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

Management of discoid lupus erythematosus and subacute cutaneous lupus erythematosus refractory to antimalarial therapy

Management of discoid lupus erythematosus and subacute cutaneous lupus erythematosus refractory to antimalarial therapy
Literature review current through: Jan 2024.
This topic last updated: Dec 07, 2022.

INTRODUCTION — Discoid lupus erythematosus (DLE) and subacute cutaneous lupus erythematosus (SCLE) are variants of cutaneous lupus erythematosus (cutaneous LE) that may occur independently or as manifestations of systemic lupus erythematosus (SLE). DLE most commonly occurs on the head and is characterized by well-defined, inflammatory plaques that evolve into atrophic, disfiguring scars (picture 1A-B). SCLE typically presents with inflamed, scaly papules or annular plaques on the neck, upper trunk, and arms (picture 2A-B). The symptomatic and disfiguring nature of DLE and SCLE underlies the need for appropriate treatment.

Most patients with DLE or SCLE will respond to conservative therapy with topical agents and antimalarial drugs. However, when the response to these interventions is inadequate, alternative immunomodulating or immunosuppressive drugs must be utilized (algorithm 1).

The management of refractory DLE and SCLE will be discussed here. The initial management of these disorders and the clinical manifestations of DLE and SCLE are reviewed separately.

(See "Initial management of discoid lupus erythematosus and subacute cutaneous lupus erythematosus".)

(See "Overview of cutaneous lupus erythematosus", section on 'Discoid lupus erythematosus'.)

(See "Overview of cutaneous lupus erythematosus", section on 'Subacute cutaneous lupus erythematosus'.)

TREATMENT PRINCIPLES

General approach — Most patients with cutaneous lupus erythematosus (cutaneous LE) achieve satisfactory improvement with strict photoprotection combined with topical or intralesional corticosteroids, topical calcineurin inhibitors, or oral antimalarial drugs (algorithm 1). (See "Initial management of discoid lupus erythematosus and subacute cutaneous lupus erythematosus".)

Patients requiring treatments for refractory disease are those who have not improved adequately with antimalarial therapy. Appropriate subsequent therapies for these patients include methotrexate, oral retinoids, mycophenolate mofetil, and dapsone. Selection among these four therapies should involve consideration of patient comorbidities. Case reports suggest that oral retinoids may be particularly useful in treating hypertrophic forms of DLE. (See 'After failure of antimalarial therapy' below.)

When remission does not occur despite these interventions, thalidomide, lenalidomide, or intravenous immune globulin (IVIG) can be used to achieve remission. Because of the potential adverse effects of thalidomide and lenalidomide and the high cost of IVIG, these therapies are generally used temporarily, with the goal of maintaining improvement with other therapies. (See 'Remission-inducing therapy' below.)

Additional medications have been used for DLE and SCLE. Limited efficacy data or the potential for severe adverse effects generally limit use to disease that has failed to respond to other therapies for refractory disease. (See 'Other therapies' below.)

Continuation of hydroxychloroquine — Hydroxychloroquine is typically continued during treatment with therapies for refractory disease [1]. Quinacrine can be discontinued prior to the addition of these therapies in patients who did not experience at least partial improvement with the addition of quinacrine to hydroxychloroquine therapy [1].

Limitations — Data on the efficacy of treatments for refractory DLE and SCLE are primarily limited to small open-label studies, retrospective reviews, case series, and case reports. The scarcity of high-quality studies stems from a lack of pharmaceutical, governmental, and foundational support for large, multicenter, controlled trials in cutaneous LE, as well as the relative rarity of refractory cutaneous LE.

In addition, the historical absence of standardized, objective measures of disease activity in cutaneous LE has made it difficult to systematically interpret the results of much of the published literature [2]. The Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) was developed and validated as a tool to assess disease activity and skin damage in cutaneous LE [3,4]. CLASI is now being used in a number of clinical trials and should improve the quality and applicability of data from studies of cutaneous LE [5]. A revised version of CLASI has also been proposed [6].

AFTER FAILURE OF ANTIMALARIAL THERAPY

Treatment selection — Patients with persistent disease activity despite antimalarial therapy can be treated with methotrexate, oral retinoids, mycophenolate mofetil, or dapsone [7]. Among these agents, methotrexate and acitretin have the strongest evidence for efficacy for DLE and SCLE.

In general, we favor methotrexate based upon the data supporting the use of this agent as well as reports of patients in whom periods of disease remission occurred following the discontinuation of methotrexate therapy. Oral retinoids (acitretin and isotretinoin) are much more expensive than methotrexate, and in our experience, relapses occur quickly following discontinuation. (See 'After failure of antimalarial therapy' above.)

Treatment tolerability also influences the selection of the most appropriate medication:

MethotrexateMethotrexate can cause serious toxicity in patients with renal insufficiency or pre-existing liver disease. (See 'Methotrexate' below.)

Acitretin – Although methotrexate, mycophenolate mofetil, and all oral retinoids are teratogenic, acitretin is an unfavorable choice for females of childbearing potential due to the long period during which females must abstain from pregnancy following treatment with the drug (three years). (See 'Oral retinoids' below.)

DapsoneDapsone cannot be given to patients with sulfonamide allergies and must be used with caution in patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency. (See 'Dapsone' below.)

Methotrexate — Methotrexate is a folic acid antagonist that has been used since the 1960s for systemic lupus erythematosus (SLE). The drug has also been shown to be effective for patients with cutaneous disease. Proposed mechanisms for benefit of methotrexate in connective tissue diseases include anti-inflammatory effects secondary to increased production of adenosine, inhibitory action on lymphocytes and neutrophils, and suppression of antibody production [8-10]:

Administration – Typical doses of methotrexate for DLE or SCLE range from 10 to 25 mg per week. Methotrexate can be administered orally, intramuscularly, or subcutaneously.

A reasonable approach to methotrexate therapy consists of an initial dose of 10 mg per week for adults. A complete blood count and liver function tests can be obtained after two weeks. If the laboratory results are normal, the dose of methotrexate may be increased by 5 mg every two weeks up to a maximum of 25 mg per week. Daily folic acid supplementation is indicated and may lessen the severity of gastrointestinal upset and oral ulceration [11].

Renal insufficiency increases the risk for toxicity, and the use of methotrexate should be carefully considered in older adult patients and those with renal disease. Dose adjustments are necessary when methotrexate is required in the setting of renal insufficiency. Alcohol abuse and pre-existing liver disease are additional relative contraindications for methotrexate therapy. Methotrexate is teratogenic and should be avoided in females for one to three cycles preceding pregnancy and in males for three months prior to an attempt to conceive. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Methotrexate'.)

Improvement with methotrexate is rapid, generally becoming evident within the first two to four weeks of therapy. Lesion clearance occurs at an average of six to eight weeks [12].

Efficacy – Treatment with methotrexate was beneficial for cutaneous lupus erythematosus (cutaneous LE) in a six-month, randomized trial in which 37 patients with SLE were treated with either methotrexate (15 to 20 mg/week) or placebo [13]. Compared with the patients who were given placebo, those who took methotrexate were less likely to have signs of active cutaneous LE (malar rash or DLE) at the end of the trial. The percentage of patients with cutaneous LE decreased from 60 to 16 percent in the group treated with methotrexate but increased from 76 to 84 percent in the placebo group. The authors did not distinguish between the response rates in patients with malar rash versus DLE.

The use of methotrexate specifically for refractory DLE and SCLE is supported by the results of retrospective studies and case reports [8,12,14-16]. In one retrospective study, 43 patients with cutaneous LE that was refractory to other systemic agents (including 16 subjects with SCLE and 12 subjects with DLE) were treated with 7.5 to 25 mg/week of oral or intravenous methotrexate [12]. All patients except one exhibited improvement in skin lesions, with the best clinical improvement noted in patients with SCLE or localized DLE. Another retrospective review of 12 patients with various types of cutaneous LE found similar success with methotrexate (10 to 25 mg/week) in refractory disease [16]. Complete or partial responses occurred in 9 out of the 10 patients with DLE or SCLE. Five of the patients with cutaneous LE who responded to treatment also had long-lasting remissions after cessation of methotrexate therapy.

Adverse effects – Adverse effects of methotrexate include gastrointestinal upset, oral ulcers, bone marrow suppression, hepatotoxicity, pulmonary fibrosis, and renal toxicity. (See "Major side effects of low-dose methotrexate".)

Monitoring for hepatic, hematologic, and renal compromise before and during therapy is warranted in all patients treated with methotrexate.

Oral retinoids — Oral retinoids can be effective for the treatment of DLE and SCLE [17-24]. Knowledge of the anti-inflammatory properties of retinoids and the beneficial effects of these drugs on epidermal keratinization prompted the use of these agents in cutaneous LE.

Case reports suggest that oral retinoids may be particularly useful in treating hypertrophic forms of DLE [17,19,20,25,26]:

AdministrationAcitretin and isotretinoin are the retinoids used for DLE and SCLE. When used in the treatment of cutaneous LE, acitretin and isotretinoin are usually prescribed in doses of 0.2 to 1 mg/kg/day [1]. We typically use acitretin for patients who are not of childbearing potential.

The response to retinoid therapy is usually rapid, occurring within the first two to six weeks of treatment [18,19,23]. However, our clinical experiences and that of others indicate that relapses often occur quickly once the drug is stopped [23].

Retinoids are potent teratogens and must be used with caution in females of childbearing age. Although acitretin has a shorter half-life than etretinate (two days), it can be reversely metabolized to etretinate, thereby prolonging the risk for teratogenicity. Due to its short elimination half-life, isotretinoin is a better choice for patients of childbearing potential; strict contraceptive practices must be adhered to during and for one month after cessation of isotretinoin. Three years of strict pregnancy prevention is necessary following the cessation of acitretin therapy. (See "Oral isotretinoin therapy for acne vulgaris", section on 'Adverse effects'.)

Efficacy – Examples of studies that suggest benefit of oral retinoid therapy include:

AcitretinAcitretin appeared effective in treating cutaneous LE in a 12-week, pilot study of 14 patients with DLE and 6 patients with SCLE. All patients with SCLE and 9 out of the 14 patients with DLE achieved complete clearing or marked reduction of lesions by the end of the study [19]. Seven patients who previously had not responded well to antimalarials or systemic glucocorticoids were among the responders to acitretin.

Acitretin (50 mg/day) was subsequently compared with hydroxychloroquine (400 mg/day) in an eight-week, randomized trial of 58 patients with DLE or SCLE and was determined to have similar efficacy as hydroxychloroquine. Complete clearing or marked improvement occurred in 46 percent of patients treated with acitretin and in 50 percent of patients treated with hydroxychloroquine [22]. Seven patients who had previously failed antimalarial therapy were included in the acitretin group (n = 28). However, the authors did not report response rates specific for this group. Adverse effects were more frequent with acitretin therapy.

IsotretinoinIsotretinoin has a shorter elimination half-life than acitretin and etretinate, and favorable responses have been documented in patients with DLE and SCLE in an open-label study and case reports [21,23,24]. In the open-label study, 8 out of 10 patients with SCLE or DLE that was refractory to sunscreens, topical corticosteroids, and antimalarials completed a 16-week course of isotretinoin therapy (80 mg/day) [21]. All eight patients achieved a response designated as excellent. Two patients were lost to follow-up prior to the first two-week, follow-up visit.

Alitretinoin – Improvement in DLE and SCLE with oral alitretinoin (30 mg per day) has been reported in a few patients who failed treatment with a variety of other therapies [27]. Further study is necessary to evaluate the efficacy and safety of alitretinoin when used for cutaneous LE. Oral alitretinoin is not commercially available in the United States.

Adverse effects – The most common side effects of isotretinoin and acitretin include hypertriglyceridemia and xerosis, both of which resolve with drug discontinuation. Elevated transaminases, skeletal hyperostosis, and, rarely, leukopenia may also occur with systemic retinoids. Retinoids are teratogenic and should be avoided during pregnancy and within specified periods of proximity to pregnancy (one month after cessation of isotretinoin and three years after cessation of acitretin).

Mycophenolate mofetil — Mycophenolate mofetil is an immunosuppressant that reversibly inhibits inosine monophosphate dehydrogenase, an enzyme necessary for de novo purine synthesis. Because it preferentially targets cells that are dependent on de novo purine synthesis for deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) production (eg, B and T cells), it has a more favorable toxicity profile than many other immunosuppressants:

AdministrationMycophenolate mofetil is typically given in doses of 1 to 3 g per day. Administering the drug in two divided doses during the day may help to minimize gastrointestinal symptoms. Treatment is usually started at 500 mg twice daily and subsequently increased up to 1.5 g twice daily, depending upon the response and tolerability [1,28,29]. Initial responses to mycophenolate mofetil are often evident within one to three months [30,31].

Efficacy – Evidence supporting the use of mycophenolate mofetil for SCLE and DLE is limited to an open-label study [32], retrospective studies [31,33], and case reports [30,34-37]:

In the open-label study, 10 patients with SCLE that was refractory to topical corticosteroids or antimalarials were treated with mycophenolate mofetil (1440 mg/day) for three months [32]. Reductions in disease activity were statistically significant (Cutaneous Lupus Erythematosus Disease Area and Severity Index [CLASI] decreased from 10.8±6.0 to 2.9±2.6).

A retrospective study (n = 73) that compared outcomes after mycophenolate mofetil therapy with methotrexate therapy in patients with SCLE or DLE did not find a statistically significant difference in response rates but suggested a need for larger studies to clarify whether mycophenolate mofetil might be more effective for DLE [33]. The mean response rate among all patients was 64.8 percent (95% CI 54.2-75.4 percent).

In a separate retrospective study, the addition of mycophenolate mofetil to existing treatment regimens led to at least partial disease improvement in all of 24 females with cutaneous LE refractory to antimalarial therapy who were treated with mycophenolate mofetil for at least three months (19 with DLE, three with SCLE, one with DLE/SCLE overlap, and one with tumid lupus erythematosus) [31]. Final doses ranged from 1500 to 3500 mg/day (average final dose 2750 mg/day), and the mean time to initial treatment response was 2.76 months.

In contrast, a retrospective study of seven patients with a variety of skin manifestations of SLE (including three patients with DLE and one patient with SCLE) who failed to improve with or tolerate other systemic therapies found that mycophenolate mofetil (2000 to 3000 mg/day) was poorly effective in treating skin disease [38]. Five out of seven patients did not respond to treatment. One of the two patients who had a partial response to treatment was a patient with DLE who responded initially but subsequently experienced a disease flare during treatment. The other responder was a patient with urticarial vasculitis.

Adverse effects – Although mycophenolate mofetil is generally well tolerated, nausea, vomiting, and diarrhea are the most common adverse reactions. Reversible cytopenias and teratogenicity are additional side effects of mycophenolate mofetil. An increased risk of malignancy has been postulated but has not been definitively demonstrated. (See "Mycophenolate: Overview of use and adverse effects in the treatment of rheumatic diseases".)

Dapsone — Dapsone has variable efficacy in the treatment of DLE and SCLE. Like other sulfonamide drugs, its antimicrobial action occurs via inhibition of the folic acid pathway. The mechanisms underlying its anti-inflammatory properties are less clear, but the efficacy of dapsone in cutaneous disease has primarily been linked to inhibitory effects on neutrophil-mediated tissue damage and neutrophil migration. Concordantly, in clinical practice, dapsone has been most useful in treating skin conditions characterized by prominent, neutrophilic infiltrates.

Dapsone therapy can result in dramatic improvement in patients with bullous lupus erythematosus, a variant of cutaneous LE that presents with neutrophilic infiltrates on biopsy [39-41]. Although neutrophils are not a prominent feature in the histopathology of DLE and SCLE, dapsone is effective for some patients:

Administration – Therapeutic doses of dapsone range from 25 to 200 mg daily. Treatment is initiated with low doses (eg, 50 mg per day), and the dose is gradually increased while closely monitoring hematologic parameters [8]. The maximum suggested dose is 1.5 mg/kg. The average time to improvement may be around two months; complete remission may take longer to achieve [42].

Patients should be assessed for G6PD deficiency prior to treatment. G6PD deficiency increases risk for dapsone-induced hemolysis. (See "Diagnosis and management of glucose-6-phosphate dehydrogenase (G6PD) deficiency", section on 'Patients at risk for G6PD deficiency who require treatment with an oxidant medication' and "Methemoglobinemia", section on 'Dapsone'.)

Efficacy – Evidence for the efficacy of dapsone in the treatment of DLE and SCLE is limited. SCLE has appeared beneficial in case reports and a retrospective study that documented improvement in six of eight patients who received dapsone (with or without other therapies) for SCLE [42-48]. Successful use of dapsone in DLE has been documented in a pilot study and retrospective review:

In the pilot study, 16 patients with DLE were given dapsone (100 mg/day) for 6 to 16 weeks [49]. At the end of the study, four patients (25 percent) were noted to have appreciable improvement in skin disease. Moderate improvement occurred in an additional four patients.

A review of the records of 33 patients with DLE who were treated with dapsone revealed excellent improvement in eight patients (24 percent) and partial effect in another eight subjects [50]. The excellent responders included two out of six patients who were treated simultaneously with dapsone and hydroxychloroquine.

Adverse effects – Adverse effects of dapsone include hemolysis and methemoglobinemia. All patients develop some degree of hemolysis, but those with G6PD deficiency are more likely to develop substantial hemolysis. Methemoglobinemia is dose dependent and is independent of G6PD status.

Idiosyncratic adverse reactions to dapsone include peripheral neuropathy, agranulocytosis, and dapsone hypersensitivity syndrome. Agranulocytosis may occur as early as three weeks into therapy and almost always occurs within the first 12 weeks of treatment [51]. Patients may present with fevers, pharyngitis, or, less commonly, signs of sepsis [52]. Fever, a morbilliform skin eruption, and hepatitis are features of the hypersensitivity syndrome. (See "Drug eruptions", section on 'Drug reaction with eosinophilia and systemic symptoms'.)

REMISSION-INDUCING THERAPY

Treatment selection — When DLE or SCLE fails to respond to methotrexate, oral retinoids, mycophenolate mofetil, and/or dapsone, thalidomide, lenalidomide, or intravenous immune globulin (IVIG) can be used to attain disease control. Certain characteristics of these therapies make them most useful as short-term, remission-inducing agents:

ThalidomideThalidomide appears highly effective for cutaneous lupus erythematosus (cutaneous LE) but has the potential for serious adverse effects, including teratogenicity, thromboembolism, and a relatively high risk of peripheral neuropathy.

Lenalidomide – Similar to thalidomide, lenalidomide appears effective for cutaneous LE but is considered teratogenic and is associated with risk for thromboembolic events.

Intravenous immune globulin – Intravenous immune globulin (IVIG) appears to be effective as a remission-inducing agent, particularly for SCLE. However, improvement following IVIG is generally short lived, and treatment with IVIG is expensive.

Thalidomide is the most extensively studied of these agents [53,54].

Maintenance treatment with conventional therapies or other systemic medications for refractory cutaneous LE can follow treatment with thalidomide, lenalidomide, or IVIG.

In the United States, obtaining and using thalidomide and lenalidomide for refractory cutaneous LE can be challenging due to safety regulations and insurance coverage. This contributes to increased use of IVIG and other therapies. (See 'Other therapies' below.)

Thalidomide — First synthesized as a sedative agent in 1954, thalidomide has had a tumultuous history. After the first description in 1961 of its association with phocomelia in infants born to females who had taken the drug during pregnancy, it was withdrawn from the pharmaceutical market worldwide in 1962. Despite these potential adverse effects, thalidomide can be highly effective for DLE and SCLE.

The mechanism of action of thalidomide in cutaneous LE is incompletely understood but is thought to involve the inhibition of tumor necrosis factor (TNF)-alpha and the suppression of ultraviolet B (UVB)-induced keratinocyte apoptosis [55]. The detection of elevated levels of TNF-alpha in lesions of SCLE supports the theory that suppression of TNF-alpha is at least partially responsible for the efficacy of thalidomide for cutaneous LE [56]. However, drugs that inhibit TNF-alpha have also been implicated in the induction of lupus-like cutaneous and systemic disease. (See "Tumor necrosis factor-alpha inhibitors: Induction of antibodies, autoantibodies, and autoimmune diseases", section on 'Autoimmune diseases' and "Drug-induced lupus".)

Other potential mechanisms of action of thalidomide include modulation of lymphocyte proliferation, cytokine production, neutrophil chemotaxis, and angiogenesis [57]:

AdministrationThalidomide has a quick onset of action; responses to thalidomide generally begin within the first month of treatment [58]. Thalidomide should be initiated at doses of 50 to 100 mg/day, and the dose should be tapered to the lowest effective dose after clinical improvement is attained [57].

In the United States, thalidomide can only be prescribed through a Risk Evaluation and Mitigation Strategy (REMS) program (www.thalomidrems.com), a registration and monitoring program for the safe use of thalidomide because of teratogenicity [59]:

Precautions – Patients should be monitored for signs or symptoms of peripheral neuropathy, an adverse effect of thalidomide, during treatment. Guidelines in the REMS program recommend that nerve conduction studies should also be performed at baseline and every six months thereafter. However, since symptoms of neuropathy may occur in patients with normal nerve conduction studies, this test should be used only as an adjunct to clinical assessment [57].

Thalidomide is associated with increased risk for thromboembolic events. Treatment with thalidomide should be considered carefully in patients with other risk factors for venous thrombosis.

After achievement of response – Although the development of adverse effects, particularly peripheral neuropathy, limits long-term treatment with thalidomide in many patients, relapse often occurs within three to four months if therapy is discontinued [57,58,60]. Thus, thalidomide is frequently used as a therapeutic bridge while awaiting the onset of action of other systemic agents.

In patients who tolerate thalidomide and cannot be transitioned to other effective therapies, thalidomide (25 to 50 mg/day or less) can be given as maintenance therapy [57]. In our clinical experience, some patients can maintain improvement on doses as low as 25 or 50 mg every three days. These patients should be monitored closely for adverse effects.

Efficacy – There are no randomized trials of thalidomide for DLE or SCLE. However, available data suggest that thalidomide can be highly effective. A systematic review and meta-analysis of observational studies evaluating the efficacy of thalidomide for cutaneous LE found an overall rate of complete or partial response to thalidomide of 90 percent (95% CI 85-94), with similar response rates among cutaneous LE subtypes [60]. The pooled rate of complete response was 64 percent (95% CI 55-73).

Adverse effects – Careful patient selection and conscientious monitoring must occur with the use of thalidomide. The most well-known and most serious toxicity of thalidomide is teratogenicity [57].

Peripheral neuropathy and thromboembolic events are additional well-recognized serious complications of thalidomide. In the systematic review and meta-analysis, the pooled rates of peripheral neuropathy and thromboembolic events were 16 percent (95% CI 9-25) and 2 percent (95% CI 1-3), respectively [60]. The pooled rate of thalidomide withdrawal related to adverse effects was 24 percent (95% CI 14-35).

A retrospective, observational study evaluating all patients with histologically confirmed cutaneous LE treated with thalidomide in five dermatology departments of French university hospitals from 1992 to 2017 revealed an overall thrombosis risk of 2.74 for 100 patient-years and a risk for arterial thrombosis and venous thrombosis of 1.72 and 1.03 for 100 patient-years, respectively [61]. The risk for all thromboembolic events was higher for patients with a history of arterial thrombosis and hypercholesterolemia and was lower for patients with a starting thalidomide dose of less than or equal to 50 mg per day and for those who received concomitant hydroxychloroquine. (See "Overview of the causes of venous thrombosis" and "Multiple myeloma: Prevention of venous thromboembolism".)

Sensory neuropathy typically precedes motor neuropathy. It is unclear whether the risk for this side effect is dose dependent [62-64]. If symptoms of neuropathy develop, the dose of thalidomide should be decreased [57]. If symptoms persist one month after dose reduction, thalidomide should be discontinued.

Other side effects of thalidomide include sedation, constipation, endocrine dysfunction, and neutropenia.

Lenalidomide — Lenalidomide is a thalidomide derivative that is a far more potent inhibitor of TNF-alpha. This drug has a lower frequency of sedation, constipation, and neuropathy than thalidomide:

Administration – Dosing of lenalidomide for adults is typically 5 to 10 mg per day [28]. Administration in the United States requires participation in a REMS program due to the teratogenic effects of lenalidomide. Responses to lenalidomide may be evident within one to three months or longer [65-69].

Efficacy – Beneficial effects of lenalidomide in refractory cutaneous LE have been reported [65-69]:

In an open-label study in which 15 females with refractory cutaneous LE (including nine with DLE and two with SCLE) were treated with lenalidomide (initially 5 mg per day and increased to 10 mg per day if no improvement), 12 patients achieved complete responses (Cutaneous Lupus Erythematosus Disease Area and Severity Index [CLASI] score = 0), including four who previously failed thalidomide [67]. The remaining patients included two patients with DLE who achieved only partial improvement (at least 50 percent improvement in the CLASI score [CLASI-50]) and one patient with DLE who was withdrawn from the study due to poor gastrointestinal tolerance of the drug. The patients with DLE and SCLE who achieved complete responses did so within a mean of 6 weeks (range 2 to 12 weeks), and the median length of treatment for all patients was 11 months. Relapses were common after the tapering or cessation of therapy.

In a smaller, six-week, open-label study, at least a four-point reduction in the CLASI score occurred in four of five patients (three with DLE and one with a mixed presentation of SCLE and tumid lupus erythematosus) who were treated with lenalidomide (5 mg per day), including two who had previously failed thalidomide therapy [66,70]. Of note, one of the responders with DLE developed symptoms of systemic lupus erythematosus (SLE) during treatment, an event that the authors postulated may have been a side effect of lenalidomide therapy. However, none of the patients in the larger, open-label study experienced a similar event [67].

In a multicenter, retrospective case series evaluating 40 patients with cutaneous LE who received lenalidomide after failure of hydroxychloroquine and at least one second-line systemic treatment, lenalidomide therapy was associated with a 20 percent reduction in the CLASI activity score (CLASI-20) in 98 percent of patients, CLASI-50 in 88 percent of patients, and a complete response (CLASI activity score of 0) in 43 percent of patients [68]. The complete response rate was decreased in active smokers.

In a retrospective review of 19 patients with cutaneous LE who had failed a median of six prior therapeutic agents, treatment with lenalidomide 5 mg per day resulted in an overall response rate of 89.5 percent (complete/nearly complete response in 12 out of 19 patients and a partial response in 5 out of 19 patients). Adverse events and serious adverse events occurred in 12 and 5 of the 19 patients, respectively [69].

Adverse effects – Examples of adverse effects of lenalidomide include teratogenicity (based upon animal studies), thrombocytopenia, and neutropenia. Thromboembolic events and hepatotoxicity have been reported, particularly in patients treated with lenalidomide and dexamethasone, as in multiple myeloma. (See "Lenalidomide: Drug information".)

Intravenous immune globulin — Intravenous immune globulin (IVIG) is a product derived from the pooled plasma of tens of thousands of donors that provides a vast array of immunoregulatory substances. The exact mechanisms of action of IVIG in the treatment of autoimmune disorders remain unknown (see "Overview of intravenous immune globulin (IVIG) therapy", section on 'Suppression of inflammatory/autoimmune processes'):

Administration – Dosing regimens for IVIG vary, although it is often administered at a dose of 2 g/kg every four to eight weeks depending upon response [71,72]. Although responses are prompt, they are typically short lived, necessitating repeated infusions to maintain benefit. Some patients may fail to respond to subsequent courses of therapy after relapse [73].

Because of the high cost of this agent and high likelihood for relapse, it should be reserved for attempting to gain rapid control of severe, refractory disease while initiating other long-term therapies.

Efficacy – Data are limited on the use of IVIG for DLE and SCLE:

The efficacy of IVIG was evaluated in an uncontrolled study of 12 patients with cutaneous LE that was refractory to other systemic therapies (including five patients with disseminated DLE and five patients with SCLE) [73]. Subjects were given a total of two doses of 1 g/kg of IVIG on consecutive days followed by 400 mg/kg monthly for up to six months. After treatment, five patients (42 percent) had more than 75 percent improvement in skin lesions, and two patients (17 percent) exhibited partial responses (50 to 75 percent improvement in skin lesions). Overall, patients with SCLE exhibited greater degrees of improvement than those with DLE, and relapse was common after the discontinuation of therapy. Several case reports have also supported the efficacy of IVIG for SCLE and DLE that had been refractory to other therapies [53,71,72,74].

In contrast, in a 12-month, open-label study of IVIG in seven patients with cutaneous LE (including five patients with SLE manifesting as malar rash and oral ulcers and two patients with SCLE), monthly infusions of IVIG at a dose of 300 mg/kg/day for five days were associated with no change in disease activity in the patients with SLE and worsening of disease in those with SCLE [75].

Adverse effects – Adverse events following IVIG infusion include hypersensitivity reactions, headache, vasculitis, aseptic meningitis, renal failure, myocardial infarction, and thrombosis. (See "Intravenous immune globulin: Adverse effects".)

OTHER THERAPIES — A number of other treatments, such as azathioprine, rituximab, and other immunomodulatory agents, have also been used in refractory DLE and SCLE. However, the evidence for the efficacy of these drugs is limited, and the range of adverse effects is wide, delegating the use of these agents to patients who fail to respond to the other therapies for refractory disease. Clofazimine has also been used for the treatment of refractory cutaneous lupus erythematosus (cutaneous LE), but the availability of this drug is limited.

Systemic corticosteroids — Because of the chronic nature of cutaneous LE, the existence of only limited evidence for the efficacy of systemic corticosteroids, and the numerous potential adverse effects of long-term therapy with systemic corticosteroids, these drugs are not generally recommended for the treatment of refractory DLE and SCLE. The role of systemic corticosteroids is generally limited to short courses during the initial treatment of extensive, severe, or scarring disease. (See "Initial management of discoid lupus erythematosus and subacute cutaneous lupus erythematosus", section on 'Systemic corticosteroids'.)

Azathioprine — Successful treatment of DLE and SCLE with azathioprine has been reported. In an open-label study, four patients with refractory SCLE and two patients with refractory DLE were treated with azathioprine (100 to 150 mg/day) and prednisone (20 to 30 mg/day) [76]. One patient with SCLE had an excellent response, with near clearing of skin lesions and a decreased oral prednisone requirement. Two additional patients with SCLE exhibited partial responses. Adverse effects led to the discontinuation of therapy in two patients, and one patient with severe, erosive, palmar DLE failed to respond to therapy. However, several case reports have supported the use of azathioprine in patients with extensive or recalcitrant DLE [77-79]. Improvement with azathioprine usually occurs within one to two months [8].

Adverse effects of azathioprine include bone marrow suppression [80], gastrointestinal upset [81-83], hepatitis, and abnormal liver function tests [80,84]. The risk of myelosuppression may be related to thiopurine methyltransferase (TPMT) activity, an enzyme involved in the metabolism of azathioprine. Assessment of the TPMT enzyme activity should be performed prior to initiating azathioprine therapy [85]. Most patients with cutaneous LE and normal TPMT activity are treated with 1 to 2.5 mg/kg/day of azathioprine. (See "Overview of pharmacogenomics", section on 'Thiopurines and polymorphisms in TPMT and NUDT15'.)

An increased incidence of lymphoproliferative malignancies has been reported in patients with rheumatoid arthritis treated with azathioprine [86,87]. However, it has not been proven that a similar risk occurs in patients treated with azathioprine for cutaneous LE. One retrospective study (n = 358) that compared the incidence of lymphoma in patients with systemic lupus erythematosus (SLE) who were treated with azathioprine versus those who had not received the drug found no significant difference in the risk for lymphoma or other malignancies between the two groups [88].

Rituximab — Originally approved in 1997 for the treatment of non-Hodgkin lymphoma, rituximab is a chimeric monoclonal antibody specific for human CD20, a membrane-associated glycoprotein highly expressed on the surface of pre-B cells and resting and activated mature B lymphocytes [89]. The binding of rituximab to CD20 results in B cell death [89].

Rituximab use is generally limited to patients with severe, extensive cutaneous LE that has failed to respond to other therapies for refractory disease. Although a few case reports document improvement in severe, refractory SCLE with rituximab therapy [90-93], other data suggest poor efficacy for DLE and variable efficacy for SCLE:

In a prospective, uncontrolled study of 82 patients with SLE, none of eight patients with chronic cutaneous LE (DLE or chilblains) at baseline and one of two patients with SCLE at baseline responded to rituximab (1 g twice daily on days 1 and 15) [94]. In addition, nine patients who lacked skin disease or had acute cutaneous LE at baseline experienced flares of chronic cutaneous LE or SCLE after rituximab therapy.

In a separate study of 17 patients treated with a rituximab-based, B cell-depleting regimen (two 1 g doses of rituximab separated by two weeks and 750 mg of cyclophosphamide given one day after the first rituximab infusion) for SLE and/or cutaneous LE, only three of eight patients with chronic cutaneous LE were in complete or partial remission six months after treatment [95]. The same endpoint was achieved by two of three patients with SCLE.

Although generally well tolerated, the adverse effects of rituximab include infections; nausea; serum sickness-like reactions [96]; and infusion-related events, including hypotension, fevers, and rigors. A boxed warning has been mandated by the US Food and Drug Administration (FDA) due to reports of fatalities from infusion reactions, severe mucocutaneous reactions, and progressive multifocal leukoencephalopathy due to JC virus in patients treated with rituximab.

Cyclosporine — Cyclosporine exerts its immunosuppressive effects via the inhibition of interleukin (IL) 2 and other cytokines. Combination therapy with cyclosporine and hydroxychloroquine was effective in improving skin lesions in a patient with SCLE and generalized lichen planus [97]. However, in other reports, cyclosporine failed to induce improvement in patients with DLE that was refractory to other therapies [98,99]. DLE has also occurred during treatment with cyclosporine for other disorders in patients without a history of cutaneous LE [100,101]. (See "Pharmacology of cyclosporine and tacrolimus".)

Belimumab — Belimumab is a monoclonal antibody that reduces B lymphocyte survival by blocking the binding of soluble human B lymphocyte stimulator to its B cell receptors. It is used for the treatment of SLE. The effect of belimumab in cutaneous LE has not been well studied. However, in a series of five patients with SLE and acute cutaneous lupus erythematosus, SCLE, and/or DLE, all had significant improvement in the Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) disease activity scores with the addition of intravenous belimumab to standard treatment [102]. The average time to clinical improvement was four months, and marked improvement occurred as early as eight weeks.

In a multicenter, retrospective, observational study of patients with cutaneous LE that was refractory to hydroxychloroquine and at least two second-line agents, treatment with belimumab 10 mg/kg every two weeks for three doses and then monthly resulted in 50 percent improvement in the CLASI activity score (CLASI-50) in 8 of 16 patients and a complete response in 3 of 16 patients [103]. CLASI-50 was observed more frequently in patients with Fitzpatrick skin phototypes IV to VI than II or III (table 1), and baseline CLASI tended to be lower in patients with complete response than without.

Serious potential adverse effects of belimumab include hypersensitivity reactions, infections, and depression. (See "Overview of the management and prognosis of systemic lupus erythematosus in adults", section on 'Approach to drug therapy'.)

Anifrolumab — Anifrolumab (a humanized immunoglobulin G1 [IgG1] monoclonal antibody that binds interferon [IFN]-alpha/beta/omega receptors and prevents type 1 IFN signaling) has emerged as a potential treatment option for refractory cutaneous LE [104-106]. Improvement in the severity of skin disease in patients with SLE was reported in a phase 3, placebo-controlled, randomized trial that assessed efficacy of anifrolumab for SLE [105]. Additionally, improvement in refractory cutaneous LE has been documented in a series of three patients with SLE, including two patients who were smokers [106]. (See "Overview of the management and prognosis of systemic lupus erythematosus in adults", section on 'Anifrolumab'.)

Additional therapies — Other therapies that have been used in the treatment of cutaneous LE include ustekinumab [107,108], phenytoin [109], sulfasalazine [110-112], cefuroxime axetil [113], danazol [114,115], Janus kinase (JAK) inhibitors [116-120], and extracorporeal photophoresis [121-123]. Despite reports of efficacy in cutaneous LE, phenytoin and sulfasalazine have also been associated with the development of drug-induced lupus. (See "Drug-induced lupus".)

Use of a novel tacrolimus 0.3% lotion was associated with reduced disease severity and hair regrowth in three patients with refractory DLE [124]. Tacrolimus 0.3% lotion is not commercially available.

Methyl aminolevulinic-photodynamic therapy (MAL-PDT) appeared effective for DLE in two case reports that described patients who failed or could not tolerate other therapies [125,126]. However, aminolevulinic acid-photodynamic therapy (ALA-PDT) was not useful in two patients with refractory DLE in another report [127].

Investigational therapy — Litifilimab, a humanized IgG1 monoclonal antibody that binds dendritic cell antigen 2 (a plasmacytoid dendritic cell-specific receptor that inhibits type 1 IFN, inflammatory cytokines, and chemokines), appeared beneficial for cutaneous LE in a phase 2 trial [128].

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

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

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

Basics topic (see "Patient education: Discoid lupus (The Basics)")

SUMMARY AND RECOMMENDATIONS

Overview – Most patients with discoid lupus erythematosus (DLE) and subacute cutaneous lupus erythematosus (SCLE) respond well to treatment with photoprotection, topical medications, and systemic antimalarial drugs (algorithm 1). Patients who fail to respond to these agents may benefit from alternative systemic therapies. (See 'Treatment principles' above.)

Treatment selection – The selection of a treatment for refractory cutaneous lupus erythematosus (cutaneous LE) is based upon consideration of drug efficacy, potential adverse effects, and treatment availability:

Choice after failure of antimalarial therapy – For patients with DLE and SCLE who fail treatment with antimalarial therapy, we suggest treatment with methotrexate before trials of other systemic agents for refractory disease (Grade 2C). Methotrexate appears efficacious for refractory cutaneous LE, can be used for long-term therapy, and is relatively inexpensive.

For patients who cannot tolerate methotrexate or do not respond to this drug, additional treatment options include an oral retinoid, mycophenolate mofetil, and dapsone. (See 'After failure of antimalarial therapy' above.)

Patients requiring remission-inducing therapies – Patients who fail to improve with methotrexate, oral retinoids, mycophenolate mofetil, and/or dapsone may require more aggressive therapies to induce remission.

For these patients, we suggest treatment with thalidomide rather than other therapies (Grade 2C). Intravenous immune globulin (IVIG) and lenalidomide are additional therapeutic options for inducing remission in patients with refractory cutaneous LE. (See 'Remission-inducing therapy' above.)

Special safety considerationsMethotrexate, oral retinoids, thalidomide, and lenalidomide are contraindicated in pregnancy due to teratogenicity. Acitretin also requires a prolonged period of pregnancy avoidance after the completion of therapy.

In the United States, both clinicians and patients must register in a Risk Evaluation and Mitigation Strategy (REMS) program prior to the initiation of isotretinoin, thalidomide, or lenalidomide. Patients treated with thalidomide should also be carefully monitored for the development of peripheral neuropathy.

Use of other therapies – Other therapies, such as azathioprine, rituximab, and other immunomodulatory agents, have been used in refractory DLE and SCLE. However, limited efficacy data and/or the potential for severe adverse effects limit the use of these drugs to disease that has not responded to other therapies or to patients for whom other therapies are contraindicated.

  1. Kuhn A, Aberer E, Bata-Csörgő Z, et al. S2k guideline for treatment of cutaneous lupus erythematosus - guided by the European Dermatology Forum (EDF) in cooperation with the European Academy of Dermatology and Venereology (EADV). J Eur Acad Dermatol Venereol 2017; 31:389.
  2. Jessop S, Whitelaw DA, Delamere FM. Drugs for discoid lupus erythematosus. Cochrane Database Syst Rev 2009; :CD002954.
  3. Albrecht J, Taylor L, Berlin JA, et al. The CLASI (Cutaneous Lupus Erythematosus Disease Area and Severity Index): an outcome instrument for cutaneous lupus erythematosus. J Invest Dermatol 2005; 125:889.
  4. Kushner CJ, Tarazi M, Gaffney RG, et al. Evaluation of the reliability and validity of the Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) in paediatric cutaneous lupus among paediatric dermatologists and rheumatologists. Br J Dermatol 2019; 180:165.
  5. Jolly M, Kazmi N, Mikolaitis RA, et al. Validation of the Cutaneous Lupus Disease Area and Severity Index (CLASI) using physician- and patient-assessed health outcome measures. J Am Acad Dermatol 2013; 68:618.
  6. Kuhn A, Meuth AM, Bein D, et al. Revised Cutaneous Lupus Erythematosus Disease Area and Severity Index (RCLASI): a modified outcome instrument for cutaneous lupus erythematosus. Br J Dermatol 2010; 163:83.
  7. Kuhn A, Ruland V, Bonsmann G. Cutaneous lupus erythematosus: update of therapeutic options part II. J Am Acad Dermatol 2011; 65:e195.
  8. Kuhn A, Specker C, Ruzicka T, Lehmann P. Methotrexate treatment for refractory subacute cutaneous lupus erythematosus. J Am Acad Dermatol 2002; 46:600.
  9. Genestier L, Paillot R, Quemeneur L, et al. Mechanisms of action of methotrexate. Immunopharmacology 2000; 47:247.
  10. Böhm I. Decrease of B-cells and autoantibodies after low-dose methotrexate. Biomed Pharmacother 2003; 57:278.
  11. Ortiz Z, Shea B, Suarez-Almazor ME, et al. The efficacy of folic acid and folinic acid in reducing methotrexate gastrointestinal toxicity in rheumatoid arthritis. A metaanalysis of randomized controlled trials. J Rheumatol 1998; 25:36.
  12. Wenzel J, Brähler S, Bauer R, et al. Efficacy and safety of methotrexate in recalcitrant cutaneous lupus erythematosus: results of a retrospective study in 43 patients. Br J Dermatol 2005; 153:157.
  13. Carneiro JR, Sato EI. Double blind, randomized, placebo controlled clinical trial of methotrexate in systemic lupus erythematosus. J Rheumatol 1999; 26:1275.
  14. Böhm L, Uerlich M, Bauer R. Rapid improvement of subacute cutaneous lupus erythematosus with low-dose methotrexate. Dermatology 1997; 194:307.
  15. Malcangi G, Brandozzi G, Giangiacomi M, et al. Bullous SLE: response to methotrexate and relationship with disease activity. Lupus 2003; 12:63.
  16. Boehm IB, Boehm GA, Bauer R. Management of cutaneous lupus erythematosus with low-dose methotrexate: indication for modulation of inflammatory mechanisms. Rheumatol Int 1998; 18:59.
  17. Shimizu S, Yasui C, Kawasaki H, Tsuchiya K. Dramatic efficacy of oral aromatic retinoid in long-standing hypertrophic lupus erythematosus. Acta Derm Venereol 2004; 84:491.
  18. Ruzicka T, Meurer M, Braun-Falco O. Treatment of cutaneous lupus erythematosus with etretinate. Acta Derm Venereol 1985; 65:324.
  19. Ruzicka T, Meurer M, Bieber T. Efficiency of acitretin in the treatment of cutaneous lupus erythematosus. Arch Dermatol 1988; 124:897.
  20. Al-Mutairi N, Rijhwani M, Nour-Eldin O. Hypertrophic lupus erythematosus treated successfully with acitretin as monotherapy. J Dermatol 2005; 32:482.
  21. Newton RC, Jorizzo JL, Solomon AR Jr, et al. Mechanism-oriented assessment of isotretinoin in chronic or subacute cutaneous lupus erythematosus. Arch Dermatol 1986; 122:170.
  22. Ruzicka T, Sommerburg C, Goerz G, et al. Treatment of cutaneous lupus erythematosus with acitretin and hydroxychloroquine. Br J Dermatol 1992; 127:513.
  23. Shornick JK, Formica N, Parke AL. Isotretinoin for refractory lupus erythematosus. J Am Acad Dermatol 1991; 24:49.
  24. Furner BB. Subacute cutaneous lupus erythematosus response to isotretinoin. Int J Dermatol 1990; 29:587.
  25. Green SG, Piette WW. Successful treatment of hypertrophic lupus erythematosus with isotretinoin. J Am Acad Dermatol 1987; 17:364.
  26. Green PJ, Pasternak S. Hypertrophic and ulcerated discoid lupus erythematosus. J Cutan Med Surg 2012; 16:453.
  27. Kuhn A, Patsinakidis N, Luger T. Alitretinoin for cutaneous lupus erythematosus. J Am Acad Dermatol 2012; 67:e123.
  28. O'Kane D, McCourt C, Meggitt S, et al. British Association of Dermatologists guidelines for the management of people with cutaneous lupus erythematosus 2021. Br J Dermatol 2021; 185:1112.
  29. Lu Q, Long H, Chow S, et al. Guideline for the diagnosis, treatment and long-term management of cutaneous lupus erythematosus. J Autoimmun 2021; 123:102707.
  30. Sadlier M, Kirby B, Lally A. Mycophenolate mofetil and hydroxychloroquine: an effective treatment for recalcitrant cutaneous lupus erythematosus. J Am Acad Dermatol 2012; 66:160.
  31. Gammon B, Hansen C, Costner MI. Efficacy of mycophenolate mofetil in antimalarial-resistant cutaneous lupus erythematosus. J Am Acad Dermatol 2011; 65:717.
  32. Kreuter A, Tomi NS, Weiner SM, et al. Mycophenolate sodium for subacute cutaneous lupus erythematosus resistant to standard therapy. Br J Dermatol 2007; 156:1321.
  33. Keyes E, Jobanputra A, Feng R, et al. Comparative responsiveness of cutaneous lupus erythematosus patients to methotrexate and mycophenolate mofetil: A cohort study. J Am Acad Dermatol 2022; 87:447.
  34. Goyal S, Nousari HC. Treatment of resistant discoid lupus erythematosus of the palms and soles with mycophenolate mofetil. J Am Acad Dermatol 2001; 45:142.
  35. Hanjani NM, Nousari CH. Mycophenolate mofetil for the treatment of cutaneous lupus erythematosus with smoldering systemic involvement. Arch Dermatol 2002; 138:1616.
  36. Schanz S, Ulmer A, Rassner G, Fierlbeck G. Successful treatment of subacute cutaneous lupus erythematosus with mycophenolate mofetil. Br J Dermatol 2002; 147:174.
  37. Bardazzi F, Gallo G, Loi C, et al. Refractory palmo-plantar discoid lupus erythematosus successfully treated with mycophenolate mofetil: Unusual localization and literature review. Dermatol Ther 2021; 34:e14743.
  38. Pisoni CN, Obermoser G, Cuadrado MJ, et al. Skin manifestations of systemic lupus erythematosus refractory to multiple treatment modalities: poor results with mycophenolate mofetil. Clin Exp Rheumatol 2005; 23:393.
  39. Hall RP, Lawley TJ, Smith HR, Katz SI. Bullous eruption of systemic lupus erythematosus. Dramatic response to dapsone therapy. Ann Intern Med 1982; 97:165.
  40. Ludgate MW, Greig DE. Bullous systemic lupus erythematosus responding to dapsone. Australas J Dermatol 2008; 49:91.
  41. Tay YK, Wong SN, Tan T. Bullous systemic lupus erythematosus--a case report and review. Ann Acad Med Singapore 1995; 24:879.
  42. Klebes M, Wutte N, Aberer E. Dapsone as Second-Line Treatment for Cutaneous Lupus Erythematosus? A Retrospective Analysis of 34 Patients and a Review of the Literature. Dermatology 2016; 232:91.
  43. McCormack LS, Elgart ML, Turner ML. Annular subacute cutaneous lupus erythematosus responsive to dapsone. J Am Acad Dermatol 1984; 11:397.
  44. Neri R, Mosca M, Bernacchi E, Bombardieri S. A case of SLE with acute, subacute and chronic cutaneous lesions successfully treated with Dapsone. Lupus 1999; 8:240.
  45. Tsutsui K, Imai T, Hatta N, et al. Widespread pruritic plaques in a patient with subacute cutaneous lupus erythematosus and hypocomplementemia: response to dapsone therapy. J Am Acad Dermatol 1996; 35:313.
  46. Fenton DA, Black MM. Low-dose dapsone in the treatment of subacute cutaneous lupus erythematosus. Clin Exp Dermatol 1986; 11:102.
  47. Holtman JH, Neustadt DH, Klein J, Callen JP. Dapsone is an effective therapy for the skin lesions of subacute cutaneous lupus erythematosus and urticarial vasculitis in a patient with C2 deficiency. J Rheumatol 1990; 17:1222.
  48. Zampeli E, Moutsopoulos HM. Dapsone: an old drug effective for subacute cutaneous lupus erythematosus. Rheumatology (Oxford) 2019; 58:920.
  49. Coburn PR, Shuster S. Dapsone and discoid lupus erythematosus. Br J Dermatol 1982; 106:105.
  50. Lindskov R, Reymann F. Dapsone in the treatment of cutaneous lupus erythematosus. Dermatologica 1986; 172:214.
  51. Hall R. Dapsone. In: Comprehensive Dermatologic Therapy, Wolverton S (Ed), WB Saunders Co, 2001. p.230.
  52. Hall RP, Mickle CP. Dapsone. In: Comprehensive Dermatologic Drug Therapy, 2nd ed, Wolverton SE (Ed), Elsevier Inc., 2007. p.240.
  53. Tenti S, Fabbroni M, Mancini V, et al. Intravenous Immunoglobulins as a new opportunity to treat discoid lupus erythematosus: A case report and review of the literature. Autoimmun Rev 2018; 17:791.
  54. Yuki EFN, Silva CA, Aikawa NE, et al. Thalidomide and Lenalidomide for Refractory Systemic/Cutaneous Lupus Erythematosus Treatment: A Narrative Review of Literature for Clinical Practice. J Clin Rheumatol 2021; 27:248.
  55. Lu KQ, Brenneman S, Burns R Jr, et al. Thalidomide inhibits UVB-induced mouse keratinocyte apoptosis by both TNF-alpha-dependent and TNF-alpha-independent pathways. Photodermatol Photoimmunol Photomed 2003; 19:272.
  56. Zampieri S, Alaibac M, Iaccarino L, et al. Tumour necrosis factor alpha is expressed in refractory skin lesions from patients with subacute cutaneous lupus erythematosus. Ann Rheum Dis 2006; 65:545.
  57. Pelle MT, Werth VP. Thalidomide in cutaneous lupus erythematosus. Am J Clin Dermatol 2003; 4:379.
  58. Cortés-Hernández J, Torres-Salido M, Castro-Marrero J, et al. Thalidomide in the treatment of refractory cutaneous lupus erythematosus: prognostic factors of clinical outcome. Br J Dermatol 2012; 166:616.
  59. System for thalidomide education and prescribing safety (STEPSTM), Starter Kit and Prescriber Info. Celegene Corporation, Warren, NJ 2001.
  60. Chasset F, Tounsi T, Cesbron E, et al. Efficacy and tolerance profile of thalidomide in cutaneous lupus erythematosus: A systematic review and meta-analysis. J Am Acad Dermatol 2018; 78:342.
  61. Cesbron E, Bessis D, Jachiet M, et al. Risk of thromboembolic events in patients treated with thalidomide for cutaneous lupus erythematosus: A multicenter retrospective study. J Am Acad Dermatol 2018; 79:162.
  62. Bastuji-Garin S, Ochonisky S, Bouche P, et al. Incidence and risk factors for thalidomide neuropathy: a prospective study of 135 dermatologic patients. J Invest Dermatol 2002; 119:1020.
  63. Briani C, Zara G, Rondinone R, et al. Positive and negative effects of thalidomide on refractory cutaneous lupus erythematosus. Autoimmunity 2005; 38:549.
  64. Ochonisky S, Verroust J, Bastuji-Garin S, et al. Thalidomide neuropathy incidence and clinico-electrophysiologic findings in 42 patients. Arch Dermatol 1994; 130:66.
  65. Shah A, Albrecht J, Bonilla-Martinez Z, et al. Lenalidomide for the treatment of resistant discoid lupus erythematosus. Arch Dermatol 2009; 145:303.
  66. Braunstein I, Goodman NG, Rosenbach M, et al. Lenalidomide therapy in treatment-refractory cutaneous lupus erythematosus: histologic and circulating leukocyte profile and potential risk of a systemic lupus flare. J Am Acad Dermatol 2012; 66:571.
  67. Cortés-Hernández J, Ávila G, Vilardell-Tarrés M, Ordi-Ros J. Efficacy and safety of lenalidomide for refractory cutaneous lupus erythematosus. Arthritis Res Ther 2012; 14:R265.
  68. Aitmehdi R, Arnaud L, Francès C, et al. Long-term efficacy and safety outcomes of lenalidomide for cutaneous lupus erythematosus: A multicenter retrospective observational study of 40 patients. J Am Acad Dermatol 2021; 84:1171.
  69. Reymann V, Bessis D, Bergeret B, et al. Efficacy and safety of low-dose oral lenalidomide in refractory cutaneous lupus erythematosus: an open series of 19 cases. J Eur Acad Dermatol Venereol 2021; 35:e113.
  70. Okon L, Rosenbach M, Krathen M, et al. Lenalidomide in treatment-refractory cutaneous lupus erythematosus: Efficacy and safety in a 52-week trial. J Am Acad Dermatol 2014; 70:583.
  71. Généreau T, Chosidow O, Danel C, et al. High-dose intravenous immunoglobulin in cutaneous lupus erythematosus. Arch Dermatol 1999; 135:1124.
  72. Lampropoulos CE, Hughes GR, D' Cruz DP. Intravenous immunoglobulin in the treatment of resistant subacute cutaneous lupus erythematosus: a possible alternative. Clin Rheumatol 2007; 26:981.
  73. Goodfield M, Davison K, Bowden K. Intravenous immunoglobulin (IVIg) for therapy-resistant cutaneous lupus erythematosus (LE). J Dermatolog Treat 2004; 15:46.
  74. Kreuter A, Hyun J, Altmeyer P, Gambichler T. Intravenous immunoglobulin for recalcitrant subacute cutaneous lupus erythematosus. Acta Derm Venereol 2005; 85:545.
  75. De Pità O, Bellucci AM, Ruffelli M, et al. Intravenous immunoglobulin therapy is not able to efficiently control cutaneous manifestations in patients with lupus erythematosus. Lupus 1997; 6:415.
  76. Callen JP, Spencer LV, Burruss JB, Holtman J. Azathioprine. An effective, corticosteroid-sparing therapy for patients with recalcitrant cutaneous lupus erythematosus or with recalcitrant cutaneous leukocytoclastic vasculitis. Arch Dermatol 1991; 127:515.
  77. Tsokos GC, Caughman SW, Klippel JH. Successful treatment of generalized discoid skin lesions with azathioprine. Its use in a patient with systemic lupus erythematosus. Arch Dermatol 1985; 121:1323.
  78. Shehade S. Successful treatment of generalized discoid skin lesions with azathioprine. Arch Dermatol 1986; 122:376.
  79. Ashinoff R, Werth VP, Franks AG Jr. Resistant discoid lupus erythematosus of palms and soles: successful treatment with azathioprine. J Am Acad Dermatol 1988; 19:961.
  80. Whisnant JK, Pelkey J. Rheumatoid arthritis: treatment with azathioprine (IMURAN (R)). Clinical side-effects and laboratory abnormalities. Ann Rheum Dis 1982; 41 Suppl 1:44.
  81. Santiago M. Diarrhoea secondary to azathioprine in two patients with SLE. Lupus 1999; 8:565.
  82. Assini JF, Hamilton R, Strosberg JM. Adverse reactions to azathioprine mimicking gastroenteritis. J Rheumatol 1986; 13:1117.
  83. Godeau B, Paul M, Autegarden JE, et al. Hypersensitivity to azathioprine mimicking gastroenteritis. Absence of recurrence with 6-mercaptopurine. Gastroenterol Clin Biol 1995; 19:117.
  84. Eisenbach C, Goeggelmann C, Flechtenmacher C, et al. Severe cholestatic hepatitis caused by azathioprine. Immunopharmacol Immunotoxicol 2005; 27:77.
  85. Snow JL, Gibson LE. The role of genetic variation in thiopurine methyltransferase activity and the efficacy and/or side effects of azathioprine therapy in dermatologic patients. Arch Dermatol 1995; 131:193.
  86. Silman AJ, Petrie J, Hazleman B, Evans SJ. Lymphoproliferative cancer and other malignancy in patients with rheumatoid arthritis treated with azathioprine: a 20 year follow up study. Ann Rheum Dis 1988; 47:988.
  87. Sutcliffe N, Smith C, Speight PM, Isenberg DA. Mucosa-associated lymphoid tissue lymphomas in two patients with rheumatoid arthritis on second-line agents, and secondary Sjögren's syndrome. Rheumatology (Oxford) 2000; 39:185.
  88. Nero P, Rahman A, Isenberg DA. Does long term treatment with azathioprine predispose to malignancy and death in patients with systemic lupus erythematosus? Ann Rheum Dis 2004; 63:325.
  89. Silverman GJ. Anti-CD20 therapy in systemic lupus erythematosus: a step closer to the clinic. Arthritis Rheum 2005; 52:371.
  90. Kieu V, O'Brien T, Yap LM, et al. Refractory subacute cutaneous lupus erythematosus successfully treated with rituximab. Australas J Dermatol 2009; 50:202.
  91. Uthman I, Taher A, Abbas O, et al. Successful treatment of refractory skin manifestations of systemic lupus erythematosus with rituximab: report of a case. Dermatology 2008; 216:257.
  92. Penha MÁ, Libório RDS, Miot HA. Rituximab in the treatment of extensive and refractory subacute cutaneous lupus erythematosus. An Bras Dermatol 2018; 93:467.
  93. Dodds M, Hobday P, Schultz B, Maguiness S. Successful treatment of severe subacute cutaneous lupus erythematosus with rituximab in an adolescent. Pediatr Dermatol 2018; 35:e189.
  94. Vital EM, Wittmann M, Edward S, et al. Brief report: responses to rituximab suggest B cell-independent inflammation in cutaneous systemic lupus erythematosus. Arthritis Rheumatol 2015; 67:1586.
  95. Hofmann SC, Leandro MJ, Morris SD, Isenberg DA. Effects of rituximab-based B-cell depletion therapy on skin manifestations of lupus erythematosus--report of 17 cases and review of the literature. Lupus 2013; 22:932.
  96. Hellerstedt B, Ahmed A. Delayed-type hypersensitivity reaction or serum sickness after rituximab treatment. Ann Oncol 2003; 14:1792.
  97. Grabbe S, Kolde G. Coexisting lichen planus and subacute cutaneous lupus erythematosus. Clin Exp Dermatol 1995; 20:249.
  98. Heule F, van Joost T, Beukers R. Cyclosporine in the treatment of lupus erythematosus. Arch Dermatol 1986; 122:973.
  99. Yell JA, Burge SM. Cyclosporin and discoid lupus erythematosus. Br J Dermatol 1994; 131:132.
  100. Obermoser G, Weber F, Sepp N. Discoid lupus erythematosus in a patient receiving cyclosporine for liver transplantation. Acta Derm Venereol 2001; 81:319.
  101. Di Lernia V, Bisighini G. Discoid lupus erythematosus during treatment with cyclosporine. Acta Derm Venereol 1996; 76:87.
  102. Vashisht P, Borghoff K, O'Dell JR, Hearth-Holmes M. Belimumab for the treatment of recalcitrant cutaneous lupus. Lupus 2017; 26:857.
  103. Salle R, Chasset F, Kottler D, et al. Belimumab for refractory manifestations of cutaneous lupus: A multicenter, retrospective observational study of 16 patients. J Am Acad Dermatol 2020; 83:1816.
  104. Merrill JT, Furie R, Werth VP, et al. Anifrolumab effects on rash and arthritis: impact of the type I interferon gene signature in the phase IIb MUSE study in patients with systemic lupus erythematosus. Lupus Sci Med 2018; 5:e000284.
  105. Morand EF, Furie R, Tanaka Y, et al. Trial of Anifrolumab in Active Systemic Lupus Erythematosus. N Engl J Med 2020; 382:211.
  106. Blum FR, Sampath AJ, Foulke GT. Anifrolumab for treatment of refractory cutaneous lupus erythematosus. Clin Exp Dermatol 2022; 47:1998.
  107. De Souza A, Ali-Shaw T, Strober BE, Franks AG Jr. Successful treatment of subacute lupus erythematosus with ustekinumab. Arch Dermatol 2011; 147:896.
  108. Mazgaj M, Picard-Dahan C, Deschamps L, et al. Successful ustekinumab treatment in a patient with psoriasis and subacute cutaneous lupus erythematosus. Int J Dermatol 2020; 59:e118.
  109. Rodriguez-Castellanos MA, Barba Rubio J, Barba Gómez JF, González Méndoza A. Phenytoin in the treatment of discoid lupus erythematosus. Arch Dermatol 1995; 131:620.
  110. Artüz F, Lenk N, Deniz N, Alli N. Efficacy of sulfasalazine in discoid lupus erythematosus. Int J Dermatol 1996; 35:746.
  111. Carmichael AJ, Paul CJ. Discoid lupus erythematosus responsive to sulphasalazine. Br J Dermatol 1991; 125:291.
  112. Delaporte E, Catteau B, Sabbagh N, et al. [Treatment of discoid lupus erythematosus with sulfasalazine: 11 cases]. Ann Dermatol Venereol 1997; 124:151.
  113. Rudnicka L, Szymańska E, Walecka I, Słowińska M. Long-term cefuroxime axetil in subacute cutaneous lupus erythematosus. A report of three cases. Dermatology 2000; 200:129.
  114. Torrelo A, España A, Medina S, Ledo A. Danazol and discoid lupus erythematosus. Dermatologica 1990; 181:239.
  115. Englert HJ, Hughes GV. Danazol and discoid lupus. Br J Dermatol 1988; 119:407.
  116. Fetter T, Smith P, Guel T, et al. Selective Janus Kinase 1 Inhibition Is a Promising Therapeutic Approach for Lupus Erythematosus Skin Lesions. Front Immunol 2020; 11:344.
  117. Wenzel J, van Holt N, Maier J, et al. JAK1/2 Inhibitor Ruxolitinib Controls a Case of Chilblain Lupus Erythematosus. J Invest Dermatol 2016; 136:1281.
  118. Klaeschen AS, Wolf D, Brossart P, et al. JAK inhibitor ruxolitinib inhibits the expression of cytokines characteristic of cutaneous lupus erythematosus. Exp Dermatol 2017; 26:728.
  119. Morand E, Pike M, Merrill JT, et al. Deucravacitinib, a Tyrosine Kinase 2 Inhibitor, in Systemic Lupus Erythematosus: A Phase II, Randomized, Double-Blind, Placebo-Controlled Trial. Arthritis Rheumatol 2023; 75:242.
  120. Bonnardeaux E, Dutz JP. Oral tofacitinib citrate for recalcitrant cutaneous lupus. JAAD Case Rep 2022; 20:61.
  121. Richter HI, Krutmann J, Goerz G. [Extracorporeal photopheresis in therapy-refractory disseminated discoid lupus erythematosus]. Hautarzt 1998; 49:487.
  122. Wollina U, Looks A. Extracorporeal photochemotherapy in cutaneous lupus erythematosus. J Eur Acad Dermatol Venereol 1999; 13:127.
  123. Boeckler P, Liu V, Lipsker D. Extracorporeal photopheresis in recalcitrant lupus erythematosus. Clin Exp Dermatol 2009; 34:e295.
  124. Milam EC, Ramachandran S, Franks AG Jr. Treatment of Scarring Alopecia in Discoid Variant of Chronic Cutaneous Lupus Erythematosus With Tacrolimus Lotion, 0.3. JAMA Dermatol 2015; 151:1113.
  125. Fernández-Guarino M, Pérez-García B, Harto A, Jaén P. Discoid lupus erythematosus: good response to treatment with photodynamic therapy. J Eur Acad Dermatol Venereol 2008; 22:1142.
  126. Debu A, Girard C, Bessis D. Discoid lupus erythematosus successfully treated by photodynamic therapy. Br J Dermatol 2015; 172:821.
  127. Romero-Maté A, Castaño-Suárez E, García-Donoso C, et al. Unsuccessful treatment of recalcitrant cutaneous discoid lupus erythematosus with photodynamic therapy. Photodermatol Photoimmunol Photomed 2010; 26:156.
  128. Werth VP, Furie RA, Romero-Diaz J, et al. Trial of Anti-BDCA2 Antibody Litifilimab for Cutaneous Lupus Erythematosus. N Engl J Med 2022; 387:321.
Topic 13775 Version 35.0

References

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