovirus disease during severe drug eruptions: report of 2 cases and retrospective study of 18 patients with drug-induced hypersensitivity syndrome. Arch Dermatol. 2009. 145:1030–1036.
115. Aota N, Shiohara T. Viral connection between drug rashes and autoimmune diseases: how autoimmune responses are generated after resolution of drug rashes. Autoimmun Rev. 2009. 8:488–494.
116. Patterson R, Dykewicz MS, Gonzalzles A, Grammer LC, Green D, Greenberger PA, McGrath KG, Walker CL. Erythema multiforme and Stevens-Johnson syndrome. Descriptive and therapeutic controversy. Chest. 1990. 98:331–336.
117. Patterson R, Grammer LC, Greenberger PA, Lawrence ID, Zeiss CR, Detjen PF, Ganz MA, Miller TP, Orfan NA, Sonenthal KR, Stoloff RS. Stevens-Johnson syndrome (SJS): effectiveness of corticosteroids in management and recurrent SJS. Allergy Proc. 1992. 13:89–95.
118. Patterson R, Miller M, Kaplan M, Doan T, Brown J, Detjen P, Grammer LC, Greenberger PA, Hogan MB, Latall J, et al. Effectiveness of early therapy with corticosteroids in Stevens-Johnson syndrome: experience with 41 cases and a hypothesis regarding pathogenesis. Ann Allergy. 1994. 73:27–34.
119. Cheriyan S, Patterson R, Greenberger PA, Grammer LC, Latall J. The outcome of Stevens-Johnson syndrome treated with corticosteroids. Allergy Proc. 1995. 16:151–155.
120. Tripathi A, Ditto AM, Grammer LC, Greenberger PA, McGrath KG, Zeiss CR, Patterson R. Corticosteroid therapy in an additional 13 cases of Stevens-Johnson syndrome: a total series of 67 cases. Allergy Asthma Proc. 2000. 21:101–105.
121. Hynes AY, Kafkala C, Daoud YJ, Foster CS. Controversy in the use of high-dose systemic steroids in the acute care of patients with Stevens-Johnson syndrome. Int Ophthalmol Clin. 2005. 45:25–48.
122. Chave TA, Mortimer NJ, Sladden MJ, Hall AP, Hutchinson PE. Toxic epidermal necrolysis: current evidence, practical management and future directions. Br J Dermatol. 2005. 153:241–253.
123. Zaki I, Patel S, Reed R, Dalziel KL. Toxic epidermal necrolysis associated with severe hypocalcaemia, and treated with cyclosporin. Br J Dermatol. 1995. 133:337–338.
124. Jarrett P, Ha T, Snow J. Toxic epidermal necrolysis and cyclosporin. Clin Exp Dermatol. 1997. 22:254.
125. Sullivan JR, Watson A. Lamotrigine-induced toxic epidermal necrolysis treated with intravenous cyclosporin: a discussion of pathogenesis and immunosuppressive management. Australas J Dermatol. 1996. 37:208–212.
126. Wolkenstein P, Latarjet J, Roujeau JC, Duguet C, Boudeau S, Vaillant L, Maignan M, Schuhmacher MH, Milpied B, Pilorget A, Bocquet H, Brun-Buisson C, Revuz J. Randomised comparison of thalidomide versus placebo in toxic epidermal necrolysis. Lancet. 1998. 352:1586–1589.
127. Trautmann A, Klein CE, Kampgen E, Brocker EB. Severe bullous drug reactions treated successfully with cyclophosphamide. Br J Dermatol. 1998. 139:1127–1128.
128. Bamichas G, Natse T, Christidou F, Stangou M, Karagianni A, Koukourikos S, Chaidemenos G, Chrysomallis F, Sombolos K. Plasma exchange in patients with toxic epidermal necrolysis. Ther Apher. 2002. 6:225–228.
129. Stella M, Cassano P, Bollero D, Clemente A, Giorio G. Toxic epidermal necrolysis treated with intravenous high-dose immunoglobulins: our experience. Dermatology. 2001. 203:45–49.
130. Tristani-Firouzi P, Petersen MJ, Saffle JR, Morris SE, Zone JJ. Treatment of toxic epidermal necrolysis with intravenous immunoglobulin in children. J Am Acad Dermatol. 2002. 47:548–552.
131. Prins C, Kerdel FA, Padilla RS, Hunziker T, Chimenti S, Viard I, Mauri DN, Flynn K, Trent J, Margolis DJ, Saurat JH, French LE. Treatment of toxic epidermal necrolysis with high-dose intravenous immunoglobulins: multicenter retrospective analysis of 48 consecutive cases. Arch Dermatol. 2003. 139:26–32.
132. Trent JT, Kirsner RS, Romanelli P, Kerdel FA. Analysis of intravenous immunoglobulin for the treatment of toxic epidermal necrolysis using SCORTEN: The University of Miami Experience. Arch Dermatol. 2003. 139:39–43.
133. Bachot N, Revuz J, Roujeau JC. Intravenous immunoglobulin treatment for Stevens-Johnson syndrome and toxic epidermal necrolysis: a prospective noncomparative study showing no benefit on mortality or progression. Arch Dermatol. 2003. 139:33–36.
134. Yip LW, Thong BY, Tan AW, Khin LW, Chng HH, Heng WJ. High-dose intravenous immunoglobulin in the treatment of toxic epidermal necrolysis: a study of ocular benefits. Eye (Lond). 2005. 19:846–853.
135. Shay E, Kheirkhah A, Liang L, Sheha H, Gregory DG, Tseng SC. Amniotic membrane transplantation as a new therapy for the acute ocular manifestations of Stevens-Johnson syndrome and toxic epidermal necrolysis. Surv Ophthalmol. 2009. 54:686–696.
136. 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. 48:1122–1128.
137. Vedove CD, Del Giglio M, Schena D, Girolomoni G. Drug-induced lupus erythematosus. Arch Dermatol Res. 2009. 301:99–105.
138. Drew RH. Antimicrobial stewardship programs: how to start and steer a successful program. J Manag Care Pharm. 2009. 15:S18–S23.
139. Chazard E, Ficheur G, Merlin B, Serrot E, Beuscart R. Adverse drug events prevention rules: multi-site evaluation of rules from various sources. Stud Health Technol Inform. 2009. 148:102–111.
140. Abookire SA, Teich JM, Sandige H, Paterno MD, Martin MT, Kuperman GJ, Bates DW. Improving allergy alerting in a computerized physician order entry system. Proc AMIA Symp. 2000. 2–6.
TOOLS
Similar articles