Content » Vol 95, Issue 5

Review

Effective Strategies for the Management of Pyoderma Gangrenosum: A Comprehensive Review

Forum PATEL1, Sarah FITZMAURICE1, Christopher DUONG1,Young HE1, Jonathan FERGUS1, Siba P. RAYCHAUDHURI2, Miki Shirakawa GARCIA1 and Emanual MAVERAKIS1,2

1Department of Dermatology, School of Medicine, University of California, Davis, and 2Veterans Affairs Northern California Health Care System, Sacramento, USA

Pyoderma gangrenosum (PG) is an inflammatory disease characterized by painful skin ulcerations with undermined and erythematous borders. The etiology of PG is not well understood, but it is generally considered to be an aberrant immune response characterized by a dermal neutrophilic infiltrate. Given the existence of only a few PG clinical trials, treatment options are largely based upon anecdotal data and small case studies. In addition to classic immunosuppressive medications, PG has been reported to respond well to the anti-TNF agents, infliximab, etanercept, and adalimumab. Newer biologics such as ustekinumab (anti-IL-23), ixekizumab (anti-IL-17) and brodalumab (anti-IL-17R) are promising given the effect of IL-17 on neutrophil migration. However, the effectiveness of these newer agents remains to be rigorously evaluated. Multi-drug regimens have not been well described in the literature but are an excellent alternative for patients with refractory disease. Herein, we provide a comprehensive review of the pathophysiology of PG and of the different treatments available for managing PG patients, including the theoretical benefit of initiating multidrug regimens. We also provide one possible treatment algorithm for patients with refractory disease and give examples of refractory PG cases successfully treated with multidrug regimens. Key words: adalimumab; biologic; infliximab; IVIG; mycophenolate mofetil; pyoderma gangrenosum.

Accepted Nov 11, 2014; Epub ahead of print Nov 12, 2014

Acta Derm Venereol

Emanual Maverakis, MD, Department of Dermatology, University of California, Davis School of Medicine, 3301 C Street, Suite 1400, Sacramento, CA 95816, USA. E-mail: emaverakis@ucdavis.edu

Most commonly, a PG lesion forms at a site of minor trauma as a tender inflammatory nodule or pustule that breaks down over time to create a necrotic ulceration, a process known as pathergy. The prototypical lesions appear on the lower extremities as painful ulcerations with raised erythematous and undermined borders. The undermined border precedes the advancement of the ulcer’s edge and is an ominous sign of worsening disease. In addition to being red, the border can sometimes be purple in color. In active lesions, macular erythema can also exist peripherally to the undermined border, or in place of it in less active lesions, especially in patients partially controlled with immunosuppressive medications. Once formed, the ulceration can increase in size symmetrically or asymmetrically by following the growth of its undermined edge or, alternatively, it can extend through the appearance of new peripherally located pustules.

The base of the ulcer is characteristically composed of excessive granulation tissue with or without neutro­philic abscesses. It usually does not extend past the underlying adipose tissue, but rare lesions involving the fascia have been reported (1). In contrast, “superficial granulomatous pyoderma” presents with superficial ulcerations that on biopsy demonstrate granulomas, plasma cells, and eosinophils (2–5).

Although PG has a predilection for the lower extremities, any body site, including the face and genitalia, can be affected. In patients who have a colostomy, the peristomal region is commonly involved (6). This is likely due to pathergy; even mild trauma can lead to large ulcerations. There are also other subtypes of PG including those with pustular, bullous, and vegetative morphologies (2, 3). Certain patient populations may be predisposed to developing a particular subtype over others and patients may have more than one subtype present simultaneously. For example, patients with PG associated with inflammatory bowel disease (IBD) often have discrete pustular lesions with or without simultaneous classic ulcerations (6, 7).

PG can also present as a paraneoplastic phenomenon, seen frequently in patients with myelodysplastic syndrome, multiple myeloma, polycythemia vera, paraproteinemia, and leukemia (8, 9). These patients can have a more atypical presentation with vesiculobullous lesions or ulcers appearing at atypical sites such as the hands (8–11).

The onset of PG can be just as variable as its clinical presentation. Some patients present with one or two slowly growing ulcers while others experience a rapid onset with the abrupt appearance of multiple rapidly enlarging ulcerations simultaenously. While waxing and waning with spontaneous resolution is possible, PG patients usually require aggressive immunosuppressive therapy to induce disease remission. The chronic nature of the disease usually requires long-term maintenance therapy to prevent relapses from occurring.

Although Brunsting and coworkers were incorrect in proposing an infectious etiology for PG, they did make several seminal observations (12, 13). For example, they noted the chronic nature of the disease and described the characteristic atrophic cigarette paper-like scars associated with the disease. They also reported that PG lesions do not respond well to debridement and skin grafting. In fact, they were the first to document pathergy, demonstrating that new, non-healing PG lesions often occur at skin graft donor sites. However, the most interesting observation made by this group was that PG commonly occurs in patients with severe diarrhea and inflammatory arthritis. Today, the link between PG and a variety of underlying inflammatory diseases has been firmly established.

PG affects patients of all ages but it is characteristically seen in patients between 20–55 years of age. Its incidence has been estimated to be 3–10/million based on a case series from tertiary-care facilities and cohort studies of patients with inflammatory bowel disease (14, 15). A recent population-based study by Langan et al. (16) reported a similar incidence of 6/million. Most frequently, PG has been associated with arthritis, IBD, and hematological disorders (12, 13, 16–19). IBD was found to have the highest association (20.2%), followed by rheumatoid arthritis (RA) (11.8%), and then hematological disorders (3.9%) (16–19). PG has also been associated with HIV, hepatitis, systemic lupus erythematosus, PAPA syndrome (pyogenic arthritis, pyoderma gangrenosum, and acne), Takayasu’s arteritis, and pregnancy (20). Thus, part of the work-up for PG includes searching for underlying co-existing conditions, and the disease management should incorporate a therapy designed to treat PG as well as its associated inflammatory disorder(s) when present. Patients can be quite ill and as a group, they have a 3-fold increase in mortality compared to general population (16).

EVALUATION

Evaluating a lesion concerning for PG should start with a thorough history to assess for associated risk factors, as mentioned above, particularly for signs of IBD and internal malignancies. A focused physical exam should be performed, making particular note of the location of the lesions, characteristics of the ulcer border, and the presence of scars from previous ulcers. Multiple ulcers on the anterior lower extremities with the characteristic undermining border and surrounding erythema are nearly pathognomonic for PG. Likewise, if the disease has been long-standing, healed atrophic “cigarette paper-like” scars are also classic. The size of the undermining border can provide some insight into how rapidly the ulcer edge will evolve.

There are no clear serologic or histological criteria to diagnose PG; it is considered to be a clinical diagnosis of exclusion. The work-up often starts with a biopsy that preferentially includes part of an ulcer border and adjacent skin. Histologic features include dermal edema, neutrophilic abscesses, and suppurative inflammation in the dermis that can extend into subcutaneous fat. Within the dermis, neutrophils loosely surround perivascular lymphocytic infiltrates. Similar changes can be seen in the setting of infection. Thus, infection needs to be ruled out with tissue cultures and special stains prior to making the diagnosis of PG. Clinically, deep fungal infections, syphilis, insect bites, leishmaniasis, and mycobacterial infections can mimic PG. One must also consider factitious ulcerations, vasculitis, parasites, venous insufficiency, antiphospholipid antibody syndrome, malignancy, and other inflammatory disorders in the differential (21, 22). In addition to obtaining a skin biopsy and cultures, workup may sometimes include colonoscopy, and extensive blood and urine screening.

Pathophysiology

PG is now considered to be an aberrant and possibly autoreactive immune response. It is most commonly categorized as a severe neutrophilic dermatosis that is characteristically challenging to diagnose and treat. There are several lines of evidence supporting an immunologic etiology of PG. For one, as mentioned above, patients often have a coexisting immune-mediated disease such as IBD or inflammatory arthritis (RA, ankylosing spondylitis, or other seronegative arthritis) (23–30). Second, patients treated with immune modifying medications for other conditions rarely develop PG; for example, there have been separate case reports of patients developing PG in the setting of infliximab or granulocyte-macrophage colony-stimulating factor (GMCSF) therapy (31, 32). Third, genetic diseases involving the immune system, such as leukocyte-adherence glycoprotein (LAD) deficiency, and PAPA syndrome, are associated with PG or PG-like lesions (33, 34). Finally, medications that target key components of the immune system are emerging as effective treatments for PG. Thus, there is strong correlative evidence in support of PG having an underlying immunologic mechanism; however, its exact pathophysiology is not well understood.

Oka et al. (35) developed an experimental model of PG in which they grafted human skin onto mice with severe combined immunodeficiency (SCID mice). After the grafts were accepted, they injected them with an adenovirus vector containing cDNA that encoded for human interleukin-8 (IL-8). This resulted in an intense perivascular infiltration of neutrophils, which after 12 h caused ulceration of the overlying epidermis. The ulcers were chronic, remaining for a few weeks; and resembled PG, both clinically and histologically. A small number of other studies also exist that support the role of IL-8 in the pathogenesis of PG (35, 36). For example, IL-8 is overexpressed in PG ulcers and serum IL-8 levels fall following successful treatment of PG with systemic therapy. In addition to IL-8, elevations of IL-1β, IL-6, interferon (IFN)-γ, G-CSF, tumour necrosis factor (TNF), matrix metallopaptidase (MMP)-9, MMP, and Elafin have all been reported (37–40). TNF is a cytokine also well known to be associated with IBD (12, 13, 17–19) and it is not surprising that the TNF-targeting biologics infliximab, etanercept and adalimumab, have all been successfully used to treat PG (41–44). Of note, TNF is also known to induce the secretion of IL-8, which is a strong chemotactic factor for neutrophils, the predominant inflammatory cell type seen in PG biopsy specimens. Keratinocytes and T cells are the two main sources of TNF in the skin. Supporting the possibility of an aberrant T-cell response driving PG, two groups have demonstrated the presence of T-cell clonal expansions in PG patients (42, 43). Although these expansions have not been further characterized (45), they likely appear early-on in the course of disease (44). We speculate that the infiltrating T cells are directed against a processed autoantigen that has yet to be determined (46–51). In addition to T cells, a variety of other cell types, including lesional dermal fibroblasts, may also overexpress IL-8 in the setting of PG (36). As will be described later, IL-17 and IL-23 are also very critical in the pathophysiology of PG, due to their role in neutrophil migration.

In some settings IL-1 can play a major role in PG. When associated with PAPA syndrome, PG can be treated with anakinra (52). Anakinra is a recombinant homologue of human IL-1RA that competitively inhibits binding of IL-1α and IL-1β to the IL-1 receptor type 1 (53). IL-1 is a proinflammatory cytokine that initially exists as inactive pro-IL-1β within inflammatory cells. Upon appropriate signaling, pro-IL-1β is cleaved to its active form by caspase-1 (54). PAPA syndrome results from mutations in the CD2 binding protein-1 (CD2BP1) known as PSTPIP1 (55). Binding of PSTPIP1 mutants to pyrin is believed to result in increased IL-1β levels via increased caspase-1 activation (56). These pathways underscore the multifactorial and complex pathogenesis of PG. Similarly, gevokizumab is a monoclonal antibody against IL-1β and is currently being tested against PG (NCT01882504).

Clinical Management of Pyoderma Gangrenosum

A paucity of studies to guide clinical decision-making

The lack of validated outcome measures makes it difficult to conduct PG clinical trials. Common outcome measures such as resolution of ulcers are not ideal for assessing response to treatment because even after the pathogenic inflammation has resolved, PG lesions may take weeks or months to heal. Also, factors not associated with the pathogenic immune response (e.g. obesity, diabetes, edema, etc.) may contribute to the inability of an ulcer to heal. The relatively low incidence of disease, 3–10 patients/million, has also made conducting randomized clinical trials problematic. Currently there are only two controlled clinical trials in PG (41, 57). However, devising an appropriate treatment strategy is essential. The opinions presented here are founded mainly upon our limited knowledge of the pathophysiology of PG, case reports in the literature, our experience with approximately two-dozen PG patients, and our experience in managing other related inflammatory disorders.

Traditional monotherapy

PG is a disfiguring disease that may require aggressive therapy for optimal control. Regardless of the therapeutic agent used, the goal of therapy is the same; reduce the aberrant inflammatory response to promote wound healing while minimizing adverse drug events. Patients with mild PG have been reported to respond well to topical tacrolimus, topical sodium cromoglycate, nicotine, 5-aminosalicylic acid, intralesional triamcinolone diacetate, and intralesional cyclosporine (58–62). However, more severe presentations warrant management with systemic medications. Oral and intravenous medications that have been reported to successfully treat PG (Table SI1) include: azathioprine, corticosteroids, sulfasalazine, dapsone, thalidomide, minocycline, clofazimine, methotrexate, mycophenolate mofetil, tacrolimus, cyclosporine, intravenous immune globulin and cyclophosphamide (23, 41, 63–78). Of these, corticosteroids and cyclosporine are the best characterized. Their rapid onset of action makes them excellent choices for initial therapy. A reasonable alternative to cyclosporine is oral tacrolimus, although it has not been well studied in the setting of PG (79). Both tacrolimus and cyclosporine inhibit TCR-mediated signal transduction pathways. However, tacrolimus is 10–100 times more potent than cyclosporine in vitro (80). Other systemic immunosuppressive agents used to treat PG are listed in Table SI1. PG is a chronic disease, which should be taken into consideration when designing a maintenance treatment regimen. For example, dermatologists tend to limit the use of cyclosporine to 1–2 years, making it unsuitable for the long-term management of PG. It is however reasonable to use this fast-acting agent initially and then to transition the patient to another agent for maintenance therapy (81).

Managing pyoderma gangrenosum with biologics

Over the past decade, new parenterally administered protein therapeutics have changed the landscape of treatment options for patients with immune-mediated diseases. These “biologic” medications are highly sophisticated proteins that target key components of the immune system (82). PG has been reported to respond well to many different biologic medications, most commonly the TNF blockers, etanercept, (83–88) adalimumab (89–94), and infliximab (41). In patients with coexisting IBD and PG, biologics have been reported to successfully treat both conditions simultaneously (90, 91, 94). Of the anti-TNF agents, etanercept is the only one that is ineffective in the treatment of IBD (95), which should be taken into consideration if the patient has concurrent IBD. Unfortunately, there has been very little consistency in the published biologic treatment regimens for PG. One group demonstrated successful treatment with infliximab 5 mg/kg initially followed by adalimumab 40 mg weekly (89). A case report showed complete resolution of recalcitrant PG after 4 months of treatment with 80 mg adalimumab every other week (90) and another group reported complete skin healing after 5 months of therapy with adalimumab 40 mg every other week (91). Other regimens include adalimumab 40 mg weekly or adalimumab 80 mg/week for 2 weeks followed by 40 mg/week thereafter (93, 94). Of note, the biologic most studied in the treatment of PG is infliximab. In a phase II randomized, double-blinded, placebo-controlled study, it was found that at 2 weeks, patients in the infliximab group (5 mg/kg every 2 weeks) had significant improvement compared to placebo (46% vs 6%, p = 0.025). The remission rate at 6 weeks was 21% (41). The relatively poor complete response rate seen in this trial underscores the treatment refractory nature of PG.

New biologics are continuously being developed and it is likely that some will prove useful in the management of PG. For example, ustekinumab blocks the common p40 subunit of IL-12 and IL-23. IL-12 is needed for the differentiation of Th1 cells and IL-23 is important for maintenance of Th17 cells. IL-17, a Th 17 cytokine, is required for neutrophil migration (96). Given the predominance of neutrophils in PG lesions, IL-23 blockade with ustekinumab is a reasonable therapeutic option. Although ustekinumab has not been well characterized in PG, two case reports have demonstrated elevated expression of IL-23 in recalcitrant PG lesions. Furthermore, investigators have demonstrated the effectiveness of ustekinumab in the management of two PG patients and the medication has shown success in other neutrophilic disorders (97–99). Other biologics such as ixekizumab (anti-IL-17) and brodalumab (anti-IL-17R) will likely be effective in treating PG, given their theoretical ability to block IL-17-dependent neutrophil migration (96) but they have yet to be tested.

IL-1 antagonists (i.e. anakinra and gevokizumab) have produced a very good response in patients with PAPA syndrome but studies have not used this drug to treat PG associated with other inflammatory conditions such as IBD. They remain to be a promising treatment option and are being tested in the clinical trials setting (NCT01882504). Certolizumab is a pegylated humanized Fab’ fragment of an anti-TNF monoclonal antibody which will likely be effective in treating PG. We anticipate that there will be many new case reports in the future demonstrating the effectiveness of additional biologics in the treatment of PG.

It can be difficult to predict long-term risks of starting patients on biologic medications (82). Rare associations between infliximab and the development of hepatosplenic T-cell lymphoma have been reported (100) and other forms of lymphoma have been reported (82, 101, 102). A large meta-analysis of RA patients treated with infliximab or adalimumab also demonstrated a small increased risk of lymphoma in patients on TNF antagonists (103). Biologics have also been rarely associated with congestive heart failure, multiple sclerosis, peripheral neuropathy, and anti-DNA antibody formation (82, 104–106). Additionally, reactivation of tuberculosis remains to be one of the more concerning complications associated with anti-TNF therapy (107). Patients need to be screened prior to the initiation of a biologic therapy and monitored serially for reactivation or de novo infection while on therapy (82).

Intravenous immunoglobulin

IVIG has also been used to treat PG (108, 109). A case series reported that 7 of 10 PG patients treated with IVIG experienced remission and 6 of the patients could be maintained in remission with repeat IVIG treatments (108). Another case series reported complete clearance of 5 out of 7 PG patients treated with IVIG and significant improvement in pain was noted in all patients (110). The excellent safety profile of IVIG makes it an appealing treatment option. Adverse drug reactions to be aware of include anaphylaxis, especially in IgA-deficient individuals; aseptic meningitis; and headaches.

Surgical management

Surgery alone has not been shown to be an effective strategy for management of PG. Trauma from surgery may induce formation of new PG ulcers or cause enlargement of the existing lesion. However, we strongly recommend that patients receive traditional wound care after they have been successfully immunosuppressed. Compression, debridement, and even skin grafting can all be options for slow healing ulcers that are entirely devoid of an inflammatory border. Importantly, the persistence of an ulcer does not necessarily mean that a PG patient has failed to respond to immunosuppressive therapy. Ulcers take time to heal following successful suppression of the aberrant pathogenic immune response. Other factors such as edema, often induced by therapy with high-dose prednisone; cytotoxic immunosuppressives, which can inhibit wound healing; diabetes, induced by prednisone therapy; and heavy bacterial colonization can delay wound healing in a patient otherwise appropriately immunosuppressed. Thus, being able to distinguish a residual poorly healing, but non-inflammatory, ulcer from an active inflammatory PG ulcer is essential, as the former does not require additional immunosuppression.

Combination therapy

Fig. 1 shows our personal clinical experience with 3 patients with refractory PG lesions successfully treated with multidrug regimens. Combination therapy for PG has not been highlighted in the literature, but it is important to consider this option for the management of patients with refractory disease. Due to the lack of controlled trials, we recommend using well-studied immunosuppressive drug combinations that are known to be effective in other immune-mediated diseases. For example, methotrexate is commonly used in conjunction with infliximab, but never with myco­phenolate mofetil. The combination of cyclosporine (or tacrolimus), myco­phenolate mofetil, and prednisone is often used to treat hematopoietic transplant patients with graft-versus-host disease. Ultimately, a multidrug regimen may be a better strategy than sequentially switching a patient from one immunosuppressive agent to another. Ideally, synergistic drug combinations would allow for better long-term management with fewer adverse drug events. For example, concomitant administration of cyclosporine and rapamycin at subtherapeutic doses reduces the drugs’ respective toxicities but not their effectiveness (111). One group reported complete resolution of PG ulcers with a regimen of intravenous cylophosphamide pulse therapy (1,000 mg/month), cyclosporine (100 mg/day), and prednisone (30 mg daily tapered to 20 mg daily after 2 months) (112). Another group reported complete response with a combination of cyclosporine (10 mg/kg), mycophenolate mofetil (2 g daily), and negative pressure dressings (113). Yet another group successfully treated an infliximab and azathioprine-resistant PG patient with a combination of adalimumab (80 mg SQ weekly), cyclosporine (3 mg/kg), prednisone (20 mg daily), and sulfasalazine (2 g daily) (90).

 

4310Fig1.tif
Fig. 1. Example of 3 PG patients that required a multi-drug regimen in order to achieve remission. Patient 1 was initially started on etanercept with no response, and eventually required combination therapy with infliximab and low dose mycophenolate mofetil to achieve complete remission. Patient 2 was immediately started on prednisone and cyclosporine with minimal response. Adalimumab was then added, allowing prednisone and cyclosporine to be titrated off. Patient was then maintained on adalimumab for over a year before she was transitioned to maintenance therapy with mycophenolate mofetil at the lowest dose required to maintain remission (500 mg twice daily). Patient 3 was started on prednisone with minimal response and later required combinational therapy with prednisone, mycophenolate mofetil and cyclosporine. After complete response was achieved and cyclosporine was titrated off, the patient was maintained on adalimumab and mycophenolate mofetil 500 mg twice daily. All 3 cases are from the University of California, Davis Rheumatology/Dermatology clinic.

Another therapeutic approach is to treat patients with combination therapy initially and then attempt to maintain remission with a less toxic regimen after the patient has been well-controlled for several months. For example, the slow onset of mycophenolate mofetil makes it a poor choice for the initial management of PG, but it may be an excellent alternative for maintenance therapy. Drugs such as prednisone and cyclosporine have fast onsets and are therefore excellent choices for a patient’s initial management. During the transition the initial medications may need to be titrated down slowly to prevent rebound as a patient is transitioned to a different drug for maintenance therapy.

CONCLUSION

Fig. S11 depicts one possible algorithm for the management of patients with PG, based on current medical knowledge (Table SI1) and our personal clinical experience (see Fig. 1). Given the adverse drug reactions associated with classical immunosuppressive medications and the severe morbidity associated with PG, biologics are an excellent therapeutic option for PG patients.

ACKNOWLEDGMENTS

EM was supported by early career awards from the Burroughs Wellcome Fund and the Howard Hughes Medical Institute.

Funding sources: This study was supported by career awards from the Howard Hughes Medical Institute and the Burroughs Wellcome Fund, EM.

The authors declare no conflicts of interest.

10981.png

1http://www.medicaljournals.se/acta/content/?doi=10.2340/00015555-2008

REFERENCES

 

1. Horvath R, Duffy P, McCormack JG. Necrotic ulceration of the skin and fascia. Clin Infect Dis 2003; 36: 869, 925–866.

2. Powell FC, Su WP, Perry HO. Pyoderma gangrenosum: classification and management. J Am Acad Dermatol 1996; 34: 395–409; quiz 410–392.

3. Wilson-Jones E, Winkelmann RK. Superficial granulomatous pyoderma: a localized vegetative form of pyoderma gangrenosum. J Am Acad Dermatol 1988; 18: 511–521.

4. Quimby SR, Gibson LE, Winkelmann RK. Superficial granulomatous pyoderma: clinicopathologic spectrum. Mayo Clin Proc 1989; 64: 37–43.

5. Dobson CM, Parslew RA, Evans S. Superficial granulomatous pyoderma treated with intravenous immunoglobulin. J Am Acad Dermatol 2003; 48: 456–460.

6. Hughes AP, Jackson JM, Callen JP. Clinical features and treatment of peristomal pyoderma gangrenosum. JAMA 2000; 284: 1546–1548.

7. O’Loughlin S, Perry HO. A diffuse pustular eruption associated with ulcerative colitis. Arch Dermatol 1978; 114: 1061–1064.

8. Duguid CM, O’Loughlin S, Otridge B, Powell FC. Paraneoplastic pyoderma gangrenosum. Australas J Dermatol 1993; 34: 17–22.

9. Maverakis E, Goodarzi H, Wehrli LN, Ono Y, Garcia MS. The etiology of paraneoplastic autoimmunity. Clin Rev Allergy Immunol 2012; 42: 135–144.

10. Bennett ML, Jackson JM, Jorizzo JL, Fleischer AB, Jr., White WL, Callen JP. 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: 37–46.

11. Ehst BD, Minzer-Conzetti K, Swerdlin A, Devere TS. Cutaneous manifestations of internal malignancy. Curr Probl Surg 2010; 47: 384–445.

12. Brunsting LA, Goeckerman WH, O’Leary PA. Pyoderma (echthyma) gangrenosum – Clinical and experimental observations in five cases occurring in adults. Arch Dermatol Syph 1930; 22: 655–680.

13. Perry HO, Brunsting LA. Pyoderma gangrenosum; a clinical study of nineteen cases. AMA Arch Derm 1957; 75: 380–386.

14. Farhi D, Cosnes J, Zizi N, Chosidow O, Seksik P, Beaugerie L, et al. Significance of erythema nodosum and pyoderma gangrenosum in inflammatory bowel diseases: a cohort study of 2402 patients. Medicine (Baltimore) 2008; 87: 281–293.

15. Vavricka SR, Brun L, Ballabeni P, Pittet V, Prinz Vavricka BM, Zeitz J, et al. Frequency and risk factors for extraintestinal manifestations in the Swiss inflammatory bowel disease cohort. Am J Gastroenterol 2011; 106: 110–119.

16. Langan SM, Groves RW, Card TR, Gulliford MC. Incidence, mortality, and disease associations of pyoderma gangrenosum in the United Kingdom: a retrospective cohort study. J Invest Dermatol 2012; 132: 2166–2170.

17. Bernstein CN, Blanchard JF, Rawsthorne P, Yu N. The prevalence of extraintestinal diseases in inflammatory bowel disease: a population-based study. Am J Gastroenterol 2001; 96: 1116–1122.

18. Tan MH, Gordon M, Lebwohl O, George J, Lebwohl MG. Improvement of Pyoderma gangrenosum and psoriasis associated with Crohn disease with anti-tumor necrosis factor alpha monoclonal antibody. Arch Dermatol 2001; 137: 930–933.

19. Veloso FT, Carvalho J, Magro F. Immune-related systemic manifestations of inflammatory bowel disease. A prospective study of 792 patients. J Clin Gastroenterol 1996; 23: 29–34.

20. Goodarzi H, Sivamani RK, Garcia MS, Wehrli LN, Craven H, Ono Y, et al. Effective strategies for the management of pyoderma gangrenosum. Adv Wound Care 2012; 1: 194–199.

21. Weenig RH, Davis MD, Dahl PR, Su WP. Skin ulcers misdiagnosed as pyoderma gangrenosum. N Engl J Med 2002; 347: 1412–1418.

22. Thomas RHM, Payne CMER, Black MM. Wegeners granulomatosis presenting as pyoderma gangrenosum. Clin Exp Dermatol 1982; 7: 523–527.

23. Ruocco E, Sangiuliano S, Gravina AG, Miranda A, Nicoletti G. Pyoderma gangrenosum: an updated review. J Eur Acad Dermatol Venereol 2009; 23: 1008–1017.

24. Powell FC, Schroeter AL, Su WPD, Perry HO. Pyoderma gangrenosum and monoclonal gammopathy. Arch Dermatol 1983; 119: 468–472.

25. Stolman LP, Rosenthal D, Yaworsky R, Horan F. Pyoderma gangrenosum and rheumatoid-arthritis. Arch Dermatol 1975; 111: 1020–1023.

26. Enright H, Jacob HS, Vercellotti G, Howe R, Belzer M, Miller W. Paraneoplastic autoimmune phenomena in patients with myelodysplastic syndromes – response to immunosuppressive Therapy. Br J Haematol 1995; 91: 403–408.

27. Daoud MS, Lust JA, Kyle RA, Pittelkow MR. Monoclonal gammopathies and associated skin disorders. J Am Acad Dermatol 1999; 40: 507–538.

28. Tay CH. Pyoderma-gangrenosum and leukemia. Arch Dermatol 1973; 108: 580–581.

29. Kois JM, Sexton FM, Lookingbill DP. Cutaneous manifestations of multiple-myeloma. Arch Dermatol 1991; 127: 69–74.

30. Mahood JM, Sneddon IB. Pyoderma gangrenosum complicating non-Hodgkins lymphoma. Br J Dermatol 1980; 102: 223–225.

31. Jaimes-Lopez N, Molina V, Arroyave JE, Vasquez LA, Ruiz AC, Castano R, et al. Development of pyoderma gangrenosum during therapy with infliximab. J Dermatol Case Rep 2009; 3: 20–23.

32. Ross HJ, Moy LA, Kaplan R, Figlin RA. Bullous pyoderma-gangrenosum after granulocyte colony-stimulating factor treatment. Cancer 1991; 68: 441–443.

33. Thakur N, Sodani R, Chandra J, Singh V. Leukocyte adhesion defect type 1 presenting with recurrent pyoderma gangrenosum. Indian J Dermatol 2013; 58: 158.

34. Vahlquist A, Håkansson LD, Rönnblom L, Karawajczyk M, Fasth A, van Gijn ME, et al. Recurrent pyoderma gangrenosum and cystic acne associated with leucocyte adhesion deficiency due to novel mutations in ITGB2: successful treatment with infliximab and adalimumab. Acta Derm Venereol 2014 Jul 4. [Epub ahead of print].

35. Oka M, Berking C, Nesbit M, Satyamoorthy K, Schaider H, Murphy G, et al. Interleukin-8 overexpression is present in pyoderma gangrenosum ulcers and leads to ulcer formation in human skin xenografts. Lab Invest 2000; 80: 595–604.

36. Oka M. Pyoderma gangrenosum and interleukin 8. Br J Dermatol 2007; 157: 1279–1281.

37. Tanaka N, Fujioka A, Tajima S, Ishibashi A, Hirose S. Elafin is induced in epidermis in skin disorders with dermal neutrophilic infiltration: interleukin-1 beta and tumour necrosis factor-alpha stimulate its secretion in vitro. Br J Dermatol 2000; 143: 728–732.

38. Saito S, Yasui K, Hosoda W, Ogawa M, Kobayashi N, Sakashita K, et al. CD30+ anaplastic large cell lymphoma complicated by pyoderma gangrenosum with increased levels of serum cytokines. Eur J Haematol 2006; 77: 251–254.

39. Kawakami T, Yamazaki M, Soma Y. Reduction of interleukin-6, interleukin-8, and anti-phosphatidylserine-prothrombin complex antibody by granulocyte and monocyte adsorption apheresis in a patient with pyoderma gangrenosum and ulcerative colitis. Am J Gastroenterol 2009; 104: 2363–2364.

40. Bister V, Makitalo L, Jeskanen L, Saarialho-Kere U. Expression of MMP-9, MMP and TNF-alpha and lack of epithelial MMP-1 and MMP-26 characterize pyoderma gangrenosum. J Cutan Pathol 2007; 34: 889–898.

41. Brooklyn TN, Dunnill MG, Shetty A, Bowden JJ, Williams JD, Griffiths CE, et al. Infliximab for the treatment of pyoderma gangrenosum: a randomised, double blind, placebo controlled trial. Gut 2006; 55: 505–509.

42. Mittal S, Milner BJ, Vickers MA. Pyoderma gangrenosum as a cause of splenomegaly and association with a T-cell clone. Clin Lab Haematol 2005; 27: 402–404.

43. Brooklyn TN, Williams AM, Dunnill MG, Probert CS. T-cell receptor repertoire in pyoderma gangrenosum: evidence for clonal expansions and trafficking. Br J Dermatol 2007; 157: 960–966.

44. Su WP, Schroeter AL, Perry HO, Powell FC. Histopathologic and immunopathologic study of pyoderma gangrenosum. J Cutan Pathol 1986; 13: 323–330.

45. Zhang W, Ono Y, Miyamura Y, Bowlus CL, Gershwin ME, Maverakis E. T cell clonal expansions detected in patients with primary biliary cirrhosis express CX3CR1. J Autoimmun 2011; 37: 71–78.

46. Maverakis E, van den Elzen P, Sercarz EE. Self-reactive T cells and degeneracy of T cell recognition: evolving concepts – from sequence homology to shape mimicry and TCR flexibility. J Autoimmun 2001; 16.

47. Sercarz EE, Maverakis E. Mhc-guided processing: binding of large antigen fragments. Nat Rev Immunol 2003; 3: 621–629.

48. Sercarz EE, Maverakis E. Recognition and function in a degenerate immune system. Mol Immunol 2004; 40: 1003–1008.

49. van den Elzen P, Menezes JS, Ametani A, Maverakis E, Madakamutil L, Tang XL, et al. Limited clonality in autoimmunity: drivers and regulators. Autoimmun Rev 2004; 3: 524–529.

50. Menezes JS, van den Elzen P, Thornes J, Huffman D, Droin NM, Maverakis E, et al. A public T cell clonotype within a heterogeneous autoreactive repertoire is dominant in driving EAE. J Clin Invest 2007; 117: 2176–2185.

51. Maverakis E. Sercarzian immunology – In memoriam. Eli E. Sercarz, 1934–2009. Cell Immunol 2012; 273: 99–108.

52. Brenner M, Ruzicka T, Plewig G, Thomas P, Herzer P. Targeted treatment of pyoderma gangrenosum in PAPA (pyogenic arthritis, pyoderma gangrenosum and acne) syndrome with the recombinant human interleukin-1 receptor antagonist anakinra. Br J Dermatol 2009; 161: 1199–1201.

53. Arend WP, Welgus HG, Thompson RC, Eisenberg SP. Biological properties of recombinant human monocyte-derived interleukin 1 receptor antagonist. J Clin Invest 1990; 85: 1694–1697.

54. Maverakis E, Miyamura Y, Bowen MP, Correa G, Ono Y, Goodarzi H. Light, including ultraviolet. J Autoimmun 2010; 34: J247–257.

55. Wise CA, Gillum JD, Seidman CE, Lindor NM, Veile R, Bashiardes S, et al. Mutations in CD2BP1 disrupt binding to PTP PEST and are responsible for PAPA syndrome, an autoinflammatory disorder. Hum Mol Genet 2002; 11: 961–969.

56. Shoham NG, Centola M, Mansfield E, Hull KM, Wood G, Wise CA, et al. Pyrin binds the PSTPIP1/CD2BP1 protein, defining familial Mediterranean fever and PAPA syndrome as disorders in the same pathway. Proc Natl Acad Sci U S A 2003; 100: 13501–13506.

57. Craig FF, Thomas KS, Mitchell EJ, Williams HC, Norrie J, Mason JM, et al. UK Dermatology Clinical Trials Network’s STOP GAP trial (a multicentre trial of prednisolone versus ciclosporin for pyoderma gangrenosum): protocol for a randomised controlled trial. Trials 2012; 13: 51.

58. Richter-Hintz D, Schuppe HC, Homey B, Lehmann P, Ruzicka T. Topical tacrolimus (FK 506) is effective in the treatment of pyoderma gangrenosum. J Am Acad Dermatol 2000; 42: 304–304.

59. Schuppe HC, Homey B, Assmann T, Martens R, Ruzicka T. Topical tacrolimus for pyoderma gangrenosum. Lancet 1998; 351: 832–832.

60. Moschell S. Pyoderma gangrenosum – a patient successfully treated with intralesional injections of steroid. Arch Dermatol 1967; 95: 121–123.

61. Jennings JL. Pyoderma gangrenosum – successful treat­ment with intralesional steroids. J Am Acad Dermatol 1983; 9: 575–580.

62. Mrowietz U, Christophers E. Clearing of pyoderma-gangrenosum by intralesional cyclosporine-A. Br J Dermatol 1991; 125: 499–499.

63. Faulds D, Goa KL, Benfield P. Cyclosporine – a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in immunoregulatory disorders. Drugs 1993; 45: 953–1040.

64. Johnson RB, Lazarus GS. Pulse Therapy – Therapeutic efficacy in the treatment of pyoderma gangrenosum. Arch Dermatol 1982; 118: 76–84.

65. Michaelsson G, Molin L, Öhman S, Gip L, Lindstrom B, Skogh M, et al. Clofazimine – new agent for treatment of pyoderma gangrenosum. Arch Dermatol 1976; 112: 344–349.

66. Curley RK, Macfarlane AW, Vickers CFH. Pyoderma gangrenosum treated with cyclosporin-A. Br J Dermatol 1985; 113: 601–604.

67. Lynch WS, Bergfeld WF. Pyoderma gangrenosum responsive to minocycline hydrochloride. Cutis 1978; 21: 535–538.

68. Gupta AK, Shear NH, Sauder DN. Efficacy of human intravenous immune globulin in pyoderma-gangrenosum. J Am Acad Dermatol 1995; 32: 140–142.

69. Thomsen K, Rothenborg HW. Clofazimine in the treatment of pyoderma gangrenosum. Arch Dermatol 1979; 115: 851–852.

70. Hohenleutner U, Mohr VD, Michel S, Landthaler M. Mycophenolate mofetil and cyclosporin treatment for recalcitrant pyoderma gangrenosum. Lancet 1997; 350: 1748–1748.

71. Crawford SE, Sherman R, Favara B. Pyoderma gangrenosum with response to cyclophosphamide therapy. J Pediatr 1967; 71: 255–258.

72. Venencie PY, Saurat JH. Pyoderma Gangrenosum in a child – treatment with thalidomide. Ann Pediatr 1982; 29: 67–69.

73. Nousari HC, Lynch W, Anhalt GJ, Petri M. The effectiveness of mycophenolate mofetil in refractory pyoderma gangrenosum. Arch Dermatol 1998; 134: 1509–1511.

74. Galun E, Flugelman MY, Rachmilewitz D. Pyoderma-gangrenosum complicating ulcerative-colitis – successful treatment with methylprednisolone pulse therapy and dapsone. Am J Gastroenterol 1986; 81: 988–989.

75. Happle R, Schiffer HP, Kovary PM. Ocular involvement in pyoderma gangrenosum. Arch Dermatol 1977; 113: 1612.

76. Medeiros CC, Colombari ML, Nassif PW, Gurgel AC, Nassif AE. [Pyoderma gangrenosum in an infant: Case report]. Dermatol Online J 2012; 18: 6 (Article in Portuguese).

77. Teitel AD. Treatment of pyoderma gangrenosum with methotrexate. Cutis 1996; 57: 326–328.

78. Shenefelt PD. Pyoderma gangrenosum associated with cystic acne and hidradenitis suppurativa controlled by adding minocycline and sulfasalazine to the treatment regimen. Cutis 1996; 57: 315–319.

79. Baumgart DC, Wiedenmann B, Dignass AU. Successful therapy of refractory pyoderma gangrenosum and periorbital phlegmona with tacrolimus (FK506) in ulcerative colitis. Inflammatory bowel diseases 2004; 10: 421–424.

80. Bierer BE, Hollander G, Fruman D, Burakoff SJ. Cyclosporin A and FK506: molecular mechanisms of immunosuppression and probes for transplantation biology. Curr Opin Immunol 1993; 5: 763–773.

81. Matis WL, Ellis CN, Griffiths CE, Lazarus GS. Treatment of pyoderma gangrenosum with cyclosporine. Arch Dermatol 1992; 128: 1060–1064.

82. Sivamani RK, Goodarzi H, Garcia MS, Raychaudhuri SP, Wehrli LN, Ono Y, et al. Biologic therapies in the treatment of psoriasis: a comprehensive evidence-based basic science and clinical review and a practical guide to tuberculosis monitoring. Clin Rev Allergy Immunol 2013; 44: 121–140.

83. Roy DB, Conte ET, Cohen D. The treatment of pyoderma gangrenosum using etanercept. J Am Acad Dermatol 2006; 54: S128–S134.

84. Rogge FJ, Pacifico M, Kang N. Treatment of pyoderma gangrenosum with the anti-TNF alpha drug – Etanercept. J Plast Reconstr Aes 2008; 61: 431–433.

85. Pastor N, Betlloch I, Pascual JC, Blanes M, Banuls J, Silvestre JF. Pyoderma gangrenosum treated with anti-TNF alpha therapy (etanercept). Clin Exp Dermatol 2006; 31: 152–153.

86. Goldenberg G, Jorizzo JL. Use of etanercept in treatment of pyoderma gangrenosum in a patient with autoimmune hepatitis. J Dermatol Treat 2005; 16: 347–349.

87. Disla E, Quayum B, Cuppari GG, Pancorbo R. Successful use of etanercept in a patient with pyoderma gangrenosum complicating rheumatoid arthritis. Jcr-J Clin Rheumatol 2004; 10: 50–52.

88. Charles CA, Leon A, Banta MR, Kirsner RS. Etanercept for the treatment of refractory pyoderma gangrenosum: a brief series. Int J Dermatol 2007; 46: 1095–1099.

89. Hubbard VG, Friedmann AC, Goldsmith P. Systemic pyoderma gangrenosum responding to infliximab and adalimumab. Br J Dermatol 2005; 152: 1059–1061.

90. Fonder MA, Cummins DL, Ehst BD, Anhalt GJ, Meyerle JH. Adalimumab therapy for recalcitrant pyoderma gangrenosum. J Burns Wounds 2006; 5: e8.

91. Pomerantz RG, Husni ME, Mody E, Qureshi AA. Adalimumab for treatment of pyoderma gangrenosum. Br J Dermatol 2007; 157: 1274–1275.

92. Heffernan MP, Anadkat MJ, Smith DI. Adalimumab treatment for pyoderma gangrenosum. Arch Dermatol 2007; 143: 306–308.

93. Jacob SE, Weisman RS, Kerdel FA. Pyoderma gangrenosum – rebel without a cure? Int J Dermatol 2008; 47: 192–194.

94. Zold E, Nagy A, Devenyi K, Zeher M, Barta Z. Successful use of adalimumab for treating fistulizing Crohn’s disease with pyoderma gangrenosum: Two birds with one stone. World J Gastroenterol 2009; 15: 2293–2295.

95. Sandborn WJ, Hanauer SB, Katz S, Safdi M, Wolf DG, Baerg RD, et al. Etanercept for active Crohn’s disease: A randomized, double-blind, placebo-controlled trial. Gastroenterology 2001; 121: 1088–1094.

96. Ye P, Rodriguez FH, Kanaly S, Stocking KL, Schurr J, Schwarzenberger P, et al. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J Exp Med 2001; 194: 519–527.

97. Guenova E, Teske A, Fehrenbacher B, Hoerber S, Adamczyk A, Schaller M, et al. Interleukin 23 expression in pyoderma gangrenosum and targeted therapy with ustekinumab. Arch Dermatol 2011; 147: 1203–1205.

98. Fahmy M, Ramamoorthy S, Hata T, Sandborn WJ. Ustekinumab for peristomal pyoderma gangrenosum. Am J Gastroenterol 2012; 107: 794–795.

99. Sharon VR, Garcia MS, Bagheri S, Goodarzi H, Yang C, Ono Y, et al. Management of recalcitrant hidradenitis suppurativa with ustekinumab. Acta Derm Venereol 2012; 92: 320–321.

100. Rosh JR, Gross T, Mamula P, Griffiths A, Hyams J. Hepatosplenic T-cell lymphoma in adolescents and young adults with Crohn’s disease: a cautionary tale? Inflamm Bowel Dis 2007; 13: 1024–1030.

101. Brown SL, Greene MH, Gershon SK, Edwards ET, Braun MM. Tumor necrosis factor antagonist therapy and lymphoma development: twenty-six cases reported to the Food and Drug Administration. Arthritis Rheum 2002; 46: 3151–3158.

102. Wolfe F, Michaud K. Biologic treatment of rheumatoid arthritis and the risk of malignancy: analyses from a large US observational study. Arthritis Rheum 2007; 56: 2886–2895.

103. Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL, Montori V. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA 2006; 295: 2275–2285.

104. Gottlieb A, Korman NJ, Gordon KB, Feldman SR, Lebwohl M, Koo JY, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: Section 2. Psoriatic arthritis: overview and guidelines of care for treatment with an emphasis on the biologics. J Am Acad Dermatol 2008; 58: 851–864.

105. Menter A, Gottlieb A, Feldman SR, Van Voorhees AS, Leonardi CL, Gordon KB, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: Section 1. Overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol 2008; 58: 826–850.

106. De Rycke L, Baeten D, Kruithof E, Van den Bosch F, Veys EM, De Keyser F. Infliximab, but not etanercept, induces IgM anti-double-stranded DNA autoantibodies as main antinuclear reactivity: biologic and clinical implications in autoimmune arthritis. Arthritis Rheum 2005; 52: 2192–2201.

107. Keane J, Gershon S, Wise RP, Mirabile-Levens E, Kasznica J, Schwieterman WD, et al. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N Engl J Med 2001; 345: 1098–1104.

108. Cummins DL, Anhalt GJ, Monahan T, Meyerle JH. Treatment of pyoderma gangrenosum with intravenous immunoglobulin. Br J Dermatol 2007; 157: 1235–1239.

109. Hagman JH, Carrozzo AM, Campione E, Romanelli P, Chimenti S. The use of high-dose immunoglobulin in the treatment of pyoderma gangrenosum. J Dermatolog Treat 2001; 12: 19–22.

110. Kreuter A, Reich-Schupke S, Stucker M, Altmeyer P, Gambichler T. Intravenous immunoglobulin for pyoderma gangrenosum. Br J Dermatol 2008; 158: 856–857.

111. Reitamo S, Spuls P, Sassolas B, Lahfa M, Claudy A, Griffiths CE. Efficacy of sirolimus (rapamycin) administered concomitantly with a subtherapeutic dose of cyclosporin in the treatment of severe psoriasis: a randomized controlled trial. Br J Dermatol 2001; 145: 438–445.

112. Rozin A, Balbir-Gurman A, Gilead L, Slodownik D. Combined therapy for pyoderma gangrenosum. Ann Rheum Dis 2004; 63: 888–889.

113. Gilmour E, Stewart D. Severe recalcitrant pyoderma gangrenosum responding to a combination of mycophenolate mofetil with cyclosporin and complicated by a mononeuritis. Br J Dermatol 2001; 144: 397–400.

114. Hawryluk EB, Penn SK, Wasko MC, Johnson JT, Ferris LK. Treatment of postsurgical pyoderma gangrenosum with a high-potency topical steroid. Ear Nose Throat J 2010; 89: E5–7.

Supplementary content
Table SI
Figure S1