OBSERVATION. (EBA) is a chronic subepidermal bullous disease

Similar documents
Rituximab in refractory autoimmune bullous diseases

Erythema gyratumrepens-like eruption in a patient with epidermolysisbullosaacquisita associated with ulcerative colitis

Epidermolysis Bullosa Acquisita: A Retrospective Clinical Analysis of 30 Cases

Epidermolysis Bullosa Acquisita

Autoimmune Diseases with Oral Manifestations

A case of bullous pemphigoid following pemphigus foliaceus

Introduction to Pemphigoid: Spectrum of Disease & Treatment

Drug Class Prior Authorization Criteria Immune Globulins

Clinical spectrum and therapeutic approach in pemphigus vulgaris

MEDICAL COVERAGE GUIDELINES ORIGINAL EFFECTIVE DATE: 08/19/14 SECTION: DRUGS LAST REVIEW DATE: LAST CRITERIA REVISION DATE: ARCHIVE DATE:

Interesting Case Series. Linear IgA Bullous Dermatosis

Background information of DIF

RITUXAN (rituximab), NONONCOLOGIC USES

Classification: 1. Infective: 2. Traumatic: 3. Idiopathic: Recurrent Aphthous Stomatitis (RAS) 4. Associated with systemic disease:

rituximab (Rituxan ), rituximab and hyaluronidase, human (Rituxan Hycela )

Pemphigus in younger age group in Bangladeshi population

Department of Dermatology, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo , Japan 2

Acquired and Inherited Bullous Diseases

Sarolta Kárpáti. Technology Transfer in Diagnostic Pathology, 5th Central European Regional Meeting May 1, 2010, Siófok

RITUXAN (rituximab and hyaluronidase human)

Title. CitationBritish Journal of Dermatology, 155(5): Issue Date Doc URL. Rights. Type. File Information

IMMUNE GLOBULIN (IVIG AND SCIG) Brand Name Generic Name Length of Authorization Bivigam IVIG Per Medical Guidelines Carimune IVIG Per Medical

Bullous Pemphigoid with Lymphocytic Colitis: A Case Report and Short Literature Review

Paraneoplastic Pemphigus in A Patient with Chronic Lymphocytic Leukemia: A Case Report

Immune Globulin. Prior Authorization

AUTOIMMUNE BLISTERING DISEASES; WINDOW TO SYSTEMIC DISEASE

Current concepts of autoimmune bullous diseases Advances in pathogenesis. Luca Borradori

NEWS RELEASE Genentech Contacts: Media: Joe St. Martin (650) Investor: Karl Mahler Thomas Kudsk Larsen (973)

EPIDERMOLYSIS BULLOSA

Rituximab in refractory autoimmune diseases: Brazilian experience with 29 patients ( )

B. Autoimmune blistering diseases

Autoimmune bullous dermatoses

Appendix to Notification Letter for rituximab and eltrombopag dated 20 February 2014

Rituxan (Rituximab) Policy

National Horizon Scanning Centre. Rituximab (MabThera) for chronic lymphocytic leukaemia. September 2007

Rituxan Hycela. Rituxan Hycela (rituximab and hyaluronidase human) Description

The CARI Guidelines Caring for Australasians with Renal Impairment. Idiopathic membranous nephropathy: use of other therapies GUIDELINES

Michael P. Heffernan, M.D San Luis Dermatology & Laser Clinic Director, US Probity Medical Research

Immunoadsorption as an alternative approach for the treatment of autoimmune disease of the nervous system

Intravenous Immune Globulin (IVIg)

Clinicopathological correlation of blistering diseases of skin

Persistence of Autoreactive IgA-Secreting B Cells Despite Multiple Immunosuppressive Medications Including Rituximab

Rituxan (rituximab) DRUG POLICY BENEFIT APPLICATION

Update: New Treatment Modalities

السكري للداء مرافقة فقاعات diabeticorum= Bullosis

GENENTECH AND BIOGEN IDEC RECEIVE A COMPLETE RESPONSE FROM FDA FOR EARLIER USE OF RITUXAN FOR RHEUMATOID ARTHRITIS

Autoimmune bullous disorders 1)

TRANSPARENCY COMMITTEE OPINION. 8 November 2006

Corporate Medical Policy

A cross-sectional study of clinical, histopathological and direct immmunofluorescence diagnosis in autoimmune bullous diseases

Immunosuppressants. Assistant Prof. Dr. Najlaa Saadi PhD Pharmacology Faculty of Pharmacy University of Philadelphia

Soliris (eculizumab) DRUG.00050

Quality of Life Assessment in Korean Patients with Pemphigus

CLINICAL MEDICAL POLICY

Nine patients with anti-neutrophil cytoplasmic antibody-positive vasculitis successfully treated with rituximab

Treatment with steroids and immunosuppressants

Coverage Criteria: Express Scripts, Inc. monograph dated 12/15/ months or as otherwise noted by indication

HEMORRHAGIC BULLOUS HENOCH- SCHONLEIN PURPURA: A CASE REPORT

Chronic Lymphocytic Leukemia Update. Learning Objectives

Inspiration for this talk. Introduction to Rituximab. Introduction to Rituximab (RTX) Introduction to Rituximab. Introduction to Rituximab

REPEATED B CELL DEPLETION IN TREATMENT OF REFRACTORY SYSTEMIC LUPUS ERYTHEMATOSUS

Immunobullous Diseases: Review and Update. May P. Chan, MD Associate Professor of Pathology and Dermatology University of Michigan

Index. derm.theclinics.com. Note: Page numbers of article titles are in boldface type.

Citation for published version (APA): Buijsrogge, J. J. A. (2011). Unusual variants of subepidermal autoimmune bullous diseases. Groningen: s.n.

I n the past, analgesics and nonsteroidal

MabThera. SC. The wait is over. MabThera delivered in just 5 minutes. SC= subcutaneous injection

The legally binding text is the original French version TRANSPARENCY COMMITTEE OPINION. 6 October 2010

AUTOIMMUNE DISORDERS IN THE ACUTE SETTING

Policy. Medical Policy Manual. Draft Revised Policy: Do Not Implement. Rituximab DESCRIPTION

Clinical Commissioning Policy Proposition: Rituximab for cytopaenia complicating primary immunodeficiency

Immunizing the Immunocompromised. Leilani T. Sanchez, MD, DPPS, DPIDSP Crowne Plaza Galleria Manila, 21 February 2013

COMPLEX CUTANEOUS LUPUS CASES PEARLS AND PITFALLS

FOR PUBLIC CONSULTATION ONLY RITUXIMAB FOR CYTOPENIA FROM PRIMARY IMMUNE DEFICIENCY

Policy Number: PHARMACY T2 Effective Date: April 1, 2018

Il Rituximab nella ITP

IN A STUDY OF PATIENTS WITH GPA & MPA RITUXIMAB REGIMEN DEMONSTRATED CLINICALLY SIGNIFICANT REDUCTION OF MAJOR RELAPSE RATE VS AZA 1

Rituximab for the treatment of Immune (Idiopathic) Thrombocytopenic Purpura (ITP)

Bullous Eruption: A Manifestation of Lupus Erythematosus

Immune tolerance, autoimmune diseases

Pharmacologyonline 1: 1-6 (2010) Case Report Ravishankar and Hiremath CIPROFLOXACIN INDUCED BULLOUS PEMPHIGOID: A CASE REPORT

Rayos Prior Authorization Program Summary

Reporting Autoimmune Diseases in Hematopoietic Stem Cell Transplantation

Discovering potential drug-targets for personalized treatment of autoimmune disorders - what we learn from epidermolysis bullosa acquisita

Immune Globulin Therapy

Clinical Policy: Rituximab (Rituxan) Reference Number: PA.CP.PHAR.260

Name of Policy: Therapeutic Apheresis, with Extracorporeal Column Immunoadsorption and Plasma Reinfusion

Pyoderma gangrenosum presenting with acute generalised haemorrhagic bullae

PHM142 Autoimmune Disorders + Idiosyncratic Drug Reactions

Egyptian Dermatology Online Journal Vol. 8 No 2: 6, December Yasmeen J Bhat*, Iffat Hasan*, Atiya Yaseen*, Hina Altaf*, Shylla Mir**

3/6/2017. Treatment of Detected Antibodies. I have financial relationship(s) with: Thoratec/St. Jude/Abbott Consultant CareDx Consultant/Speaker

RiTUXimab 375 mg/m 2 Therapy-7 day

NCCP Chemotherapy Protocol. Maintenance therapy for the treatment of follicular CD20 positive, B-cell NHL patients responding to induction therapy.

Pemphigus foliaceus: a clinical study of 32 cases in Hong Kong 32

STUDY. High Risk of Death in Elderly Patients With Extensive Bullous Pemphigoid

Primary Diagnosis: Diagnosis Code(s) (if known): Individual s Weight (lbs) (kg)

Corporate Medical Policy

Paul K. Shitabata, M.D. Dermatopathology Institute

Co-existence of Common Variable Immunodeficiency (CVID) with Idiopathic Thrombocytopenic purpura (ITP)

Original Contribution

A. Erythema multiforme and related diseases

Transcription:

OBSERVATION Clinical Response of Severe Mechanobullous Epidermolysis Bullosa Acquisita to Combined Treatment With Immunoadsorption and Rituximab (Anti-CD2 Monoclonal Antibodies) Andrea Niedermeier, MD; Rüdiger Eming, MD; Martin Pfütze, MD; Christine R. Neumann, MD; Claudia Happel; Kristian Reich, MD; Michael Hertl, MD Background: Epidermolysis bullosa acquisita (EBA) is an autoimmune bullous disorder with mucocutaneous involvement, skin fragility, and tendency to scarring. The mechanobullous form of EBA has a chronic relapsing course and is difficult to treat. We describe herein the therapeutic response of 2 patients with recalcitrant mechanobullous EBA to combined treatment with immunoadsorption and rituximab, an anti-cd2 monoclonal antibody that induces depletion of B cells in vivo. Observations: Two patients with mechanobullous EBA received combined treatment with immunoadsorption and rituximab, resulting in an almost complete clinical remission in one patient and stable disease in the other patient. In the patient with complete remission, prolonged B-cell depletion and clinical improvement with disappearance of mucocutaneous erosions paralleled the decline in titers of circulating anti basement membrane zone autoantibodies. In the other patient, combined treatment with immunoadsorption and rituximab reduced the de novo appearance of blisters but did not lead to significant improvement of gingivitis, despite depleted B cells for 6 months that remained at 5% 12 months after the last administration of rituximab, as well as a reduction in autoantibody titers. Conclusion: The patients response suggests that combined treatment with immunoadsorption and rituximab may be a valuable adjuvant treatment regimen for severe mechanobullous EBA, which is in line with recently observed beneficial effects in inflammatory EBA. Arch Dermatol. 27;143:192-198 Author Affiliations: Klinik für Dermatologie und Allergologie, Universitätsklinikum Gießen und Marburg, Philipps-Universität, Marburg (Drs Niedermeier, Eming, Pfütze, and Hertl and Ms Happel), Hautklinik und Poliklinik der Georg-August-Universität Göttingen, Göttingen (Drs Neumann and Reich), and Dermatologikum Hamburg, Hamburg (Dr Reich), Germany. EPIDERMOLYSIS BULLOSA ACquisita (EBA) is a chronic subepidermal bullous disease of the skin and mucous membranes characterized by the presence of autoantibodies against type VII collagen, a major component of anchoring fibrils. There is great diversity in the clinical presentation of the disease. At least 5 clinical subsets of EBA can be distinguished 1 : (1) classic mechanobullous presentation with spontaneous or trauma-induced blisters resembling hereditary dystrophic epidermolysis bullosa, (2) bullous pemphigoid like presentation, (3) mucous membrane pemphigoid like presentation, (4) Brunsting- Perry pemphigoid like presentation, and (5) linear IgA bullous dermatosis like disease. The classic form is especially difficult to treat. This form manifests as mechanobullous noninflammatory disease with an acral distribution and skin fragility over trauma-prone surfaces. The blisters and erosions heal with scarring and milia formation. The vesicles and tense bullae appear on noninflamed or atrophic skin, leading to erosions, crusts, scales, scars, scarring alopecia, cysts, milia, and nail dystrophy. In the more severe form, there can also be loss of nails, sclerosis of the hands and fingers, and esophageal stenosis, 2,3 reminiscent of recessive dystrophic epidermolysis bullosa. 1 The treatment of EBA is often a major therapeutic challenge. There are anecdotal reports of successful treatment with cyclosporine, 4-6 colchicine, 7,8 high- and low-dose intravenous immunoglobulins, 9 plasmapheresis in conjunction with immunoglobulins, 1 and extracorporeal photochemotherapy. 11,12 Because of the 192

rareness of the disease, controlled clinical therapeutic trials have not been performed, to our knowledge. Immunoadsorption (IA) is used in antibodymediated autoimmune disorders refractory to immunosuppressive therapy. 13-16 It is an effective adjuvant treatment to reduce circulating autoantibodies in patients in whom unacceptably high dosages of glucocorticosteroids are required for initial therapy or in whom the disease activity does not allow for sufficient tapering of the glucocorticosteroid dosage necessary to reduce adverse effects. Rituximab is a genetically engineered chimeric monoclonal antibody consisting of a human IgG 1 constant region with murine light- and heavy-chain variable regions that are specific for CD2, a molecule located on pre-b mature B lymphocytes and in most B-cell neoplasms. 17,18 Rituximab was originally designed for the treatment of B-cell neoplasms; it is approved for the first-line treatment of diffuse large B-cell, CD2, non- Hodgkin lymphoma, in combination with CHOP (cyclophosphamide, doxorubicin, vincristine [Oncovin], and prednisone) or other anthracycline-based chemotherapy regimens. It has also been used to treat various refractory autoimmune diseases, including idiopathic thrombocytopenic purpura, 19 autoimmune hemolytic anemia, 2 rheumatoid arthritis, 21 systemic lupus erythematosus, 22,23 myasthenia gravis, 24 Wegener granulomatosis, 25-27 Sjögren syndrome, 28 dermatomyositis, 29 and paraneoplastic pemphigus. 3-32 Rituximab has been recently approved for the treatment of rheumatoid arthritis by the Food and Drug Administration in combination with methotrexate in patients who are refractory to other disease-modifying antirheumatic drugs, including 1 therapy or more with tumor necrosis factor inhibitors. There are case reports on the effect of rituximab treatment in refractory pemphigus 33-49 and a report on the use of rituximab in a patient with inflammatory bullous pemphigoid like EBA. 5 Herein, we describe 2 patients with mechanobullous EBA who were successfully treated with a combination of IA followed by rituximab. METHODS PATIENTS Both patients had an unambiguous diagnosis of EBA according to the following clinical and laboratory diagnostic criteria: (1) cutaneous and mucosal superficial erosions and skin fragility with a tendency to scarring and cutaneous atrophy, (2) subepidermal loss of adherence as shown by histopathologic findings, (3) indirect immunofluorescence showing linear IgG fluorescence along the dermoepidermal junction and dermal IgG staining on skin substrate that was separated at the lamina lucida by incubation with 1M sodium chloride, and (4) detection of anti collagen VII antibodies in the serum. CLINICAL OUTCOME To date, there is no consensus about uniform criteria to grade disease severity or to define remission in autoimmune bullous diseases. An autoimmune bullous skin disorder intensity score was established to assess disease activity and therapeutic response in individual patients (M.P., unpublished data, 26). Briefly, the autoimmune bullous skin disorder intensity score consists of 3 components (skin, oral mucosa, and genital mucosa), representing the most commonly affected areas. Skin involvement was measured according to the type and extent of lesions. This score was designed for monitoring intraindividual therapeutic responses rather than comparing disease activity among individuals. Disease severity of the oral mucosa was graded on a scale from (no disease activity in any of 11 defined sites of the oral mucosa) to 11 (maximum disease activity). Atrophic skin lesions that are typically found in mechanobullous EBA were not counted as lesions. TREATMENT REGIMEN Immunoadsorption was performed according to an established protocol. 51 Briefly, IA was performed on 4 consecutive days using commercially available adsorbers (Globaffin; Fresenius Medical Care, Lexington, Mass), representing 1 treatment cycle. Each cycle was followed by a second cycle after a 4-week interval. During the entire IA treatment, patients continued receiving an immunosuppressive medication consisting of glucocorticosteroids and the glucocorticosteroidsparing adjuvant agent mycophenolate mofetil (1 g 3 times daily). After IA, rituximab was administered intravenously at a dose of 375 mg/m 2 in body surface area during 4 to 6 hours once weekly for 4 consecutive weeks. The patient was pretreated with intravenous administration of 1 mg of prednisolone-21- hydrogen succinate, 4 mg of dimethindene maleate, and 5 mg of ranitidine hydrochloride. As an oral antipyretic agent, 5 mg of acetaminophen was given 2 hours before treatment. IMMUNOLOGICAL FINDINGS Both patients showed linear IgG and complement C3 deposits at the dermoepidermal junction of perilesional skin by direct immunofluorescence; anti collagen VII IgG autoantibodies were detected by immunoblot analysis with recombinant protein of the immunodominant domain NC1 of collagen VII, produced in a baculovirus expression system (Ralf Müller, PhD, unpublished data, 26). Anti basement membrane zone antibodies were detected by indirect immunofluorescence analysis of patients blood samples using epithelial cell surfaces of monkey esophagus as substrate and sodium split human skin. The count of peripheral blood B cells was investigated by flow cytometric analysis of peripheral blood mononuclear cells using a monoclonal antibody reacting with the pan-b CD19 differentiation antigen (Department of Oncology and Hematology, University Hospitals of Marburg, Marburg). REPORT OF CASES PATIENT 1 A 67-year-old man had been diagnosed as having EBA 4 years previously. He had bullous and erosive lesions of the oral mucosa, esophagus, and nasopharynx. His hands, shanks, and feet showed tense blisters and erosions. On both feet, nail loss had occurred on most of the toes due to postinflammatory scarring (Figure 1A). The lesions on the soles significantly impaired walking. Before IA, he had already undergone several long-term immunosuppressive treatment regimens (Figure 2A). Because of unresponsiveness to these regimens, patient 1 received 2 treatment cycles of IA. After completion of IA, 193

A B PATIENT 2 A 42-year-old man who had been diagnosed as having EBA 9 years previously was admitted to the hospital because of a severe chronic course. He had blisters and skin fragility mainly on his trunk and forearms (Figure 3A). The oral mucosa had also been affected in a previous manifestation. To control the disease, he had undergone multiple immunosuppressive therapies (Figure 2B). Because of recalcitrant EBA, patient 2 received 2 treatment cycles of IA followed by rituximab infusions during 4 weeks and adjuvant immunosuppressive treatment with mycophenolate mofetil (3 g/d). The patient discontinued mycophenolate mofetil treatment after rituximab therapy. When he was seen 34 weeks later, his disease was well controlled, with few crusty erosions and atrophic lesions (Figure 3B). The clinical response was accompanied by a decline in titers of circulating autoantibodies (1:8 before treatment and 1:2 after treatment). Titers were below the detection limit at 34 weeks after rituximab therapy. Complete B-cell depletion persisted for at least 37 weeks (Figure 3C). Before IA and Rituximab Treatment C 2 16 12 8 4 15 wk After IA and Rituximab Treatment ABSIS Score for Oral Mucosa Body Surface Area Representing Disease Activity B-Cell Count 5 13 2 28 rituximab was administered during 4 weeks. Patient 1 subsequently received adjuvant immunosuppressive treatment with mycophenolate mofetil (3 g/d). The lesions on his hands improved slightly (Figure 1B), whereas the oral lesions and the lesions on his soles and feet showed no improvement. The patient s autoantibody titers were only marginally reduced (1:16 before treatment and 1:8 after treatment). His peripheral B cells were completely depleted for 6 months and remained at 5% 12 months after the last administration of rituximab (Figure 1C). 12 8 4 Severity ABSIS Score and B-Cell Count, % Figure 1. Patient 1. Partial clinical response of mechanobullous epidermolysis bullosa acquisita to combined treatment with immunoadsorption (IA) and rituximab. A, The patient had blisters and erosions on his hands, feet, and oral mucosa. B, At 15 weeks after completion of combined treatment with IA and rituximab therapy, marked improvement of the lesions on the dorsal aspect of his hands was notable, while erosions of the oral mucosa remained largely unaffected. C, Data are shown before and at 15 weeks after combined treatment with IA and rituximab therapy. ABSIS indicates autoimmune bullous skin disorder intensity. The light gray arrows each indicate 1 treatment cycle of IA; black arrow, 1 treatment cycle of rituximab consisting of 375 mg/m 2 body surface area weekly on 4 consecutive weeks. COMMENT Epidermolysis bullosa acquisita is a chronic subepidermal blistering disease associated with humoral autoimmunity to type VII collagen, an integral part of anchoring fibrils that are important components of the dermoepidermal junction. The pathogenic relevance of collagen VII specific autoantibodies in EBA has been recently shown in an animal model. 52 Epidermolysis bullosa acquisita is refractory to many immunosuppressive treatments. At present, no controlled clinical therapeutic studies exist for this disease, to our knowledge. In a recent systematic review of the literature, it was stated that definitive conclusions for the treatment of EBA 53 cannot be drawn. The bullous pemphigoid like inflammatory presentation of EBA seems to be more responsive to immunosuppression than the classic mechanobullous form, 1 which has been reported to be refractory to systemic corticosteroids, oral azathioprine, methotrexate, and cyclophosphamide. 9,54 Immunoadsorption has increasingly been used to decrease autoantibody levels in autoimmune disorders refractory to established immunosuppressive agents. Reports on the successful use of IA in systemic lupus erythematosus, 14 Sjögren syndrome, 13 severe bullous pemphigoid, 16 and diseases of the pemphigus group 15,55 have been published. For patients with severe pemphigus, a treatment protocol was recently published that induced prolonged clinical improvement of mucosal and cutaneous lesions and was accompanied by a dramatic reduction in serum IgG autoantibodies. 51 Therefore, IA may be an efficient technique to rapidly remove circulating autoantibodies as the pathogenic agent in EBA. In the previous study, 51 the use of an adsorber system (Globaffin) as an adjuvant treatment in 4 patients with pemphigus vulgaris and in 2 patients with pemphigus foliaceus was investigated. The peptide matrix of the adsorber binds to IgG and circulating immune complexes with high affinity 194

A B Prednisolone Azathioprine Azathioprine Dapsone Cyclosporine Dapsone Cyclophosphamide Pulse Methotrexate, 25 mg / wk ECP Dexamethasone Pulse Therapy Cyclosporine Leflunomide Mycophenolate Mofetil 52 14 156 28 Methotrexate IVIG Dapsone Dexamethasone Pulse Therapy Cyclophosphamide Pulse Mycophenolate Mofetil 52 28 26 312 364 416 IVIG Cyclosporine IA Leflunomide IA Rituximab Rituximab Figure 2. Both patients received the following treatment regimens before combined treatment with immunoadsorption (IA) (1 treatment cycle on 4 consecutive days) and rituximab (375 mg/m 2 in body surface area once weekly for 4 consecutive weeks) (arrows): extracorporeal photopheresis (ECP), dexamethasone sodium phosphate pulse (5-1 mg during 3 days every 5 weeks), cyclophosphamide pulse (5-6 mg/m 2 every 4 weeks in patient 1 and an unknown dosage in patient 2), cyclosporine (3.5 mg/d per kg of body weight in patient 1 and 2.8-1.5 mg/d per kilogram in patient 2), dapsone (1 mg/d), leflunomide (2 mg/d), intravenous immunoglobulins (IVIG) (1-2 g/kg in patient 1 and 2 g/kg in patient 2), methotrexate (25 mg/wk in patient 1 and an unknown dosage in patient 2), azathioprine (1-2 mg/d per kilogram), and mycophenolate mofetil (1-3 g/d). The complete regimens are shown for patient 1 (A) and for patient 2 (B), who also received an unknown dosage of prednisolone. and with lower affinity to IgA and IgM. Its binding characteristics are similar to those of protein A Sepharose. 56 In the IA study, 51 all 6 treated patients tolerated IA well and showed no symptoms of allergic reactions or cardiovascular dysfunction; the adsorber system effectively reduced anti desmoglein 1 and desmoglein 3 reactive IgG by a mean of 5% to 7% per IA cycle consisting of 4 consecutive IA treatments. In an earlier study by Schmidt et al, 55 5 patients with severe pemphigus were treated with IA using Staphylococcus aureus protein A columns. The treatment schedule consisted of IA treatment on 3 consecutive days; a fourth IA treatment was given on day 8, followed by up to 19 IA treatments during intervals of 1 to 4 weeks. Protein A IA effectively reduced anti desmoglein 1 and desmoglein 3 IgG by a mean of 76%, correlating with a good clinical response in all patients. 55 In a study by Lüftl et al, 15 a tryptophan-linked polyvinylalcohol adsorber system was applied. Three patients with acute onset and 6 patients with recalcitrant pemphigus received 2 IA treatments each during 3 days, followed by an intravenous prednisolone pulse. The tryptophan-linked polyvinylalcohol adsorber led to a 3% decrease in desmoglein-reactive autoantibodies, which was accompanied by significant clinical improvement. Findings from these studies, as well as other case reports, 57,58 suggest that IA is an efficacious and safe treatment for severe and therapy-resistant bullous autoimmune disorders. In contrast to the IA protocols by Schmidt et al 55 and by Frost et al, 58 we do not favor frequent and successive use of IA. In our view, IA represents an efficacious method that rapidly removes circulating autoantibodies as the pathogenic agent in blistering autoimmune disorders like pemphigus or EBA. However, for sustained remission of the disease, sufficient immunosuppressive treatment after IA is important to prevent rebounding autoantibody synthesis by autoreactive B cells and longlived plasma cells. To achieve long-term arrest of production of pathogenic antibodies, we administer rituximab according to an established protocol previously used in patients with pemphigus and the inflammatory form of EBA. 33-41,43,45-49 Rituximab is administered by slow infusion during several hours. The standard regimen consists of 4 infusions (1 course) of rituximab with a dose of 375 mg/m 2 at weekly intervals following premedication with an analgesic (such as acetaminophen) and an antihistamine or a corticosteroid. Systemic infusion reactions are frequently observed in patients with lymphoma, probably owing to a high load of abnormal B cells. 59,6 Approximately 5% of patients treated with rituximab experience infusionrelated adverse reactions, including cytokine release syndrome. These are accompanied by hypotension and bronchospasm in about 1% of patients. Severe cytokine release syndrome has been reported to occur mostly in patients with lymphoma 6,61 ; however, a few patients have been described who were treated for indications other than lymphoma. A common feature was development of severe reactions during the first infusion, particularly dyspnoe, hypoxia, or severe bronchospasm. 6 In autoimmune diseases, these adverse reactions seem to be less of a problem. A pricking sensation in the throat that occurs 3 to 6 minutes after the start of the infusion is a common feature and has been interpreted as penetration of rituximab into the Waldeyer ring. 21 The absence of normal B cells for several months has not been associated with a significant increase in infectious risk. 62 Total 195

A Before IA and Rituximab Treatment C 8 6 4 2 5 8 37 B 34 wk After IA and Rituximab Treatment ABSIS Score for Oral Mucosa Body Surface Area Representing Disease Activity B-Cell Count 12 8 4 Severity ABSIS Score and B-Cell Count, % Figure 3. Patient 2. Marked clinical response of mechanobullous epidermolysis bullosa acquisita to combined treatment with immunoadsorption (IA) and rituximab. A, The patient had extensive blisters and erosions on his trunk and hands. Top inset, Detail of fresh erosions on the trunk. Bottom inset, Detail of fresh lesion on the index finger of the right hand. B, At 8 months after completion of combined IA and rituximab therapy, the patient had almost complete clinical remission, with postinflammatory atrophic hyperpigmentations on his chest and a few crusty erosions. Top inset, Detail of healed erosions and remaining scars. C, The data are shown before and at 34 weeks after combined treatment with IA and rituximab. ABSIS indicates autoimmune bullous skin disorder intensity. The light gray arrows each indicate 1 treatment cycle of IA; black arrow, 1 treatment cycle of rituximab consisting of 375 mg/m 2 body surface area weekly on 4 consecutive weeks. IgG levels and antitetanus titers are unaffected, whereas IgM levels are reduced to the lower end of the normal range with rituximab therapy. 63 In patients with pemphigus treated with rituximab, treatment is generally well tolerated. Most adverse reactions occur during the first infusion and consist of occasional dyspnea, nausea, fever, chills, and hypotension. 6 These reactions usually diminish with subsequent infusions and can be well controlled by premedication with acetaminophen and antihistamines. However, case reports exist of patients with pemphigus or other autoimmune bullous diseases that describe serious adverse effects, such as pneumonia, septic arthritis, 33 sepsis, 4 fatal Pneumocystis carinii pneumonia, 35 deep venous thrombosis, 5 and hypogammaglobulinemia. 43 Based on previous observations in patients with recalcitrant pemphigus vulgaris, 51 the suppression of de novo autoantibody production may be critical for a prolonged clinical remission of EBA. In support of this, patient 2 in our study with mechanobullous EBA who was recalcitrant to various immunosuppressive treatments (Figure 2) showed a remarkable clinical response to combined treatment with IA and rituximab. Although reaching complete B-cell depletion, patient 1 did not similarly benefit from the combined treatment, but we believe that both treatments contributed to the patient s improvement. In the short term, IA decreased the pathologic circulating autoantibodies, and long-term treatment with rituximab led to clinical remission. We believe that the lack of detectable antibodies to collagen VII in patient 2 reflects the excellent clinical response to combined treatment with IA and rituximab and indicates that spontaneous remission was not responsible for the improvement of EBA. To our knowledge, this is the first publication on successful therapeutic control of the mechanobullous form of EBA by combined treatment with IA and rituximab. It is known from the literature that the NC1 domain of type VII collagen constitutes the major immunodominant epitopes that are targeted by most EBA serum samples. 64-66 In recent studies, 52,67 the pathogenicity of autoantibodies to the NC1 domain of collagen VII was shown in animal models. Therefore, it is likely that circulating antibodies against the NC1 domain of collagen VII may correlate with the disease activity of patients with EBA. Based on the good response in patient 2 and the stable disease in patient 1, the combination of IA and rituximab treatment may be a novel therapeutic option for patients with severe long-standing refractory EBA. Accepted for Publication: July 19, 26. Correspondence: Andrea Niedermeier, MD, Klinik für Dermatologie und Allergologie, Universitätsklinikum Gießen und Marburg, Philipps-Universität, Deutschhausstrasse 9, 3537 Marburg, Germany (a-niedermeier@web.de). Author Contributions: Dr Niedermeier had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis. Study concept and design: Niedermeier, Eming, Pfütze, and Hertl. Acquisition of data: Niedermeier, Eming, Pfütze, and Happel. Analysis and interpretation of data: Niedermeier, Neumann, Happel, Reich, and Hertl. Drafting of the manuscript: Niedermeier, Pfütze, and Hertl. Critical revision of the manuscript for important intellectual content: Niedermeier, Eming, Neumann, Happel, Reich, and Hertl. Obtained funding: Hertl. Administrative, technical, and material support: Niedermeier, Eming, Pfütze, Happel, and Reich. Study supervision: Neumann and Hertl. Financial Disclosure: None reported. Funding/Support: This study was supported in part by grants 24.12.1 from the Wilhelm-Sander-Stiftung (Dr Hertl) and He162/8-1; 82 from the Deutsche Forschungsgemeinschaft (Dr Hertl). 196

REFERENCES 1. Hallel-Halevy D, Nadelman C, Chen M, Woodley DT. Epidermolysis bullosa acquisita: update and review. Clin Dermatol. 21;19:712-718. 2. Stewart MI, Woodley DT, Briggaman RA. Epidermolysis bullosa acquisita and associated symptomatic esophageal webs. Arch Dermatol. 1991;127:373-377. 3. Harman KE, Whittam LR, Wakelin SH, Black MM. Severe, refractory epidermolysis bullosa acquisita complicated by an oesophageal stricture responding to intravenous immune globulin. Br J Dermatol. 1998;139:1126-1127. 4. Khatri ML, Benghazeil M, Shafi M. Epidermolysis bullosa acquisita responsive to cyclosporin therapy. J Eur Acad Dermatol Venereol. 21;15:182-184. 5. Burger J, Gmur J, Bruckner-Tuderman L. Epidermolysis bullosa acquisita, a rare late complication of allogeneic bone marrow transplantation? Bone Marrow Transplant. 1992;9:139-141. 6. Maize JC Jr, Cohen JB. Cyclosporine controls epidermolysis bullosa acquisita co-occurring with acquired factor VIII deficiency. Int J Dermatol. 25;44:692-694. 7. Cunningham BB, Kirchmann TT, Woodley D. Colchicine for epidermolysis bullosa acquisita. J Am Acad Dermatol. 1996;34:781-784. 8. Arora KP, Sachdeva B, Singh N, Bhattacharya SN. Remission of recalcitrant epidermolysis bullosa acquisita (EBA) with colchicine monotherapy. J Dermatol. 25; 32:114-119. 9. Kofler H, Wambacher-Gasser B, Topar G, et al. Intravenous immunoglobulin treatment in therapy-resistant epidermolysis bullosa acquisita. J Am Acad Dermatol. 1997;36:331-335. 1. Engineer L, Ahmed AR. Emerging treatment for epidermolysis bullosa acquisita. J Am Acad Dermatol. 21;44:818-828. 11. Gordon KB, Chan LS, Woodley DT. Treatment of refractory epidermolysis bullosa acquisita with extracorporeal photochemotherapy. Br J Dermatol. 1997; 136:415-42. 12. Bloching M, Dippel E, Jovanovic S, Hess M, Zouboulis CC. Manifestation der Epidermolysis bullosa acquisita (EBA) im HNO-Gebiet. HNO. 1999;47:497-51. 13. Bohm M, Dorner T, Knebel F, Bruns A, Jochmann N, Baumann G. Longlasting effects of immunoadsorption in severe Sjogren s syndrome [letter]. Ann Rheum Dis. 24;63:214-215. 14. Stummvoll GH, Aringer M, Smolen JS, et al. IgG immunoadsorption reduces systemic lupus erythematosus activity and proteinuria: a long term observational study. Ann Rheum Dis. 25;64:115-121. 15. Luftl M, Stauber A, Mainka A, Klingel R, Schuler G, Hertl M. Successful removal of pathogenic autoantibodies in pemphigus by immunoadsorption with a tryptophan-linked polyvinylalcohol adsorber. Br J Dermatol. 23;149:598-65. 16. Herrero-Gonzalez JE, Sitaru C, Klinker E, Brocker EB, Zillikens D. Successful adjuvant treatment of severe bullous pemphigoid by tryptophan immunoadsorption. Clin Exp Dermatol. 25;3:519-522. 17. Tedder TF, Engel P. CD2: a regulator of cell-cycle progression of B lymphocytes. Immunol Today. 1994;15:45-454. 18. Gottenberg JE, Guillevin L, Lambotte O, et al. Tolerance and short term efficacy of rituximab in 43 patients with systemic autoimmune diseases. Ann Rheum Dis. 25;64:913-92. 19. Koulova L, Alexandrescu D, Dutcher JP, O Boyle KP, Eapen S, Wiernik PH. Rituximab for the treatment of refractory idiopathic thrombocytopenic purpura (ITP) and thrombotic thrombocytopenic purpura (TTP): report of three cases. Am J Hematol. 25;78:49-54. 2. Shanafelt TD, Madueme HL, Wolf RC, Tefferi A. Rituximab for immune cytopenia in adults: idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, and Evans syndrome. Mayo Clin Proc. 23;78:134-1346. 21. Edwards JC, Leandro MJ, Cambridge G. B lymphocyte depletion therapy with rituximab in rheumatoid arthritis. Rheum Dis Clin North Am. 24;3:393-43, viii. 22. Looney RJ, Anolik J, Sanz I. Treatment of SLE with anti-cd2 monoclonal antibody. Curr Dir Autoimmun. 25;8:193-25. 23. Eisenberg R. SLE: rituximab in lupus. Arthritis Res Ther. 23;5:157-159. 24. Wylam ME, Anderson PM, Kuntz NL, Rodriguez V. Successful treatment of refractory myasthenia gravis using rituximab: a pediatric case report. J Pediatr. 23;143:674-677. 25. Sneller MC. Rituximab and Wegener s granulomatosis: are B cells a target in vasculitis treatment? Arthritis Rheum. 25;52:1-5. 26. Kallenbach M, Duan H, Ring T. Rituximab induced remission in a patient with Wegener s granulomatosis. Nephron Clin Pract. 25;99:c92-c96. 27. Ferraro AJ, Day CJ, Drayson MT, Savage CO. Effective therapeutic use of rituximab in refractory Wegener s granulomatosis. Nephrol Dial Transplant. 25; 2:622-625. 28. Ahmadi-Simab K, Lamprecht P, Nolle B, Ai M, Gross WL. Successful treatment of refractory anterior scleritis in primary Sjogren s syndrome with rituximab. Ann Rheum Dis. 25;64:187-188. 29. Levine TD. Rituximab in the treatment of dermatomyositis: an open-label pilot study. Arthritis Rheum. 25;52:61-67. 3. Heizmann M, Itin P, Wernli M, Borradori L, Bargetzi MJ. Successful treatment of paraneoplastic pemphigus in follicular NHL with rituximab: report of a case and review of treatment for paraneoplastic pemphigus in NHL and CLL. Am J Hematol. 21;66:142-144. 31. Borradori L, Lombardi T, Samson J, Girardet C, Saurat JH, Hugli A. Anti-CD2 monoclonal antibody (rituximab) for refractory erosive stomatitis secondary to CD2 follicular lymphoma associated paraneoplastic pemphigus. Arch Dermatol. 21;137:269-272. 32. Schadlow MB, Anhalt GJ, Sinha AA. Using rituximab (anti-cd2 antibody) in a patient with paraneoplastic pemphigus. J Drugs Dermatol. 23;2:564-567. 33. Dupuy A, Viguier M, Bedane C, et al. Treatment of refractory pemphigus vulgaris with rituximab (anti-cd2 monoclonal antibody). Arch Dermatol. 24; 14:91-96. 34. Wenzel J, Bauer R, Bieber T, Tuting T. Successful rituximab treatment of severe pemphigus vulgaris resistant to multiple immunosuppressants. Acta Derm Venereol. 25;85:185-186. 35. Morrison LH. Therapy of refractory pemphigus vulgaris with monoclonal anti- CD2 antibody (rituximab). J Am Acad Dermatol. 24;51:817-819. 36. Goebeler M, Herzog S, Brocker EB, Zillikens D. Rapid response of treatmentresistant pemphigus foliaceus to the anti-cd2 antibody rituximab. Br J Dermatol. 23;149:899-91. 37. Virgolini L, Marzocchi V. Anti-CD2 monoclonal antibody (rituximab) in the treatment of autoimmune diseases: successful result in refractory pemphigus vulgaris: report of a case. Haematologica. 23;88:ELT24. 38. Cooper HL, Healy E, Theaker JM, Friedmann PS. Treatment of resistant pemphigus vulgaris with an anti-cd2 monoclonal antibody (rituximab). Clin Exp Dermatol. 23;28:366-368. 39. Herrmann G, Hunzelmann N, Engert A. Treatment of pemphigus vulgaris with anti-cd2 monoclonal antibody (rituximab). Br J Dermatol. 23;148:62-63. 4. Salopek TG, Logsetty S, Tredget EE. Anti-CD2 chimeric monoclonal antibody (rituximab) for the treatment of recalcitrant, life-threatening pemphigus vulgaris with implications in the pathogenesis of the disorder. J Am Acad Dermatol. 22;47:785-788. 41. Arin MJ, Engert A, Krieg T, Hunzelmann N. Anti-CD2 monoclonal antibody (rituximab) in the treatment of pemphigus. Br J Dermatol. 25;153:62-625. 42. Belgi AS, Azeez M, Hoyle C, Williams RE. Response of pemphigus vulgaris to anti-cd2 antibody therapy (rituximab) may be delayed [letter]. Clin Exp Dermatol. 26;31:143. 43. Schmidt E, Herzog S, Brocker EB, Zillikens D, Goebeler M. Long-standing remission of recalcitrant juvenile pemphigus vulgaris after adjuvant therapy with rituximab. Br J Dermatol. 25;153:449-451. 44. Kong HH, Prose NS, Ware RE, Hall RP III. Successful treatment of refractory childhood pemphigus vulgaris with anti-cd2 monoclonal antibody (rituximab). Pediatr Dermatol. 25;22:461-464. 45. Niedermeier A, Wörl P, Barth S, Schuler G, Hertl M. Delayed response of oral pemphigus vulgaris to rituximab treatment. Eur J Dermatol. 26;16:266-27. 46. Esposito M, Capriotti E, Giunta A, Bianchi L, Chimenti S. Long-lasting remission of pemphigus vulgaris treated with rituximab. Acta Derm Venereol. 26;86: 87-89. 47. Pitarch G, Sanchez-Carazo JL, Pardo J, Torrijos A, Roche E, Fortea JM. Treatment of severe refractory pemphigus vulgaris with rituximab [in Spanish]. Actas Dermosifiliogr. 26;97:48-51. 48. Barnadas M, Roe E, Brunet S, et al. Therapy of paraneoplastic pemphigus with rituximab: a case report and review of literature. J Eur Acad Dermatol Venereol. 26;2:69-74. 49. Cecchi R, Gasperini U. Severe pemphigus vulgaris treated with rituximab (Mabthera). J Dermatol. 25;32:862-864. 5. Schmidt E, Benoit S, Brocker EB, Zillikens D, Goebeler M. Successful adjuvant treatment of recalcitrant epidermolysis bullosa acquisita with anti-cd2 antibody rituximab. Arch Dermatol. 26;142:147-15. 51. Eming R, Rech J, Barth S, et al. Prolonged clinical remission of patients with severe pemphigus upon rapid removal of desmoglein-reactive autoantibodies by immunoadsorption. Dermatology. 26;212:177-187. 52. Sitaru C, Mihai S, Otto C, et al. Induction of dermal-epidermal separation in mice by passive transfer of antibodies specific to type VII collagen. J Clin Invest. 25; 115:87-878. 53. Kirtschig G, Murrell D, Wojnarowska F, Khumalo N. Interventions for mucous 197

membrane pemphigoid/cicatricial pemphigoid and epidermolysis bullosa acquisita: a systematic literature review. Arch Dermatol. 22;138:38-384. 54. Chen M, Hallel-Halevy D, Nadelman C, Woodley D. Epidermolysis bullosa acquisita. In: Hertl M, ed. Autoimmune Diseases of the Skin. Wien, Germany: Springer- Verlag; 25:19-132. 55. Schmidt E, Klinker E, Opitz A, et al. Protein A immunoadsorption: a novel and effective adjuvant treatment of severe pemphigus. Br J Dermatol. 23;148: 1222-1229. 56. Ronspeck W, Brinckmann R, Egner R, et al. Peptide based adsorbers for therapeutic immunoadsorption. Ther Apher Dial. 23;7:91-97. 57. Ogata K, Yasuda K, Matsushita M, Kodama H. Successful treatment of adolescent pemphigus vulgaris by immunoadsorption method. J Dermatol. 1999; 26:236-239. 58. Frost N, Messer G, Fierlbeck G, Risler T, Lytton SD. Treatment of pemphigus vulgaris with protein A immunoadsorption: case report of long-term history showing favorable outcome. Ann N Y Acad Sci. 25;151:591-596. 59. Plosker GL, Figgitt DP. Rituximab: a review of its use in non-hodgkin s lymphoma and chronic lymphocytic leukaemia. Drugs. 23;63:83-843. 6. Arin MJ, Hunzelmann N. Anti B-cell directed immunotherapy (rituximab) in the treatment of refractory pemphigus: an update. Eur J Dermatol. 25;15:224-23. 61. Winkler U, Jensen M, Manzke O, Schultz H, Dihl V, Engert A. Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-cd2 monoclonal antibody (rituximab, IDEC-C2B8). Blood. 1999;94:2217-2224. 62. McLaughlin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chimeric anti-cd2 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol. 1998;16:2825-2833. 63. Looney RJ. B cell targeted therapy in diseases other than rheumatoid arthritis. J Rheumatol Suppl. 25;73:25-3. 64. Gammon WR, Murrell DF, Jenison MW, et al. Autoantibodies to type VII collagen recognize epitopes in a fibronectin-like region of the noncollagenous (NC1) domain. J Invest Dermatol. 1993;1:618-622. 65. Lapiere JC, Woodley DT, Parente MG, et al. Epitope mapping of type VII collagen: identification of discrete peptide sequences recognized by sera from patients with acquired epidermolysis bullosa. J Clin Invest. 1993;92:1831-1839. 66. Jones DA, Hunt SW III, Prisayanh PS, Briggaman RA, Gammon WR. Immunodominant autoepitopes of type VII collagen are short, paired peptide sequences within the fibronectin type III homology region of the noncollagenous (NC1) domain. J Invest Dermatol. 1995;14:231-235. 67. Woodley DT, Chang C, Saadat P, Ram R, Liu Z, Chen M. Evidence that anti type VII collagen antibodies are pathogenic and responsible for the clinical, histological, and immunological features of epidermolysis bullosa acquisita. J Invest Dermatol. 25;124:958-964. ARCHIVES Feature Free color publication if color illustrations enhance the didactic value of the article. 198