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

Pyoderma gangrenosum: Treatment and prognosis

Pyoderma gangrenosum: Treatment and prognosis
Literature review current through: Jan 2024.
This topic last updated: Dec 04, 2023.

INTRODUCTION — Pyoderma gangrenosum (PG) is an uncommon inflammatory and ulcerative skin disorder characterized histopathologically by the accumulation of neutrophils in the skin. The most common presentation of PG is the rapid development of one or more painful, purulent ulcers with undermined borders on sites of normal or traumatized skin (picture 1A-C).

Although multiple local and systemic therapies have been utilized for PG, high-quality efficacy studies are lacking for most interventions. Topical corticosteroids, systemic corticosteroids, and cyclosporine are common initial therapies (algorithm 1).

The management and prognosis of PG will be reviewed here. The pathogenesis, clinical features, and diagnosis of PG are reviewed separately. (See "Pyoderma gangrenosum: Pathogenesis, clinical features, and diagnosis".)

TREATMENT PRINCIPLES — In general, after the diagnosis of PG is made, patients are managed with a combination of pharmacologic therapies that suppress the inflammatory process and wound care measures that optimize the environment for wound healing. Although initial signs of improvement may be evident within days of the start of some treatments, weeks to months are often required to achieve complete ulcer healing [1,2].

Major principles of treatment include:

Importance of confirming the diagnosis – The careful exclusion of causes of cutaneous ulceration (eg, vascular occlusion disorders, venous disease, vasculitis, malignancy, cutaneous infection, trauma, and other ulcerative inflammatory disorders) prior to treatment is critical. Some therapies utilized for PG can be ineffective or harmful in other diseases. (See "Pyoderma gangrenosum: Pathogenesis, clinical features, and diagnosis", section on 'Diagnosis' and "Pyoderma gangrenosum: Pathogenesis, clinical features, and diagnosis", section on 'Differential diagnosis'.)

Limited efficacy data – Due to a paucity of high-quality data on interventions for PG, definitive guidelines for patient management are lacking. The approach to the treatment of PG is primarily guided by small uncontrolled studies and reports of clinical experience [3]. (See 'Treatment selection' below.)

Role of disease severity in initial treatment selection – The severity of PG influences our choice of initial therapy. Although topical therapies successfully induce healing in some patients with limited disease, patients with more extensive PG generally require systemic therapy to arrest disease activity. (See 'Limited disease' below and 'More extensive disease and treatment-resistant limited disease' below.)

Role of adjunctive systemic therapy – Systemic administration of glucocorticoids or cyclosporine is our preferred initial treatment for extensive PG. However, monotherapy can be insufficient, and long-term treatment with these drugs is associated with risk for serious adverse effects. We frequently incorporate other systemic immunomodulatory drugs (eg, infliximab, adalimumab, mycophenolate mofetil) to reduce exposure to systemic glucocorticoids and cyclosporine and promote healing of PG. (See 'Subsequent management' below.)

Impact of concomitant PG-associated diseases PG can occur in association with a variety of disorders, such as inflammatory bowel disease, inflammatory arthritis, malignancy, and hematologic disease. The presence of an associated disorder often influences the selection of adjunctive or subsequent treatments for PG, contributing to the selection of treatments that have benefit in both PG and the associated disease. (See 'Prognosis' below and 'Subsequent management' below and "Pyoderma gangrenosum: Pathogenesis, clinical features, and diagnosis", section on 'Associated disorders'.)

Assessing treatment response – Stabilization of disease progression and reductions in pain and visible signs of inflammation (eg, violaceous discoloration at the wound border) are important early signs of response. Subsequently, the wound base begins to fill with granulation tissue, and the wound diameter progressively shrinks. Documentation of the size, number, and location of ulcers and serial clinical photographs are useful for following the response to therapy.

Wound management Wound care is an important component of the management of PG. Key principles include wound dressing, monitoring for infection, avoidance of trauma, and judicious use of surgery. (See 'Wound management' below.)

TREATMENT SELECTION

Limited disease — Limited PG may be considered PG presenting as a single small, superficial (eg, <3 cm) ulcer (picture 1A); a few small, superficial ulcers; or a solitary plaque of vegetative PG. (See "Pyoderma gangrenosum: Pathogenesis, clinical features, and diagnosis", section on 'Subtypes'.)

Initial therapy — Topical corticosteroid therapy is our preferred initial treatment for limited PG (algorithm 1). Wound care and pain management are additional important components of treatment. (See 'Topical corticosteroids' below and 'Wound management' below and 'Pain management' below.)

Topical corticosteroids

Administration and precautions We typically utilize a high-potency topical corticosteroid (group 1 or 2 (table 1)), such as clobetasol 0.05% ointment. We instruct patients to apply the topical corticosteroid once or twice daily to the inflamed ulcer periphery. Initial signs of response to topical corticosteroid therapy (eg, stabilization of disease progression and reduced pain) are expected within two to four weeks. If there is a response, treatment is continued until complete healing.

Intralesional injection of corticosteroids (eg, triamcinolone acetonide) has also been used for PG. However, some clinicians, including ourselves, avoid intralesional injections due to concern for the induction of pathergy. Concentrations of triamcinolone between 6 and 40 mg/mL have been utilized [4]. The corticosteroid is injected circumferentially into the ulcer periphery. (See "Intralesional corticosteroid injection" and "Pyoderma gangrenosum: Pathogenesis, clinical features, and diagnosis", section on 'Common features'.)

Cutaneous atrophy, hypopigmentation, and suppression of the hypothalamic-pituitary axis are examples of adverse effects of topical and intralesional corticosteroid therapy. Adverse effects are reviewed in greater detail separately. (See "Topical corticosteroids: Use and adverse effects", section on 'Adverse effects' and "Intralesional corticosteroid injection", section on 'Adverse effects and pitfalls'.)

Efficacy Although topical corticosteroids are frequently prescribed as primary and adjunctive therapies in PG, data on the efficacy of local corticosteroid therapy are limited [5-8]. Examples of studies assessing efficacy include:

A multicenter prospective cohort study that evaluated outcomes of topical therapy in 66 patients with PG supports benefit of topical corticosteroid treatment [7]. Most patients (74 percent) received clobetasol propionate 0.05%; the remaining patients received topical tacrolimus 0.03% or 0.1% ointment. At six months, 28 of the 64 patients (44 percent) for whom information on healing was available had healed on topical therapy alone, including 20 of 47 (43 percent) treated with clobetasol and 5 of 10 (50 percent) treated with topical tacrolimus. The median time to healing was 145 days. Initial ulcer size was a predictor of time to healing.

In a prospective cohort study that included 49 patients with PG treated with clobetasol propionate 0.05%, 20 (43 percent) achieved healing within six months [7]. The median time to healing was 136 days.

Benefit of intralesional corticosteroid therapy in PG is documented in case reports [6,9-12].

Insufficient response to initial therapy — When topical corticosteroid therapy does not improve limited PG within two to four weeks, we often switch to tacrolimus 0.1% ointment or start oral dapsone or oral minocycline (algorithm 1). Although efficacy data are limited, the relatively favorable adverse effect profiles of these therapies prompt us to try them prior to proceeding to immunosuppressive therapies. (See 'More extensive disease and treatment-resistant limited disease' below.)

Topical tacrolimus

Administration and precautions Topical tacrolimus, a topical calcineurin inhibitor, is commercially available as a 0.03% and 0.1% ointment. We typically prescribe tacrolimus 0.1% ointment and instruct patients to apply the ointment to the inflamed ulcer periphery once to twice daily. Signs of improvement may be evident within the first few days to weeks of treatment. However, several weeks to a few months of treatment may be necessary for complete ulcer healing [13-15].

Topical tacrolimus is usually well tolerated; occasionally, patients experience mild burning sensations at the site of application [14].

Efficacy Topical tacrolimus in concentrations of 0.03% to 0.3% has demonstrated efficacy for PG in multiple case reports, a few prospective case series, and uncontrolled studies [5,7,13,16]. In one uncontrolled study, a compounded formulation of 0.3% tacrolimus in a carmellose sodium paste appeared to be effective for peristomal PG [8]. Seven of 11 patients treated with the tacrolimus formulation and 5 of 13 patients treated with clobetasol 0.05% lotion or ointment achieved complete healing. In a prospective cohort study that included 10 patients with PG treated with topical tacrolimus, five (50 percent) achieved healing with six months, with a median time to healing of 161 days [7].

Other calcineurin inhibitors may have benefit for PG, but fewer data are available on these agents. A case report documented improvement with pimecrolimus 1% cream [17]. Improvement with topical or intralesional cyclosporine has been described in a few patients [18-21].

Oral dapsone

Administration and precautions – Our typical target dose for dapsone ranges from 100 to 150 mg per day. We typically start treatment for adults at 50 mg per day and subsequently increase the dose to 100 mg per day after 7 to 10 days (if laboratory tests detect no significant abnormalities).

Signs of improvement usually occur within two to four weeks of an optimal dose. If there is a poor response at 100 mg per day and the patient is tolerating dapsone, we consider increasing the dose to 150 mg per day. If there is no evidence of response within four weeks at this dose, we stop dapsone. Hematologic adverse events can be a limiting factor for increasing the dose of dapsone.

Examples of adverse effects of dapsone include hemolysis, agranulocytosis, methemoglobinemia, dapsone hypersensitivity syndrome, and peripheral neuropathy (table 2). Patients require laboratory monitoring for hematologic and liver toxicity.

Patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency are at increased risk for hematologic toxicity. Screening for G6PD deficiency should precede dapsone therapy. (See "Diagnosis and management of glucose-6-phosphate dehydrogenase (G6PD) deficiency".)

Efficacy – In case reports and small series, dapsone has been associated with improvements in PG when administered as monotherapy or as a glucocorticoid-sparing agent [9,22-29]. In one retrospective study of 27 patients who received dapsone for at least four weeks as a component of PG therapy, a complete or partial response was documented for 16 and 81 percent of patients, respectively, with an average time to initial response of approximately five weeks [29].

Oral minocycline

Administration and precautions – Typical dosing for oral minocycline for adults is 100 mg twice daily. Some clinicians utilize doxycycline as an alternative to minocycline because of a more favorable adverse effect profile; however, most published reports of benefit involve use of minocycline [22]. If there is no evidence of response within six to eight weeks, we stop minocycline.

Examples of potential adverse effects of minocycline include dizziness, skin discoloration, photosensitivity, and a lupus-like syndrome. Similar to other tetracyclines, minocycline should not be administered to pregnant individuals or children under the age of nine years due to adverse effects on tooth development.

Efficacy – Efficacy data for minocycline are limited. Case reports describe improvement in PG when used as monotherapy or as a glucocorticoid-sparing agent [22,30-39].

Refractory limited disease — When limited PG does not respond adequately to topical corticosteroids, topical calcineurin inhibitors, oral dapsone, or oral minocycline, we proceed to interventions utilized for more extensive disease (algorithm 1). (See 'More extensive disease and treatment-resistant limited disease' below.)

More extensive disease and treatment-resistant limited disease — We take a different approach to the treatment of PG that exceeds our description of limited disease (ie, more than a few small [eg, <3 cm] superficial ulcers or a solitary plaque of vegetative PG). This extent of PG generally requires systemic therapy to calm the inflammatory process to allow for wound healing. Systemic therapy is also appropriate for limited PG that responds insufficiently to other treatments. (See 'Limited disease' above.)

Initial treatment — Our preferred initial treatment for extensive PG is a systemic glucocorticoid or oral cyclosporine because of a rapid onset of anti-inflammatory effects (algorithm 1). We select the most tolerable drug based on the patient-specific factors, such as comorbidities. Most often, we begin with a systemic glucocorticoid, reserving cyclosporine for patients who may not tolerate systemic glucocorticoid therapy.

Because long-term treatment with these therapies is often associated with serious adverse effects, we often incorporate other systemic immunosuppressive therapies within the first few weeks of treatment. (See 'Subsequent management' below and "Major adverse effects of systemic glucocorticoids" and "Pharmacology of cyclosporine and tacrolimus", section on 'Side effects'.)

Wound care and pain management are additional important components of treatment. (See 'Wound management' below.)

Topical corticosteroids and topical tacrolimus are often used as adjuncts to systemic treatment. (See 'Topical corticosteroids' above and 'Topical tacrolimus' above.)

Systemic glucocorticoids

Administration and precautions – Our typical initial regimen for adults is 0.5 to 1.5 mg/kg per day of oral prednisone or its equivalent, with a maximum dose of 60 mg of prednisone per day. In very aggressive or painful disease, intravenous pulse corticosteroids (methylprednisolone 1 g per day for one to five days) can be used as initial treatment [40-42]. Patients are subsequently transitioned to an oral corticosteroid.

The response to systemic glucocorticoids is usually rapid; stabilization of the disease is often evident within the first week [43]. Pain also tends to improve soon after the initiation of treatment.

We aim to taper the glucocorticoid as soon as feasible because of the potential for serious adverse effects with long-term systemic glucocorticoid treatment. For most patients, this prompts us to add an additional systemic agent within the first few weeks of treatment. (See 'Subsequent management' below.)

Systemic glucocorticoid therapy is associated with a wide variety of potential adverse effects (table 3). Systemic glucocorticoid therapy should include consideration of measures to reduce glucocorticoid-associated bone loss. (See "Major adverse effects of systemic glucocorticoids" and "Prevention and treatment of glucocorticoid-induced osteoporosis".)

Efficacy Although multiple case reports, case series, and our clinical experience support the use of systemic glucocorticoids for PG [6,22], the findings of a single-blind randomized trial (n = 112) that compared prednisolone monotherapy (initial dose 0.75 mg/kg per day, up to 75 mg per day) with cyclosporine 4 mg/kg per day (up to 400 mg per day) showed that systemic glucocorticoid therapy alone may be insufficient for many patients [44]. After six months, there was only a 47 percent rate of complete ulcer healing in both groups. The pace of improvement during the first six weeks of treatment was similar between the groups, suggesting a comparable time to onset of action. Rates of adverse reactions were also similar in both groups, but serious reactions, notably serious infections, were more frequent in the prednisolone group.

Systemic cyclosporine

Administration and precautions Our typical initial regimen for adults is 4 to 5 mg/kg of cyclosporine per day [45]. As with oral glucocorticoids, the response is usually rapid. Pain often improves within a few days, and other signs of improvement are expected within the first four weeks.

We aim to taper cyclosporine as soon as feasible because of concern for adverse effects. For most patients, this prompts us to add an additional systemic agent within the first few weeks of treatment. (See 'Subsequent management' below.)

Examples of adverse effects of cyclosporine include renal toxicity, hypertension, and increased risk for infections and malignancy. Adverse effects are reviewed in greater detail separately. Periodic monitoring of renal function and blood pressure are indicated. Contraindications for cyclosporine include concurrent malignancy, uncontrolled hypertension, uncontrolled infections. (See "Pharmacology of cyclosporine and tacrolimus", section on 'Side effects'.)

Efficacy A single-blind randomized trial (n = 112) that compared cyclosporine with prednisolone found complete healing after six months in approximately one-half of patients treated with cyclosporine and a similar time to onset of action between cyclosporine and prednisolone [44]. (See 'Systemic glucocorticoids' above.)

In addition, several retrospective analyses and case reports suggest a beneficial effect of cyclosporine [9,22-24,44,46,47].

Subsequent management

Role and implementation — In our experience, most patients with extensive PG require many months of systemic treatment to achieve healing. Systemic therapies other than glucocorticoids and cyclosporine (eg, infliximab, adalimumab, mycophenolate mofetil) may play an important role in achieving healing of PG and reducing risk for adverse effects related to long-term systemic glucocorticoid or cyclosporine therapy.

Timing Many of the immunosuppressive therapies preferred for long-term treatment have a slower onset of action than systemic glucocorticoids and cyclosporine. Therefore, we often add these therapies early, usually within the first few weeks of systemic glucocorticoid or cyclosporine treatment. This supports tapering of glucocorticoids and cyclosporine within a reasonable period.

An occasional exception is the patient who experiences very rapid ulcer healing during the first few weeks of systemic glucocorticoid or cyclosporine therapy. If a need for prolonged systemic treatment (eg, more than two to three months for oral glucocorticoids or more than three to six months for cyclosporine) appears unlikely, we often defer the addition of another systemic treatment.

Treatment selection Infliximab, adalimumab, or mycophenolate mofetil are our preferred long-term therapies for PG because of data that suggest efficacy for PG. However, other factors can influence the selection of treatment, such as comorbidities, patient preference, or treatment availability. For example, infliximab or adalimumab may be preferred over mycophenolate mofetil for a patient with Crohn disease or ulcerative colitis because of the benefit of biologic tumor necrosis factor (TNF) inhibitors for inflammatory bowel disease and PG, and contraindications to immunosuppression may lead to the selection of intravenous immune globulin (IVIG) over other therapies. In other scenarios, a biologic interleukin (IL) 17 or IL-23 inhibitor may be preferred because of efficacy for a comorbidity. (See 'Infliximab' below and 'Mycophenolate mofetil' below and 'Other interventions' below.)

Tapering and discontinuation of systemic glucocorticoids and cyclosporine For patients receiving a systemic glucocorticoid (or cyclosporine) plus another systemic agent, we typically begin to taper the glucocorticoid or cyclosporine once disease progression has stopped and clear signs of improvement are evident (reductions in pain, visible inflammation, and wound size). For patients receiving a systemic glucocorticoid or cyclosporine alone because of a high likelihood of achieving complete healing relatively quickly, we begin to taper the drug after achievement of complete healing.

We aim to taper and discontinue glucocorticoids within 4 to 10 weeks and monitor patients closely for continued improvement. Tapering glucocorticoids too rapidly may precipitate disease flares [43].

We typically taper cyclosporine over a period that is twice as long as the duration of cyclosporine therapy (eg, taper over five to six months for patients with a three-month duration of cyclosporine therapy).

Infliximab

Administration and precautions – The treatment regimens for infliximab and the time to response to infliximab vary considerably across case reports and studies [48]. The optimal treatment regimen has not been determined. We typically administer a 5 mg/kg intravenous infusion of infliximab at weeks 0, 2, and 6, followed by infusions every six to eight weeks [45]. In our experience, initial signs of response are usually evident within 8 to 12 weeks.

Potential adverse effects of infliximab include infusion reactions, infections, demyelinating disease, and heart failure. The side effects of TNF-alpha inhibitors are discussed in greater detail separately. (See "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects".)

Efficacy The use of infliximab, a chimeric antibody against TNF-alpha, in PG is supported by a placebo-controlled randomized trial of 30 adults with PG, many of whom were concomitantly treated with topical or systemic therapy (most often oral prednisolone) [49]. At the start of the trial, patients were given either a single 5 mg/kg infusion of infliximab (n = 13) or placebo (n = 17). After two weeks, more patients in the infliximab group had clinical signs of improvement (46 versus 6 percent).

Patients in both groups who did not exhibit improvement at week 2 were then given open-label infliximab (a 5 mg/kg single dose). At week 6, 20 of the 29 patients who received infliximab (69 percent) had improved, and 6 patients achieved complete remission (21 percent). There was no significant difference in the response to treatment among patients with (n = 18) and without (n = 11) inflammatory bowel disease. Long-term follow-up was not performed.

In addition to the randomized trial, other studies suggest efficacy of infliximab [50,51]. As an example, a multicenter retrospective study of 13 patients with refractory PG and inflammatory bowel disease treated with 5 mg/kg of infliximab (1 to 24 infusions over zero to four years and in conjunction with other systemic therapies) found that all 13 patients achieved complete healing. The mean times to initial response and complete healing were 11 days (range 2 to 30 days) and 86 days (range 7 to 210 days), respectively [51]. Ten patients required periodic infusions every 4 to 12 weeks to maintain the clinical response. All patients who were taking systemic glucocorticoids were able to discontinue glucocorticoid therapy.

Adalimumab

Administration and precautions – As with infliximab, adalimumab treatment regimens vary [52-55]. Our typical approach begins with a 40 mg dose of adalimumab every other week. Signs of improvement usually become evident within 8 to 12 weeks. In our experience, a subsequent increase to once-weekly dosing is often required to achieve complete healing.

Potential adverse effects include injection site reactions, infections, demyelinating disease, and heart failure. The side effects of TNF-alpha inhibitors are discussed in greater detail separately. (See "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects".)

Efficacy – An open-label study of 22 adults with PG treated with adalimumab (at doses of 160 mg at week 0, followed by 80 mg at week 2, and then 40 mg every other week starting at week 4) suggests benefit of adalimumab [56]. Sixteen patients (73 percent) also received oral corticosteroids (≤10 mg per day) for PG or other indications. At week 26, 12 patients achieved complete healing of the target ulcer (55 percent, 95% CI 32-76 percent). The mean time to healing of the target ulcer was 115 days. Adalimumab has also been associated with ulcer healing in reports of individual patients [52-55].

Mycophenolate mofetil

Administration and precautions Typical dosing of mycophenolate mofetil for PG in adults is 2 to 3 g per day. Initial signs of response are expected within 8 to 12 weeks.

Examples of adverse effects of mycophenolate mofetil include gastrointestinal symptoms and bone marrow suppression. Adverse effects are reviewed in greater detail separately. (See "Mycophenolate: Overview of use and adverse effects in the treatment of rheumatic diseases", section on 'Adverse effects'.)

Efficacy – Multiple cases of improvement in PG with mycophenolate mofetil have been reported [9,24,57-62]. Mycophenolate mofetil appeared beneficial in a retrospective study of 26 patients with PG treated with mycophenolate mofetil and prednisolone (with or without other immunomodulatory agents) for at least one month [62]. Improvement in disease severity during treatment was noted in 22 patients (85 percent). Moreover, in a retrospective study of seven patients treated with mycophenolate mofetil for at least two months, four of seven had complete healing, including two patients for whom mycophenolate mofetil was the only systemic therapy given for PG [61].

Refractory disease — When our typical approach to treatment is insufficient, we proceed to other therapies. Because efficacy data are limited, selection of a next-line treatment is primarily based upon consideration of patient comorbidities, patient preference, and treatment availability. (See 'Other interventions' below.)

Cessation of therapy — Once complete healing of lesions has occurred, treatment cessation can be attempted. Systemic therapy should be gradually tapered and stopped over the course of several months rather than abruptly discontinued.

Prolonged treatment is necessary for some patients. In a series of 42 patients with PG who were followed for a median of 26.5 months, 56 percent of the 34 patients who remained alive during the follow-up period required continuing pharmacologic therapy, and 44 percent were able to maintain complete remission without therapy [23].

WOUND MANAGEMENT — The goal of wound care in PG is to create an optimal environment for wound healing.

Local care — Wounds should be cleansed gently with tepid sterile saline or a mild antiseptic prior to dressing changes [2]. Wound dressings that promote a moist wound environment and do not adhere to the wound base are preferred, as they may be beneficial for healing [63]. (See "Basic principles of wound management", section on 'Importance of moisture'.)

The selection of a specific type of wound dressing depends on the nature of the wound and patient and clinician preferences. As an example, patients with wounds that are highly exudative may benefit from the use of more absorptive wound dressings, such as alginates to avoid tissue maceration [64]. (See "Basic principles of wound management", section on 'Wound dressings'.)

In addition to the area of ulceration or erosion, attention should also be paid to the surrounding skin. Use of a barrier cream or ointment, such as zinc oxide paste or petrolatum, may help to prevent skin breakdown at the wound edge [63,64].

Patients should be monitored for clinical signs of wound infection (eg, fever, warmth, swelling, malodor, lymphangitic streaks, increased drainage, or pain) and should be treated appropriately with antibiotics if infection occurs [64].

Avoidance of trauma — Pathergy (exacerbation or new development of lesions at sites of trauma) can occur in PG. Thus, unnecessary traumatic insults to the wound, such as the use of wet to dry dressings and the application of caustic substances (eg, silver nitrate), should be avoided [2,64]. (See "Pyoderma gangrenosum: Pathogenesis, clinical features, and diagnosis".)

If surgery for another indication is necessary in a patient with active PG, we encourage the continuation of PG treatment to the greatest degree feasible and halt tapering of PG therapies in the perioperative period and until the surgical wound has healed. Ideally, elective surgical procedures should be deferred until complete healing of PG has been achieved.

Role of surgery and other interventions — Due to the potential for pathergy, the role of surgery for wound management in PG is controversial [65]. Surgical procedures are considered only in select cases, such as those in which accumulation of necrotic tissue presents a risk for infection or where vital tissues such as tendons or ligaments are exposed in the ulcer bed [6,45].

Although multiple cases in which PG worsened following surgical intervention have been reported [1,66], documentation of cases in which gentle debridement, skin flaps or grafts [67-70], split-thickness grafts with negative pressure wound therapy [71], and the application of bioengineered keratinocyte autografts [58] or allogenic cultured dermal substitutes [72] appeared to be beneficial in PG exists. In a review of the literature that identified cases of surgical intervention in patients with active PG, postoperative disease progression was identified in only 17 percent of patients [65].

If surgery for a PG ulcer will be performed, we advise delaying surgery until a period of good disease control (ie, resolution of erythema or violaceous color at the wound border and cessation of progression of disease). Patients should be concomitantly treated with systemic PG therapy [73].

Although stoma closure can lead to resolution of peristomal PG, recurrence of PG may be common after relocation or revision of a stoma. In a retrospective chart review of 44 patients with peristomal PG, 10 of 15 patients (67 percent) who underwent relocation or revision of a stoma developed recurrent PG [74].

Hyperbaric oxygen has been reported to be of benefit for wound healing in a few patients with PG, but data are insufficient to recommend the routine use of this therapy [75-77].

PAIN MANAGEMENT — Ulcers of PG are often associated with significant pain that appears out of proportion to the clinical appearance of lesions. Although pain may improve during treatment, some patients require the use of narcotic agents to manage discomfort. Consultation with a pain management specialist may be beneficial [63]. (See "Approach to the management of chronic non-cancer pain in adults".)

OTHER INTERVENTIONS — Various other interventions have been utilized for PG, but limited efficacy data, unfavorable adverse effect profiles, cost, or availability generally restrict their use to refractory disease and scenarios in which a particular treatment may provide joint benefit for PG and a comorbidity. Examples of these interventions include other biologic agents (adalimumab, interleukin [IL] 17 inhibitors, IL-23 inhibitors), other conventional immunosuppressants (methotrexate, azathioprine), intravenous immune globulin (IVIG), alkylating agents, and other treatments.

Other biologic TNF inhibitors – In our experience, infliximab and adalimumab are more efficacious than other tumor necrosis factor (TNF) inhibitors, although benefit of other TNF inhibitors has been reported. Improvement in PG with etanercept (at a dose of 25 to 50 mg twice weekly) has also been reported in a small retrospective series and case report [78,79]. There are case reports describing benefit of other biologic TNF-alpha inhibitors, such as certolizumab pegol [80,81] and golimumab [82].

Biologic IL-17 and IL-23 inhibitors – Case reports, most describing biologic therapy as an adjunctive treatment for PG in patients with concomitant inflammatory bowel disease, suggest benefit of ustekinumab, an anti-interleukin (IL) 12/23 inhibitor [83-85], as well as the anti-IL-23 biologic agents tildrakizumab [86], guselkumab [87], and risankizumab [88]. The use of anti-IL-17 biologics for PG is uncertain, with both cases of induction and clearance of PG in secukinumab [89,90], ixekizumab [91,92], and brodalumab [93,94].

Conventional immunosuppressantsMethotrexate [95] and azathioprine [23,96] have appeared beneficial as adjunctive or glucocorticoid sparing in case reports and small case series [97]. Similar to mycophenolate mofetil, these drugs have a delayed onset of action (usually 8 to 12 weeks).

Typical adult dosing for methotrexate is 10 to 30 mg per week; azathioprine is usually prescribed at doses ranging from 100 to 300 mg per day [45]. Thiopurine methyltransferase (TPMT) enzyme activity guides appropriate dosing for azathioprine. Both drugs have the potential for serious adverse effects. (See "Major side effects of low-dose methotrexate" and "Pharmacology and side effects of azathioprine when used in rheumatic diseases", section on 'Adverse effects' and "Thiopurines: Pretreatment testing and approach to therapeutic drug monitoring for adults with inflammatory bowel disease", section on 'Assessing TPMT enzyme activity'.)

IVIG – Intravenous immune globulin (IVIG) is typically given as a total dose of 2 g/kg every four weeks. The total dose is divided and administered over the course of two to five days.

Efficacy data for IVIG for PG are limited. A systematic review of cases and case series documenting treatment of refractory PG with IVIG (often in conjunction with systemic glucocorticoids) found complete responses in 26 of 49 patients (53 percent) and complete or partial responses in 43 of 49 patients (88 percent) [98]. The mean time to initial response was 3.5 weeks, and patients were treated for an average of 5.9 months. Limitations of the review included potential publication bias given that most data were derived from case reports and a paucity of long-term, follow-up data in the included patients. Moreover, failures to respond to retreatment with IVIG upon disease relapse have occurred. Well-designed prospective trials are necessary to clarify the efficacy of IVIG for PG. (See "Overview of intravenous immune globulin (IVIG) therapy".)

Cyclophosphamide and chlorambucil – Although a few case reports and uncontrolled studies document improvement in refractory PG with pulsed intravenous cyclophosphamide [99-101] or chlorambucil [102-104], concern for myelosuppression and other severe adverse effects limits the use of these alkylating agents. In an uncontrolled study of nine patients with PG treated with monthly infusions of cyclophosphamide (at a dose of 500 mg/m2 for up to six treatments), seven patients achieved complete remission, one achieved partial remission, and one failed to respond to therapy [99]. Four patients achieved complete healing prior to the third treatment. Two patients relapsed within three months after the cessation of therapy.

Chlorambucil appeared to be effective for recalcitrant PG in an uncontrolled study of six patients who were treated with chlorambucil (at a dose of 2 to 4 mg per day) with or without prednisone therapy [102]. Improvement was noted in six to eight weeks, and healing of ulcers occurred within 2 to 10 months in all patients. All patients who were also receiving prednisone for PG were able to discontinue prednisone therapy. Prolonged remissions (four to nine years) were sustained in four patients who discontinued chlorambucil after 6 to 24 months of therapy.

Other agents – Multiple other interventions have been reported to be of benefit in individual patients. Examples of topical therapies include sodium cromoglycate [9,105,106], nicotine [107-109], benzoyl peroxide [110,111], 5-aminosalicylic acid [112], nitrogen mustard [113], and platelet-derived growth factor [114,115].

Other biologic agents with reports of apparent benefit include the IL-1 inhibitor anakinra [116], the IL-1-beta inhibitor canakinumab [117], the IL-36 receptor antagonist spesolimab [118], and the IL-6 inhibitor tocilizumab [119]. Janus kinase (JAK) inhibitors with reports of apparent benefit include baricitinib [120,121] and tofacitinib [122-125].

Additional systemic agents that may be beneficial include clofazimine [23,126], colchicine [127,128], interferon alfa [129], melphalan [130], mercaptopurine [131], metronidazole [132], potassium iodide [133], sulfasalazine [24], tacrolimus [134], and thalidomide [22,135,136]. Apheresis of leukocytes [137-141] and plasmapheresis [101] have also been used for treatment.

PROGNOSIS — Follow-up studies of patients with PG suggest that with treatment, more than 50 percent of patients achieve complete wound healing within one year:

In an Australian series of 26 patients hospitalized for PG, 6 of 12 patients available for follow-up six months after hospital discharge had complete ulcer healing [142].

In a series with 64 patients with classic PG and 22 patients with bullous (atypical) PG, the mean time to complete remission (wound healing and absence of active disease) was 11.5±11.1 months in patients with classic PG and 9±13.7 months in patients with bullous PG [1]. Overall, 68 percent of patients achieved complete remission within six months, and 95 percent were in remission within three years.

The effect of treatment of associated underlying medical disorders on disease activity in patients with PG is variable, as the course of PG does not always parallel that of the related underlying disease [9]. Improvement in PG has been reported in some patients following proctocolectomy for inflammatory bowel disease [143]. However, PG may also occur peristomally or in other locations following proctocolectomy [9,144-148].

PG typically heals with scars, which can be disfiguring. New lesions may develop during or after healing of other lesions. Relapses can appear after long periods of disease quiescence [149].

PG may be associated with increased risk for death. In a retrospective cohort study that included 3386 patients with PG and over 67,000 controls, all-cause mortality was higher in patients with PG (adjusted hazard ratio 2.122, 95% CI 1.971-2.285) [150]. Increased mortality rates were also observed for diseases within a wide variety of organ systems.

Various factors may contribute to increased mortality in patients with PG, and death may occur due to underlying associated disorders, particularly in patients with malignancy, or due to complications directly related to ulcers or immunosuppressive therapy (eg, sepsis) [23,151]. Other potential contributory factors, such as effects related to an underlying chronic inflammatory state, have also been proposed [150].

Due to the association between bullous PG and hematologic disease, patients with bullous PG who present without an identifiable underlying hematologic disorder should be followed closely [45].

PREVENTION — There are no specific measures that have been shown to prevent the initial development of PG. In patients with an existing diagnosis, avoidance of trauma may help to reduce the development of new lesions. If patients with PG require surgery for other indications, wound closure with subcuticular stitches and close postoperative follow-up with a dermatologist are suggested.

Some authors have suggested that administering systemic glucocorticoids during and for two weeks or more after surgery may help to prevent the development of new lesions in patients with aggressive PG [2]. However, the impact of this intervention has not been formally studied.

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: Pyoderma gangrenosum".)

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 topics (see "Patient education: Pyoderma gangrenosum (The Basics)")

SUMMARY AND RECOMMENDATIONS

Disease overview – Pyoderma gangrenosum (PG) is an uncommon inflammatory and ulcerative disorder that may occur independently or in association with a variety of systemic diseases (picture 1A-C). (See 'Introduction' above.)

Treatment principles – The approach to the treatment of PG involves consideration of various factors, such as disease severity and comorbidities. Topical and systemic therapies that suppress the inflammatory response are the mainstays of treatment. Wound care and pain management are additional important components of treatment. The role of surgery in PG is controversial. (See 'Treatment principles' above and 'Wound management' above and 'Pain management' above and 'Role of surgery and other interventions' above.)

Response assessment – The response to PG therapy is assessed clinically. Typical early signs of response are the stabilization of disease progression, reduced visible signs of inflammation (eg, violaceous discoloration of the wound border), and reduced pain. Subsequently, the wound base begins to fill with granulation tissue, and the wound diameter progressively shrinks. Documentation of the size, number, and location of ulcers and serial clinical photographs are useful for following the response to therapy. (See 'Treatment principles' above.)

Approach to treatment – Efficacy data on treatments for PG are limited. The severity of PG influences the initial approach to treatment (algorithm 1).

Limited disease

-Initial treatment – For patients with limited PG (a single or few small [eg, <3 cm], superficial ulcers or a solitary plaque of vegetative PG), we suggest initial treatment with a super high-potency or high-potency topical corticosteroid (group 1 or 2 (table 1)) (Grade 2C). (See 'Limited disease' above.)

-Subsequent management of limited disease – For limited disease that responds well to topical corticosteroid therapy, treatment is continued until complete healing. For limited disease that does not respond sufficiently to topical corticosteroids, we suggest topical tacrolimus 0.1% ointment, oral minocycline, or oral dapsone for next-line treatment rather than systemic immunosuppressive therapies (Grade 2C). If the response remains insufficient, we proceed to therapies for extensive disease. (See 'Insufficient response to initial therapy' above and 'Refractory limited disease' above.)

Extensive disease

-Initial treatment – For patients with more extensive PG, we suggest initial treatment with oral prednisone (Grade 2C). Oral cyclosporine is a reasonable alternative for patients who are less likely to tolerate systemic glucocorticoid therapy. (See 'More extensive disease and treatment-resistant limited disease' above.)

-Subsequent management of patients receiving a systemic glucocorticoid or cyclosporine – Treatment with prednisone or cyclosporine alone is not sufficient for many patients, and long-term treatment with prednisone or cyclosporine is associated with risk for serious adverse effects. For most patients, we suggest the addition of infliximab, adalimumab, or mycophenolate mofetil during the first few weeks of prednisone or cyclosporine treatment (Grade 2C). Tapering of prednisone or cyclosporine may begin once disease progression has stopped and clear signs of improvement are evident. If the response is inadequate, we proceed to other treatments. (See 'Subsequent management' above and 'Other interventions' above.)

Some patients may benefit from a different approach (eg, addition of a different systemic agent that may also improve a particular PG-associated comorbidity or avoidance of immunosuppression due to comorbidities). In addition, PG that responds very rapidly to prednisone or cyclosporine and appears likely to heal completely within several weeks may not require the addition of another systemic agent. (See 'Other interventions' above and 'Subsequent management' above.)

Prognosis – It is estimated that with treatment, more than one-half of patients with PG achieve wound healing within one year, and almost all patients achieve remission with longer follow-up. However, relapses can occur after long periods of disease remission. (See 'Prognosis' above.)

  1. Bennett ML, Jackson JM, Jorizzo JL, et al. Pyoderma gangrenosum. A comparison of typical and atypical forms with an emphasis on time to remission. Case review of 86 patients from 2 institutions. Medicine (Baltimore) 2000; 79:37.
  2. Powell FC, Hackett BC, Wallach D. Pyoderma gangrenosum. In: Fitzpatrick's Dermatology in General Medicine, 8th ed, Goldsmith LA, Katz SI, Gilchrest BA, et al (Eds), McGraw-Hill Companies, Inc., 2012. Vol 1, p.371.
  3. Maronese CA, Pimentel MA, Li MM, et al. Pyoderma Gangrenosum: An Updated Literature Review on Established and Emerging Pharmacological Treatments. Am J Clin Dermatol 2022; 23:615.
  4. Prajapati V, Man J, Brassard A. Pyoderma gangrenosum: common pitfalls in management and a stepwise, evidence-based, therapeutic approach. J Cutan Med Surg 2009; 13 Suppl 1:S2.
  5. Le Cleach L, Moguelet P, Perrin P, Chosidow O. Is topical monotherapy effective for localized pyoderma gangrenosum? Arch Dermatol 2011; 147:101.
  6. Reichrath J, Bens G, Bonowitz A, Tilgen W. Treatment recommendations for pyoderma gangrenosum: an evidence-based review of the literature based on more than 350 patients. J Am Acad Dermatol 2005; 53:273.
  7. Thomas KS, Ormerod AD, Craig FE, et al. Clinical outcomes and response of patients applying topical therapy for pyoderma gangrenosum: A prospective cohort study. J Am Acad Dermatol 2016; 75:940.
  8. Lyon CC, Stapleton M, Smith AJ, et al. Topical tacrolimus in the management of peristomal pyoderma gangrenosum. J Dermatolog Treat 2001; 12:13.
  9. Hughes AP, Jackson JM, Callen JP. Clinical features and treatment of peristomal pyoderma gangrenosum. JAMA 2000; 284:1546.
  10. Dohan FC. Schizophrenia and neuroactive peptides from food. Lancet 1979; 1:1031.
  11. Jennings JL. Pyoderma gangrenosum: successful treatment with intralesional steroids. J Am Acad Dermatol 1983; 9:575.
  12. Moschella SL. Pyoderma gangrenosum. A patient successfully treated with intralesional injections of steroid. Arch Dermatol 1967; 95:121.
  13. Altieri M, Vaziri K, Orkin BA. Topical tacrolimus for parastomal pyoderma gangrenosum: a report of two cases. Ostomy Wound Manage 2010; 56:56.
  14. Marzano AV, Trevisan V, Lazzari R, Crosti C. Topical tacrolimus for the treatment of localized, idiopathic, newly diagnosed pyoderma gangrenosum. J Dermatolog Treat 2010; 21:140.
  15. Kimble RM, Tickler AK, Nicholls VS, Cleghorn G. Successful topical tacrolimus (FK506) therapy in a child with pyoderma gangrenosum. J Pediatr Gastroenterol Nutr 2002; 34:555.
  16. Larsen CG, Thyssen JP. Pustular penile pyoderma gangrenosum successfully treated with topical tacrolimus ointment. Acta Derm Venereol 2012; 92:104.
  17. Bellini V, Simonetti S, Lisi P. Successful treatment of severe pyoderma gangrenosum with pimecrolimus cream 1%. J Eur Acad Dermatol Venereol 2008; 22:113.
  18. Mrowietz U, Christophers E. Clearing of pyoderma gangrenosum by intralesional cyclosporin A. Br J Dermatol 1991; 125:499.
  19. Wenzel J, Gerdsen R, Phillipp-Dormston W, et al. Topical treatment of pyoderma gangraenosum. Dermatology 2002; 205:221.
  20. Theissen U, Luger TA, Schwarz T. [Successful topical administration of cyclosporin A in pyoderma gangraenosum]. Hautarzt 1996; 47:132.
  21. Azizan NZ, Gangaram HB, Hussein SH. A novel therapy for the treatment of pyoderma gangrenosum. Med J Malaysia 2008; 63:51.
  22. Binus AM, Qureshi AA, Li VW, Winterfield LS. Pyoderma gangrenosum: a retrospective review of patient characteristics, comorbidities and therapy in 103 patients. Br J Dermatol 2011; 165:1244.
  23. von den Driesch P. Pyoderma gangrenosum: a report of 44 cases with follow-up. Br J Dermatol 1997; 137:1000.
  24. Vidal D, Puig L, Gilaberte M, Alomar A. Review of 26 cases of classical pyoderma gangrenosum: clinical and therapeutic features. J Dermatolog Treat 2004; 15:146.
  25. Fukuhara K, Urano Y, Kimura S, et al. Pyoderma gangrenosum with rheumatoid arthritis and pulmonary aseptic abscess responding to treatment with dapsone. Br J Dermatol 1998; 139:556.
  26. Teasley LA, Foster CS, Baltatzis S. Sclerokeratitis and facial skin lesions: a case report of pyoderma gangrenosum and its response to dapsone therapy. Cornea 2007; 26:215.
  27. Brown RE, Lay L, Graham D. Bilateral pyoderma gangrenosum of the hand: treatment with dapsone. J Hand Surg Br 1993; 18:119.
  28. Galun E, Flugelman MY, Rachmilewitz D. Pyoderma gangrenosum complicating ulcerative colitis: successful treatment with methylprednisolone pulse therapy and dapsone. Am J Gastroenterol 1986; 81:988.
  29. Din RS, Tsiaras WG, Li DG, Mostaghimi A. Efficacy of Systemic Dapsone Treatment for Pyoderma Gangrenosum: A Retrospective Review. J Drugs Dermatol 2018; 17:1058.
  30. Farrell AM, Black MM, Bracka A, Bunker CB. Pyoderma gangrenosum of the penis. Br J Dermatol 1998; 138:337.
  31. Shenefelt PD. Pyoderma gangrenosum associated with cystic acne and hidradenitis suppurativa controlled by adding minocycline and sulfasalazine to the treatment regimen. Cutis 1996; 57:315.
  32. Boulinguez S, Bernard P, Bedane C, et al. Pyoderma gangrenosum complicating Cogan's syndrome. Clin Exp Dermatol 1998; 23:286.
  33. Langan SM, Powell FC. Vegetative pyoderma gangrenosum: a report of two new cases and a review of the literature. Int J Dermatol 2005; 44:623.
  34. Vandevyvere K, Luyten FP, Verschueren P, et al. Pyoderma gangrenosum developing during therapy with TNF-alpha antagonists in a patient with rheumatoid arthritis. Clin Rheumatol 2007; 26:2205.
  35. Sachs D, Su L, Dlugosz A. Verrucous annular ulcerated hip plaques. Diagnosis: superficial granulomatous pyoderma form of pyoderma gangrenosum. Arch Dermatol 2000; 136:1263.
  36. Miralles ES, Nuñez M, Pérez B, Ledo A. Minocycline hydrochloride hyperpigmentation complicating treatment of pyoderma gangrenosum. J Dermatol 1994; 21:965.
  37. Reynolds NJ, Peachey RD. Response of atypical bullous pyoderma gangrenosum to oral minocycline hydrochloride and topical steroids. Acta Derm Venereol 1990; 70:538.
  38. Davies MG, Piper S. Pyoderma gangrenosum: successful treatment with minocycline. Clin Exp Dermatol 1981; 6:219.
  39. Lynch WS, Bergfeld WF. Pyoderma gangrenosum responsive to minocycline hydrochloride. Cutis 1978; 21:535.
  40. Prystowsky JH, Kahn SN, Lazarus GS. Present status of pyoderma gangrenosum. Review of 21 cases. Arch Dermatol 1989; 125:57.
  41. Yamauchi T, Ishida K, Iwashima Y, et al. Successful treatment of pyoderma gangrenosum that developed in a patient with myelodysplastic syndrome. J Infect Chemother 2003; 9:268.
  42. Aseni P, Di Sandro S, Mihaylov P, et al. Atypical presentation of pioderma gangrenosum complicating ulcerative colitis: rapid disappearance with methylprednisolone. World J Gastroenterol 2008; 14:5471.
  43. Chow RK, Ho VC. Treatment of pyoderma gangrenosum. J Am Acad Dermatol 1996; 34:1047.
  44. Ormerod AD, Thomas KS, Craig FE, et al. Comparison of the two most commonly used treatments for pyoderma gangrenosum: results of the STOP GAP randomised controlled trial. BMJ 2015; 350:h2958.
  45. Callen JP, Jackson JM. Pyoderma gangrenosum: an update. Rheum Dis Clin North Am 2007; 33:787.
  46. Hasselmann DO, Bens G, Tilgen W, Reichrath J. Pyoderma gangrenosum: clinical presentation and outcome in 18 cases and review of the literature. J Dtsch Dermatol Ges 2007; 5:560.
  47. Turner RB, Emer JJ, Weill M, et al. Rapid resolution of pyoderma gangrenosum after treatment with intravenous cyclosporine. J Am Acad Dermatol 2010; 63:e72.
  48. Adişen E, Oztaş M, Gürer MA. Treatment of idiopathic pyoderma gangrenosum with infliximab: induction dosing regimen or on-demand therapy? Dermatology 2008; 216:163.
  49. Brooklyn TN, Dunnill MG, Shetty A, et al. Infliximab for the treatment of pyoderma gangrenosum: a randomised, double blind, placebo controlled trial. Gut 2006; 55:505.
  50. Mooij JE, van Rappard DC, Mekkes JR. Six patients with pyoderma gangrenosum successfully treated with infliximab. Int J Dermatol 2013; 52:1418.
  51. Regueiro M, Valentine J, Plevy S, et al. Infliximab for treatment of pyoderma gangrenosum associated with inflammatory bowel disease. Am J Gastroenterol 2003; 98:1821.
  52. Hubbard VG, Friedmann AC, Goldsmith P. Systemic pyoderma gangrenosum responding to infliximab and adalimumab. Br J Dermatol 2005; 152:1059.
  53. Alkhouri N, Hupertz V, Mahajan L. Adalimumab treatment for peristomal pyoderma gangrenosum associated with Crohn's disease. Inflamm Bowel Dis 2009; 15:803.
  54. Reddick CL, Singh MN, Chalmers RJ. Successful treatment of superficial pyoderma gangrenosum associated with hidradenitis suppurativa with adalimumab. Dermatol Online J 2010; 16:15.
  55. Cariñanos I, Acosta MB, Domènech E. Adalimumab for pyoderma gangrenosum associated with inflammatory bowel disease. Inflamm Bowel Dis 2011; 17:E153.
  56. Yamasaki K, Yamanaka K, Zhao Y, et al. Adalimumab in Japanese patients with active ulcers of pyoderma gangrenosum: Twenty-six-week phase 3 open-label study. J Dermatol 2020; 47:1383.
  57. Nousari HC, Lynch W, Anhalt GJ, Petri M. The effectiveness of mycophenolate mofetil in refractory pyoderma gangrenosum. Arch Dermatol 1998; 134:1509.
  58. Wollina U, Karamfilov T. Treatment of recalcitrant ulcers in pyoderma gangrenosum with mycophenolate mofetil and autologous keratinocyte transplantation on a hyaluronic acid matrix. J Eur Acad Dermatol Venereol 2000; 14:187.
  59. Gilmour E, Stewart DG. Severe recalcitrant pyoderma gangrenosum responding to a combination of mycophenolate mofetil with cyclosporin and complicated by a mononeuritis. Br J Dermatol 2001; 144:397.
  60. Daniels NH, Callen JP. Mycophenolate mofetil is an effective treatment for peristomal pyoderma gangrenosum. Arch Dermatol 2004; 140:1427.
  61. Eaton PA, Callen JP. Mycophenolate mofetil as therapy for pyoderma gangrenosum. Arch Dermatol 2009; 145:781.
  62. Li J, Kelly R. Treatment of pyoderma gangrenosum with mycophenolate mofetil as a steroid-sparing agent. J Am Acad Dermatol 2013; 69:565.
  63. Miller J, Yentzer BA, Clark A, et al. Pyoderma gangrenosum: a review and update on new therapies. J Am Acad Dermatol 2010; 62:646.
  64. Ahronowitz I, Harp J, Shinkai K. Etiology and management of pyoderma gangrenosum: a comprehensive review. Am J Clin Dermatol 2012; 13:191.
  65. Haag CK, Bacik L, Latour E, et al. Perioperative management of pyoderma gangrenosum. J Am Acad Dermatol 2020; 83:369.
  66. Barańska-Rybak W, Kakol M, Naesstrom M, et al. A retrospective study of 12 cases of pyoderma gangrenosum: why we should avoid surgical intervention and what therapy to apply. Am Surg 2011; 77:1644.
  67. Kim DW, Lee BI, Park SH. Accelerated healing of pyoderma gangrenosum in Behçet patient treated with cyclosporine and split thickness skin graft. Ann Plast Surg 2008; 61:552.
  68. Classen DA, Thomson C. Free flap coverage of pyoderma gangrenosum leg ulcers. J Cutan Med Surg 2002; 6:327.
  69. Zakhireh M, Rockwell WB, Fryer RH. Stabilization of pyoderma gangrenosum ulcer with oral cyclosporine prior to skin grafting. Plast Reconstr Surg 2004; 113:1417.
  70. Rozen SM, Nahabedian MY, Manson PN. Management strategies for pyoderma gangrenosum: case studies and review of literature. Ann Plast Surg 2001; 47:310.
  71. Pichler M, Larcher L, Holzer M, et al. Surgical treatment of pyoderma gangrenosum with negative pressure wound therapy and split thickness skin grafting under adequate immunosuppression is a valuable treatment option: Case series of 15 patients. J Am Acad Dermatol 2016; 74:760.
  72. Toyozawa S, Yamamoto Y, Nishide T, et al. Case report: a case of pyoderma gangrenosum with intractable leg ulcers treated by allogeneic cultured dermal substitutes. Dermatol Online J 2008; 14:17.
  73. Alam M, Grossman ME, Schneiderman PI, et al. Surgical management of pyoderma gangrenosum: case report and review. Dermatol Surg 2000; 26:1063.
  74. Barbosa NS, Tolkachjov SN, El-Azhary RA, et al. Clinical features, causes, treatments, and outcomes of peristomal pyoderma gangrenosum (PPG) in 44 patients: The Mayo Clinic experience, 1996 through 2013. J Am Acad Dermatol 2016; 75:931.
  75. Thomas CY Jr, Crouch JA, Guastello J. Hyperbaric oxygen therapy for pyoderma gangrenosum. Arch Dermatol 1974; 110:445.
  76. Davis JC, Landeen JM, Levine RA. Pyoderma gangrenosum: skin grafting after preparation with hyperbaric oxygen. Plast Reconstr Surg 1987; 79:200.
  77. Mazokopakis EE, Kofteridis DP, Pateromihelaki AT, et al. Improvement of ulcerative pyoderma gangrenosum with hyperbaric oxygen therapy. Dermatol Ther 2011; 24:134.
  78. Charles CA, Leon A, Banta MR, Kirsner RS. Etanercept for the treatment of refractory pyoderma gangrenosum: a brief series. Int J Dermatol 2007; 46:1095.
  79. Roy DB, Conte ET, Cohen DJ. The treatment of pyoderma gangrenosum using etanercept. J Am Acad Dermatol 2006; 54:S128.
  80. Pender TM, Ayandibu G, Van Voorhees AS. Certolizumab for the treatment of localized pyoderma gangrenosum associated with Crohn's disease: A case report. Dermatol Ther 2020; 33:e14352.
  81. Cinotti E, Labeille B, Perrot JL, et al. Certolizumab for the treatment of refractory disseminated pyoderma gangrenosum associated with rheumatoid arthritis. Clin Exp Dermatol 2014; 39:750.
  82. Diotallevi F, Campanati A, Radi G, et al. Pyoderma gangrenosum successfully treated with golimumab: Case report and review of the literature. Dermatol Ther 2019; 32:e12928.
  83. Guenova E, Teske A, Fehrenbacher B, et al. Interleukin 23 expression in pyoderma gangrenosum and targeted therapy with ustekinumab. Arch Dermatol 2011; 147:1203.
  84. Fahmy M, Ramamoorthy S, Hata T, Sandborn WJ. Ustekinumab for peristomal pyoderma gangrenosum. Am J Gastroenterol 2012; 107:794.
  85. Phillips FM, Verstockt B, Sebastian S, et al. Inflammatory Cutaneous Lesions in Inflammatory Bowel Disease Treated With Vedolizumab or Ustekinumab: An ECCO CONFER Multicentre Case Series. J Crohns Colitis 2020; 14:1488.
  86. John JM, Sinclair RD. Tildrakizumab for treatment of refractory pyoderma gangrenosum of the penis and polymyalgia rheumatica: Killing two birds with one stone. Australas J Dermatol 2020; 61:170.
  87. Reese AM, Erickson K, Reed KB, Ortega-Loayza AG. Modified dose of guselkumab for treatment of pyoderma gangrenosum. JAAD Case Rep 2022; 21:38.
  88. Burgdorf B, Schlott S, Ivanov IH, Dissemond J. Successful treatment of a refractory pyoderma gangrenosum with risankizumab. Int Wound J 2020; 17:1086.
  89. Orita A, Hoshina D, Hirosaki K. Pyoderma gangrenosum caused by secukinumab successfully treated with risankizumab: a case report and literature review. Clin Exp Dermatol 2022; 47:1372.
  90. Moreno García M, Madrid González M, Prada Lobato JM. Secukinumab for pyoderma gangrenosum: A case report. Med Clin (Barc) 2019; 152:246.
  91. Pollack IR, Wolner ZJ, Hammett J, Swerlick RA. Pyoderma gangrenosum in a patient on ixekizumab. JAAD Case Rep 2021; 16:152.
  92. Kao AS, King AD, Daveluy S. Successful treatment of cabozantinib-induced pyoderma gangrenosum with ixekizumab therapy: A case report. Dermatol Ther 2022; 35:e15716.
  93. Sadik CD, Thieme M, Zillikens D, Terheyden P. First emergence of pyoderma gangraenosum, palmoplantar pustulosis and sacroiliitis in a psoriasis patient associated with switching from secukinumab to brodalumab. J Eur Acad Dermatol Venereol 2019; 33:e406.
  94. Tee MW, Avarbock AB, Ungar J, Frew JW. Rapid resolution of pyoderma gangrenosum with brodalumab therapy. JAAD Case Rep 2020; 6:1167.
  95. Teitel AD. Treatment of pyoderma gangrenosum with methotrexate. Cutis 1996; 57:326.
  96. Sardar P, Guha P, Das NK, et al. Ulcerative pyoderma gangrenosum in mixed connective tissue disorder: a rare association and role of azathioprine in the management. Indian J Dermatol 2011; 56:600.
  97. Schmidt C, Wittig BM, Moser C, et al. Cyclophosphamide pulse therapy followed by azathioprine or methotrexate induces long-term remission in patients with steroid-refractory Crohn's disease. Aliment Pharmacol Ther 2006; 24:343.
  98. Song H, Lahood N, Mostaghimi A. Intravenous immunoglobulin as adjunct therapy for refractory pyoderma gangrenosum: systematic review of cases and case series. Br J Dermatol 2018; 178:363.
  99. Reynoso-von Drateln C, Perla-Navarro AV, Gamez-Nava JI, et al. Intravenous cyclophosphamide pulses in pyoderma gangrenosum: an open trial. J Rheumatol 1997; 24:689.
  100. Zonana-Nacach A, Jiménez-Balderas FJ, Martínez-Osuna P, Mintz G. Intravenous cyclophosphamide pulses in the treatment of pyoderma gangrenosum associated with rheumatoid arthritis: report of 2 cases and review of the literature. J Rheumatol 1994; 21:1352.
  101. Kaminska R, Ikäheimo R, Hollmen A. Plasmapheresis and cyclophosphamide as successful treatments for pyoderma gangrenosum. Clin Exp Dermatol 1999; 24:81.
  102. Burruss JB, Farmer ER, Callen JP. Chlorambucil is an effective corticosteroid-sparing agent for recalcitrant pyoderma gangrenosum. J Am Acad Dermatol 1996; 35:720.
  103. Callen JP, Case JD, Sager D. Chlorambucil--an effective corticosteroid-sparing therapy for pyoderma gangrenosum. J Am Acad Dermatol 1989; 21:515.
  104. Resnik BI, Rendon M, Kerdel FA. Successful treatment of aggressive pyoderma gangrenosum with pulse steroids and chlorambucil. J Am Acad Dermatol 1992; 27:635.
  105. de Cock KM, Thorne MG. The treatment of pyoderma gangrenosum with sodium cromoglycate. Br J Dermatol 1980; 102:231.
  106. Tamir A, Landau M, Brenner S. Topical treatment with 1% sodium cromoglycate in pyoderma gangrenosum. Dermatology 1996; 192:252.
  107. Patel GK, Rhodes JR, Evans B, Holt PJ. Successful treatment of pyoderma gangrenosum with topical 0.5% nicotine cream. J Dermatolog Treat 2004; 15:122.
  108. Wolf R. Nicotine for pyoderma gangrenosum. Arch Dermatol 1998; 134:1071.
  109. Kanekura T, Kanzaki T. Successful treatment of pyoderma gangrenosum with nicotine chewing gum. J Dermatol 1995; 22:704.
  110. Nguyen LQ, Weiner J. Treatment of pyoderma gangrenosum with benzoyl peroxide. Cutis 1977; 19:842.
  111. Vereecken P, Wautrecht JC, De Dobbeleer G, Heenen M. A case of pyoderma gangrenosum stabilized with lymecycline, topical benzoyl peroxide and treated by autograft. Dermatology 1997; 195:50.
  112. Sanders CJ, Hulsmans RF. Successful treatment of pyoderma gangrenosum with topical 5-aminosalicylic acid. Cutis 1993; 51:262.
  113. Tsele E, Yu RC, Chu AC. Pyoderma gangrenosum--response to topical nitrogen mustard. Clin Exp Dermatol 1992; 17:437.
  114. Braun-Falco M, Stock K, Ring J, Hein R. Topical platelet-derived growth factor accelerates healing of myelodysplastic syndrome-associated pyoderma gangrenosum. Br J Dermatol 2002; 147:829.
  115. Freedman BM, Oplinger EH. Use of becaplermin in progressive limb-threatening pyoderma gangrenosum. Adv Skin Wound Care 2002; 15:180.
  116. O'Connor C, Gallagher C, Hollywood A, et al. Anakinra for recalcitrant pyoderma gangrenosum. Clin Exp Dermatol 2021; 46:1558.
  117. Kolios AG, Maul JT, Meier B, et al. Canakinumab in adults with steroid-refractory pyoderma gangrenosum. Br J Dermatol 2015; 173:1216.
  118. Guénin SH, Khattri S, Lebwohl MG. Spesolimab use in treatment of pyoderma gangrenosum. JAAD Case Rep 2023; 34:18.
  119. Lee WS, Choi YJ, Yoo WH. Use of tocilizumab in a patient with pyoderma gangrenosum and rheumatoid arthritis. J Eur Acad Dermatol Venereol 2017; 31:e75.
  120. Scheinberg M, Machado LA, M Castro LG, et al. Successful treatment of ulcerated pyoderma gangrenosum with baricitinib, a novel JAK inhibitor. J Transl Autoimmun 2021; 4:100099.
  121. Nasifoglu S, Heinrich B, Welzel J. Successful therapy for pyoderma gangrenosum with a Janus kinase 2 inhibitor. Br J Dermatol 2018; 179:504.
  122. Kochar B, Herfarth N, Mamie C, et al. Tofacitinib for the Treatment of Pyoderma Gangrenosum. Clin Gastroenterol Hepatol 2019; 17:991.
  123. Orfaly VE, Kovalenko I, Tolkachjov SN, et al. Tofacitinib for the treatment of refractory pyoderma gangrenosum. Clin Exp Dermatol 2021; 46:1082.
  124. Sedano R, Jairath V. Tofacitinib for the Treatment of Three Immune-mediated Conditions in One Patient: Ulcerative Colitis, Pyoderma Gangrenosum, and Alopecia Areata. Inflamm Bowel Dis 2021; 27:e65.
  125. Salmón Olavarría P, Rubio Iturria S, Nantes Castillejo Ó. Tofacitinib, a useful option for the treatment of pyoderma gangrenosum in an ulcerative colitis patient. Rev Esp Enferm Dig 2021; 113:733.
  126. Mensing H. Clofazimine in dermatitis ulcerosa (pyoderma gangrenosum). Open clinical trial. Dermatologica 1988; 177:232.
  127. Paolini O, Hébuterne X, Flory P, et al. Treatment of pyoderma gangrenosum with colchicine. Lancet 1995; 345:1057.
  128. Kontochristopoulos GJ, Stavropoulos PG, Gregoriou S, Zakopoulou N. Treatment of Pyoderma gangrenosum with low-dose colchicine. Dermatology 2004; 209:233.
  129. Smith JB, Shenefelt PD, Soto O, Valeriano J. Pyoderma gangrenosum in a patient with cryoglobulinemia and hepatitis C successfully treated with interferon alfa. J Am Acad Dermatol 1996; 34:901.
  130. Jones RR, Kobza Black A, Donaghy M, et al. Defective monocyte function in pyoderma gangrenosum with IgG kappa paraproteinaemia. Clin Exp Immunol 1983; 52:685.
  131. East-Innis A, Desnoes R, Thame K, et al. Pyoderma gangrenosum associated with osteomyelitis in a paediatric patient: a case report. West Indian Med J 2005; 54:207.
  132. Sheldon DG, Sawchuk LL, Kozarek RA, Thirlby RC. Twenty cases of peristomal pyoderma gangrenosum: diagnostic implications and management. Arch Surg 2000; 135:564.
  133. Richardson JB, Callen JP. Pyoderma gangrenosum treated successfully with potassium iodide. J Am Acad Dermatol 1993; 28:1005.
  134. Jolles S, Niclasse S, Benson E. Combination oral and topical tacrolimus in therapy-resistant pyoderma gangrenosum. Br J Dermatol 1999; 140:564.
  135. Kim TH, Oh SY, Myung SC. Pyoderma gangranosum of the penis. J Korean Med Sci 2009; 24:1200.
  136. Federman GL, Federman DG. Recalcitrant pyoderma gangrenosum treated with thalidomide. Mayo Clin Proc 2000; 75:842.
  137. Seishima M, Mizutani Y, Shibuya Y, et al. Efficacy of granulocyte and monocyte adsorption apheresis for three cases of refractory pyoderma gangrenosum. Ther Apher Dial 2007; 11:177.
  138. Fujimoto E, Fujimoto N, Kuroda K, Tajima S. Leukocytapheresis treatment for pyoderma gangrenosum. Br J Dermatol 2004; 151:1090.
  139. Kanekura T, Maruyama I, Kanzaki T. Granulocyte and monocyte adsorption apheresis for pyoderma gangrenosum. J Am Acad Dermatol 2002; 47:320.
  140. Okuma K, Mitsuishi K, Hasegawa T, et al. A case report of steroid and immunosuppressant-resistant pyoderma gangrenosum successfully treated by granulocytapheresis. Ther Apher Dial 2007; 11:387.
  141. Watanabe Y, Yamada H. Leukocyte adsorption apheresis for the treatment of pyoderma gangrenosum. J Dermatol 2008; 35:792.
  142. Saracino A, Kelly R, Liew D, Chong A. Pyoderma gangrenosum requiring inpatient management: a report of 26 cases with follow up. Australas J Dermatol 2011; 52:218.
  143. Powell FC, Schroeter AL, Su WP, Perry HO. Pyoderma gangrenosum: a review of 86 patients. Q J Med 1985; 55:173.
  144. Abdelrazeq AS, Lund JN, Leveson SH. Pouchitis-associated pyoderma gangrenosum following restorative proctocolectomy for ulcerative colitis. Eur J Gastroenterol Hepatol 2004; 16:1057.
  145. Pishori T, Qureshi AH. Post-colectomy peristomal pyoderma gangrenosum. J Coll Physicians Surg Pak 2005; 15:121.
  146. Yanaru-Fujisawa R, Matsumoto T, Nakamura S, et al. Granulocyte apheresis for pouchitis with arthritis and pyoderma gangrenosum after restorative proctocolectomy for ulcerative colitis: a case report. Inflamm Bowel Dis 2005; 11:780.
  147. Holmlund DE, Wählby L. Pyoderma gangrenosum after colectomy for inflammatory bowel disease. Case report. Acta Chir Scand 1987; 153:73.
  148. Cox NH, Peebles-Brown DA, MacKie RM. Pyoderma gangrenosum occurring 10 years after proctocolectomy for ulcerative colitis. Br J Hosp Med 1986; 36:363.
  149. Ruocco E, Sangiuliano S, Gravina AG, et al. Pyoderma gangrenosum: an updated review. J Eur Acad Dermatol Venereol 2009; 23:1008.
  150. Lee S, Lee JY, Ju HJ, et al. Association of All-Cause and Cause-Specific Mortality Risks With Pyoderma Gangrenosum. JAMA Dermatol 2023; 159:151.
  151. Kaffenberger BH, Hinton A, Krishna SG. The impact of underlying disease state on outcomes in patients with pyoderma gangrenosum: A national survey. J Am Acad Dermatol 2018; 79:659.
Topic 83623 Version 16.0

References

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