Alopecia Areata: a Comprehensive Review of Pathogenesis and Management

Alopecia areata is a common hair loss condition that is characterized by acute onset of non-scarring hair loss in usually sharply defined areas ranging from small patches to extensive or less frequently diffuse involvement. Depending on its acuity and extent, hair loss is an important cause of anxiety and disability. The current understanding is that the condition represents an organ-specific autoimmune disease of the hair follicle with a genetic background. Genome-wide association studies provide evidence for the involvement of both innate and acquired immunity in the pathogenesis, and mechanistic studies in mouse models of alopecia areata have specifically implicated an IFN-γ-driven immune response, including IFNγ, IFNγ-induced chemokines and cytotoxic CD8 T cells as the main drivers of disease pathogenesis. A meta-analysis of published trials on treatment of alopecia areata states that only few treatments have been well evaluated in randomized trials. Nevertheless, depending on patient age, affected surface area and disease duration, an empiric treatment algorithm can be designed with corticosteroids and topical immunotherapy remaining the mainstay of therapy. The obviously limited success of evidence-based therapies points to a more important complexity of hair loss. At the same time, the complexity of pathogenesis offers opportunities for the development of novel targeted therapies. New treatment opportunities based on the results of genome-wide association studies that implicate T cell and natural killer cell activation pathways are paving the way to new approaches in future clinical trials. Currently, there are ongoing studies with the CTLA4-Ig fusion protein abatacept, anti-IL15Rβ monoclonal antibodies and the Janus kinase inhibitors tofacitinib, ruxolitinib and baricitinib. Ultimately, the options available for adapting to the disease rather than treating it in an effort to cure may also be taken into consideration in selected cases of long-standing or recurrent small spot disease.

Similar content being viewed by others

Alopecia Areata: Current Treatments and New Directions

Article 22 August 2023

Alopecia Areata

Chapter © 2023

Alopecia Areata

Chapter © 2017

Avoid common mistakes on your manuscript.

Introduction

Alopecia areata (AA) is a common hair loss condition with a lifetime prevalence of approximately 2% [1] that is characterized by acute onset of non-scarring hair loss in usually sharply defined areas [2]. Some patients lose hair in only a small patch, while others have more extensive or less frequently diffuse involvement [3]. Any hair-bearing area can be affected, but the most noticeable are the scalp, the beard area and the eyebrows.

History

Hair is a power symbol, as the Biblical Samson teaches us in Judges 16. Hair falling out in dreams is interpreted as loss of power and control. The hair loss dream is typically a manifestation of a stressful situation. Ultimately, hair dreams represent a situation that is beyond one’s control. The same type of helpless feeling is experienced in waking life when hair is suddenly lost.

AA has obviously existed at all times, and is referred to by Roman encyclopaedist Aulus Cornelius Celsus (25 bc –50 ad ) in his celebrated treatise on medicine (“De Medicina”) in the chapter “De capillis fluentibus”, or can be spotted in great works of art such as in Francisco Goya’s (1774–1828) “Il Matrimonio” in the Museo del Prado in Madrid, Spain.

Epidemiology

Depending on its acuity and extent, hair loss is obviously an important cause of anxiety and disability. At any given time, AA is found in 0.1 to 0.2% of the population [4], with all ethnic backgrounds appearing to be similarly affected, and no difference between the sexes [1]. The onset of AA is before the age of 40 years in 70–80% of those affected; however, a significant proportion (48%) will present with the condition during their first and second decades, making AA the most common cause of hair loss in otherwise healthy children [5]. General practitioners are often called upon as primary care takers, but often are not familiar with the condition and may fail to provide patients with the appropriate management or to refer them to the dermatologist.

Clinical Presentation

The most frequent clinical presentation of AA is in single or multiple patches. The characteristic patch of AA is usually round or oval, and is completely bald and smooth. Occasionally, AA may progress to complete baldness, which is referred to as alopecia (areata) totalis (AT). When the entire body suffers from complete hair loss, it is referred to as alopecia (areata) universalis (AU). Ophiasis is a form of AA characterized by the loss of hair in the shape of a wave at the circumference of the head (Fig. 1), and was originally referred to by Celsus. Its name derives from the ancient Greek word for snake (“ophis”), because of the snake-like shape of the pattern of hair loss.

The skin within the patches is inconspicuous, and hair follicles are intact. Toward the periphery of the area of active hair loss, one can usually note characteristic, smaller 3- to 4-mm exclamation mark hairs and/or black dots (Fig. 2a). They basically represent dystrophic hairs that have fractured tips due to an inhibition of cell division in the hair matrix at the level of the hair bulb. Severe inhibition leads to fracturing of the hair shaft before emergence from the scalp and then appears as a black dot (or cadaverized hair). The exclamation mark appearance describes a damaged strand of hair that is thicker at its distal end and notably thinner proximally as it enters the scalp. Exclamation mark hairs typically are found in acute forms of AA, and are not seen in patients with long-standing areas of hair loss. The latter rather presents with yellow dots within the affected scalp. Visualized at a higher magnification (×10) than the naked eye with help of a simple handheld dermatoscope (Heine Delta®, DermoGenius®, DermLite PRO HR® or DermLite DL3®), they present as quite monomorphous, though variably sized, yellow, round dots that are devoid of hair or contain vellus hair (Fig. 2b). They represent dilated follicular infundibula that contain sebaceous and keratinous material [6].

Hair recovery in AA usually occurs as a uniform process with thin white hair emerging first, followed by healthy pigmented hair. At times, it may occur in a concentric targetoid hair re-growth pattern [7], or in tufts of white hair [8].

Acute Diffuse and Total Alopecia of the Female Scalp

In 2002, Sato-Kawamura et al. [21, 22] suggested naming a peculiar type of inflammatory non-cicatricial alopecia that is characterized by marked female predominance and a uniquely short clinical course acute diffuse and total alopecia of the female scalp. It is basically identical with a subtype of AA presenting with diffuse hair loss as originally proposed in the German literature by Braun-Falco and Zaun 40 years earlier in 1962 [23]. Diffuse alopecia areata or alopecia areata incognita, yet another synonymous designation proposed by Rebora in 1987 [24], represents an uncommon variety of AA characterized by diffuse hair shedding in the absence of typical patches.

Marie Antoinette and Thomas More Syndrome

Marie Antoinette syndrome designates the condition in which scalp hair suddenly turns white [25]. The name alludes to the French Queen Marie Antoinette (1755–1793), whose hair allegedly turned white the night before her last walk to the guillotine during the French Revolution. Although the actual incidence is rare, this stigmatizing phenomenon has captured the storytellers’ imagination like few other afflictions as a sign of grave sorrow. The first documented case of sudden hair whitening can be traced back to the Talmud (83 ad ), which describes a scholar who was appointed chief of the main Israeli Talmudic academy at the young age of 17 years. Upon his wife’s concern that he looked too youthful for the position, he developed 18 rows of white hair [26].

History also records that the hair of the English martyr Sir Thomas More (1478–1535) turned white overnight in the Tower of London before his execution [27]. Thomas More was beheaded in 1535 after having refused to sign the Act of Supremacy that declared King Henry VIII of England Supreme Head of the Church of England over the Pope. More modern accounts refer to the turning white of hair in survivors of bomb attacks during World War II [28, 29].

Although Marie Antoinette was originally chosen as eponym for the syndrome, Thomas More who turned white in 1535 ought to have the right of seniority over the Queen of France who succumbed to the same fate only in 1793. Since there seems to be no other particular reason for favouring Marie Antoinette over Thomas More, it seems logical to use the term Marie Antoinette syndrome for the condition afflicting women and Thomas More syndrome for men.

Today, the syndrome is interpreted as an acute episode of diffuse AA in which the very sudden whitening of hair is caused by the preferential loss of pigmented hairs. As early as 1901, immunology research pioneer and Nobel Prize Laureate (1908) in Medicine for his work on phagocytosis, Elie Metchnikoff (1845–1916), believed that whitening of hair was related to pigment absorbed and carried away by phagocytes that he observed in the vicinity of dystrophic hair follicles and called pigmentophages [30]. In 1981, Guin et al. [31] reported a lady whose hair turned white over 3 months with hair thinning but no patches of alopecia. Immunofluorescence studies demonstrated immune deposits along the follicular epithelium, suggesting an underlying immunological mechanism. Finally, case reports of sudden whitening of hair in association with easily plucked hairs, exclamation mark hairs and patchy alopecia typical for AA, and in co-existence with other autoimmune phenomena, such as vitiligo, supported the immune theory [32]. Ultimately, these observations have led some authorities to speculate that the autoimmune target in AA may be related to the hair follicle melanin pigment system [33]. McBride and Bergfeld [34] described mosaic hair colour changes in two patients with extensive AA, and suggested that these processes may result from localized immunological reactions against hair follicle melanocytes in a multifocal pattern with subsequent changes in hair colour. In contrast to total whitening of hair, this mechanism would account better for the pattern of 18 rows of white hair originally described in the Talmud.

Why Henry IV of France’s Hair Did Not Turn White on St. Bartholomew’s Day

Although early accounts of overnight whitening of hair are substantiated by more recent case reports in the scientific literature, it may be suspected that the phenomenon has been used both as a literary stylistic means in fiction, with the aim of dramatizing, and in historical accounts with the claim of authenticity [35]. Scrutinizing the case of Henry III of Navarra (later King Henry IV of France), whose hair allegedly turned white overnight on the occasion of the horrors of St. Bartholomew’s Day massacre in 1572, Navarini and Trüeb [36] found inconsistencies between Henry’s biography and the current pathophysiological understanding of the condition. Since a pre-requisite for overnight whitening of hair would be that the individual in question has a salt-and-pepper pattern with pigmented hairs interspersed with white hairs and the pigmented hairs selectively shed while the white hairs remain, it seems quite improbable that Henry III of Navarre’s hair should have turned white so rapidly, as he was only 19 years old at the time in question.

Autoimmune Pathogenesis

Today, AA is considered an organ-specific autoimmune disease of the hair follicle with a genetic background.

Genetic factors have been implicated with the reports of familial incidence of AA in a range between 10 and 27% [37,38,39]. Among first-degree relatives of 348 severely affected patients, van der Steen et al. [40] found that one of the parents was affected in 7%. Among the sibs, 3% had AA, while AA was present in 2% of the children. Ultimately, Martinez-Mir et al. [41] suggested that AA fits the pattern of a complex genetic trait based on its prevalence in the population of approximately 2%, a tenfold increased risk for first-degree relatives of affected individuals with no clear Mendelian pattern of inheritance within affected families, a concordance in monozygotic twins of 55% [42] and a Gaussian distribution of severity that ranges from patchy localized hair loss on the scalp to the complete absence of hair on the body. With regard to the genetic susceptibility to AA and the disease severity, Price and Colombe [43] originally suggested several genes associated with the human leukocyte antigen (HLA) class II alleles: HLA-DQ3 appeared to be the general susceptibility allele for AA, while patients with long-standing disease patterns, namely long-term patchy AA and long-term AT or AU, could be differentiated by their particular HLA associations with HLA antigens DR4, DR11 and DQ7.

The hair follicle is an immune-privileged site, with low levels of major histocompatibility complex (MHC) expression [44]. It is assumed that AA results from a breakdown in immune privilege with the subsequent assault on the follicle on the level of the bulb by CD8 + T lymphocytes [45]. T lymphocytes have been shown to be oligoclonal and autoreactive [46], and are predominantly present in the peribulbar inflammatory infiltrate. The peribulbar lymphocytic infiltrate induces hair follicle keratinocytes to undergo apoptosis that results in inhibition of cell division within the hair matrix and of hair shaft production. Depending on the severity of inflammation and apoptosis, the net result is a telogen-type effluvium and/or dystrophic anagen effluvium.

Notably, AA occurs in association with other autoimmune diseases, such as autoimmune thyroid disease, in between 8 and 27% [37, 47,48,49,50], and vitiligo in between 4 and 9% [2, 47, 51, 52] depending on author, and with respective circulating anti-thyroid and anti-parietal cell autoantibodies [50, 53, 54]. Finally, autoantibodies to follicular components have been detected. Tobin et al. [55] tested 39 AA sera and 27 control sera by Western immunoblotting for antibodies to 6 M urea-extractable proteins of normal anagen scalp hair follicles. At serum diluted 1:80, all AA subjects (100%), while only 44% of the control individuals had antibodies directed to one or more antigens of approximately 57, 52, 50, 47 or 44 kD. The incidence of antibodies to individual hair follicle antigens in AA was up to seven times more frequent than in control sera and their level up to 13 times greater and was statistically significant for all five antigens. Tissue specificity analysis indicated that these antigens were selectively expressed in hair follicles. Candidate autoantigens that have been identified include the 44/46 kDa hair-specific keratin (expressed in the precortical zone of anagen hair follicles) and trichohyalin (an important intermediate filament-associated protein) expressed in the inner root sheath of the growing hair follicle [56].

Ultimately, studies have demonstrated that AA scalp skin grafted on to nude mice with severe combined immunodeficiency grow hair and that infiltrating lymphocytes in the graft are lost, while it is possible to induce AA in human scalp explants on these mice by injecting T lymphocytes with scalp homogenate [57].

The C3H/HeJ mouse is an inbred laboratory strain that spontaneously develops an adult-onset disease that resembles adult-onset alopecia areata in humans. Sundberg et al. [58] identified 4 genetic susceptibility loci on mouse chromosomes 8, 9, 15 and 17, wherein the chromosome 17 locus corresponds to HLA orthologs. C3H/HeJ mice with hair loss have been shown to have circulating antibodies to hair follicles similar to those present in humans with AA [59], corroborating that these mice represent a fitting model for human AA, and supporting the hypothesis that AA results from an abnormal autoimmune response to the hair follicle [59]. However, to what extent B cells are involved in the pathogenesis of AA, and whether antibodies to hair follicle-specific proteins [56] contribute to disease or only represent an autoimmune epiphenomenon, remains to be elucidated.

Increased reactive oxygen species and high cellular stress level have been identified in AA [60,61,62], and studied in its relationship with disease severity, duration, recurrence and pattern [63]. Antioxidant/oxidant balance perturbation is a common feature in autoimmune, emotional and environmental stress. Whether these changes play a role in pathogenesis of AA or again only result from the inflammatory process requires further investigation.

Genome-wide association studies performed by Petukhova et al. [64] demonstrated an association with genomic regions containing several genes controlling the activation and proliferation of regulatory T cells, cytotoxic T lymphocyte-associated antigen 4 (CTLA4), interleukin (IL)-2/IL-21, IL-2 receptor A (IL-2RA; CD25) and Eos (also known as Ikaros family zinc finger 4 (IKZF4)), as well as the HLA region. Furthermore, they found a region of strong association within the cytomegalovirus UL16 binding protein (ULBP) gene cluster on chromosome 6q25.1, encoding activating ligands of the natural killer cell receptor NKG2D. By probing the role of ULBP-3 in disease pathogenesis, they showed that its expression in lesional scalp from patients with AA was markedly upregulated in the hair follicle dermal sheath during active disease. This study provides evidence for the involvement of both innate and acquired immunity in the pathogenesis of AA. Furthermore, the authors have defined the genetic underpinnings of AA, placing it within the context of shared pathways among autoimmune diseases, and implicating a novel disease mechanism, the upregulation of ULBP ligands, in triggering autoimmunity.

The possibility that cytomegalovirus (CMV) may be involved in the pathogenesis of AA was originally proposed by Skinner et al. [65] after detection of CMV DNA sequences using polymerase chain reaction in scalp biopsies of AA patients; however, this hypothesis was not confirmed by subsequent authors [66,67,68].

A recent genome-wide meta-analysis in AA resolved the HLA associations and revealed two new susceptibility loci. Betz et al. [69] performed a meta-analysis of research on AA by combining data from their genome-wide association studies, and replication with supplemented ImmunoChip data for a total of 3253 cases and 7543 controls. The strongest region of association is the MHC, where they fine mapped four independent effects, all implicating HLA-DR as a key etiologic driver. Outside the MHC, they identified two novel loci that exceed the threshold of statistical significance. Candidate susceptibility gene expression analysis in these regions demonstrated expression in relevant immune cells and the hair follicle. These findings uncover new molecular pathways disrupted in AA, including autophagy/apoptosis, transforming growth factor beta/Tregs and Janus kinase (JAK) signalling.

Notably, mechanistic studies in the mouse models of AA have specifically implicated an interferon (IFN)γ-driven immune response, including IFNγ, IFNγ-induced chemokines and cytotoxic CD8 T cells as the main drivers of disease pathogenesis [70].

Finally, Fischer et al. [71] investigated the contribution of copy number variants (CNVs) to AA, a prominent class of genomic variants involved in autoimmune disorders. Again, a genome-wide and a candidate gene focused CNV analysis were performed in a cohort of 585 AA patients and 1340 controls of Central European origin. A nominally significant association with AA was found in the genes MCHR2 and MCHR2-AS1, implicated in melanin-concentrating hormone (MCH) signalling. This might indicate a relationship between AA, pigmentation and MCH signalling, consistent with the originally perplexing phenomena of 18 rows of white hair growing in the Talmud scholar, the Marie Antoinette and Thomas More syndromes, and, in general, why the hair of AA patients frequently shows a change in colour to white upon re-growing after an acute episode of AA.

Co-morbidities

For centuries, physicians propagated the viability of a complex approach in the diagnosis and treatment of disease, while today, we stress on specification. This brought up the question of how to wholly evaluate the state of an individual who suffers from a number of conditions simultaneously. Ultimately, Alvan R. Feinstein came out in 1970 with the concept of co-morbidity, which has been defined as presence of one or more additional diseases co-occurring with a primary disease; or the effect of such additional diseases, whereby the additional disorder may also be a behavioural or mental disorder [72].

The interrelations of disease, age and drug pathomorphism greatly affect the clinical presentation and progress of the primary nosology, character and severity of complications, impair the patient’s life quality and impede the diagnostic and remedial process. The presence of co-morbidity must therefore be taken into account when selecting the algorithm of diagnosis and treatment plans for any given, including trichological disease. Ultimately, the dermatologist participates with the other medical disciplines in the diagnosis and treatment of all types of hair problems as they relate to systemic disease.

While AA occurs in approximately 0.1–0.2% of the general population, it is found in approximately 9% of the patients with Down syndrome [73].

Since AA represents an autoimmune disease, it is not surprising that an association exists with other autoimmune conditions [47,48,49,50,51,52,53,54]. Moreover, more recently additional co-morbid conditions have been identified that may have a disease-modifying effect and therefore must be included in the treatment plan (Table 1).

The art of using corticosteroids for treatment of AA is maximizing efficacy and minimizing toxicity.

Single patches of AA are best treated with intralesional triamcinolone acetonide (ITA) in a concentration between 2.5 (eyebrows and beard area) and a maximum of 10 mg/ml (scalp), depending on the localization, by jet injector [135] or insulin syringe (frontal and temporal regions, eyebrows, beard area) at 4–6-week intervals, as long as needed (Fig. 5a–c). ITA has proven to have a better efficacy in the treatment of localized AA than topical treatment, either with betamethasone valerate [136, 137] or with tacrolimus [137]. Since patchy AA is the most prevalent form of the disease, and ITA is the most frequently practiced treatment for this condition, Samrao et al. [138] conducted a study on patients with AA treated in this way for at least 20 months to evaluate the effect on bone mineral density using dual-energy x-ray absorptiometry (DXA). Fifty percent of the patients had abnormal DXA results. Patients with the following risk factors were more likely to be affected: age older than 50 years, body mass index less than 18.5 kg/m 2 , lack of weight-bearing exercise, smoking history, postmenopausal status, past stress fracture, family history of osteopenia or osteoporosis and a cumulative ITA dose of greater than 500 mg.

In an attempt to circumvent side effects related to the use of corticosteroids, Chu et al. [139] evaluated the benefit of different concentrations (2.5, 5 and 10 mg/ml) of ITA in AA, and did not find any difference in re-growth of hair between the different concentrations, enabling injection of larger surfaces at lower concentrations and lesser cumulative doses of triamcinolone acetonide.

Acute and widespread AA (>30% surface area) are best treated with systemic corticosteroid therapy, either orally or intravenously (Fig. 6a, b). Again, in an attempt to circumvent side effects, pulsed administration has been proposed for treatment of AA: Sharma [140] originally proposed 300 mg oral prednisolone pulses at 4-week intervals, for a minimum of four doses or until cosmetically acceptable hair growth was obtained. 58.3% of the patients with widespread AA showed cosmetically acceptable hair growth. Response was evident on average after 2 to 3 months of therapy. In a subsequent study, Sharma and Muralidhar [141] reported efficacy and safety of monthly oral corticosteroid pulse therapy also for treatment of young patients (up to 18 years of age), including children. Children aged less than 12 years received betamethasone sodium phosphate as soluble equivalent to prednisolone 5 mg/kg body weight every month. Side effects were minimal and included transient giddiness, headache and epigastric burning. Subsequently, Kar et al. [142] confirmed in the first placebo-controlled study the usefulness of oral prednisolone pulse therapy in AA. Agarwal et al. [143] alternatively suggested twice weekly 5 mg betamethasone oral pulse therapy on two consecutive days per week for a total duration of 12 weeks.

To determine the effectiveness of intravenous pulse therapy, much in the same manner as for treatment of other autoimmune diseases, Friedli et al. [144] originally performed an open prospective study of patients with rapid and extensive hair loss (>30% scalp area) for less than 1 year (first occurrence or relapse). Two hundred fifty-milligramme intravenous methylprednisolone (IV-MPPT) was administered twice a day on three consecutive days. A single series of IV-MPPT was well tolerated and appeared to be effective in patients with rapidly progressing extensive multifocal AA, but not those with ophiasic and AU. Subsequently, Nakajima [145] confirmed the efficacy of IV-MPPT in a larger study of patients aged >15 years with AA. With IV-MPPT (500 mg methylprednisolone on three consecutive days, in 3 cycles, 4 weeks apart) within 6 months of disease onset, remission rates were 88% for multilocular AA with surface area 50% and 21.4% in AT/U. Performed after 6 months of disease onset, the remission rate decreased to 15.8%. Im et al. [146] studied the outcome and prognostic factors in patients with severe AA treated with IV-MPPT on three consecutive days. All of the patients had rapid and extensive hair loss with the bald area exceeding 50% of the scalp. Seventy percent showed terminal hair growth, and 41.4% complete recovery with acceptable cosmetic results. The prognostic factors that influenced successful outcome of IV-MPPT for AA were disease duration before treatment in relation to the type of AA. A good response was obtained for all types of AA with a duration of 3 months or less before treatment, and for the multifocal type of AA, with a duration of

Smith et al. [148] investigated the outcome of IV-MPPT in severe childhood AA with short disease duration. Five patients had AT/U and 13 had multilocular AA. All patients underwent 2 or 3 cycles of IV-MPPT at monthly intervals (maximum 500 mg/day on three consecutive days). Within 7 months of last IV-MPPT session, 10 of 18 children had good response, with eight showing improvement within the first 4 months. Seven of the initial 10 good responders experienced relapses, with an estimated median time to relapse of 8 months. Therefore, even early in the course of disease, IV-MPPT did not convincingly affect the long-term outcome of severe AA in the children age group. IV-MPPT was generally well tolerated. One patient experienced transient mood changes during the first cycle; three patients complained of a metallic taste during the infusions. Acne vulgaris was aggravated in two teenage patients.

For treatment of widespread and long-standing (severe and refractory) AA, Tosti et al. [149] evaluated topical clobetasol. With 0.05% topical clobetasol ointment under occlusion (Saran wrap) on six consecutive nights per week, over 6 months re-growth of hair was achieved in AT/U in 17.8% (Fig. 7a, b). Negative prognostic features for treatment success were positive family history for AA, first manifestation of disease before the age of 10 years and association with atopic disease or autoimmune thyroid disease. Folliculitis and acne were the frequent side effects. Despite positive clinical results obtained using clobetasol ointment with occlusive dressing, this approach has a low patient compliance, especially in patients with residual hair. Therefore, in a subsequent study performed again by Tosti et al. [150], 0.05% clobetasol foam was applied twice daily on five consecutive days per week and proved to also be effective, safe and well tolerated, with good cosmetic acceptance and patient compliance (Fig. 8a, b). Folliculitis occurred in a smaller proportion of patients. No significant modifications in cortisol and ACTH blood levels were observed.

Topical immunotherapy with allergic contact sensitizers has been used from the 1970s to treat dermatological conditions assumed to result from an altered immunological state. Dinitrochlorobenzene (DNCB) is a synthetic compound that works as a haptene by combining with proteins to become a complete antigen resulting in an allergic contact dermatitis upon sensitization, and has predominantly been used for the study of experimental allergic contact dermatitis in animals. The use of DNCB to treat extensive AA was originally reported to be successful in 1976 based on two cases in whom half of the bare scalp was treated successfully [151]. The underlying mode of action is hypothesized to be related to factors inherent to the late phase of allergic contact dermatitis. These supposedly modulate the T cell-mediated mechanisms underlying AA, including involvement of cytokines and growth factors, such as IL1 beta [152], while the therapeutic effect mediated by contact sensitizers is assumed to be mediated by counteracting pro-inflammatory cytokines such as TNF-alpha, IL-10 or transforming growth factor (TGF)-beta 1 [153].

Due to mutagenicity in the Ames test, DNCB was substituted with squaric acid dibutyl ester (SADBE) by Happle et al. [154] who originally tested the compound dissolved in acetone and applied weekly to one side of the head, the other side serving as control. In 46 of 53 patients with extensive or total AA treated in this manner (87%), hair re-grew either exclusively on the treated side or considerably faster and denser on this side. In some patients, continuous treatment failed to maintain the response. Persistent response was observed in 37 patients (70%).

At present, yet another contact sensitizer, diphenylcyclopropenone (DPCP), is considered as the agent of choice for topical immunotherapy of AA [155]. DPCP is more stable in acetone and is relatively cheaper than SADBE [156]. Nevertheless, in a comparative study of SADBE and DPCP for the treatment of AA, SABDE proved to be more efficacious and to have lower frequencies of side effects than DPCP [157]. Again, Happle et al. [158] introduced the use of DPCP for topical immunotherapy of AA, and their positive results were soon confirmed by other authors [159, 160]. In a retrospective study of 68 patients with severe AA (>40% scalp hair loss) treated for at least 5 months with topical DPCP, Pericin and Trüeb [161] found an overall response rate of 70.6% with 30.9% complete remission and 39.7% partial remission (Fig. 9a–c). Among the investigated prognostic factors for the outcome of DPCP therapy, the extent of hair loss before therapy was the main predictor for therapeutic success: total remission rates for multilocular AA were 43.8%, for subtotal AA and ophiasis 33.3% and for AT/U 21.4%, irrespective of disease duration. A long period of therapy is required and may increase the percentage of responders, especially in AT/U. Finally, DPCP therapy is associated with a high relapse rate. Therefore, maintenance therapy is recommended to reduce the risk of relapse. Subsequent authors confirmed that topical immunotherapy with DPCP is effective for treatment of widespread and long-standing AA, and is usually well tolerated [162,163,164,165,166,167,168,169,170]. Its efficacy has also been demonstrated in children [171, 172]. Addition of oral fexofenadine, an H1-receptor antagonist, may partially inhibit the itch of contact dermatitis induced by DCP in patients with alopecia areata [173]. Moreover, Inui et al. [174] suggested that oral fexofenadine may enhance the efficacy of contact immunotherapy for extensive AA in patients with an atopic background. Poor accessibility, high cost of the compound and poor patient compliance are major drawbacks of topical immunotherapy of AA.

In an analogy to other lymphocyte-mediated autoimmune diseases, Joly [175] proposed the use of methotrexate (MTX) alone or in combination with low-dose oral corticosteroids for the treatment of AT/U with an overall success rate of 64%. Best results are achieved with subcutaneous MTX at the maximal dosage of 30 mg weekly in combination with 20 mg oral prednisone daily: re-growth of hair begins within 2 to 4 months of this regimen (Fig. 10a–f). Lasting improvement required continuous treatment in most cases, though with a significant lower dosage of MTX after gradually tapering the oral corticosteroid, once hair re-growth has set in [176]. Ultimately, drug toxicities are to be carefully weighed out against treatment benefit, and therapeutic drug monitoring following the respective guidelines is strongly recommended.

Other Therapies

The original report of successful treatment of a case of AU with subcutaneous efalizumab by Kaelin et al. [177] could subsequently not be confirmed by a 3- to 6-month placebo-controlled trial with efalizumab in a cohort of 62 patients with moderate-to-severe AA [178]. Efalizumab is a recombinant humanized monoclonal antibody designed to treat autoimmune diseases by binding to the CD11a subunit of lymphocyte function-associated antigen 1 and inhibiting lymphocyte activation and cell migration out of blood vessels into tissues. Efalizumab was associated with fatal brain infections [179] and therefore withdrawn from the market in 2009.

Namazi [180] originally proposed the use of statins for treatment of a variety of dermatologic conditions characterized by ingress of activated leucocytes into the skin, including AA. The 3-hydroxy-3-methylglutaryl co-enzyme A reductase inhibitors (statins) are currently used for reducing atherogenesis and cardiovascular morbidity, but there is increasing evidence that they may also have immunomodulatory activities. Robins [181] reported the original indicator case of hair re-growth in a patient with AU following initiation of simvastatin and ezetimibe therapy. Later, Ali and Martin [182] reported two additional patients with treatment-refractory AA that benefited from treatment with a combination of ezetimibe and simvastatin, in addition to the continuation of intralesional corticosteroid injections. In this report, the putative immunomodulatory effects of statins in relation to the known pathophysiology underlying AA are discussed. Lattouf et al. [183] reported a case series of 29 AA patients with 40–70% scalp involvement treated with simvastatin/ezetimibe 40 mg/10 mg daily. Nineteen completed 24 weeks of treatment, and 14 of 19 were judged responders. Upon completion of the initial 24 weeks of treatment, the responders were randomized into a group of seven patients who continued treatment for an additional 24 weeks, or into a group of seven patients who stopped medication. In the former group, five of seven continued with hair growth or had stable disease, while in the latter group, five of seven patients relapsed. These findings could not be reproduced by subsequent authors who failed to demonstrate efficacy of simvastatin/ezetimibe in 20 [184] respectively 12 patients with AA [185].

Therefore, as a general rule, single case observations or limited case series of successful treatment of a condition like AA, where spontaneous remissions may occur, need to be confirmed by large placebo-controlled trials.

Topical anthralin is a contact irritant that has traditionally been used in the treatment of AA for decades. Unlike DPCP, it creates an irritant versus allergic contact dermatitis of the scalp, and its mode of action in AA is therefore less sophisticated. Nevertheless, small studies have demonstrated results in 25–75% of the patients depending again on the severity of involvement [186, 187]. The treatment is predominantly used in children as an alternative to corticosteroids due to its low risks. A major drawback is the risk of permanent purple discoloration of textiles and bathtubs when used at home.

Topical minoxidil is a hair growth-promoting agent predominantly used for treatment of androgenetic alopecia. In contrast to corticosteroids and immunomodulatory agents, which act on inflammation, minoxidil acts mainly to promote hair growth, and is therefore used in combination with other treatment modalities, such as topical anthralin [188]. Olsen et al. suggested that the use of topical minoxidil may help reducing hair loss after tapering corticosteroids for treatment of AA [189]. It seems reasonable to add topical minoxidil when androgenetic alopecia represents a co-morbidity of AA.

Since cyclosporine (CyS) inhibits the activation of helper T cells that are pathogenic in AA, Gupta et al. [190] originally treated patients with AA with oral CyS 6 mg/kg/day for 12 weeks. Three patients had AU, one had AT and two had patchy AA of the scalp. Hair re-growth in the scalp of all patients occurred between the second and the fourth week of therapy, followed by hair re-growth of the face and body. Overall, the site of best response was the scalp. Cosmetically acceptable terminal hair re-growth on the scalp occurred in three of six patients. Significant hair loss, however, occurred in all patients within 3 months of discontinuation of cyclosporine treatment. Clinical response correlated with changes in immune cell infiltration of the hair follicles. The degree of terminal hair re-growth correlated significantly with decreases in follicular epithelial human lymphocyte antigen-DR and intercellular adhesion molecule-1 expression, T cells, helper/inducer (CD4) T cells, suppressor/cytotoxic (CD8) T cells and Langerhans cells (CD1+DR+) from the hair follicles during cyclosporine therapy. Moreover, hypertrichosis is one of the common side effects of orally administered CyS, encouraging investigators to use the drug in the treatment of AA, but the reports on this subject have been controversial [191]. More recently, combination regimens of oral CyS with low-dose corticosteroids have been found to be effective for treatment of severe AA [192]. Nevertheless, long-term toxicities and risks linked to immunosuppression limit the usefulness of CyS for treatment of AA. Moreover, comparison of IV-MPPT with combination therapy using CyS with low-dose corticosteroid demonstrated that IV-MPPT may be the better treatment option, at least in patients with severe AA of the multipatch type [193].

Other therapies that have not established themselves or lost favour in the treatment of AA include thymopentin [194, 195], isoprinosine [196,197,198], fumaric acid esters [199, 200], phototherapy (PUVA) [201, 202] and hypnotherapy [203].

Finally, platelet-rich plasma (PRP) is blood plasma that has been enriched with platelets. As a concentrated source of autologous platelets, PRP contains and releases through degranulation several growth factors and cytokines. These include platelet-derived growth factor, TGF-beta, fibroblast growth factor, insulin-like growth factors 1 and 2, vascular endothelial growth factor, epidermal growth factor EGF, IL-8 and keratinocyte growth factor KGF. PRP has gained popularity among a limited number of dermatologists with a primary commercial interest, though the use and clinical validation of the method for diverse dermatologic conditions is still in progress. Results of basic science and preclinical trials have yet to be confirmed in large-scale controlled clinical trials. A single, double-blind, placebo- and active-controlled, half-head study to evaluate the effects of PRP on AA has been performed [204]: 45 patients with AA were randomized to receive intralesional injections of PRP, ITA or placebo on one half of their scalp. The other half was not treated. Three treatments were given for each patient, with intervals of 1 month. The end points were hair re-growth, hair dystrophy as measured by dermoscopy, burning or itching sensation and cell proliferation as measured by Ki-67 evaluation. Patients were followed for 1 year. PRP was found to increase hair re-growth significantly and to decrease hair dystrophy and burning or itching sensation compared with ITA or placebo. Ki-67 levels, which served as markers for cell proliferation, were significantly higher with PRP. No side effects were noted during treatment. However, in an era, where a more comprehensive understanding of the immunologic basis of AA has opened the venue to more targeted treatments, the proposal of PRP with its poorly defined mode of action rather represents an intellectual step backwards.

Targeted Therapies

New treatment opportunities based on the results of genome-wide association studies that implicate T cell and natural killer cell activation pathways are paving the way to new approaches in future clinical trials for AA. Currently, there are ongoing studies with the CTLA4-Ig fusion protein abatacept (blocks co-stimulation of T cells), anti-IL15Rβ monoclonal antibodies (block activation of CD8 + T cells) [205] and JAK inhibitors (block signal transduction at the IL-15 receptor) [206, 207].

Craiglow and King [208] originally reported hair growth in a patient with AU treated with the JAK 1 and 3 inhibitor tofacitinib for plaque psoriasis. Xing et al. [209] subsequently reported successful treatment of three patients with AA with the oral JAK 1 and 2 inhibitor ruxolitinib. The patients achieved near-complete hair re-growth within a few months, suggesting the potential clinical utility of JAK inhibition for treatment of AA. Jabbari et al. [210] reported reversal of AA in a patient treated with the oral JAK 1 and 2 inhibitor baracitinib for chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE) syndrome, which represents yet another condition characterized by prominent IFN signatures. Ultimately, Liu et al. [211] conducted a retrospective study of patients age 18 years or older with AA with at least 40% scalp hair loss treated with tofacitinib. The primary end point was the percent change in the Severity of Alopecia Tool (SALT) score during treatment. Ninety patients met inclusion criteria. Of 65 potential responders to therapy, defined as those with alopecia totalis or alopecia universalis with duration of current episode of disease of 10 years or less or alopecia areata, 77% achieved a clinical response, with 58% of the patients achieving greater than 50% change in SALT score over 4 to 18 months of treatment. Patients with AA experienced a higher percent change in SALT score than did patients with AT/U (81.9 vs 59.0%). Tofacitinib was well tolerated, and there were no serious adverse events. In an attempt to evaluate the benefit and adverse effects of the tofacitinib in a series of adolescent patients with AA, Craiglow et al. [212] reviewed the records of 13 adolescent patients aged 12–17 years with AA treated with tofacitinib. Nine patients experienced clinically significant hair re-growth with a median percent change in SALT score of 93% at an average of 6.5 months of treatment. Adverse events were mild.

Nevertheless, at this time point, affordability of the drug for long-term treatment, sustainability of treatment result [213] and liability of the physician prescribing the drug off label [214] are major obstacles to the treatment of AA with JAK inhibitors, unless patients are enrolled in ongoing clinical studies with either of JAK inhibitors.

Concluding Remarks

In general, any treatment chosen for AA should fulfil the following criteria: remission rates superior to the spontaneous remission rates of AA, proof of efficacy in half side treatment of AT/U and a good safety profile with minimal toxicity. Depending on patient age, surface area, disease duration and co-morbidities, an individual treatment plan can be designed that is successful in a significant proportion of patients (Fig. 4). Figures 5, 6, 7, 8, 9, 10, 11 and 12 illustrate successful treatments of AA following the current lines of therapy. The complexity of its pathogenesis offers opportunities for the development of novel targeted therapies for AA. Ultimately, the options available for adapting to the disease rather than treating it in an effort to cure may also be taken into consideration in selected cases of long-standing or recurrent small spot disease.

Abbreviations

Cytotoxic T lymphocyte-associated antigen 4

Ikaros family zinc finger 4

UL16 binding protein

Copy number variants

Melanine concentrating hormone

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy-syndrome

Autoimmune regulator gene

Tumour necrosis factor

Intralesional triamcinolone acetonide

Dual-energy x-ray absorptiometry

Transforming growth factor

Squaric acid dibutyl ester

Severity of Alopecia Tool

References

  1. Safavi KH, Muller SA, Suman VJ, Moshell AN, Melton LJ III. (1995) Incidence of alopecia areata in Olmsted County, Minnesota, 1975 through 1989. Mayo Clin Proc 70(7):628–633 ArticleCASPubMedGoogle Scholar
  2. Muller SA, Winkelmann RK (1963) Alopecia areata. An evaluation of 736 patients. Arch Dermatol 88:290–297 ArticleCASPubMedGoogle Scholar
  3. Finner AM (2011) Alopecia areata: clinical presentation, diagnosis, and unusual cases. Dermatol Ther 24(3):348–354 ArticlePubMedGoogle Scholar
  4. Safavi K (1992) Prevalence of alopecia areata in the First National Health and Nutrition Examination Survey. Arch Dermatol 128(5):702 ArticleCASPubMedGoogle Scholar
  5. Kyriakis KP, Paltatzidou K, Kosma E, Sofouri E, Tadros A, Rachioti E (2009) Alopecia areata prevalence by gender and age. J Eur Acad Dermatol Venereol 23(5):572–573 ArticleCASPubMedGoogle Scholar
  6. Inui S, Nakajima T, Nakagawa K, Itami S (2008) Clinical significance of dermoscopy in alopecia areata: analysis of 300 cases. Int J Dermatol 47(7):688–693 ArticlePubMedGoogle Scholar
  7. Delorenze LM, Gavazzoni-Dias MFR, Teixeira MS, Aide MK (2016) Concentric polycyclic regrowth pattern in alopecia areata. Int J Trichol 8(1):35–37 ArticleGoogle Scholar
  8. Elston DM, Clayton AS, Meffert JJ, McCollough ML (2000) Migratory poliosis: a forme fruste of alopecia areata? J Am Acad Dermatol 42(6):1076–1077 ArticleCASPubMedGoogle Scholar
  9. Tosti A, Morelli R, Bardazzi F, Peluso AM (1994) Prevalence of nail abnormalities in children with alopecia areata. Pediatr Dermatol 11(2):112–115 ArticleCASPubMedGoogle Scholar
  10. Sharma VK, Dawn G, Muralidhar S, Kumar B (1998) Nail changes in 1000 Indian patients with alopecia areata. J Eur Acad Dermatol Venereol 10(2):189–191 ArticleCASPubMedGoogle Scholar
  11. Kasumagic-Halilovic E, Prohic A (2009) Nail changes in alopecia areata: frequency and clinical presentation. J Eur Acad Dermatol Venereol 23(2):240–241 ArticleCASPubMedGoogle Scholar
  12. Samman P (1979) Trachyonychia (rough nails). Br J Dermatol 101(6):701–705 ArticleCASPubMedGoogle Scholar
  13. Tosti A, Fanti PA, Morelli R, Bardazzi F (1991) Trachyonychia associated with alopecia areata: a clinical and pathologic study. J Am Acad Dermatol 25(2 Pt 1):266–270 ArticleCASPubMedGoogle Scholar
  14. Tosti A, Bardazzi F, Piraccini BM, Fanti PA, Cameli N, Pileri S (1995) Is trachyonychia, a variety of alopecia areata, limited to the nails? J Invest Dermatol 104(5 Suppl):27S–28S ArticleCASPubMedGoogle Scholar
  15. Dotz WI, Lieber CD, Vogt PJ (1985) Leukonychia punctata and pitted nails in alopecia areata. Arch Dermatol 121(11):1452–1454 ArticleCASPubMedGoogle Scholar
  16. Shelley WB (1980) The spotted lunula. A neglected nail sign associated with alopecia areata. J Am Acad Dermatol 2(5):385–387 ArticleCASPubMedGoogle Scholar
  17. Bergner T, Donhauser G, Ruzicka T (1992) Red lunulae in severe alopecia areata. Acta Derm Venereol 72(3):203–205 CASPubMedGoogle Scholar
  18. Tosti A, Pazzaglia M, Venturi M, Chiacchio NC Jr (2013) Severe onycholysis in a card illusionist with alopecia areata universalis. Int J Trichol 5(2):81–82 ArticleGoogle Scholar
  19. Horn RT Jr, Odom RB (1980) Twenty-nail dystrophy of alopecia areata. Arch Dermatol 116(5):573–574 ArticlePubMedGoogle Scholar
  20. De Waard-van der Spek FB, Oranje AP, De Raeymaecker DM, Peereboom-Wynia JD (1989) Juvenile versus maturity-onset alopecia areata—a comparative retrospective clinical study. Clin Exp Dermatol 14(6):429–433 ArticlePubMedGoogle Scholar
  21. Sato-Kawamura M, Aiba S, Tagami H (2002) Acute diffuse and total alopecia of the female scalp. A new subtype of diffuse alopecia areata that has a favorable prognosis. Dermatology 205(4):367–373 ArticlePubMedGoogle Scholar
  22. Lew BL, Shin MK, Sim WY (2009) Acute diffuse and total alopecia: a new subtype of alopecia areata with a favorable prognosis. J Am Acad Dermatol 60(1):85–93 ArticlePubMedGoogle Scholar
  23. Braun-Falco O, Zaun H (1962) Über die Beteiligung des gesamten Capillitiums bei Alopecia areata. Hautarzt 13:342–348 Google Scholar
  24. Rebora A (1987) Alopecia areata incognita: a hypothesis. Dermatologica 174(5):214–218 ArticleCASPubMedGoogle Scholar
  25. Navarini AA, Nobbe S, Trüeb RM (2009) Marie Antoinette syndrome. Arch Dermatol 145(6):65 ArticleGoogle Scholar
  26. Goldenhersh MA (1992) Rapid whitening of the hair first reported in the Talmud: possible mechanisms of this intriguing phenomenon. Am J Dermatopathol 14:367–368 ArticleCASPubMedGoogle Scholar
  27. Trüeb RM, Navarini AA (2010) Thomas More syndrome. Dermatology 220(1):55–56 ArticlePubMedGoogle Scholar
  28. Hoffmann E (1957) Sudden turning gray of the hair caused by fright, canities subita psychogenica. Z Haut Geschlechtskr 22:74–78 CASPubMedGoogle Scholar
  29. Ephraim A (1959) On sudden or rapid whitening of the hair. AMA Arch Derm 79:228–236 ArticleCASPubMedGoogle Scholar
  30. Metchnikoff E (1901) On the process of hair turning white. Proc R Soc Lond 69:156 ArticleGoogle Scholar
  31. Guin JD, Kumar V, Petersen BH (1981) Immunofluorescence findings in rapid whitening of scalp hair. Arch Dermatol 117:576–578 ArticleCASPubMedGoogle Scholar
  32. Helm F, Milgrom H (1970) Can scalp hair suddenly turn white? A case of canities subita. Arch Dermatol 102:102–103 ArticleCASPubMedGoogle Scholar
  33. Paus R, Slominski A, Czarnetzki BM (1993) Is alopecia areata, an autoimmune response against melanogenesis-related proteins, exposed by abnormal MHC class I expression in the anagen hair bulb? Yale J Biol Med 66:541–554 CASPubMedPubMed CentralGoogle Scholar
  34. McBride AK, Bergfeld WF (1990) Mosaic hair color changes in alopecia areata. Cleve Clin J Med 157:354–356 ArticleGoogle Scholar
  35. Xavier S (1934) Sudden blanching of the hair as described in literature. Chron Med 41:116–120 Google Scholar
  36. Navarini AA, Trüeb RM (2010) Why Henry III of Navarre’s hair probably did not turn white overnight. Int J Trichol 2:2–4 ArticleGoogle Scholar
  37. Muller SA, Winkelmann RK (1963) Alopecia areata. Arch Dermatol:290–297
  38. DeWeert K, Temmerman L, Kint A (1984) Alopecia areata. A clinical study. Dermatologica 168:224–229 ArticleCASGoogle Scholar
  39. De Waard van der Spek FB, Oranje AP, De-Raeymaecker DM, Peereboom-Wynia JDR (1989) Junvenile versus maturity-onset alopecia areata: a comparative retrospective clinical study. Clin Exp Dermatol 14:429–433 ArticlePubMedGoogle Scholar
  40. an der Steen P, Traupe H, Happle R, Boezeman J, Strater R, Hamm H (1992) The genetic risk for alopecia areata in first degree relatives of severely affected patients: an estimate. Acta Derm Venerol 72:373–375 PubMedGoogle Scholar
  41. Martinez-Mir A, Zlotogorski A, Gordon D, Petukhova L, Mo J, Gilliam TC, Londono D, Haynes C, Ott J, Hordinsky M, Nanova K, Norris D, Price V, Duvic M, Christiano AM (2007) Genomewide scan for linkage reveals evidence of several susceptibility loci for alopecia areata. Am J Hum Genet 80:316–328 ArticleCASPubMedPubMed CentralGoogle Scholar
  42. Jackow C, Puffer N, Hordinsky M, Nelson J, Tarrand J, Duvic M (1998) Alopecia areata and cytomegalovirus infection in twins: genes versus environment? J Am Acad Derm 38:418–425 ArticleCASPubMedGoogle Scholar
  43. Price VH, Colombe BW (1996) Heritable factors distinguish two types of alopecia areata. Dermatol Clin 14(4):679–689 ArticleCASPubMedGoogle Scholar
  44. Paus R, Ito N, Takigawa M, Ito T (2003) The hair follicle and immune privilege. J Investig Dermatol Symp Proc 8(2):188–194 ArticlePubMedGoogle Scholar
  45. Paus R, Bertolini M (2013) The role of hair follicle immune privilege collapse in alopecia areata: status and perspectives. Investig Dermatol Symp Proc 16(1):S25–S27 ArticleCASGoogle Scholar
  46. Dressel D, Brütt CH, Manfras B, Zollner TM, Wunderlich A, Böhm BO, Boehncke W (1997) Alopecia areata but not androgenetic alopecia is characterized by a restricted and oligoclonal T-cell receptor-repertoire among infiltrating lymphocytes. J Cutan Pathol 24(3):164–168 ArticleCASPubMedGoogle Scholar
  47. Cunliffe WJ, Hall R, Newell DJ, Stevenson CJ (1968) Vitiligo, thyroid disease and autoimmunity. Br J Dermatol 80(3):135–139 ArticleCASPubMedGoogle Scholar
  48. Cunliffe WJ, Hall R, Stevenson CJ, Weightman D (1969) Alopecia areata, thyroid disease and autoimmunity. Br J Dermatol 81(12):877–881 ArticleCASPubMedGoogle Scholar
  49. Puavilai S, Puavilai G, Charuwichitratana S, Sakuntabhai A, Sriprachya-Anunt S (1994) Prevalence of thyroid diseases in patients with alopecia areata. Int J Dermatol 33(9):632–633 ArticleCASPubMedGoogle Scholar
  50. Seyrafi H, Akhiani M, Abbasi H, Mirpour S, Gholamrezanezhad A (2005) Evaluation of the profile of alopecia areata and the prevalence of thyroid function test abnormalities and serum autoantibodies in Iranian patients. BMC Dermatol 5:11 ArticlePubMedPubMed CentralGoogle Scholar
  51. Krishnaram AS, Saigal A, Adityan B (2013) Alopecia areata—vitiligo overlap syndrome: an emerging clinical variant. Indian J Dermatol Venereol Leprol. 79(4):535–537 ArticleCASPubMedGoogle Scholar
  52. Walker A, Mesinkovska NA, Boncher J, Tamburro J, Bergfeld WF (2015) Colocalization of vitiligo and alopecia areata presenting as poliosis. J Cutan Pathol 42(2):150–154 ArticlePubMedGoogle Scholar
  53. Friedmann PS (1981) Alopecia areata and auto-immunity. Br J Dermatol 105(2):153–157 ArticleCASPubMedGoogle Scholar
  54. Kumar B, Sharma VK, Sehgal S (1995) Antismooth muscle and antiparietal cell antibodies in Indians with alopecia areata. Int J Dermatol 34(8):542–545 ArticleCASPubMedGoogle Scholar
  55. Tobin DJ, Orentreich N, Fenton DA, Bystryn JC (1994) Antibodies to hair follicles in alopecia areata. J Invest Dermatol 102(5):721–724 ArticleCASPubMedGoogle Scholar
  56. Tobin DJ (2003) Characterization of hair follicle antigens targeted by the anti-hair follicle immune response. J Investig Dermatol Symp Proc 8(2):176–181 ArticleCASPubMedGoogle Scholar
  57. Gilhar A, Ullmann Y, Berkutzki T, Assy B, Kalish RS (1998) Autoimmune hair loss (alopecia areata) transferred by T lymphocytes to human scalp explants on SCID mice. J Clin Invest 101(1):62–67 ArticleCASPubMedPubMed CentralGoogle Scholar
  58. Sundberg JP, Silva KA, Li R, Cox GA, King LE (2004) Adult-onset alopecia areata is a complex polygenic trait in the C3H/HeJ mouse model. J Invest Derm 123:294–297 ArticleCASPubMedGoogle Scholar
  59. Tobin DJ, Sundberg JP, King LE Jr, Boggess D, Bystryn JC (1997) Autoantibodies to hair follicles in C3H/HeJ mice with alopecia areata-like hair loss. J Invest Dermatol 109(3):329–333 ArticleCASPubMedGoogle Scholar
  60. Koca R, Armutcu F, Altinyazar C, Gürel A (2005) Evaluation of lipid peroxidation, oxidant/antioxidant status, and serum nitric oxide levels in alopecia areata. Med Sci Monit 11(6):CR296–CR299 CASPubMedGoogle Scholar
  61. Bakry OA, Elshazly RM, Shoeib MA, Gooda A (2014) Oxidative stress in alopecia areata: a case-control study. Am J Clin Dermatol 15(1):57–64 ArticlePubMedGoogle Scholar
  62. Prie BE, Voiculescu VM, Ionescu-Bozdog OB, Petrutescu B, Iosif L, Gaman LE, Clatici VG, Stoian I, Giurcaneanu C (2015) Oxidative stress and alopecia areata. J Med Life 8:43–46 PubMedPubMed CentralGoogle Scholar
  63. Yenin JZ, Serarslan G, Yönden Z, Ulutaş KT (2015) Investigation of oxidative stress in patients with alopecia areata and its relationship with disease severity, duration, recurrence and pattern. Clin Exp Dermatol 40(6):617–621 ArticleCASPubMedGoogle Scholar
  64. Petukhova L, Duvic M, Hordinsky M, Norris D, Price V, Shimomura Y, Kim H, Singh P, Lee A, Chen WV, Meyer KC, Paus R, Jahoda CA, Amos CI, Gregersen PK, Christiano AM (2010) Genome-wide association study in alopecia areata implicates both innate and adaptive immunity. Nature 466:113–117 ArticleCASPubMedPubMed CentralGoogle Scholar
  65. Skinner RB Jr, Light WH, Leonardi C, Bale GF, Rosenberg EW (1995) A molecular approach to alopecia areata. J Invest Dermatol 104(5 Suppl):3S–4S ArticlePubMedGoogle Scholar
  66. Tosti A, La Placa M, Placucci F, Gentilomi G, Venturoli S, Zerbini M, Musiani M (1996) No correlation between cytomegalovirus and alopecia areata. J Invest Dermatol 107(3):443 ArticleCASPubMedGoogle Scholar
  67. García-Hernández MJ, Torres MJ, Palomares JC, Rodríguez-Pichardo A, Aznar J, Camacho F (1998) No evidence of cytomegalovirus DNA in alopecia areata. J Invest Dermatol 110(2):185 ArticlePubMedGoogle Scholar
  68. Offidani A, Amerio P, Bernardini ML, Feliciani C, Bossi G (2000) Role of cytomegalovirus replication in alopecia areata pathogenesis. J Cutan Med Surg 4(2):63–65 ArticleCASPubMedGoogle Scholar
  69. Betz RC, Petukhova L, Ripke S, Huang H, Menelaou A et al (2015) Genome-wide meta-analysis in alopecia areata resolves HLA associations and reveals two new susceptibility loci. Nat Commun 22(6):5966 ArticleCASGoogle Scholar
  70. Alli R, Nguyen P, Boyd K, Sundberg JP, Geiger TL (2012) A mouse model of clonal CD8 + T lymphocyte-mediated alopecia areata progressing to alopecia universalis. J Immunol 188(1):477–486 ArticleCASPubMedGoogle Scholar
  71. Fischer J, Degenhardt F, Hofmann A, Redler S, Basmanav FB, Heilmann-Heimbach S, Hanneken S, Giehl KA Wolff H, Moebus S, Kruse R, Lutz G, Blaumeiser B, Böhm M, Garcia Bartels N, Blume-Peytavi U, Petukhova L, Christiano AM, Nöthen MM, Betz R 2016. Genomewide analysis of copy number variants in alopecia areata in a Central European cohort reveals association with MCHR2. Exp Dermatol. 16
  72. Feinstain AR (1970) The pr-therapeutic classification of co-morbidity in chronic disease. J Chronic Dis 23(7):455–468 ArticleGoogle Scholar
  73. Brown AC, Olkowski ZL, McLaren JR, Kutner MH (1977) Alopecia areata and vitiligo associated with Down’s syndrome. (Letter) ArchDerm 113:1296 CASGoogle Scholar
  74. Ikeda T (1965) A new classification of alopecia areata. Dermatologica 131(6):421–445 ArticleCASPubMedGoogle Scholar
  75. Chu SY, Chen YJ, Tseng WC, Lin MW, Chen TJ, Hwang CY, Chen CC, Lee DD, Chang YT, Wang WJ, Liu HN (2011) Comorbidity profiles among patients with alopecia areata: the importance of onset age, a nationwide population-based study. J Am Acad Dermatol 65(5):949–956 ArticlePubMedGoogle Scholar
  76. Lee NR, Kim BK, Yoon NY, Lee SY, Ahn SY, Lee WS (2014) Differences in comorbidity profiles between early-onset and late-onset alopecia areata patients: a retrospective study of 871 Korean patients. Ann Dermatol 26(6):722–726 ArticlePubMedPubMed CentralGoogle Scholar
  77. Barahmani N, Schabath MB, Duvic M, National Alopecia Areata Registry (2009) History of atopy or autoimmunity increases risk of alopecia areata. J Am Acad Dermatol 61(4):581–591 ArticlePubMedGoogle Scholar
  78. Mohan GC, Silverberg JI (2015) Association of vitiligo and alopecia areata with atopic dermatitis: a systematic review and meta-analysis. JAMA Dermatol 151(5):522–528 ArticlePubMedGoogle Scholar
  79. Corazza GR, Andreani ML, Venturo N et al (1995) Celiac disease and alopecia areata: report of a new association. Gastroenterology 109(4):1333–1337 ArticleCASPubMedGoogle Scholar
  80. Bondavalli P, Quadri G, Parodi A, Rebora A (1998) Failure of gluten-free diet in celiac disease-associated alopecia areata. Acta Derm Venereol 78(4):319 ArticleCASPubMedGoogle Scholar
  81. Naveh Y, Rosenthal E, Ben-Arieh Y, Etzioni A (1999) Celiac disease-associated alopecia in childhood. J Pediatr 134(3):362–364 ArticleCASPubMedGoogle Scholar
  82. Viola F, Barbato M, Formisano M et al (1999) Reappearance of alopecia areata in a coeliac patient during an unintentional challenge with gluten. Minerva Gastroenterol Dietol 45(4):283–285 CASPubMedGoogle Scholar
  83. Bardella MT, Marino R, Barbareschi M et al (2000) Alopecia areata and coeliac disease: no effect of a gluten-free diet on hair growth. Dermatology 2:108–110 ArticleGoogle Scholar
  84. Fessatou S, Kostaki M, Karpathios T (2003) Coeliac disease and alopecia areata in childhood. J Paediatr Child Health 39(2):152–154 ArticleCASPubMedGoogle Scholar
  85. Zampetti M, Filippetti R (2008) Alopecia areata and celiac disease. G Ital Dermatol Venereol 143(2):168 CASPubMedGoogle Scholar
  86. Ertekin V, Tosun MS, Erdem T (2014) Screening of celiac disease in children with alopecia areata. Indian J Dermatol 59(3):317 ArticlePubMedPubMed CentralGoogle Scholar
  87. Kantor J, Kessler LJ, Brooks DG, Cotsarelis G (2003) Decreased serum ferritin is associated with alopecia in women. J Invest Dermatol 121(5):985–988 ArticleCASPubMedGoogle Scholar
  88. Park H, Kim CW, Kim SS, Park CW (2009) The therapeutic effect and the changed serum zinc level after zinc supplementation in alopecia areata patients who had a low serum zinc level. Ann Dermatol 21:142–146 ArticleCASPubMedPubMed CentralGoogle Scholar
  89. Bhat YJ, Manzoor S, Khan AR, Qayoom S (2009) Trace element levels in alopecia areata. Indian J Dermatol Venereol Leprol 75(1):29–31 ArticlePubMedGoogle Scholar
  90. Abdel Fattah NS, Atef MM, Al-Qaradaghi SM (2016) Evaluation of serum zinc level in patients with newly diagnosed and resistant alopecia areata. Int J Dermatol 55(1):24–29 ArticleCASPubMedGoogle Scholar
  91. Jin W, Zheng H, Shan B, Wu Y 2017. Changes of serum trace elements level in patients with alopecia areata: ameta-analysis. J Dermatol 2
  92. Yilmaz N, Serarslan G, Gokce C (2012) Vitamin D concentration are decreased in patients with alopecia areata. Vitam Trace Elem 1:105–109 ArticleCASGoogle Scholar
  93. d’Ovidio R, Vessio M, d’Ovidio FD (2013) Reduced level of 25-hydroxyvitamin D in chronic/relapsing alopecia areata. Dermatoendocrinol 5(2):271–273 ArticlePubMedPubMed CentralCASGoogle Scholar
  94. Aksu Cerman A, Sarikaya Solak S, Kivanc AI (2014) Vitamin D deficiency in alopecia areata. Br J Dermatol 170(6):1299–1304 ArticleCASPubMedGoogle Scholar
  95. Bakry OA, El Farargy SM, El Shafiee MK, Soliman A (2016) Serum vitamin D in patients with alopecia areata. Indian Dermatol Online J 7(5):371–377 ArticlePubMedPubMed CentralGoogle Scholar
  96. Delamere FM, Sladden MM, Dobbins HM, Leonardi-Bee J (2008) Interventions for alopecia areata. Cochrane Database Syst Rev 16(2):CD004413 Google Scholar
  97. Collins SM, Dominguez M, Ilmarinen T, Costigan C, Irvine AD (2006) Dermatological manifestations of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy syndrome. Br J Dermatol 154(6):1088–1093 ArticleCASPubMedGoogle Scholar
  98. Finnish-German APECED Consortium (1997) An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nat Genet 17:399–403 ArticleGoogle Scholar
  99. Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, Krohn KJE, Lalioti MD, Mullis PE, Antonarakis SE, Kawasaki K, Asakawa S, Ito F, Shimizu N (1997) Positional cloning of the APECED gene. Nat Genet 17:393–398 ArticleCASPubMedGoogle Scholar
  100. Betterle C, Greggio NA, Volpato M (1998) Autoimmune polyglandular syndrome type 1. J Clin Endocr Metab 83:1049–1055 ArticleCASPubMedGoogle Scholar
  101. Ferre EM, Rose SR, Rosenzweig SD, Burbelo PD, et al. 2016 Redefined clinical features and diagnostic criteria in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. JCI Insight 1(13)
  102. Rodriguez TA, Duvic M, National Alopecia Areata Registry (2008) Onset of alopecia areata after Epstein-Barr virus infectious mononucleosis. J Am Acad Dermatol 59(1):137–139 ArticlePubMedGoogle Scholar
  103. Chu CH, Cheng YP, Chan JY (2016) Alopecia areata after vaccination: recurrence with rechallenge. Pediatr Dermatol 33(3):e218–e219 ArticlePubMedGoogle Scholar
  104. Stewart MI, Smoller BR (1993) Alopecia universalis in an HIV-positive patient: possible insight into pathogenesis. J Cutan Pathol 20(2):180–183 ArticleCASPubMedGoogle Scholar
  105. Sereti I, Sarlis NJ, Arioglu E, Turner ML, Mican JM (2001) Alopecia universalis and Graves’ disease in the setting of immune restoration after highly active antiretroviral therapy. AIDS 15:138–140 ArticleCASPubMedGoogle Scholar
  106. Kunz M, Seifert B, Trüeb RM (2009) Seasonality of hair shedding in healthy women complaining of hair loss. Dermatology 219(2):105–110 ArticlePubMedGoogle Scholar
  107. d’Ovidio R, d’Ovidio F (1995) Recidive stagionali dell’Alopecia Areata. G Ital Dermatol Venereol 295:130–134 Google Scholar
  108. Thompson JM, Li T, Park MK, Qureshi AA, Cho E (2016) Estimated serum vitamin D status, vitamin D intake, and risk of incident alopecia areata among US women. Arch Dermatol Res 308(9):671–676 ArticleCASPubMedGoogle Scholar
  109. Koo JY, Shellow WV, Hallman CP, Edwards JE (1994) Alopecia areata and increased prevalence of psychiatric disorders. Int J Dermatol 33(12):849–845 ArticleCASPubMedGoogle Scholar
  110. García-Hernández MJ, Ruiz-Doblado S, Rodriguez-Pichardo A, Camacho F (1999) Alopecia areata, stress and psychiatric disorders: a review. J Dermatol 26(10):625–632 ArticlePubMedGoogle Scholar
  111. Trüeb RM, Cavegn B (1996) Trichotillomania in connection with alopecia areata. Cutis 58:67–70 PubMedGoogle Scholar
  112. Trüeb RM (2010) Involvement of scalp and nails in lupus erythematosus. Lupus 19(9):1078–1086 ArticlePubMedGoogle Scholar
  113. Piraccini BM, Broccoli A, Starace M, Gaspari V, D’Antuono A, Dika E, Patrizi A (2015) Hair and scalp manifestations in secondary syphilis: epidemiology, clinical features and trichoscopy. Dermatology 231(2):171–176 ArticlePubMedGoogle Scholar
  114. Ettefagh L, Nedorost S, Mirmirani P (2004) Alopecia areata in a patient using infliximab: new insights into the role of tumor necrosis factor on human hair follicles. Arch Dermatol 140:1012 ArticlePubMedGoogle Scholar
  115. Garcia Bartels N, Lee HH, Worm M, Burmester GR, Sterry W, Blume-Peytavi U (2006) Development of alopecia areata universalis in a patient receiving adalimumab. Arch Dermatol 142:1654–1655 ArticlePubMedGoogle Scholar
  116. Chaves Y, Duarte G, Ben-Said B, Tebib J, Berard F, Nicolas JF (2008) Alopecia areata universalis during treatment of rheumatoid arthritis with anti-TNF-alpha antibody (adalimumab). Dermatology 217:380 ArticleCASPubMedGoogle Scholar
  117. Pelivani N, Hassan AS, Braathen LR, Hunger RE, Yawalkar N (2008) Alopecia areata universalis elicited during treatment with adalimumab. Dermatology 216:320–323 ArticleCASPubMedGoogle Scholar
  118. Fabre C, Dereure O (2008) Worsening alopecia areata and de novo occurrence of multiple halo nevi in a patient receiving infliximab. Dermatology 216:185–186 ArticleCASPubMedGoogle Scholar
  119. Pan Y, Rao NA (2009) Alopecia areata during etanercept therapy. Ocul Immunol Inflamm 17:127–129 ArticlePubMedGoogle Scholar
  120. Kirshen C, Kanigsberg N (2009) Alopecia areata following adalimumab. J Cutan Med Surg 13:48–50 ArticlePubMedGoogle Scholar
  121. Hernández MV, Nogués S, Ruiz-Esquide V, Alsina M, Cañete JD, Sanmartí R (2009) Development of alopecia areata after biological therapy with TNF-alpha blockers: description of a case and review of the literature. Clin Exp Rheumatol 27:892–893 PubMedGoogle Scholar
  122. Doyle LA, Sperling LC, Baksh S, Lackey J, Thomas B, Vleugels RA, Qureshi AA, Velazquez EF (2011) Psoriatic alopecia/alopecia areata-like reactions secondary to anti-tumor necrosis factor-α therapy: a novel cause of noncicatricial alopecia. Am J Dermatopathol 33:161–166 ArticlePubMedGoogle Scholar
  123. Tauber M, Buche S, Reygagne P et al (2014) Alopecia areata occurring during anti-TNF therapy: a national multicenter prospective study. J Am Acad Dermatol 70:1146–1148 ArticlePubMedGoogle Scholar
  124. Tosti A, Bellavista S, Iorizzo M (2006) Alopecia areata: a long term follow-up study of 191 patients. J Am Acad Dermatol 55(3):438–441 ArticlePubMedGoogle Scholar
  125. Tan E, Tay YK, Goh CL, Chin GY (2002) The pattern and profile of alopecia areata in Singapore—a study of 219 Asians. Int J Dermatol 41(11):748–753 ArticlePubMedGoogle Scholar
  126. Yang S, Yang J, Liu JB, Wang HY, Yang Q, Gao M et al (2004) The genetic epidemiology of alopecia areata in China. Br J Dermatol 151(1):16–23 ArticleCASPubMedGoogle Scholar
  127. Goh C, Finkel M, Christos PJ, Sinha AA (2006) Profile of 513 patients with alopecia areata: associations of disease subtypes with atopy, autoimmune disease and positive family history. J Eur Acad Dermatol Venereol 20(9):1055–1060 ArticleCASPubMedGoogle Scholar
  128. Brajac I, Tkalcic M, Dragojević DM, Gruber F (2003) Roles of stress, stress perception and trait-anxiety in the onset and course of alopecia areata. J Dermatol 30(12):871–878 ArticlePubMedGoogle Scholar
  129. Güleç AT, Tanriverdi N, Dürü C, Saray Y, Akçali C (2004) The role of psychological factors in alopecia areata and the impact of the disease on the quality of life. Int J Dermatol 43(5):352–356 ArticlePubMedGoogle Scholar
  130. Manolache L, Benea V (2007) Stress in patients with alopecia areata and vitiligo. J Eur Acad Dermatol Venereol 21(7):921–928 ArticlePubMedGoogle Scholar
  131. Walker SA, Rothman S (1950) Alopecia areata. A statistical study and consideration of endocrine influences. J Invest Dermatol 14(6):403–413 ArticleCASPubMedGoogle Scholar
  132. Meachen GN, Provis FL (1912) Alopecia areata et totalis cured by pregnancy, and relapsing with the re-establishment of the menses. Proc R Soc Med 5(Dermatol Sect):152–154 CASPubMedPubMed CentralGoogle Scholar
  133. Bakkerr JH, Seelen JC, Stolte LA, Verboom E (1956) Alopecia areata. Acta Endocrinol 23(1):60–71 Google Scholar
  134. Trüeb RM 2015. The difficult hair loss patient. Guide to successful management of alopecia and related conditions. Springer International Publishing AG Switzerland : pp. 89ff
  135. Abell E, Munro DD (1973) Intralesional treatment of alopecia areata with triamcinolone acetonide by jet injector. Br J Dermatol 88:55–59 ArticleCASPubMedGoogle Scholar
  136. Devi M, Rashid A, Ghafoor R (2015) Intralesional triamcinolone acetonide versus topical betamethasone valerate in the management of localized alopecia areata. J Coll Physicians Surg Pak 25(12):860–862 PubMedGoogle Scholar
  137. Kuldeep C, Singhal H, Khare AK, Mittal A, Gupta LK, Garg A (2011) Randomized comparison of topical betamethasone valerate foam, intralesional triamcinolone acetonide and tacrolimus ointment in management of localized alopecia areata. Int J Trichol 3(1):20–24 ArticleGoogle Scholar
  138. Samrao A, Fu JM, Harris ST, Price VH (2013) Bone mineral density in patients with alopecia areata treated with long-term intralesional corticosteroids. J Drugs Dermatol 12(2):e36–e40 PubMedGoogle Scholar
  139. Chu TW, AlJasser M, Alharbi A, Abahussein O, McElwee K, Shapiro J (2015) Benefit of different concentrations of intralesional triamcinolone acetonide in alopecia areata: an intrasubject pilot study. J Am Acad Dermatol 73(2):338–340 ArticlePubMedGoogle Scholar
  140. Sharma VK (1996) Pulsed administration of corticosteroids in the treatment of alopecia areata. Int J Dermatol 35(2):133–136 ArticleCASPubMedGoogle Scholar
  141. Sharma VK, Muralidhar S (1998) Treatment of widespread alopecia areata in young patients with monthly oral corticosteroid pulse. Pediatr Dermatol 15(4):313–317 ArticleCASPubMedGoogle Scholar
  142. Kar BR, Handa S, Dogra S, Kumar B (2005) Placebo-controlled oral pulse prednisolone therapy in alopecia areata. J Am Acad Dermatol 52(2):287–290 ArticlePubMedGoogle Scholar
  143. Agarwal A, Nath J, Barua KN (2006) Twice weekly 5 mg betamethasone oral pulse therapy in the treatment of alopecia areata. J Eur Acad Dermatol Venereol 20(10):1375–1376 ArticleCASPubMedGoogle Scholar
  144. Friedli A, Labarthe MP, Engelhardt E, Feldmann R, Salomon D, Saurat JH (1998) Pulse methylprednisolone therapy for severe alopecia areata: an open prospective study of 45 patients. J Am Acad Dermatol 39:597–602 ArticleCASPubMedGoogle Scholar
  145. Nakajima T, Inui S, Itami S (2007) Pulse corticosteroid therapy for alopecia areata: study of 139 patients. Dermatology 215:320–324 ArticleCASPubMedGoogle Scholar
  146. Im M, Lee SS, Lee Y et al (2011) Prognostic factors in methylprednisolone pulse therapy for alopecia areata. J Dermatol 38(8):767–772 ArticleCASPubMedGoogle Scholar
  147. Shreberk-Hassidim R, Ramot Y, Gilula Z, Zlotogorski A (2016) A systematic review of pulse steroid therapy for alopecia areata. J Am Acad Dermatol 74(2):372–374 ArticlePubMedGoogle Scholar
  148. Smith A, Trüeb RM, Theiler M, Hauser V, Weibel L (2015) High relapse rates despite early intervention with intravenous methylprednisolone pulse therapy for severe childhood alopecia areata. Pediatr Dermatol 32(4):481–487 ArticlePubMedGoogle Scholar
  149. Tosti A, Piraccini BM, Pazzaglia M, Vincenzi C (2003) Clobetasol propionate 0.05% under occlusion in the treatment of alopecia totalis/universalis. J Am Acad Dermatol 49:96–98 ArticlePubMedGoogle Scholar
  150. Tosti A, Iorizzo M, Botta GL, Milani M (2006) Efficacy and safety of a new clobetasol propionate 0.05% foam in alopecia areata: a randomized, double-blind placebo-controlled trial. J Eur Acad Dermatol Venereol 20:1243–1247 ArticlePubMedGoogle Scholar
  151. Rosenberg EW, Drake L (1976) Society transactions. Arch Dermatol 112:25 ArticleGoogle Scholar
  152. Philpott MP, Sanders DA, Bowen J, Kealey T (1996) Effects of interleukines, colony-stimulating factor and tumor necrosis factor on human hair follicle growth in vitro: a possible role for interleukin-1 and tumor necrosis factor-a in alopecia areata. Br J Dermatol 135:942–948 ArticleCASPubMedGoogle Scholar
  153. Hoffmann R, Happle R (1996) Topical immunotherapy in alopecia areata. What, how, and why? Dermatol Clin 14(4):739–744 ArticleCASPubMedGoogle Scholar
  154. Happle R, Kalveram KJ, Büchner U, Echternacht-Happle K, Göggelmann W, Summer KH (1980) Contact allergy as therapeutic tool for alopecia areata: application of squaric acid dibutylester. Dermatologica 161:289–297 ArticleCASPubMedGoogle Scholar
  155. Shapiro J (1993) Topical immunotherapy in the treatment of chronic severe alopecia areata. Dermatol Clin 11(3):611–617 CASPubMedGoogle Scholar
  156. Wilkerson MG, Henkin J, Wilkin JK, Smith RG (1985) Squaric acid and esters: analysis for contaminants and stability in solvents. J Am Acad Dermatol 13:229–234 ArticleCASPubMedGoogle Scholar
  157. Tiwary AK, Mishra DK, Chaudhary SS (2016) Comparative study of efficacy and safety of topical squaric acid dibutylester and diphenylcyclopropenone for the treatment of alopecia areata. N Am J Med Sci 8(6):237–242 ArticlePubMedPubMed CentralGoogle Scholar
  158. Happle R, Hausen BM, Wiesner-Menzel L (1983) Diphencyprone in the treatment of alopecia areata. Acta Derm Venereol 63(1):49–52 CASPubMedGoogle Scholar
  159. Hull SM, Norris J (1988) Diphencyprone in the treatment of long-standing alopecia areata. Br J Dermatol 119(3):367–374 ArticleCASPubMedGoogle Scholar
  160. Monk B (1989) Induction of hair growth in alopecia totalis with diphencyprone sensitization. Clin Exp Dermatol 14(2):154–157 ArticleCASPubMedGoogle Scholar
  161. Pericin M, Trüeb RM (1998) Topical immunotherapy of severe alopecia areata with diphenylcyclopropenone: evaluation of 68 cases. Dermatology 196:418–421 ArticleCASPubMedGoogle Scholar
  162. Buckley DA, du Vivier AW (1999) Topical immunotherapy in dermatology. Int J Clin Pract 53(2):130–137 CASPubMedGoogle Scholar
  163. Bolduc C, Shapiro J (2000) DPCP for the treatment of alopecia areata. Skin Therapy Lett 5(5):3–4 CASPubMedGoogle Scholar
  164. Galadari I, Rubaie S, Alkaabi J, Galadari H (2003) Diphenylcyclopropenone (diphencyprone, DPCP) in the treatment of chronic severe alopecia areata (AA). Eur Ann Allergy Clin Immunol 35(10):397–401 CASPubMedGoogle Scholar
  165. Cotellessa C, Peris K, Caracciolo E, Mordenti C, Chimenti S (2001) The use of topical diphenylcyclopropenone for the treatment of extensive alopecia areata. J Am Acad Dermatol 44(1):73–76 ArticleCASPubMedGoogle Scholar
  166. Aghaei S (2005) Topical immunotherapy of severe alopecia areata with diphenylcyclopropenone (DPCP): experience in an Iranian population. BMC Dermatol 5:6 ArticlePubMedPubMed CentralCASGoogle Scholar
  167. Sotiriadis D, Patsatsi A, Lazaridou E, Kastanis A, Vakirlis E, Chrysomallis F (2007) Topical immunotherapy with diphenylcyclopropenone in the treatment of chronic extensive alopecia areata. Clin Exp Dermatol 32(1):48–51 CASPubMedGoogle Scholar
  168. El-Zawahry BM, Bassiouny DA, Khella A, Zaki N (2010) Five-year experience in the treatment of alopecia areata with DPC. J Eur Acad Dermatol Venereol 24(3):264–269 ArticleCASPubMedGoogle Scholar
  169. Ohlmeier MC, Traupe H, Luger TA, Böhm M (2012) Topical immunotherapy with diphenylcyclopropenone of patients with alopecia areata—a large retrospective study on 142 patients with a self-controlled design. J Eur Acad Dermatol Venereol 26(4):503–507 ArticleCASPubMedGoogle Scholar
  170. Chiang KS, Mesinkovska NA, Piliang MP, Bergfeld WF (2015) Clinical efficacy of diphenylcyclopropenone in alopecia areata: retrospective data analysis of 50 patients. J Investig Dermatol Symp Proc 17(2):50–55 ArticleCASPubMedGoogle Scholar
  171. Hull SM, Pepall L, Cunliffe W (1991) Alopecia areata in children: response to treatment with diphencyprone. Br J Dermatol 125(2):164–168 ArticleCASPubMedGoogle Scholar
  172. Schuttelaar ML, Hamstra JJ, Plinck EP, Peereboom-Wynia JD, Vuzevski VD, Mulder PG, Oranje AP (1996) Alopecia areata in children: treatment with diphencyprone. Br J Dermatol 135(4):581–585 ArticleCASPubMedGoogle Scholar
  173. Katagiri K, Arakawa S, Hatano Y, Fujiwara S (2006) Fexofenadine, an H1-receptor antagonist, partially but rapidly inhibits the itch of contact dermatitis induced by diphenylcyclopropenone in patients with alopecia areata. J Dermatol 33(2):75–79 ArticleCASPubMedGoogle Scholar
  174. Inui S, Nakajima T, Toda N, Itami S (2009) Fexofenadine hydrochloride enhances the efficacy of contact immunotherapy for extensive alopecia areata: retrospective analysis of 121 cases. J Dermatol 36(6):323–327 ArticleCASPubMedGoogle Scholar
  175. Joly P (2006) The use of methotrexate alone or in combination with low doses of oral corticosteroids in the treatment of alopecia totalis or universalis. J Am Acad Dermatol 55:632–636 ArticlePubMedGoogle Scholar
  176. Chartaux E, Joly P (2010) Long-term follow-up of the efficacy of methotrexate alone or in combination with low doses of oral corticosteroids in the treatment of alopecia areata totalis or universalis. Ann Dermatol Venereol 137(8–9):507–513 ArticleCASPubMedGoogle Scholar
  177. Kaelin U, Hassan AS, Braathen LR, Yawalkar N (2006) Treatment of alopecia areata partim universalis with efalizumab. J Am Acad Dermatol 55(3):529–532 ArticlePubMedGoogle Scholar
  178. Price VH, Hordinsky MK, Olsen EA et al (2008) Subcutaneous efalizumab is not effective in the treatment of alopecia areata. J Am Acad Dermatol 58:395–402 ArticlePubMedGoogle Scholar
  179. Major E (2010) Progressive multifocal leukoencephalopathy in patients on immunomodulatory therapies. Annu Rev Med 61(1):35–47 ArticleCASPubMedGoogle Scholar
  180. Namazi MR (2004) Statins: novel additions to the dermatologic arsenal? Exp Dermatol 13(6):337–339 ArticleCASPubMedGoogle Scholar
  181. Robins DN (2007) Case reports: alopecia universalis: hair growth following initiation of simvastatin and ezetimibe therapy. J Drugs Dermatol 6(9):946–947 PubMedGoogle Scholar
  182. Ali A, Martin JM IV. (2010) Hair growth in patients alopecia areata totalis after treatment with simvastatin and ezetimibe. J Drugs Dermatol 9(1):62–64 PubMedGoogle Scholar
  183. Lattouf C, Jimenez JJ, Tosti A et al (2015) Treatment of alopecia areata with simvastatin/ezetimibe. J Am Acad Dermatol 72(2):359–361 ArticlePubMedGoogle Scholar
  184. Loi C, Starace M, Piraccini BM (2016) Alopecia areata (AA) and treatment with simvastatin/ezetimibe: experience of 20 patients. J Am Acad Dermatol 74(5):e99-e ArticleGoogle Scholar
  185. Freitas-Gouveia M, Trüeb RM (2017) Unsuccessful treatment of alopecia areata with simvastatin/ezetimibe: experience in 12 patients. Skin Appendage Disord 3:156–160 ArticlePubMedGoogle Scholar
  186. Schmoeckel C, Weissmann I, Plewig G, Braun-Falco O (1979) Treatment of alopecia areata by anthralin-induced dermatitis. Arch Dermatol 115(10):1254–1255 ArticleCASPubMedGoogle Scholar
  187. Fiedler-Weiss VC, Buys CM (1987) Evaluation of anthralin in the treatment of alopecia areata. Arch Dermatol 123(11):1491–1493 ArticleCASPubMedGoogle Scholar
  188. Fiedler VC, Wendrow A, Szpunar GJ, Metzler C, DeVillez RL (1990) Treatment-resistant alopecia areata. Response to combination therapy with minoxidil plus anthralin. Arch Dermatol 126(6):756–759 ArticleCASPubMedGoogle Scholar
  189. Olsen EA, Carson SC, Turney EA (1992) Systemic steroids with or without 2% topical minoxidil in the treatment of alopecia areata. Arch Dermatol 128(11):1467–1467 ArticleCASPubMedGoogle Scholar
  190. Gupta AK, Ellis CN, Cooper KD, Nickoloff BJ, Ho VC, Chan LS et al (1990) Oral cyclosporine for the treatment of alopecia areata. A clinical and immunohistochemical analysis. J Am Acad Dermatol 22:242–250 ArticleCASPubMedGoogle Scholar
  191. Paquest P, Arrase-Estrada J, Pierard GE (1992) Oral cyclosporine and alopecia areata. Dermatology 185:314–315 ArticleGoogle Scholar
  192. Lee D, Oh DJ, Kim JW, Park SW, Oh MK, Sung HS, Hwang SW (2008) Treatment of severe alopecia areata: combination therapy using systemic cyclosporine a with low dose corticosteroids. Ann Dermatol 20(4):172–178 ArticlePubMedPubMed CentralGoogle Scholar
  193. Yeo IK, Ko EJ, No YA et al (2015) Comparison of high-dose corticosteroid pulse therapy and combination therapy using oral cyclosporine with low-dose corticosteroid in severe alopecia Areata. Ann Dermatol 27(6):676–681 ArticlePubMedPubMed CentralGoogle Scholar
  194. Tosti A, Manuzzi P, Gasponi A (1988) Thymopentin in the treatment of severe alopecia areata. Dermatologica 177(3):170–174 ArticleCASPubMedGoogle Scholar
  195. Orecchia G (1989) Thymopentin in the treatment of alopecia areata. Dermatologica 178(4):231 ArticleCASPubMedGoogle Scholar
  196. Lowy M, Ledoux-Corbusier M, Achten G, Wybran J (1985) Clinical and immunologic response to Isoprinosine in alopecia areata and alopecia universalis: association with autoantibodies. J Am Acad Dermatol 12(1 Pt 1):78–84 ArticleCASPubMedGoogle Scholar
  197. Thiers BH (1991) Isoprinosine treatment of alopecia areata. J Invest Dermatol 96(5):72S–73S ArticleCASPubMedGoogle Scholar
  198. Georgala S, Katoulis AC, Befon A, Georgala K, Stavropoulos PG (2006) Inosiplex for treatment of alopecia areata: a randomized placebo-controlled study. Acta Derm Venereol 86(5):422–424 ArticleCASPubMedGoogle Scholar
  199. Mrowietz U, Christophers E, Altmeyer P (1999) Treatment of severe psoriasis with fumaric acid esters: scientific background and guidelines for therapeutic use. The German Fumaric Acid Ester Consensus Conference. Br J Dermatol 141(3):424–429 ArticleCASPubMedGoogle Scholar
  200. Venten I, Hess N, Hirschmüller A, Altmeyer P, Brockmeyer N (2006) Treatment of therapy-resistant alopecia areata with fumaric acid esters. Eur J Med Res 11(7):300–305 CASPubMedGoogle Scholar
  201. Healy E, Rogers S (1993) PUVA treatment for alopecia areata—does it work? A retrospective review of 102 cases. Br J Dermatol 129(1):42–44 ArticleCASPubMedGoogle Scholar
  202. Taylor CR, Hawk JL (1995) PUVA treatment of alopecia areata partialis, totalis and universalis: audit of 10 years’ experience at St John’s Institute of Dermatology. Br J Dermatol 133(6):914–918 ArticleCASPubMedGoogle Scholar
  203. Willemsen R, Haentjens P, Roseeuw D, Vanderlinden J (2010) Hypnosis in refractory alopecia areata significantly improves depression, anxiety, and life quality but not hair regrowth. J Am Acad Dermatol 62(3):517–518 ArticlePubMedGoogle Scholar
  204. Trink A, Sorbellini E, Bezzola P, Rodella L, Rezzani R, Ramot Y, Rinaldi F (2013) A randomized, double-blind, placebo- and active-controlled, half-head study to evaluate the effects of platelet-rich plasma on alopecia areata. Br J Dermatol 169(3):690–694 ArticleCASPubMedGoogle Scholar
  205. Waldmann TA (2013) The biology of IL-15: implications for cancer therapy and the treatment of autoimmune disorders. J Invest Dermatol Symp Proc 16:S28–S30 ArticleCASGoogle Scholar
  206. Falto-Aizpurua L, Choudhary S, Tosti A (2014) Emerging treatments in alopecia. Expert Opin Emerg Drugs 19(4):545–556 ArticleCASPubMedGoogle Scholar
  207. Divito SJ, Kupper TS (2014) Inhibiting Janus kinases to treat alopecia areata. Nat Med 20(9):989–990 ArticleCASPubMedGoogle Scholar
  208. Craiglow BG, King BA (2014) Killing two birds with one stone: oral tofacitinib reverses alopecia areata universalis in a patient with plaque psoriais. J Invest Dermatol 134(12):2988–2990 ArticleCASPubMedGoogle Scholar
  209. Xing L, Dai Z, Jabbari A et al (2014) Alopecia areata is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition. Nat Med 20(9):1043–1049 ArticleCASPubMedPubMed CentralGoogle Scholar
  210. Jabbari A, Dai Z, Xing L, Cerise JE, Ramot Y, Berkun Y, Sanchez GA, Goldbach-Mansky R, Christiano AM, Clynes R, Zlotogorski A (2015) Reversal of alopecia areata following treatment with the JAK1/2 inhibitor baricitinib. EBioMedicine 2(4):351–355 ArticlePubMedPubMed CentralGoogle Scholar
  211. Liu LY, Craiglow BG, Dai F, King BA (2017) Tofacitinib for the treatment of severe alopecia areata and variants: a study of 90 patients. J Am Acad Dermatol 76(1):22–28 ArticleCASPubMedGoogle Scholar
  212. Craiglow BG, Liu LY, King BA (2017) Tofacitinib for the treatment of alopecia areata and variants in adolescents. J Am Acad Dermatol 76(1):29–32 ArticleCASPubMedGoogle Scholar
  213. Anzengruber F, Maul JT, Kamarachev J, Trüeb RM, French LE, Navarini AA (2016) Transient efficacy of tofacitinib in alopecia areata universalis. Case Rep Dermatol 8(1):102–106 ArticlePubMedPubMed CentralGoogle Scholar
  214. Brunne V, Mertins G, Reimann G, Brockmeyer NH (2004) Off-label use in dermatological practice. The conflict between professional duty and legal requirements. Hautarzt 55(8):727–734 ArticleCASPubMedGoogle Scholar

Author information

Authors and Affiliations

  1. Center for Dermatology and Hair Diseases Professor Trüeb and University of Zurich, Zurich, Switzerland Ralph M. Trüeb
  2. Department of Dermatology, Universidade Federal Fluminense, Centro de Ciências Médicas, Hospital Universitário Antonio Pedro, Niterói, Rio de Janeiro, Brazil Maria Fernanda Reis Gavazzoni Dias
  1. Ralph M. Trüeb