Back to Journals » Clinical, Cosmetic and Investigational Dermatology » Volume 15

Strategies to Improve Outcomes of Bullous Pemphigoid: A Comprehensive Review of Clinical Presentations, Diagnosis, and Patients’ Assessment

Authors Ruggiero A , Megna M, Villani A, Comune R, Fabbrocini G, di Vico F

Received 25 February 2022

Accepted for publication 7 April 2022

Published 14 April 2022 Volume 2022:15 Pages 661—673

DOI https://doi.org/10.2147/CCID.S267573

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Jeffrey Weinberg



Angelo Ruggiero,1 Matteo Megna,1 Alessia Villani,1 Rosita Comune,2 Gabriella Fabbrocini,1 Francesca di Vico1

1Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, 80131, Italy; 2Section of Radiology and Radiotherapy, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, 80138, Italy

Correspondence: Angelo Ruggiero, Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, Napoli, 80131, Italy, Tel +39 081 7462457, Fax +39 081 7462442, Email [email protected]

Abstract: Bullous pemphigoid (BP) is the most frequent autoimmune bullous disease mainly affecting elderlies. Diagnosis usually results from clinical features, histological examination, and the quantification of circulating typical autoantibodies, due to its higher incidence in elderly patients, bullous pemphigoid treatment and management still represents a challenge due to the higher frequency of several comorbidities in this group of patients, which may also be linked to a reduced tolerance to BP treatments. Hence, an early diagnosis and a prompt correct treatment are mandatory to reach better clinical outcomes and improve as much as possible BP outcomes. Herein, we carried out a comprehensive literature review about the known clinical presentations, diagnosis, assessment and monitoring procedures used in daily clinical practice in patients with BP, to better define strategies to improve as much as possible BP clinical outcomes.

Keywords: bullous pemphigoid, pemphigoid, review, monitoring, assessment, prognosis

Introduction

Bullous pemphigoid (BP) is the most frequent autoimmune bullous disease mainly affecting elderlies.1 It has been recently reported a notably increasing in incidence rates which may be related to the higher aging of population, drugs use, and increasing in diagnostic sensibility also in non-bullous presentations which were frequently underdiagnosed in the past.2 Diagnosis usually results from clinical features, histological examination, and the quantification of circulating typical autoantibodies.3 However, especially in the first phases of the disease, typical clinical features may be lacking, resulting in a late diagnosis and consequently treatment. Moreover, due to its higher incidence in elderly patients, BP treatment and management still represents a challenge due to the higher frequency of several comorbidities in this group of patients. Indeed, regarding BP prognosis, clinical risk factors linked to a higher mortality include older age, and neurological disorders with a higher incidence in elderly patients, such as dementia (including Alzheimer’s disease). Furthermore, several comorbidities may also be linked to a reduced tolerance to BP treatments. Hence, an early diagnosis and a prompt correct treatment are mandatory to reach better clinical outcomes and improve as much as possible BP outcomes. Herein, we carried out a literature review about the known clinical features, diagnosis, assessment and monitoring procedures used in routine clinical practice in patients with BP, to better define strategies to improve as much as possible BP clinical outcomes.

Materials and Methods

A comprehensive review of the English-language medical literature was performed using PubMed, Ovid, Scopus, Embase, and Cochrane Library databases from their inception to 01 December 2021, using Medical Subject Headings (mesh) terms (if applicable) and medical terms for the concepts of BP diagnosis, treatment, assessment, management, and prognosis improvement. Search strategy to identify articles was performed using the following research terms: “bullous pemphigoid”, “bullous”, “bullous pemphigoid clinical features”, “bullous pemphigoid management”, “bullous pemphigoid assessment”, “bullous pemphigoid management”, bullous pemphigoid treatment”, “bullous pemphigoid prognosis”, and combinations thereof. Search involved all fields including title, abstract, keywords, and full text. Clinical and epidemiological studies, review and systematic review regarding BP diagnosis, assessment, and management were included. Papers published from the start of time through December 2021 and from all origins were considered. Therapies and management strategies that could be categorized as traditional Chinese medicine, herbal medicine or Ayurveda/Ayurvedic medicine have been excluded. The article is based on previously conducted studies.

Clinical Features

The BP clinical presentation can be highly variable, especially in the early stages of the disease or in atypical variations that lack the usual blistering lesions.4,5 First signs and symptoms in the non-bullous phase, are commonly nonspecific, usually presenting with pruritus alone or accompanied by several types of lesions, including papular, urticarial, and eczematous lesions: these non-specific manifestations results in a difficult diagnosis. Moreover, this non-specific stage may last from just few days to several months and in some cases, pruritus is the sole indicator of BP.6,7 The development of more typical vesicles and bullae is the main characteristic of the bullous stage of BP: in classic BP tense bullae, varying from 1 to 3 cm of diameter, and usually appearing on erythematous or normal skin.8 Blisters turn into degraded, crusty areas which commonly heal without leaving scars. Milia, hyper- or hypopigmentation, and other post-inflammatory alterations may occur at this stage. The flexor surfaces upper and lower limbs, lower abdomen, and axillae are among the most involved sites.9 The involvement of the mucosae may be verified in only 10–30% of individuals, with oral mucosae being the most frequently involved site,4,5,10,11 while very rarely, nasal, conjunctival, pharyngeal, esophageal, and anogenital mucosae have been reported. BP typically presents with a chronic-remitting course, being self-limited in a few years. Although skin or mucosal lesions are rarely fatal, patients have a six-fold greater death rate than a healthy, age-matched population.12

Non-Bullous Cutaneous Pemphigoid

Typical bullae do not appear in many cases, and the early non-specific lesions, as indicated above, may represent the only sign of BP. The term “non bullous cutaneous pemphigoid”13 has been used in the literature to describe this non-classical presentation. Up to 20% of BP patients have been reported to have this atypical subtype.14,15 Different subtypes of this form have been described: i) eczematous; ii) urticarial; iii) pemphigoid nodularis. Plaques, papules, or nodules, with intense pruritus characterize the eczematous variant. The urticarial variant is characterized by pruritic urticarial papules and plaques.16 Prurigo-like nodules of the distal extremities, frequently in elderly patients, characterize rarer form named “Pemphigoid nodularis”. Bullae, if present, can occur before or after the onset of nodular lesions, appearing at affected or on unaffected skin.14

Other Rare Presentations

The vesicular BP form is characterized by multi-clustered tense vesicles with a symmetric distribution, which mimics the dermatitis herpetiformis lesions.17 Dyshidrosiform pemphigoid is identified by pompholyx-like vesicles that develop in the palmoplantar region and then extend to other body segments.18 Clinically, pemphigoid vegetans is described with purulent and vegetative lesions in intertriginous regions.19 Another uncommon presentation of BP, named “Erythrodermic BP” is characterised by erythroderma with or without blistering.20 Lichen planus pemphigoids is the clinical and histological manifestation of a lichen planus-BP overlap.21 It typically affects young people with a relatively benign course. Although there is no clear link between BP and cancer, erythematous and more figurate lesions than classic BP forms, may be a symptom of an undiagnosed cancer.22 Brunsting-Perry pemphigoid, mostly described in elderly males, is distinguished by the presence of recurring blisters that typically present only on the head and neck. In this case, the lesions frequently heal with atrophic scars.23 Vulvar involvement is unusual as a localized variation of the disease,24 which is distinguished by recurring, non-scarring vesicles and erosions limited to the vulva, well responding to topical steroids.25 Furthermore, other rare presentations have been described in literature. Qiu et al described an interesting and rare presentation of BP in a patient treated with pembrolizumab. Particularly, in this case, the patient presented a widespread eruption of tense bullae on an erythematous base and erosions, mimicking toxic epidermal necrolysis (TEN).26 Another atypical presentation has been reported with palmoplantar keratoderma (PPK) in BP patients. Although dyshidrosiform pemphigoid has been shown to involve palmoplantar region, the typical histological features of these patients (lack of vesicular lesions) may categorize PPK as a novel and different presentations of BP.27 Lichen planus pemphigoides (LPP) is a rare condition characterized by the association of blistering disease, such as BP, and lichenoid skin changes. It usually presents with tense blisters and lichenoid plaques, with typical histological features (the demonstration of autoantibody deposition along the dermal-epidermal junctional zone in perilesional skin biopsies) representing the gold standard to reach the diagnosis. LPP has been considered for long time as a variant of lichen or bullous diseases, growing evidence showed it may be considered as a separate disease entity.28 A new clinical variant has been described in patients presenting typical clinical features of acquired reactive perforating dermatosis (ARPD) coexisting with BP, called perforating like BP. In these described cases, patients firstly presented with papules and nodules with a central keratotic plaque, developing point blisters only later. Both diabetes mellitus and haemodialysis have been suggested as causative in almost all reported cases. Interestingly, histological features were common with typical presentations of BP, showing BP180 autoantibodies were found to bind at the dermo-epidermal junction.29 Finally, another rare clinical presentation has been described by Mahmoudi et al, which reported the atypical case of a patient presenting BP with linear lesions. Particularly, the patient reported a history of intense pruritus and generalized non-inflammatory bullous lesions, with a linear arrangement. Interestingly, no circulating anti-BP180 or anti-BP230 antibodies were found, while the patient showed only IgG antibodies directed exclusively against the 120-kDa LAD-1 antigen.30

Risk Factors and Linked Conditions

Aging is widely recognised as the most important risk factor for BP.31–33 Indeed, BP is mostly a disease of elderly patients, with a reported onset around 75 years, and a clear female preponderance.32–34 BP is less common in children, presenting clinical features comparable to adult BP, except for the more frequent and predominant involvement of acral regions. Childhood BP typically shows a better prognosis than adult forms, quickly improving when treatment is started.35 Several neurologic illnesses have been related to an increased risk of BP. Indeed, several studies in the last ten years have highlighted the link between BP and psychiatric or neurologic diseases.36–48 Including all, 22–46% of BP patients have been reported to present at least one neurologic disorder. Indeed, BP has been significantly associated with dementia (especially Alzheimer’s disease), Parkinson’s disease, epilepsy, cerebrovascular disease, and psychiatric disorders.36,39,41–43,48 In most reported cases, BP occurred after the beginning of the neurological condition, with onset intervals ranging from a few months to more than 5 years.40 Furthermore, BP has been linked to degenerative neurological diseases42 including Parkinson’s and Alzheimer’s, which may involve immunological pathways. The delayed development of BP, in later stages of these diseases, may be probably linked to BP230 neuronal variants, recognised as non-self-structures after the rupture of immune tolerance caused by the neuronal degeneration described in some neurological degenerative disorders.49,50 Hence, these data indicate that neurologic disorders may represent an independent risk factor for BP. Many drugs have been linked to BP.43,51–55 Indeed, more than 50 drugs have been linked to the development of BP based on single case reports, including diuretics (eg, furosemide), antibiotics (eg, ciprofloxacin, amoxicillin), analgesics, potassium iodide, D-penicillamine, biologic therapy such as anti-tumor necrosis factor drugs and the anti-diabetic dipeptidyl peptidase-4. Although some reported drugs may represent a trigger for BP development and recurrence, more studies are needed to better clarify the exact mechanisms through which drugs may induce BP. Because both BP and cancer are diseases of the elderly, the relationship between BP and malignancy may be linked to the older age of patients. Furthermore, multiple case reports have described BP in connection with a variety of malignancies for years, with a number of these cases demonstrating a similar clinical course between internal malignancy and BP prognosis.56 BP has been linked to several different disorders, including several psoriasis and lichen planus, with bullae frequently observed on psoriatic plaques or lichenoid papules, suggesting a Koebner phenomenon. Indeed, burns, trauma, and ultraviolet irradiation may trigger BP in some patients.56 BP has been observed in individuals with several autoimmune diseases, including thyroiditis (both Hashimoto and Grave diseases), rheumatoid arthritis, lupus erythematosus, dermatomyositis, autoimmune neutropenia or thrombocytopenia, and vitiligo. These associations may be linked to a hereditary predisposition to develop autoimmune disorders. However, a recent case–control study found no higher incidence of autoimmune diseases in patients with BP.57 Because glycation of proteins at the dermal–epidermal interface may enhance their immunogenicity, a link between BP and diabetes mellitus has been hypothesized. Further case–control studies39–43,58 did not support this association, and the high occurrence of diabetes in BP patients may be due to the higher frequency of BP in elderly patients.

Although the genetic background of BP has not been completely clarified, recent evidence suggests a genetic background. Indeed, several studies59–66 found the HLA-DQB1*0301 in patients with BP, which may have a role in the development of BP.62 Furthermore, recent findings showed that other gene allotypes may play a role in the genetic background of BP, including mitochondrial anomalies (MT-ATP8 gene), specific cytochrome P450 isoenzyme (CYP2D6), and an over expression of specific Fc receptor subtype (FcRIIIa) on immune cells.66–70

Diagnosis

The diagnosis of BP is based on typical clinical features, the finding of specific self-antibodies (indirect immunofluorescence (IF) or enzyme-linked immunosorbent assay (ELISA), and skin biopsy (direct IF microscopy)). Particularly, specific clinical characteristics and positive direct IF microscopy represent the most useful tools for BP diagnosis. In a small percentage of patients (about 10%) who have both indirect IF microscopy and ELISA negative results, additional immunopathological examinations may be required to detect the typical BP autoantibodies (anti-BP180, and anti-BP230).71 Furthermore, most patients also show hyper-eosinophilia, and/or an elevation of total IgE levels.72

Clinical Criteria

BP should be suspected in older individuals who have generalized pruritus with or without prominent bullae. The first patients’ evaluation should include an accurate anamnesis, looking for comorbidities and risk factors, physical examination, the assessment of BP severity, based on the use of widely recognised scores, such as the BP Disease Activity Index (BPDAI).73–76 In most typical forms, the sparing of the head and neck, and mucous membranes hint at BP diagnosis. A comprehensive and accurate medical history is mandatory, evaluating the presence and the severity of pruritus, the date of symptoms start and their progression, any comorbidities, malignancies, and recent token medication (within the last 1–6 months). The prospective therapy of choice is influenced by associated cardiovascular illnesses and immunological deficits.77,78 Tight, often transparent skin blisters surrounded by erythematous or urticarial plaques and moderate-to-severe itching are symptoms of BP.4,5 A French analysis showed that the diagnosis of BP may be confirmed with high sensitivity (90% and specificity (83%), in patients with a subepidermal blistering disease and linear epidermal basement membrane deposits of IgG or C3 if these criteria are present: i) no head and neck involvement; ii) no mucosal involvement; iii) age over 70 years.79–81 These clinical criteria, however, need an history of recent blisters, which are reported to be absent in about 20% of patients at the time of diagnosis.33,34,82 Hence, in patients with non-bullous manifestations, such as in those presenting urticarial, erosive, or eczematous lesions, a prominent role to reach BP diagnosis is played by direct IF microscopy evaluation and the detection of typical serum antibodies (anti-BP180, and anti-BP-230).83–85

Immunofluorescence (IF) Microscopy

Direct IF microscopy of perilesional, nonbullies skin, represents the gold standard for autoimmune bullous disorders diagnosis. Although it is not specific, direct IF is the most sensitive diagnostic tool for BP.86 It shows fine, linear, continuous IgG (IgG4 and IgG1) and/or complement C3 deposits along with the epidermal basement membrane (BM). IgA and IgE, even if less commonly, can also exhibit a similar pattern. A biopsy sample of early bullae can detect subepidermal blistering with inflammatory infiltration, mainly consisting of eosinophils, neutrophils, and mononuclear cells of the upper dermis, whereas the cavity of the bullae includes a net of fibrin with a heterogeneous cellular infiltrate.4,84,85

Direct IF microscopy examinations of perilesional skin following treatment with 0.09% NaCl solution may guide the differential diagnosis between BP and other autoimmune blistering diseases such epidermolysis bullosa acquisita, mucous membrane pemphigoid, and anti-p200 pemphigoid. The incubation of the tissue in 1 mol/L NaCl solution induces an artificial blister at the BM, leading the skin to be tested for tissue-bound autoantibodies. BP is compatible with immune deposits seen in the BM of the blister’s roof. In DIF microscopy, the “n-serrated” pattern of BP may assist distinguish it from epidermolysis bullosa acquisita, which may have an u-serrated pattern.87

Immune deposits can be seen on either the epidermal or both the epidermal and dermal sides of the split in BP.88 Close examination of the linear fluorescence pattern at the BM helps distinguish BP (“n-serrated” staining pattern) from epidermolysis bullosa acquisita (“u-serrated” staining pattern).89

The presence of serum IgG autoantibodies binding the BM is shown by indirect IF microscopy (IIF), leading to find immune deposits on the epidermal side of the blister in BP. Salt-split skin (of healthy human skin) is the most specific IIF substrate for BP: it was demonstrated to have a 100% specificity for BP.86,90 The salt-split normal human skin is currently the preferred substrate for IIF tests and has replaced the use of other substrates, such as the intact human skin and monkey esophagus, which showed to be not adequate for the differential diagnosis between BP and other blistering disorders. As a result, it is recommended that every patient with clinical suspect of BP undergoes IIF examination using human salt-split skin as substrate.78 In 60–80% of patients, circulating anti-basement membrane autoantibodies of the IgG class and, less commonly, of the IgA and IgE classes are found.11,91–96 These autoantibodies usually bind to the epidermal side of saline-separated normal human skin or, less commonly, to both the epidermal and dermal sides.11,94,95

Enzyme-Linked Immunosorbent Assays (ELISA)

The enzyme-linked immunoassay (ELISA) reveals the existence of circulating antibodies targeting NC16A, BP180, and BP230. Autoantibody serum levels have been observed to correlate with disease activity,97 and high levels of anti-BP180 NC16A have been linked to an increased risk of relapsing.98 Thus, ELISA can be employed to adjust the treatment choice.99 Low blood levels of anti-BP180 or anti-BP230 have been found in around 4% of patients who do not have BP, particularly those with pruritic dermatoses.100,101 Direct immunofluorescence microscopy is indicated in these cases to rule out BP.90

ELISAs that use recombinant proteins that include portions of the BP antigens (the C-terminus of BP230 or BP180, and NC16A domain of BP180) allow for the quick identification of autoantibodies in the blood of BP patients.11,91,102–108 Anti-BP180 antibodies are found in 72–93% of BP patients using commercially available ELISA,88,99–104 instead anti-BP230 autoantibodies are detected in 57–63% of BP patients.93,96,109–112 Furthermore, while serum levels of anti-BP180 autoantibodies have been linked to disease activity,93,109,113 anti-BP230 autoantibodies values did not correspond with disease activity,16 although the latter did appear to be related with localized forms of BP.103,109 In daily clinical practice, the use of ELISA for BP230 protein shows low sensitivity (5–10%), being indicated in case of a negative BP180-ELISA.50,93,109 Immunoblot and immunoprecipitation techniques showed that 60–100% of the serum from BP patients included IgG targeting BP180 and/or BP230;92,114–119 this serum may also include IgA and IgE autoantibodies. Nowadays ELISA has largely supplanted the technically more difficult immunoblot and immunoprecipitation procedures. These traditional, but not entirely standardized, immunochemical approaches are only used situations of BP, including ELISA BP230-negative blood samples, or BP180-negative.

Diagnosis of Bullous Pemphigoid: Practical Approach

The diagnosis of BP may be a challenge, especially in the non-bullous phase, due to non-specific manifestations which may mimic a variety of diseases, such as contact dermatitis, prurigo, drug reactions, urticaria, scabies, and acquired non-autoimmune blistering disorders (such as Stevens-Johnson syndrome, dyshidrotic eczema, bullous arthropod bites, bullous drug eruptions), which are usually differentiated by clinical history, pathologic features, and direct IF microscopy. The pemphigus disease, dermatitis herpetiformis, and paraneoplastic pemphigus are differentiated from BP with no difficulties, basing the differential diagnosis on typical clinical and direct IF findings. In elderlies presenting typical generalized blistering eruption, the diagnosis is easily made using clinical criteria and direct IF. On the other hand, atypical and localized BP forms typically does not present the clinical criteria to confirm BP diagnosis.66,79 Hence, in these cases, the diagnosis must be confirmed by further examinations, including direct IF microscopy examination.

Furthermore, in case of BP suspect, it is mandatory to consider other autoimmune subepidermal blistering diseases with the same linear BM deposits pattern by direct IF microscopy. Particularly, patients presenting with both cutaneous and oral manifestations, the differentiation of BP from mucous membrane pemphigoid may be a challenge. Hence, to reach the right diagnosis, it is important to evaluate the limited skin involvement, the scarring mucosal tendency and the characterization of circulating autoantibodies.120–126 Moreover, BP should be differentiated from the inflammatory form of epidermolysis bullosa acquisita, which may be excluded with IIF examination, and the characterization of serum antibodies (against type VII collagen in epidermolysis bullosa acquisita).127–131

In patients with non-bullous manifestations, further differential diagnosis should be considered, such as in patients presenting vesicles, urticarial lesions, and plaques. These patients have reported to be frequently younger, of Japanese origins, and frequently affected also by psoriasis.6 These patients typically present autoantibodies that specifically bind the dermal side of salt-split human skin.7,132 Finally, another difficult challenge among BP patients is the categorization elderly patients presenting generalized pruritus (with or without skin lesions), in which anti-BP180 and/or BP230 are found, but IF microscopy examination remains negative.96,133–135 It is important to remark that the finding of serum antiBP-180/230 antibodies should not be considered as a diagnostic criterion for BP in the absence of typical direct IF microscopy findings.82 However, a follow-up of these patients may be useful to detect early BP symptoms, because some of these patients, may develop BP and could be thought of as having pemphigoid incipient.

BIOCHIP Method

The BIOCHIP (Dermatology Mosaic 7, EUROIMMUN, Lubeck, Germany) method, represents a new multiplex IF technique reported in the serological diagnosis of BP.136 Particularly, this technique combines the screening of many autoantibodies and target antigen-specific substrates in a single incubation field, leading the simultaneous processing of the most common autoimmune bullous disorders’ antibodies with a single investigation.137–139 Hence, through the presence of multiple antigens a single incubation field, this novel approach led to screen patients for most common autoimmune bullous disease autoantibody using a single investigation, providing a more efficient and cost-saving way to investigate for these conditions, and resulting particularly useful to provide a timely diagnosis in cases where a prompt treatment is required.136 To date, the BIOCHIP has already been validated, showing high specificity and sensitivity in seven studies analysing its use in the diagnosis of BP.136–141

Treatment

BP is a chronic disease showing phase remissions and spontaneous exacerbations, linked with an important impact on patients’ quality of life. Treatment choice is usually driven by both the severity of the disease and the general health conditions and comorbidities of patients. Indeed, due to higher comorbidity rate in BP patients, which are most frequently elderly patients, one of the most important objectives to control both itchy symptoms and skin lesions, avoiding as much as possible patients’ exposure to the potential AEs linked with chosen treatment. Although a standard treatment duration is still unestablished, BP patients are frequently treated for about 6–12 months (including a maintenance phase with low-dose oral or topical steroids), except in corticosteroid-refractory cases, where a longer treatment may be required.109 Systemic corticosteroids have been considered the first-line treatment for long time, especially in generalized forms.71 Particularly, oral prednisone (0.5–1 mg/kg/day, progressively over a period of 6–9 months) is widely considered the most used treatment, leading to reach disease control usually within 2 weeks. The main problem, linked with the use of systemic steroids in elderly patients, is represented by the significant high rate of adverse effects, which may result in a higher mortality and increased adverse than topical clobetasol propionate 0.05%.71 Indeed, based on the results two randomized controlled trials, potent topical steroids (clobetasol propionate 0.05% cream) should be considered as first-line treatment whenever possible, with similar clinical outcomes and lower AEs rate than systemic treatments.75,76 Immunosuppressive drugs, such as azathioprine, methotrexate, and mycophenolate mofetil, should be considered to reduce systemic steroid dosage, and as second-line treatment in case of failure or contraindications to systemic steroids.71,90 Among immunosuppressive drugs, azathioprine (1–3 mg/kg/day orally) is the most frequently used in the management of BP.50 Other reported treatments that have been reported in the treatment of BP include i) the combination of nicotinamide (at the dosage of 500–2500 mg/day) and tetracyclines (200 mg/day orally); ii) topical immunomodulators (tacrolimus ointment); iii) and few cases treated with biological drugs, including rituximab, an anti-CD20, and etanercept an anti-TNFα. However, these treatments showed contrasting results and their role in the treatment of BP is still not well clarified.142–145

Monitoring

Since systemic therapies may be necessary for a long time in BP patients, the severity of the disease should be thoroughly evaluated at baseline and follow-up sessions to evaluate clinical response to treatment as well as any AEs during follow-ups. Both clinical and biological monitoring should be considered. Regarding clinical monitoring, it is currently advised that disease severity and treatment response should be assessed using BP-specific criteria and outcome measures. BP Disease Area Index (BPDAI) score is the most used and accepted score to evaluate disease severity.74 This score considers all the following aspects: body sites affected (with separate scores for skin and mucosal involvements), the type of lesions (differentiated in transient and non-transient), the number of the lesions (considering blisters, eczematous plaques, urticarial lesions), and the severity of itch (using a separate a separate subjective component).74 BPDAI score was established by an international BP definitions committee, resulting relevant for both clinical practice and clinical trials, in order to use a unified score and better compare clinical outcomes during treatments and to improve BP follow-up for physicians.

While clinical monitoring was well established, the biological monitoring, which is frequently based on the use of serological tests (ELISA-BP180), to guide and evaluate treatments is still not established. Indeed, although the correlation between BP activity and BP180 ELISA levels has been reported, the practical and clinical significance of ELISA values during treatments is still not well been clarified. However, it has been shown that the serum concentration of anti-BP180 antibodies, evaluated with ELISA studies, correlates with BP severity.11,33,91 Furthermore, the fluctuation of anti-BP180 levels between baseline, day 60, and day 150 may be used as treatment outcomes predictor.146 Indeed, even a small decrease of these levels between baseline and day 60 has been associated with a higher rate of BP relapse within the first year of treatment.147 Moreover, other indicators found to be linked of later relapse of BP have been found in positive direct IF findings and high BP180-NC16A ELISA title.148 Indeed, a recent study found that relapses did not occur when the ELISA values decreased to 40% of initial values, although they were still not within the normal range.149

Conclusions

Although our knowledge of BP pathophysiology increased in recent years, mortality and morbidity rates are still high in this disease. Indeed, several large studies showed that even with early treatment, BP patients have a prognosis comparable with a diagnosis of end-stage heart disease, reporting a mortality rate of up to 40% of patients dying within 12 months from the diagnosis.150 This may be linked to both patients’ comorbidities and immunosuppressive drugs used in BP treatments.150 An early diagnosis, and consequently correct management, monitoring, and assessment of patients suffering from BP are crucial to improving as much as possible the prognosis of these patients. As shown, many factors may trigger BP an accurate anamnesis focused on the recognition of a possible trigger factor can improve prognosis by promptly removing it. Furthermore, most BP patients are elderly, suffering from multiple comorbidities; hence, it is important to choose the best treatment option also consider other diseases and reduce as much as possible side effects which may be linked to the prescribed treatments. It results in primary relevance also the correct assessment of disease severity. Indeed, using widely approved scores, such as BPDAI, to assess disease severity at baseline and each follow-up visit, may improve the management of the disease by choosing the right treatment at the right dosage, trying to reduce patients’ exposition to possible adverse events which may be linked, for example, to higher drugs’ dosage. Near to the clinical monitoring, also biological monitoring is frequently required in clinical practice. Indeed, even if a standardized definition of the best serum concentration of BP-180 antibodies levels is to reach during treatments, biological monitoring may be useful during follow-ups, due to the link between an elevation of these levels and BP flare-up. Another important aspect to consider in the management of BP patients, especially in elderlies, is to carefully check the eventual burden of any comorbidities during treatments.

In conclusion, the management of BP is still considered a challenge in dermatology, due to the high mortality rates and difficulties in treating elderly patients. However, early recognition of the disease, correct treatment choice, and monitoring during follow-up of both BP and patients’ comorbidities may certainly improve patients’ outcomes.

Acknowledgments

None of the authors received any financial support for the writing of this manuscript.

Disclosure

G. Fabbrocini acted as a speaker or consultant for AbbVie, Amgen, Eli Lilly, Janssen, Leo-Pharma, Almyrall, Novartis, and UCB. M. Megna acted as a speaker or consultant for AbbVie, Eli Lilly, Janssen, Leo-Pharma, and Novartis. None of the other contributing authors have any conflict of interest, including specific financial interests of relationships and affiliation relevant to the subject matter or discussed materials in the manuscript.

References

1. Miyamoto D, Santi CG, Aoki V, Maruta CW. Bullous pemphigoid. An Bras Dermatol. 2019;94(2):133–146. doi:10.1590/abd1806-4841.20199007

2. Schmidt E, Zillikens D. Pemphigoid diseases. Lancet. 2013;381:320–332. doi:10.1016/S0140-6736(12)61140-4

3. Lamberts A, Meijer JM, Jonkman MF. Nonbullous pemphigoid: a systematic review. J Am Acad Dermatol. 2018;78:989–995.e2. doi:10.1016/j.jaad.2017.10.035

4. Korman N. Bullous pemphigoid. J Am Acad Dermatol. 1987;16:907–924. doi:10.1016/S0190-9622(87)70115-7

5. Di Zenzo G, Marazza G, Borradori L. Bullous pemphigoid: physiopathology, clinical features and management. Adv Dermatol. 2007;23:257–288. doi:10.1016/j.yadr.2007.07.013

6. Groth S, Recke A, Vafia K, et al. Development of a simple ELISA for the detection of autoantibodies in anti-P200 pemphigoid. Br J Dermatol. 2011;164:76–82. doi:10.1111/j.1365-2133.2010.10056.x

7. Dainichi T, Koga H, Tsuji T, et al. From anti-P200 pemphigoid to anti laminin gamma1 pemphigoid. J Dermatol. 2010;37:231–238. doi:10.1111/j.1346-8138.2009.00793.x

8. Cozzani E, Gasparini G, Burlando M, Drago F, Parodi A. Atypical presentations of bullous pemphigoid: clinical and immunopathological aspects. Autoimmun Rev. 2015;14:438–445. doi:10.1016/j.autrev.2015.01.006

9. Walsh SR, Hogg D, Mydlarski PR. Bullous pemphigoid: from bench to bedside. Drugs. 2005;65:905–926. doi:10.2165/00003495-200565070-00002

10. Fuertes de Vega I, Iranzo-Fernández P, Mascaró-Galy JM. Bullous pemphigoid: clinical practice guidelines. Actas Dermosifiliogr. 2014;105(4):328–346. doi:10.1016/j.ad.2012.10.022

11. Di Zenzo G, Thoma-Uszynski S, Fontao L, et al. Multicenter prospective study of the humoral autoimmune response in bullous pemphigoid. Clin Immunol. 2008;128:415–426. doi:10.1016/j.clim.2008.04.012

12. Joly P, Baricault S, Sparsa A, et al. Incidence and mortality of bullous pemphigoid in France. J Invest Dermatol. 2012;132:1998–2004. doi:10.1038/jid.2012.35

13. Bakker CV, Terra JB, Jonkman MF. Toward a practical renaming of bullous pemphigoid and all its variants-reply. JAMA Dermatol. 2014;150:459–460. doi:10.1001/jamadermatol.2014.54

14. Di Zenzo G, Della Torre R, Zambruno G, Borradori L. Bullous pemphigoid: from the clinic to the bench. Clin Dermatol. 2012;30:3–16. doi:10.1016/j.clindermatol.2011.03.005

15. Jeong SJ, Lee CW. Bullous pemphigoid: persistent lesions of eczematous/urticarial erythemas. Cutis. 1995;56:225–226.

16. Bakker CV, Terra JB, Pas HH, Jonkman MF. Bullous pemphigoid as pruritus in the elderly: a common presentation. JAMA Dermatol. 2013;149:950–953. doi:10.1001/jamadermatol.2013.756

17. Yasuda M, Miyachi Y, Utani A. Two cases of dyshidrosiform pemphigoid with different presentations. Clin Exp Dermatol. 2009;34:151–153. doi:10.1111/j.1365-2230.2008.03083.x

18. Komine M, Nashiro K, Asahina A, et al. Vesicular pemphigoid. Int J Dermatol. 1992;31:868–870. doi:10.1111/j.1365-4362.1992.tb03546.x

19. Goldscheider I, Herzinger T, Varga R, et al. Childhood lichen planus pemphigoides: report of two cases treated successfully with systemic glucocorticoids and dapsone. Pediatr Dermatol. 2014;31:751–753. doi:10.1111/pde.12214

20. Alonso-Llamazares J, Dietrich SM, Gibson LE. Bullous pemphigoid presenting as exfoliative erythroderma. J Am Acad Dermatol. 1998;39:827–830. doi:10.1016/S0190-9622(98)70358-5

21. Suda-Takayanagi T, Hara H, Ohyama B, Hashimoto T, Terui T. A case of pemphigoid vegetans with autoantibodies against both BP180 and BP230 antigens. J Am Acad Dermatol. 2011;64:206–208. doi:10.1016/j.jaad.2009.06.078

22. Jedlickova H, Niedermeier A, Zgažarová S, Hertl M. Brunsting-Perry pemphigoid of the scalp with antibodies against laminin 332. Dermatology. 2011;222:193–195. doi:10.1159/000322842

23. Gilmour E, Bhushan M, Griffiths CE. Figurate erythema with bullous pemphigoid: a true paraneoplastic phenomenon? Clin Exp Dermatol. 1999;24:446–448. doi:10.1046/j.1365-2230.1999.00528.x

24. Bağcı IS, Horváth ON, Ruzicka T, Sárdy M. Bullous pemphigoid. Autoimmun Rev. 2017;16(5):445–455. doi:10.1016/j.autrev.2017.03.010

25. Farrell AM, Kirtschig G, Dalziel KL, et al. Childhood vulval pemphigoid: a clinical and immunopathological study of five patients. Br J Dermatol. 1999;140:308–312. doi:10.1046/j.1365-2133.1999.02668.x

26. Qiu C, Shevchenko A, Hsu S. Bullous pemphigoid secondary to pembrolizumab mimicking toxic epidermal necrolysis. JAAD Case Rep. 2020;6(5):400–402. doi:10.1016/j.jdcr.2020.03.003

27. Sadeghi B, Nili A, Tavakolpour S, Balighi K, Daneshpazhooh M, Mahmoudi H. Concomitant bullous pemphigoid and palmoplantar keratoderma: a report of three cases and review of literature. Dermatol Ther. 2020;33(6):e14481. doi:10.1111/dth.14481

28. Hübner F, Langan EA, Recke A. Lichen planus pemphigoides: from lichenoid inflammation to autoantibody-mediated blistering. Front Immunol. 2019;10:1389. doi:10.3389/fimmu.2019.01389

29. Schauer F, Kern JS, Virtic O, et al. A new clinical variant of acquired reactive perforating dermatosis-like bullous pemphigoid. Br J Dermatol. 2019;180(1):231–232. doi:10.1111/bjd.17146

30. Mahmoudi H, Toosi R, Kamyab K, Zillikens D, Schmidt E, Daneshpazhooh M. Bullous pemphigoid with linear lesions and antibodies exclusively against the soluble ectodomain of BP180 (LAD-1). J Dtsch Dermatol Ges. 2019;17(9):933–935.

31. Langan SM, Smeeth L, Hubbard R, et al. Bullous pemphigoid and pemphigus vulgaris incidence and mortality in the UK: population based cohort study. BMJ. 2008;337:a180. doi:10.1136/bmj.a180

32. Jung M, Kippes W, Messer G, et al. Increased risk of bullous pemphigoid in male and very old patients: a population-based study on incidence. J Am Acad Dermatol. 1999;41:266–268. doi:10.1016/S0190-9622(99)70061-7

33. Joly P, Benichou J, Lok C, et al. Prediction of survival for patients with bullous pemphigoid: a prospective study. Arch Dermatol. 2005;141:691–698. doi:10.1001/archderm.141.6.691

34. Marazza G, Pham HC, Scha¨rer L, et al. Incidence of bullous pemphigoid and pemphigus in Switzerland: a 2-year prospective study. Br J Dermatol. 2009;161:861–868. doi:10.1111/j.1365-2133.2009.09300.x

35. de la Fuente S, Hernández-Martín Á, de Lucas R, et al. Postvaccination bullous pemphigoid in infancy: report of three new cases and literature review. Pediatr Dermatol. 2013;30:741–744. doi:10.1111/pde.12231

36. Cordel N, Chosidow O, Hellot M-F, et al. Neurological disorders in patients with bullous pemphigoid. Dermatology. 2007;215:187–191. doi:10.1159/000106574

37. Jedlickova H, Hlubinka M, Pavlik T, et al. Bullous pemphigoid and internal diseases: a case–control study. Eur J Dermatol. 2010;20:96–101. doi:10.1684/ejd.2010.0805

38. Chosidow O, Doppler V, Bensimon G, et al. Bullous pemphigoid and amyotrophic lateral sclerosis: a new clue for under standing the bullous disease? Arch Dermatol. 2000;136:521–524. doi:10.1001/archderm.136.4.521

39. Chen YJ, Wu CY, Lin MW, et al. Comorbidity profiles among patients with bullous pemphigoid: a nationwide population-based study. Br J Dermatol. 2011;165:593–599. doi:10.1111/j.1365-2133.2011.10386.x

40. Stinco G, Codutti R, Scarbolo M, Valent F, Patrone P. A retrospective epidemiological study on the association of bullous pemphigoid and neurological diseases. Acta Derm Venereol. 2005;85:136–139. doi:10.1080/00015550410024481

41. Taghipour K, Chi -C-C, Vincent A, Groves RW, Venning V, Wojnarowska F. The association of bullous pemphigoid with cerebrovascular disease and dementia: a case–control study. Arch Dermatol. 2010;146:1251–1254. doi:10.1001/archdermatol.2010.322

42. Brick K, Weaver CH, Savica R, et al. A population-based study of the association between bullous pemphigoid and neurologic disorders. J Am Acad Dermatol. 2014;71:1191–1197. doi:10.1016/j.jaad.2014.07.052

43. Casas-de-la-asuncion E, Ruano-Ruiz J, Rodriguez-Martin AM, et al. Association between bullous pemphigoid and neurologic diseases: a case–control study in Portuguese patients. Actas Dermosifiliogr. 2014;105:860–865. doi:10.1016/j.ad.2014.04.013

44. Langan SM, Groves RW, West J. The relationship between neurological disease and bullous pemphigoid: a population based case–control study. J Invest Dermatol. 2011;131:631–636. doi:10.1038/jid.2010.357

45. Bastuji-Garin S, Joly P, Lemordant P, et al. Risk factors for bullous pemphigoid in the elderly. A prospective case–control study. J Invest Dermatol. 2011;131:637–643. doi:10.1038/jid.2010.301

46. Texeira VB, Cabral R, Brites MM, et al. Bullous pemphigoid and comorbidities: a case–control study in Portuguese patients. An Bras Dermatol. 2014;89:274–279. doi:10.1590/abd1806-4841.20142516

47. Chevalier V, Barbe C, Reguiai Z, et al. Impact of neurological diseases on the prognosis of bullous pemphigoid: a retrospective study of 178 patients [in French]. Ann Dermatol Ve´ne´re´ol. 2016;143:179–186. doi:10.1016/j.annder.2015.12.016

48. Seppanen A, Suuronen T, Hofmann SC, et al. Distribution of collagen XVII in the human brain. Brain Res. 2007;1158:50–56. doi:10.1016/j.brainres.2007.04.073

49. Lai YC, Yew YW, Lambert WC. Bullous pemphigoid and its association with neurological diseases: a systematic review and meta-analysis. J Eur Acad Dermatol Venereol. 2016;30:2007–2015. doi:10.1111/jdv.13660

50. Kirtschig G, Middleton P, Bennett C, et al. Interventions for bullous pemphigoid. Cochrane Database Syst Rev. 2010;(10):CD002292. doi:10.1002/14651858.CD002292.pub3

51. Stravopoulos PG, Soura E, Antoniou C. Drug-induced pemphigoid. J Eur Acad Dermatol. 2014;28:1133–1140. doi:10.1111/jdv.12366

52. Vassilieva S. Drug-induced pemphigoid: bullous and cicatricial. Clin Dermatol. 1998;16:379–387. doi:10.1016/S0738-081X(98)00008-X

53. Bastuji-Garin S, Joly P, Picard-Dahan C, et al. Drugs associated with bullous pemphigoid. A case–control study. Arch Dermatol. 1996;132:272–276. doi:10.1001/archderm.1996.03890270044006

54. Bene J, Moulis G, Bennani I, et al. Bullous pemphigoid and dipeptidyl peptidase IV inhibitors: a case-noncase study in the French Pharmacovigilance database. Br J Dermatol. 2016;175(2):296–301. doi:10.1111/bjd.14601

55. Lloyd-Lavery A, Chi CC, Wojnarowska F, Taghipour K. The associations between bullous pemphigoid and drug use: a UK case–control study. JAMA Dermatol. 2013;149:58–62. doi:10.1001/2013.jamadermatol.376

56. Ruocco E, Wolf R, Caccavale S, et al. Bullous pemphigoid: associations and management guidelines: facts and controversies. Clin Dermatol. 2013;31:400–412. doi:10.1016/j.clindermatol.2013.01.007

57. Taylor G, Venning V, Wojnarowska F, et al. Bullous pemphigoid and autoimmunity. J Am Acad Dermatol. 1993;29:181–184. doi:10.1016/0190-9622(93)70164-O

58. Oh DD, Zhao CY, Murrell DF. A review of case–control studies on the risk factors for the development of autoimmune blistering diseases. J Eur Acad Dermatol. 2016;30:595–603. doi:10.1111/jdv.13386

59. Delgado JC, Turbay D, Yunis EJ, et al. A common major histocompatibility complex class II allele HLA-DQB1* 0301 is present in clinical variants of pemphigoid. Proc Natl Acad Sci USA. 1996;93:8569–8571. doi:10.1073/pnas.93.16.8569

60. Setterfield J, Theron J, Vaughan RW, et al. Mucous membrane pemphigoid: HLA-DQB1*0301 is associated with all clinical sites of involvement and may be linked to antibasement membrane IgG production. Br J Dermatol. 2001;145:406–414. doi:10.1046/j.1365-2133.2001.04380.x

61. Chan LS, Hammerberg C, Cooper KD. Significantly increased occurrence of HLADQB1*0301 allele in patients with ocular cicatricial pemphigoid. J Invest Dermatol. 1997;108:129–132. doi:10.1111/1523-1747.ep12332352

62. Zakka LR, Keskin DB, Reche P, Ahmed AR. Relationship between target antigens and major histocompatibility complex (MHC) class II genes in producing two pathogenic antibodies simultaneously. Clin Exp Immunol. 2010;162:224–236. doi:10.1111/j.1365-2249.2010.04239.x

63. Okazaki A, Miyagawa S, Yamashina Y, Kitamura W, Shirai T. Polymorphisms of HLADR and -DQ genes in Japanese patients with bullous pemphigoid. J Dermatol. 2000;27:149–156. doi:10.1111/j.1346-8138.2000.tb02141.x

64. Drouet M, Delpuget-Bertin N, Vaillant L, et al. HLA-DRB1 and HLA-DQB1 genes in susceptibility and resistance to cicatricial pemphigoid in French Caucasians. Eur J Dermatol. 1998;8:330–333.

65. Büdinger L, Borradori L, Yee C, et al. Identification and characterization of autoreactive T cell responses to bullous pemphigoid antigen 2 in patients and healthy controls. J Clin Invest. 1998;102:2082–2089. doi:10.1172/JCI3335

66. Joly P, Courville P, Lok C, et al. Clinical criteria for the diagnosis of bullous pemphigoid: a reevaluation according to immunoblot analysis of patient sera. Dermatology. 2004;208:16–20. doi:10.1159/000075040

67. Rychlik-Sych M, Barańska M, Wojtczak A, et al. The impact of the CYP2D6 gene polymorphism on the risk of pemphigoid. Int J Dermatol. 2015;54:1396–1401. doi:10.1111/ijd.12967

68. Hirose M, Schilf P, Benoit S, et al. Polymorphisms in the mitochondrially encoded ATP synthase 8 gene are associated with susceptibility to bullous pemphigoid in the German population. Exp Dermatol. 2015;24:715–717. doi:10.1111/exd.12732

69. Weisenseel P, Martin S, Partsch K, Messer G, Prinz JC. Relevance of the low-affinity type of the Fcgamma-receptor iiia-polymorphism in bullous pemphigoid. Arch Dermatol Res. 2007;299:163–164. doi:10.1007/s00403-007-0755-8

70. Grando SA. The mitochondrion is a common target of disease pathophysiology in pemphigus and pemphigoid. Exp Dermatol. 2015;24:655–656. doi:10.1111/exd.12772

71. Bernard P, Antonicelli F. Bullous pemphigoid: a review of its diagnosis, associations and treatment. Am J Clin Dermatol. 2017;18(4):513–528. doi:10.1007/s40257-017-0264-2

72. Yayli S, Pelivani N, Beltraminelli H, et al. Detection of linear ige deposits in bullous pemphigoid and mucous membrane pemphigoid: a useful clue for diagnosis. Br J Dermatol. 2011;165:1133–1137. doi:10.1111/j.1365-2133.2011.10481.x

73. Joly P, Roujeau JC, Benichou J, et al. A comparison of two regimens of topical corticosteroids in the treatment of patients with bullous pemphigoid: a multicenter randomized study. J Invest Dermatol. 2009;129:1681–1687. doi:10.1038/jid.2008.412

74. Murrell DF, Daniel BS, Joly P, et al. Definitions and out come measures for bullous pemphigoid: recommendations by an international panel of experts. J Am Acad Dermatol. 2012;66:479–485. doi:10.1016/j.jaad.2011.06.032

75. Joly P, Roujeau JC, Benichou J, et al. A comparison of oral and topical corticosteroids in patients with bullous pemphigoid. N Engl J Med. 2002;346:321–327. doi:10.1056/NEJMoa011592

76. Levy-Sitbon C, Barbe C, Plee J, et al. Assessment of bullous pemphigoid disease area index during treatment: a prospective study of 30 patients. Dermatology. 2014;229:116–122. doi:10.1159/000362717

77. Feliciani C, Joly P, Jonkman MF, et al. Management of bullous pemphigoid: the European Dermatology Forum consensus in collaboration with the European Academy of Dermatology and Venereology. Br J Dermatol. 2015;172(4):867–877. doi:10.1111/bjd.13717

78. Eming R, Sticherling M, Hofmann SC, et al. S2k guidelines for the treatment of pemphigus vulgaris/foliaceus and bullous pemphigoid. J Dtsch Dermatol Ges. 2015;13:833–844. doi:10.1111/ddg.12606

79. Vaillant L, Bernard P, Joly P, et al. Evaluation of clinical criteria for diagnosis of bullous pemphigoid. Arch Dermatol. 1998;134:1075–1080. doi:10.1001/archderm.134.9.1075

80. Bernard P, Vaillant L, Labeille B, et al. Incidence and distribution of subepidermal autoimmune bullous skin diseases in three French regions. Bullous Diseases French Study Group. Arch Dermatol. 1995;131:48–52. doi:10.1001/archderm.1995.01690130050009

81. Zillikens D, Wever S, Roth A, et al. Incidence of autoimmune subepidermal blistering dermatoses in a region of central Germany. Arch Dermatol. 2005;131:957–958. doi:10.1001/archderm.131.8.957

82. Schmidt E, Della Torre R, Borradori L. Clinical features and practical diagnosis of bullous pemphigoid. Clin Dermatol. 2011;29:427–438. doi:10.1016/j.det.2011.03.010

83. Lipsker D, Borradori L. Bullous pemphigoid: what are you? Urgent need of definitions and diagnostic criteria. Dermatology. 2010;221:131–134. doi:10.1159/000316104

84. Della Torre R, Combescure C, Cortes B, et al. Clinical presentation and diagnostic delay in bullous pemphigoid: a prospective nationwide cohort. Br J Dermatol. 2012;167:1111–1117. doi:10.1111/j.1365-2133.2012.11108.x

85. Di Zenzo G, Grosso F, Terracina M, et al. Characterization of the anti-BP180 autoantibody reactivity profile and epitope mapping in bullous pemphigoid patients. J Invest Dermatol. 2004;122:103–110. doi:10.1046/j.0022-202X.2003.22126.x

86. Sárdy M, Kostaki D, Varga R, Peris K, Ruzicka T. Comparative study of direct and indirect immunofluorescence and of bullous pemphigoid 180 and 230 enzyme-linked immunosorbent assays for diagnosis of bullous pemphigoid. J Am Acad Dermatol. 2013;69:748–753. doi:10.1016/j.jaad.2013.07.009

87. Vodegel RM, Jonkman MF, Pas HH, de Jong MC. U-serrated immunodeposition pattern differentiates type VII collagen targeting bullous diseases from other subepidermal bullous autoimmune diseases. Br J Dermatol. 2004;151:112–118. doi:10.1111/j.1365-2133.2004.06006.x

88. Terra JB, Meijer JM, Jonkman MF, Diercks GFH. The n- vs. U-serration is a learnable criterion to differentiate pemphigoid from epidermolysis bullosa acquisita in direct immunofluorescence serration-pattern analysis. Br J Dermatol. 2013;169:100–105. doi:10.1111/bjd.12308

89. Gammon WR, Kowalewski C, Chorzelski TP, et al. Direct immunofluorescence studies of sodium chloride-separated skin in the differential diagnosis of bullous pemphigoid and epidermolysis bullosa acquisita. J Am Acad Dermatol. 1990;22:664–670. doi:10.1016/0190-9622(90)70094-X

90. Zillikens D, Rose PA, Balding SD, et al. Tight clustering of extracellular BP180 epitopes recognized by bullous pemphigoid autoantibodies. J Invest Dermatol. 1997;109:573–579. doi:10.1111/1523-1747.ep12337492

91. Bernard P, Bedane C, Prost C, et al. Bullous pemphigoid. Guidelines for the diagnosis and treatment. Centres de référence des maladies bulleuses auto-immunes. Société Française de Dermatologie [in French]. Ann Dermatol Venereol. 2011;138:247–251. doi:10.1016/j.annder.2011.01.009

92. Charneux J, Lorin J, Vitry F, et al. Usefulness of BP230 and BP180-NC16a enzyme-linked immunosorbent assays in the initial diagnosis of bullous pemphigoid: a retrospective study of 138 patients. Arch Dermatol. 2011;147:286–291. doi:10.1001/archdermatol.2011.23

93. Di Zenzo G, Thoma-Uszynski S, Calabresi V, et al. Demonstration of epitope-spreading phenomena in bullous pemphigoid: results of a prospective multicenter study. J Invest Dermatol. 2011;131:2271–2280. doi:10.1038/jid.2011.180

94. Gammon WR, Fine JD, Forbes M, et al. Immunofluorescence on split skin for the detection and differentiation of basement membrane zone autoantibodies. J Am Acad Dermatol. 1992;27:79–87. doi:10.1016/0190-9622(92)70161-8

95. Kelly SE, Wojnarowska F. The use of chemically split tissue in the detection of circulating anti-basement membrane zone antibodies in bullous pemphigoid and cicatricial pemphigoid. Br J Dermatol. 1988;118:31–40. doi:10.1111/j.1365-2133.1988.tb01747.x

96. Baaker C, Terra JB, Jonkman MF, Jonkman MF. Bullous pemphigoid as pruritus in the elderly. A common presentation. JAMA Dermatol. 2014;149:950–953.

97. Schmidt E, Obe K, Bröcker EB, Zillikens D. Serum levels of autoantibodies to BP180 correlate with disease activity in patients with bullous pemphigoid. Arch Dermatol. 2000;136:174–178. doi:10.1001/archderm.136.2.174

98. Bernard P, Reguiai Z, Tancrede-Bohin E, et al. Risk factors for relapse in patients with bullous pemphigoid in clinical remission: a multicenter, prospective, cohort study. Arch Dermatol. 2009;145:537–542. doi:10.1001/archdermatol.2009.53

99. Otten JV, Hashimoto T, Hertl M, Payne AS, Sitaru C. Molecular diagnosis in autoimmune skin blistering conditions. Curr Mol Med. 2014;14:69–95. doi:10.2174/15665240113136660079

100. Wieland CN, Comfere NI, Gibson LE, Weaver AL, Krause PK, Murray JA. Anti bullous pemphigoid 180 and 230 antibodies in a sample of unaffected subjects. Archdermatol. 2010;146:21–25.

101. Hofmann SC, Tamm K, Hertl M, Borradori L. Diagnostic value of an enzyme-linked immunosorbent assay using BP180 recombinant proteins in elderly patients with pruritic skin disorders. Br J Dermatol. 2003;149:910–912. doi:10.1046/j.1365-2133.2003.05603.x

102. Giudice GJ, Wilske KC, Anhalt GJ, et al. Development of an ELISA to detect anti-BP180 autoantibodies in bullous Bullous Pemphigoid pemphigoid and herpes gestationis. J Invest Dermatol. 1994;102:878–881. doi:10.1111/1523-1747.ep12382738

103. Kobayashi M, Amagai M, Kuroda-Kinoshita K, et al. BP180 ELISA using bacterial recombinant NC16a protein as a diagnostic and monitoring tool for bullous pemphigoid. J Dermatol Sci. 2002;30:224–232. doi:10.1016/S0923-1811(02)00109-3

104. Zillikens D, Mascaro JM, Rose PA, et al. A highly sensitive enzyme-linked immunosorbent assay for the detection of circulating anti-BP180 autoantibodies in patients with bullous pemphigoid. J Invest Dermatol. 1997;109:679–683. doi:10.1111/1523-1747.ep12338088

105. Sakuma-Oyama Y, Powell AM, Oyama N, et al. Evaluation of a BP180- NC16a enzyme-linked immunosorbent assay in the initial diagnosis of bullous pemphigoid. Br J Dermatol. 2004;151:126–131. doi:10.1111/j.1365-2133.2004.06082.x

106. Tsuji-Abe Y, Akiyama M, Yamanaka Y, et al. Correlation of clinical severity and ELISA indices for the NC16A domain of BP180 measured using BP180 ELISA kit in bullous pemphigoid. J Dermatol Sci. 2005;37:145–149. doi:10.1016/j.jdermsci.2004.10.007

107. Thoma-Uszynski S, Uter W, Schwietzke S, et al. BP230- and BP180-specific auto-antibodies in bullous pemphigoid. J Invest Dermatol. 2004;122:1413–1422. doi:10.1111/j.0022-202X.2004.22603.x

108. Barnadas MA, Rubiales MV, Gonza´lez MJ, et al. Enzyme linked immunosorbent assay (ELISA) and indirect immunofluorescence testing in a bullous pemphigoid and pemphigoid gestationis. Int J Dermatol. 2008;47:1245–1249. doi:10.1111/j.1365-4632.2008.03824.x

109. Kromminga A, Sitaru C, Hagel C, et al. Development of an ELISA for the detection of autoantibodies to BP230. Clin Immunol. 2004;111:146–152. doi:10.1016/j.clim.2003.12.007

110. Daniel BS, Borradori L, Hall RP, Murrell DF. Evidence-based management of bullous pemphigoid. Dermatol Clin. 2011;29:613–620. doi:10.1016/j.det.2011.06.003

111. Le Sache´-de Peufeilhoux L, Ingen-Housz-Oro S, Hue S, et al. The value of BP230 enzyme-linked immunosorbent assay in the diagnosis and immunological follow-up of bullous pemphigoid. Dermatology. 2012;224:154–159. doi:10.1159/000337545

112. Tampoia M, Lattanzi V, Zucano A, et al. Evaluation of a new ELISA assay for detection of BP230 autoantibodies in bullous pemphigoid. Ann NY Acad Sci. 2009;1173:15–20. doi:10.1111/j.1749-6632.2009.04630.x

113. Roussel A, Benichou J, Randriamanantany ZA, et al. Enzyme linked immunosorbent assay for the combination of bullous pemphigoid antigens 1 and 2 in the diagnosis of bullous pemphigoid. Arch Dermatol. 2011;147:293–298. doi:10.1001/archdermatol.2011.21

114. Giudice GJ, Emery DJ, Diaz LA. Cloning and primary structural analysis of the bullous pemphigoid autoantigen BP180. J Invest Dermatol. 1992;99:243–250. doi:10.1111/1523-1747.ep12616580

115. Stanley JR, Tanaka T, Mueller S, et al. Isolation of complementary DNA for bullous pemphigoid antigen by use of patients’ autoantibodies. J Clin Invest. 1988;82:1864–1870. doi:10.1172/JCI113803

116. Bernard P, Didierjean L, Denis F, et al. Heterogeneous bullous pemphigoid antibodies: detection and characterization by immunoblotting when absent by indirect immunofluorescence. J Invest Dermatol. 1989;92:171–175. doi:10.1111/1523-1747.ep12276689

117. Labib RS, Anhalt GJ, Patel HP, et al. Molecular heterogeneity of the bullous pemphigoid antigens as detected by immunoblotting. J Immunol. 1986;136:1231–1235.

118. Gohestani R, Kanitakis J, Nicolas JF, et al. Comparative sensitività of indirect immunofluorescence to immunoblot assay for the detection of circulating antibodies to bullous pemphigoid antigens 1 and 2. Br J Dermatol. 1996;135:74–79. doi:10.1111/j.1365-2133.1996.tb03611.x

119. Bernard P, Aucouturier P, Denis F, et al. Immunoblot analysis of IgG subclasses of circulating antibodies in bullous pemphigoid. Clin Immunol Immunopathol. 1990;54:484–494. doi:10.1016/0090-1229(90)90060-4

120. Bernard P, Antonicelli F, Bedane C, et al. Prevalence and clinical significance of anti-Laminin 332 autoantibodies detected by a novel enzyme-linked immunosorbent assay in mucous membrane pemphigoid. JAMA Dermatol. 2013;149:533–540. doi:10.1001/jamadermatol.2013.1434

121. Chan LS, Ahmed AR, Anhalt GJ, et al. The first International consensus on mucous membrane pemphigoid: definition, diagnostic criteria, pathogenic factors, medical treatment, and prognostic indicators. Arch Dermatol. 2002;138:370–379. doi:10.1001/archderm.138.3.370

122. Schmidt E, Skrobek C, Kromminga A, et al. Cicatricial pemphigoid: IgA and IgG autoantibodies target epitopes on both intra- and extracellular domains of bullous pemphigoid antigen 180. Br J Dermatol. 2001;145:778–783. doi:10.1046/j.1365-2133.2001.04471.x

123. Ghohestani RF, Nicolas JF, Rousselle P, Claudy AL. Diagnostic value of indirect immunofluorescence on sodium chloride-split skin in differential diagnosis of subepidermal autoimmune bullous dermatoses. Arch Dermatol. 1997;133:1102–1107. doi:10.1001/archderm.1997.03890450048006

124. Oyama N, Setterfield JF, Powell AM, et al. Bullous pemphigoid antigen II (BP180) and its soluble extracellular domains are major autoantigens in mucous membrane pemphigoid: the pathogenic relevance to HLA class II alleles and disease severity. Br J Dermatol. 2006;154:90–98. doi:10.1111/j.1365-2133.2005.06998.x

125. Yeh SW, Usman AQ, Ahmed AR. Profile of autoantibody to basement membrane zone proteins in patients with mucous membrane pemphigoid: long-term follow up and influence of therapy. Clin Immunol. 2004;112:268–272. doi:10.1016/j.clim.2004.04.010

126. Ludwig R. Clinical presentation, pathogenesis, diagnosis, and treatment of epidermolysis bullosa acquisita. ISRN Dermatol. 2013;2013:812029. doi:10.1155/2013/812029

127. Bruch-Gerharz D, Hertl M, Ruzicka T. Mucous membrane pemphigoid: clinical aspects, immunopathological features and therapy. Eur J Dermatol. 2007;17:191–200. doi:10.1684/ejd.2007.0148

128. Komorowski L, Mu¨ller R, Vorobyev A. Sensitive and specific assays for routine serological diagnosis of epidermolysis bullosa acquisita. J Am Acad Dermatol. 2013;68:e89–95. doi:10.1016/j.jaad.2011.12.032

129. Chen M, Kim GH, Prakash I, Woodley DT. Epidermolysis bullosa acquisita: autoimmunity against to anchoring fibril collagen. Autoimmunity. 2012;45:91–101. doi:10.3109/08916934.2011.606450

130. De Jong MC, Bruins S, Heeres K, et al. Bullous pemphigoid and epidermolysis bullosa acquisita. Differentiation by fluorescence overlay antigen mapping. Arch Dermatol. 1996;132:151–157. doi:10.1001/archderm.1996.03890260053008

131. Yaoita H, Briggaman RA, Lawley TH, Provost TT, Katz SI. Epidermolysis bullosa acquisita: ultrastructural and immunological studies. J Invest Dermatol. 1981;76:288–292. doi:10.1111/1523-1747.ep12526124

132. Danichi T, Kurono S, Ohyama B, et al. Anti-laminin gamma-1 pemphigoid. Proc Natl Acad Sci USA. 2009;106:2800–2805. doi:10.1073/pnas.0809230106

133. Meijer JM, Lamberts A, Pas HH, Jonkman MF. Significantly higher prevalence of circulating bullous pemphigoid-specific IgG autoantibodies in elderly patients with a non-bullous skin disorder. Br J Dermatol. 2015;173:1274–1276. doi:10.1111/bjd.13874

134. Van Beek N, Dohse A, Riechert F, et al. Serum autoantibodies against the dermal-epidermal junction in patients with chronic pruritic disorders, elderly individuals and blood donors prospectively recruited. Br J Dermatol. 2014;170:943–947. doi:10.1111/bjd.12739

135. Rieckoff-Cantoni L, Bernard P, Didierjean L, et al. Frequency of bullous pemphigoid-like antibodies as detected by Western blotting in pruritic dermatoses. Arch Dermatol. 1992;128:791–794. doi:10.1001/archderm.1992.01680160075007

136. Yang A, Xuan R, Murrell DF. A new indirect immunofluorescence BIOCHIP method for the serological diagnosis of bullous pemphigoid: a review of literature. Australas J Dermatol. 2019;60(3):e173–e177. doi:10.1111/ajd.13034

137. Damoiseaux J, van Rijsingen M, Warnemunde N, et al. Autoantibody detection in bullous pemphigoid: clinical evaluation of the EUROPLUSTM Dermatology Mosaic. J Immunol Methods. 2012;382:76–80. doi:10.1016/j.jim.2012.05.007

138. Ozkesici B, Mutlu D, Donmez L, et al. The value of the BIOCHIP mosaic-based indirect immunofluorescence technique in the diagnosis of pemphigus and bullous pemphigoid in Turkish patients. Acta Dermatovenerol Croat. 2017;25:202–209.

139. Gornowicz-Porowska J, Seraszek-Jaros A, Bowszyc-Dmochowska M, et al. Accuracy of molecular diagnostics in pemphigus and bullous pemphigoid: comparison of commercial and modified mosaic indirect immunofluorescence tests as well as enzyme-linked immunosorbent assays. Postepy Dermatol Alergol. 2017;34:21–27. doi:10.5114/ada.2017.65617

140. Zarian H, Saponeri A, Michelotto A, et al. Biochip technology for the serological diagnosis of bullous pemphigoid. ISRN Dermatol. 2012;2012:237802. doi:10.5402/2012/237802

141. van Beek N, Rentzsch K, Probst C, et al. Serological diagnosis of autoimmune bullous skin diseases: prospective comparison of the BIOCHIP mosaic-based indirect immunofluorescence technique with the conventional multi-step single test strategy. Orphanet J Rare Dis. 2012;7:49. doi:10.1186/1750-1172-7-49

142. Ko MJ, Chu CY. Topical tacrolimus therapy for localized bullous pemphigoid. Br J Dermatol. 2003;149:1079–1081. doi:10.1111/j.1365-2133.2003.05611.x

143. Yamauchi PS, Lowe NJ, Gindi V. Treatment of coexisting bullous pemphigoid and psoriasis with the tumour necrosis factor antagonist etanercept. J Am Acad Dermatol. 2006;54:S121–2. doi:10.1016/j.jaad.2005.10.055

144. Kasperkiewicz M, Shimanovitch I, Ludwig R, et al. Rituximab for treatment-refractory pemphigus and pemphigoid. J Am Acad Dermatol. 2011;65:552–558. doi:10.1016/j.jaad.2010.07.032

145. Schmidt E, Seitz CS, Benoit S, et al. Rituximab in autoimmune bullous diseases: mixed responses and adverse effects. Br J Dermatol. 2007;156:352–356. doi:10.1111/j.1365-2133.2006.07646.x

146. Cai SC, Lim YL, Li W, et al. Anti-BP180 NC16A titres as an indicator of disease activity and outcome in Asian with bullous pemphigoid. Ann Acad Med Singap. 2015;44:119–126.

147. Fichel F, Barbe C, Joly P, et al. Clinical and immunologic factors associated with bullous pemphigoid relapse during the first year of treatment: a multicenter, prospective study. JAMA Dermatol. 2014;150:25–33. doi:10.1001/jamadermatol.2013.5757

148. Ingen-Housz-Oro S, Plee J, Belmondo T, et al. Positive direct immunofluorescence is of better value than ELISA-BP180 and ELISA-BP230 values for the prediction of relapse after treatment cessation in bullous pemphigoid: a retrospective study of 97 patients. Dermatology. 2015;231:50–55. doi:10.1159/000381143

149. Izumi T, Ichiki Y, Esaki C, Kitajima Y. Monitoring of ELISA for anti-BP180 antibodies: clinical and therapeutic analysis of steroid-treated patients with bullous pemphigoid. J Dermatol. 2004;31(5):383–391. doi:10.1111/j.1346-8138.2004.tb00689.x

150. Swerlick RA, Korman NJ. Bullous pemphigoid: what is the prognosis? J Invest Dermatol. 2004;122(5):XVII–XVIII. doi:10.1111/j.0022-202X.2004.22538.x

Creative Commons License © 2022 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.