Skip to main content
Erschienen in: Advances in Therapy 7/2017

Open Access 23.06.2017 | Review

The Changing Landscape of Alopecia Areata: The Translational Landscape

verfasst von: Etienne C. E. Wang, Angela M. Christiano

Erschienen in: Advances in Therapy | Ausgabe 7/2017

Abstract

Recent genetic and preclinical studies have increased our understanding of the immunopathogenesis of alopecia areata (AA). This has allowed expedited development of targeted therapies for the treatment of AA, and a paradigm shift in our approach and understanding of autoimmunity and the hair follicle. The synergy between preclinical studies, animal models, and translational studies has led to unprecedented advances in the treatment options for AA, ultimately benefiting patients who have had little recourse. In this review, we summarize the scientific field of contemporary AA research, and look forward to potential new technologies and developments.
Hinweise

Enhanced content

To view enhanced content for this article go to http://​www.​medengine.​com/​Redeem/​C818F0606DABA121​.

Introduction

Alopecia areata (AA) is one of the most prevalent autoimmune diseases in the USA, affecting approximately 5.3 million people, including both sexes across all ethnicities, with a lifetime risk of 1.7% [1]. This is comparable to the estimated 2% lifetime incidence of AA globally, which comes with a significant healthcare burden [2]. Despite this high prevalence, there have been no evidence-based treatments for AA as recently as 5 years ago. Recent scientific and clinical efforts have uncovered novel insights into the pathogenesis of AA, which have led to the development of new translational treatments. This recent progress has revived the “translational landscape” of AA, in terms of its etiology, treatment options, and prognosis, and has given new hope to the millions of patients suffering from the disease. In this brief review, we will survey the rapidly evolving landscape of AA, and provide an overview of how basic research, in collaboration with clinical dermatology, has transformed the future prospects for treatment of this devastating and highly prevalent disease. Treatment algorithms are out of the scope of this review, and will be discussed elsewhere. This article is based on previously conducted studies and does not involve any new studies of human or animal subjects performed by any of the authors.

Pathogenesis

Historically, AA has been classified as an autoimmune disease of the hair follicle due to the characteristic T cell accumulation at the hair bulb, also known as the histological “swarm of bees”. However, a detailed description of this immune infiltrate remained unexplored. Little was known about the initial trigger, factors required for maintenance, and identity of the elusive autoantigen(s). Recent fundamental and basic science studies have greatly improved our understanding of AA and its possible causes.

Genetic Basis of AA

In the past decade, genome-wide association studies (GWAS) and genetic linkage studies have been central to the elucidation of the pathogenesis of AA. Compared to GWAS of other autoimmune and immune-mediated diseases, the “hits” obtained from the AA GWAS have yielded a higher number of meaningful candidate genes that have shed light on the mechanisms of disease in AA [3], providing new therapeutic targets.
In our first GWAS for AA, which included 1054 AA patients from a comprehensive registry (National Alopecia Areata Registry) in North America, we identified eight regions of the genome that were significantly associated with AA [4]. Like other autoimmune conditions, the human leukocyte antigen (HLA) locus was significantly represented in the AA GWAS, supporting previous candidate gene association studies [5, 6]. Interestingly, the second highest peak uncovered in our GWAS, and confirmed in other cohorts, mapped to the ULBP6/ULBP3 gene locus on chromosome 6q. Functional studies confirmed that ULBP3 was aberrantly upregulated as a danger signal on the hair follicles of AA patients [4, 7], which contributed to the collapse of immune privilege, and the recruitment of cytotoxic CD8+ NKG2D+ T cells to carry out destruction of the hair follicle. We subsequently showed that this subset of T cells was both necessary and sufficient for the development of AA. Meta-analysis of GWAS studies in AA was conducted on the combined North American and European AA cohorts [8], and confirmed the significance of these susceptibility loci, as well as identifying several new loci.
Applying pathway and network analyses (e.g., Gene Ontology term enrichment and proteomic interactions) to the GWAS data has also uncovered sets of genes that contribute to pathogenic processes. This type of analysis grouped AA with other autoimmune diseases like type 1 diabetes, rheumatoid arthritis (RA), and celiac disease. Pathways that are implicated in the pathogenesis of this group of autoimmune diseases include antigen processing and presentation, co-stimulatory pathways, and JAK-STAT signaling [9]. These associations led to a re-analysis of the underlying immunopathogenesis of AA, and to a consideration of employing new therapies that target these pathways specifically.

Immunopathogenesis

Adaptive immune responses were traditionally classified into Th1 and Th2 responses, depending on the components of the cytokine milieu in the diseased state. This binary view of the immune response has been shown to be insufficient to account for the complex immune profiles of many diseases. Historically, AA was assigned to the Th1 category owing to its significant interferon-gamma (IFN-γ) signature. However, AA has also been epidemiologically associated with atopy [10], a classic Th2 condition defined by a predominance of IL-4 and IL-13. While measuring cytokine transcripts in circulating immune cells in AA tend to favor a Th1 profile [11, 12], lesional skin in AA exhibited a more mixed profile [13]. Gene expression profiles of AA lesional skin have also uncovered mixed immune response signatures [14]. This intriguing finding suggests that the immune response in AA may be heterogenous, and may fluctuate and evolve within a single patient through time, or may even signal distinct disease subtypes that may require different therapeutic approaches.

Autoantigens of the Hair Follicle

A few autoimmune diseases (i.e., autoimmune thyroiditis, celiac disease, rheumatoid arthritis) have well-characterized autoantigens that are the known targets for the immune system. Most other autoimmune conditions and immune-mediated diseases, including AA and psoriasis, do not yet have any autoantigen that is clearly associated with their pathogenesis. The hair follicle is regarded as a relatively immune-privileged site [15], with generally low expression of MHC class I molecules and “danger signals” that are overexpressed in disease (e.g., ULBP6/3). In order to sequester hair follicle antigens from the immune system, particularly during the hair cycle when significant tissue remodeling occurs, the hair follicle microenvironment needs to be precisely regulated. Some studies have suggested that failure of this mechanism leads to a “collapse of immune privilege” and sensitization of circulating T cells to keratinocyte and/or melanocyte peptides, which might precipitate AA [16]. Autoantibodies specific for hair follicle proteins such as trichohyalin and certain keratins have been detected in both mouse and human cases of AA [17, 18], but more work remains to expand the repertoire of discovered AA autoantigens and their relevance to disease pathogenesis.

Preclinical Studies and Animal Models

Basic science and preclinical research are not frequently appreciated when discussing the clinical aspects of a disease, but nonetheless they provide the bedrock of innovation that drives the technology and progress that fuels novel therapies.

Animal Model of AA

A well-validated animal model can make a significant impact in the study of human disease. In the case of AA, the C3H/HeJ mouse was found to spontaneously develop an immune-mediated hair loss that resembled human AA both histopathologically and immunologically, albeit at a low frequency, as the mice age [19]. This phenotype was found to be transferrable with skin grafts from an affected mouse to younger, congenic recipients [20]. This model revolutionized the field of AA research by establishing a reliable animal model that closely recapitulated the human disease. Recently, this process has been reproduced with transfer of skin-draining lymph node cells, abrogating the need for invasive surgical procedures [21].
Humanized models of AA have also been attempted. By grafting normal human scalp skin onto immunocompromised mice, and transferring peripheral blood monocytes from an unrelated healthy donor enriched for the pathogenic NKG2D+ population, one can precipitate an autoimmune type hair loss which may have similarities to AA, although the tissues are derived from normal healthy donors [22].

Targeting Effector Cytokines/Chemokines in Animal Models of AA

As knowledge accumulates on the immunopathogenesis of AA, researchers have been able to systematically target and investigate the roles of upstream and downstream effectors of the system. The identification of the pathogenic subset of NKG2D+ CD8+ cytotoxic T cells led to the characterization of the cytokine milieu that they secrete. This subset of T cells was found to respond to IFN-induced chemokines CXCL9/10/11, released by the hair follicle itself. Antagonism of their common chemokine receptor (CXCR3) on T cells was subsequently shown to be sufficient to prevent onset of AA in a mouse model [23].

Diagnosis and Disease Prognostication

AA is normally a straightforward clinical diagnosis to make, particularly when it presents in its classical form with focal or multifocal patches of acute non-scarring alopecia, and a positive hair-pull test. Occasionally, a skin biopsy might be required to distinguish diffuse AA from other conditions such as female androgenetic alopecia or telogen effluvium. In these cases, AA is unequivocally associated with the histopathological finding of a peribulbar immune infiltrate of cytotoxic T cells (the so-called swarm of bees). However, newer non-invasive technologies discussed below have been adopted to facilitate in diagnosis of difficult cases.

Trichoscopy and Reflectance Confocal Microscopy

Dermatoscopy has been an indispensable tool for the management of melanocytic lesions, and training to detect melanoma amongst dermatology residents has reached a very high standard. Thus, most, if not all, dermatologists are equipped with a state-of-the-art dermatoscope, which has recently been adopted for diagnosing hair disorders [24]. In addition to the classical “exclamation mark hairs” which are found at the periphery of AA lesions, dermatoscopic, or trichoscopic features of AA include yellow dots, which correlate with dystrophic follicular epithelium and sebaceous glands [25]. Digital videodermatoscopy has also been employed for the diagnosis and follow-up of hair disorders in many clinics. Reflectance confocal microscopy is another tool that may be employed for non-invasive diagnosis of AA, with the “yellow dots” clearly visible as degenerated follicles and, in some cases, even the immune infiltrate can be visualized [26].

Stratification of Patients with ALADIN Gene Expression Profiling

The era of personalized medicine has brought with it a vast body of large datasets related to sequencing, genotyping, and gene expression. Not only do we have information from GWAS hits that may suggest potential new druggable targets but RNA-Seq and microarray data from individual patients are beginning to provide insight into the molecular differences in disease pathogenesis from patient to patient. Integrating the relative expression of genes in the interferon (IFN), cytotoxic T cell (CTL), and keratinocyte (KER) clusters, a new metric known as the Alopecia Areata Disease Activity Index (ALADIN) has been developed [14]. ALADIN is specific to AA, and correlates with disease activity, making it a suitable biomarker for assessment of clinical response to treatment, and to identify patients who may or may not respond to certain therapeutic modalities [14].

Treatment

Treatment modalities for AA are diverse and to date have not been reliably effective. Until recently, the treatments for AA have been generally non-specific and employ a “scorched earth” strategy of either suppressing (intralesional and topical corticosteroids) or stimulating (topical immunotherapy, excimer laser therapy) the immune system, with varying results. Very few well-designed randomized controlled clinical trials have been conducted to justify their widespread adoption. These have been reviewed extensively elsewhere [27, 28]. For this review, we will focus on the innovative new therapies that have been developed for the treatment of AA.

Janus Kinase Inhibitors

A significant breakthrough in the treatment of AA has been the discovery of the importance of the JAK-STAT pathway in the initiation and the maintenance of the diseased state (see “Pathogenesis”). This provided the rationale to use small molecule JAK inhibitors, which have been FDA-approved for other diseases (tofacitinib for rheumatoid arthritis and ruxolitinib for myelofibrosis), for the treatment of AA. Treatment-resistant patients with extensive disease, alopecia universalis or totalis, were found in open-label trials to respond with full regrowth of hair after a twice-daily regimen of ruxolitinib within 3–5 months. This effect was supported by case reports that reported JAK inhibitors reversing co-incidental AA in patients being treated for other diseases [29, 30]. Further clinical trials are underway for systemic tofacitinib and ruxolitinib for the treatment of AA [31, 32], as well as topical formulations of the JAK inhibitors.

Co-stimulatory Blockade

Cytotoxic T lymphocyte antigen (CTLA) 4 is a negative regulator of T cells, attenuating the immune response by blocking co-stimulatory signals by interfering with the interaction between CD28 on T cells and CD80/CD86 on antigen-presenting cells. Its dysfunction has been associated with several autoimmune and immune-mediated conditions like thyroiditis [33], rheumatoid arthritis [34], and inflammatory bowel disease [35]. Single nucleotide polymorphisms in the CTLA4 gene have also been associated with AA in some populations [36]. Its location on chromosome 2q33.2 was also among the GWAS hits for AA [4]. Abatacept, a recombinant fusion protein (CTLA4-Ig), has been an effective and successful treatment for RA and other autoimmune diseases [37]. Preclinical studies have also shown that abatacept is effective in preventing the mouse model of AA if given before the onset of disease [38], providing rationale and promise for its efficacy in human AA patients. Clinical trials are underway to investigate its role in AA (ClinicalTrials.gov NCT02018042).

Simvastatin/Ezetimibe

Several case reports [39, 40] and a small case–control study [41] have suggested that simvastatin/ezetimibe, a common treatment for hyperlipidemia, may have immunomodulatory effects that might benefit AA patients. Considering atherosclerosis to be a disease of the adaptive immune system [42], this drug combination has been postulated to have effects on antigen presentation, lymphocyte trafficking, and regulatory T cell induction. Inevitably, clinicians will be testing this treatment off-label on their most recalcitrant cases of AA, since simvastatin/ezetimibe has an acceptable safety record. Larger randomized controlled studies are required to confirm its efficacy.

Biologic Therapies for AA

Similar preclinical studies are directed at other pathways that intersect with the pathogenic NKG2D+ CD8+ T cell subset, one of which may eventually translate into the clinical realm. Since anti-TNF (tumor necrosis factor) therapies have uniformly failed in AA [43] and have been reported to precipitate AA in some patients [44], there has been a resurgence of interest in testing other biological modalities in the setting of AA. Preclinical studies have established the roles of IL-15 [45], IFN-γ [46], and IL-2 [47] in the pathogenesis of AA in mice, and specific therapies targeting these pathways are being investigated. Low-dose IL-2, which has been used to stimulate expansion of regulatory T cells in the treatment of autoimmune disease such as type 1 diabetes [48], has been used in AA with modest results [49].
As alluded to before, the apparent heterogeneity of the immune response in AA has led to speculation that Th2 and even Th17 immune pathways may be involved in the development of the disease. This stems from gene expression data and cytokine profiling of a small and probably heterogenous group of patients [13]. A subsequent small case series showed that patients with a higher inflammatory score measured with ALADIN responded well to ustekinumab [50], a humanized monoclonal antibody to the p40 subunit that is central to the IL-12/IL-23 signaling pathways of the Th17 response. Further work to deconstruct the components of the immune response in AA to determine the heterogeneity and spectrum of disease phenotypes that may present in different patients will help to tailor treatment strategies in a precise manner. This personalized approach will maximize the efficacy of treatment, while minimizing the potential adverse effects and drug exposures.

Conclusions

As our knowledge and understanding of the immune system progress, we have been able to dissect and target increasingly precise components of the immune response. With this understanding also comes the revelation that the complexities of the immune system cannot be explained by simple dichotomies, and that every patient is different, and requires a precise, personalized treatment. Our conceptual understanding of autoimmunity, illustrated here with the case of AA, is evolving to encompass new concepts such that the entire approach of diagnosis, prognosis, and management is evolving along with it. The coming years will herald new exciting therapies for our patients, targeting pathways that we may not even have suspected previously, which will in turn bring us more questions and new perceptions of this common, devastating disease.

Acknowledgements

This work was funded by NIH Grant P50 AR070588-01. No sponsorship was received for the article processing charges. All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval for the version to be published.

Disclosures

Dr Christiano is a consultant to Aclaris Therapeutics. Dr Wang has nothing to disclose.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not involve any new studies of human or animal subjects performed by any of the authors.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Open Access

This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (https://​creativecommons.​org/​licenses/​by/​4.​0), which permits use, duplication, adaptation, distribution, and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Safavi KH, et al. Incidence of alopecia areata in Olmsted County, Minnesota, 1975 through 1989. Mayo Clin Proc. 1995;70(7):628–33.CrossRefPubMed Safavi KH, et al. Incidence of alopecia areata in Olmsted County, Minnesota, 1975 through 1989. Mayo Clin Proc. 1995;70(7):628–33.CrossRefPubMed
2.
Zurück zum Zitat Villasante Fricke AC, Miteva M. Epidemiology and burden of alopecia areata: a systematic review. Clin Cosmet Investig Dermatol. 2015;8:397–403.PubMedPubMedCentral Villasante Fricke AC, Miteva M. Epidemiology and burden of alopecia areata: a systematic review. Clin Cosmet Investig Dermatol. 2015;8:397–403.PubMedPubMedCentral
3.
Zurück zum Zitat Petukhova L, Christiano AM. The genetic architecture of alopecia areata. J Investig Dermatol Symp Proc. 2013;16(1):S16–22.CrossRefPubMed Petukhova L, Christiano AM. The genetic architecture of alopecia areata. J Investig Dermatol Symp Proc. 2013;16(1):S16–22.CrossRefPubMed
4.
Zurück zum Zitat Petukhova L, et al. Genome-wide association study in alopecia areata implicates both innate and adaptive immunity. Nature. 2010;466(7302):113–7.CrossRefPubMedPubMedCentral Petukhova L, et al. Genome-wide association study in alopecia areata implicates both innate and adaptive immunity. Nature. 2010;466(7302):113–7.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Colombe BW, et al. HLA class II antigen associations help to define two types of alopecia areata. J Am Acad Dermatol. 1995;33(5 Pt 1):757–64.PubMed Colombe BW, et al. HLA class II antigen associations help to define two types of alopecia areata. J Am Acad Dermatol. 1995;33(5 Pt 1):757–64.PubMed
6.
Zurück zum Zitat Barahmani N, et al. Major histocompatibility complex class I chain-related gene A polymorphisms and extended haplotypes are associated with familial alopecia areata. J Invest Dermatol. 2006;126(1):74–8.CrossRefPubMed Barahmani N, et al. Major histocompatibility complex class I chain-related gene A polymorphisms and extended haplotypes are associated with familial alopecia areata. J Invest Dermatol. 2006;126(1):74–8.CrossRefPubMed
7.
Zurück zum Zitat Moftah NH, et al. ULBP3: a marker for alopecia areata incognita. Arch Dermatol Res. 2016;308(6):415–21.CrossRefPubMed Moftah NH, et al. ULBP3: a marker for alopecia areata incognita. Arch Dermatol Res. 2016;308(6):415–21.CrossRefPubMed
8.
Zurück zum Zitat Betz RC, et al. Genome-wide meta-analysis in alopecia areata resolves HLA associations and reveals two new susceptibility loci. Nat Commun. 2015;6:5966.CrossRefPubMedPubMedCentral Betz RC, et al. Genome-wide meta-analysis in alopecia areata resolves HLA associations and reveals two new susceptibility loci. Nat Commun. 2015;6:5966.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Petukhova L, Christiano AM. Functional interpretation of genome-wide association study evidence in alopecia areata. J Invest Dermatol. 2016;136(1):314–7.CrossRefPubMedPubMedCentral Petukhova L, Christiano AM. Functional interpretation of genome-wide association study evidence in alopecia areata. J Invest Dermatol. 2016;136(1):314–7.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Kuwano Y, et al. Serum chemokine profiles in patients with alopecia areata. Br J Dermatol. 2007;157(3):466–73.CrossRefPubMed Kuwano Y, et al. Serum chemokine profiles in patients with alopecia areata. Br J Dermatol. 2007;157(3):466–73.CrossRefPubMed
12.
Zurück zum Zitat Sadeghi S, et al. Study of Th1/Th2 balance in peripheral blood mononuclear cells of patients with alopecia areata. Acta Microbiol Immunol Hung. 2015;62(3):275–85.CrossRefPubMed Sadeghi S, et al. Study of Th1/Th2 balance in peripheral blood mononuclear cells of patients with alopecia areata. Acta Microbiol Immunol Hung. 2015;62(3):275–85.CrossRefPubMed
13.
Zurück zum Zitat Suarez-Farinas M, et al. Alopecia areata profiling shows TH1, TH2, and IL-23 cytokine activation without parallel TH17/TH22 skewing. J Allergy Clin Immunol. 2015;136(5):1277–87.CrossRefPubMed Suarez-Farinas M, et al. Alopecia areata profiling shows TH1, TH2, and IL-23 cytokine activation without parallel TH17/TH22 skewing. J Allergy Clin Immunol. 2015;136(5):1277–87.CrossRefPubMed
15.
Zurück zum Zitat Paus R, Bertolini M. The role of hair follicle immune privilege collapse in alopecia areata: status and perspectives. J Investig Dermatol Symp Proc. 2013;16(1):S25–7.CrossRefPubMed Paus R, Bertolini M. The role of hair follicle immune privilege collapse in alopecia areata: status and perspectives. J Investig Dermatol Symp Proc. 2013;16(1):S25–7.CrossRefPubMed
16.
Zurück zum Zitat Wang EH, et al. Identification of autoantigen epitopes in alopecia areata. J Invest Dermatol. 2016;136(8):1617–26. Wang EH, et al. Identification of autoantigen epitopes in alopecia areata. J Invest Dermatol. 2016;136(8):1617–26.
17.
Zurück zum Zitat Tobin DJ, et al. Autoantibodies to hair follicles in C3H/HeJ mice with alopecia areata-like hair loss. J Invest Dermatol. 1997;109(3):329–33.CrossRefPubMed Tobin DJ, et al. Autoantibodies to hair follicles in C3H/HeJ mice with alopecia areata-like hair loss. J Invest Dermatol. 1997;109(3):329–33.CrossRefPubMed
18.
Zurück zum Zitat Leung MC, et al. Trichohyalin is a potential major autoantigen in human alopecia areata. J Proteome Res. 2010;9(10):5153–63.CrossRefPubMed Leung MC, et al. Trichohyalin is a potential major autoantigen in human alopecia areata. J Proteome Res. 2010;9(10):5153–63.CrossRefPubMed
19.
Zurück zum Zitat Sundberg JP, et al. C3H/HeJ mouse model for alopecia areata. J Invest Dermatol. 1995;104(5 Suppl):16S–7S.CrossRefPubMed Sundberg JP, et al. C3H/HeJ mouse model for alopecia areata. J Invest Dermatol. 1995;104(5 Suppl):16S–7S.CrossRefPubMed
20.
Zurück zum Zitat McElwee KJ, et al. Experimental induction of alopecia areata-like hair loss in C3H/HeJ mice using full-thickness skin grafts. J Invest Dermatol. 1998;111(5):797–803.CrossRefPubMed McElwee KJ, et al. Experimental induction of alopecia areata-like hair loss in C3H/HeJ mice using full-thickness skin grafts. J Invest Dermatol. 1998;111(5):797–803.CrossRefPubMed
21.
Zurück zum Zitat Wang EH, et al. Transfer of alopecia areata to C3H/HeJ mice using cultured lymph node-derived cells. J Invest Dermatol. 2015;135(10):2530–2.CrossRefPubMed Wang EH, et al. Transfer of alopecia areata to C3H/HeJ mice using cultured lymph node-derived cells. J Invest Dermatol. 2015;135(10):2530–2.CrossRefPubMed
22.
Zurück zum Zitat Gilhar A, Keren A, Paus R. A new humanized mouse model for alopecia areata. J Investig Dermatol Symp Proc. 2013;16(1):S37–8.CrossRefPubMed Gilhar A, Keren A, Paus R. A new humanized mouse model for alopecia areata. J Investig Dermatol Symp Proc. 2013;16(1):S37–8.CrossRefPubMed
23.
Zurück zum Zitat Dai Z, et al. CXCR3 blockade inhibits T cell migration into the skin and prevents development of alopecia areata. J Immunol. 2016;197(4):1089–99.CrossRefPubMedPubMedCentral Dai Z, et al. CXCR3 blockade inhibits T cell migration into the skin and prevents development of alopecia areata. J Immunol. 2016;197(4):1089–99.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Estefan J, et al. Alopecia areata–Part II: diagnosis and pathology. Skinmed. 2015;13(2):121–6.PubMed Estefan J, et al. Alopecia areata–Part II: diagnosis and pathology. Skinmed. 2015;13(2):121–6.PubMed
25.
Zurück zum Zitat Tosti A, et al. The role of scalp dermoscopy in the diagnosis of alopecia areata incognita. J Am Acad Dermatol. 2008;59(1):64–7.CrossRefPubMed Tosti A, et al. The role of scalp dermoscopy in the diagnosis of alopecia areata incognita. J Am Acad Dermatol. 2008;59(1):64–7.CrossRefPubMed
26.
Zurück zum Zitat Ardigo M, et al. Reflectance confocal microscopy of the yellow dot pattern in alopecia areata. Arch Dermatol. 2011;147(1):61–4.CrossRefPubMed Ardigo M, et al. Reflectance confocal microscopy of the yellow dot pattern in alopecia areata. Arch Dermatol. 2011;147(1):61–4.CrossRefPubMed
27.
Zurück zum Zitat Hordinsky M, Donati A. Alopecia areata: an evidence-based treatment update. Am J Clin Dermatol. 2014;15(3):231–46.CrossRefPubMed Hordinsky M, Donati A. Alopecia areata: an evidence-based treatment update. Am J Clin Dermatol. 2014;15(3):231–46.CrossRefPubMed
28.
Zurück zum Zitat Hordinsky MK. Current treatments for alopecia areata. J Investig Dermatol Symp Proc. 2015;17(2):44–6.CrossRefPubMed Hordinsky MK. Current treatments for alopecia areata. J Investig Dermatol Symp Proc. 2015;17(2):44–6.CrossRefPubMed
29.
Zurück zum Zitat Pieri L, Guglielmelli P, Vannucchi AM. Ruxolitinib-induced reversal of alopecia universalis in a patient with essential thrombocythemia. Am J Hematol. 2015;90(1):82–3.CrossRefPubMed Pieri L, Guglielmelli P, Vannucchi AM. Ruxolitinib-induced reversal of alopecia universalis in a patient with essential thrombocythemia. Am J Hematol. 2015;90(1):82–3.CrossRefPubMed
30.
Zurück zum Zitat Higgins E, et al. Use of ruxolitinib to successfully treat chronic mucocutaneous candidiasis caused by gain-of-function signal transducer and activator of transcription 1 (STAT1) mutation. J Allergy Clin Immunol. 2015;135(2):551–3.CrossRefPubMed Higgins E, et al. Use of ruxolitinib to successfully treat chronic mucocutaneous candidiasis caused by gain-of-function signal transducer and activator of transcription 1 (STAT1) mutation. J Allergy Clin Immunol. 2015;135(2):551–3.CrossRefPubMed
31.
Zurück zum Zitat Kennedy Crispin M, et al. Safety and efficacy of the JAK inhibitor tofacitinib citrate in patients with alopecia areata. JCI Insight. 2016;1(15):e89776.CrossRefPubMedPubMedCentral Kennedy Crispin M, et al. Safety and efficacy of the JAK inhibitor tofacitinib citrate in patients with alopecia areata. JCI Insight. 2016;1(15):e89776.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Mackay-Wiggan J, et al. Oral ruxolitinib induces hair regrowth in patients with moderate-to-severe alopecia areata. JCI Insight. 2016;1(15):e89790.CrossRefPubMedPubMedCentral Mackay-Wiggan J, et al. Oral ruxolitinib induces hair regrowth in patients with moderate-to-severe alopecia areata. JCI Insight. 2016;1(15):e89790.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Patel H, et al. Association of cytotoxic T-lymphocyte antigen 4 (CTLA4) and thyroglobulin (TG) genetic variants with autoimmune hypothyroidism. PLoS One. 2016;11(3):e0149441.CrossRefPubMedPubMedCentral Patel H, et al. Association of cytotoxic T-lymphocyte antigen 4 (CTLA4) and thyroglobulin (TG) genetic variants with autoimmune hypothyroidism. PLoS One. 2016;11(3):e0149441.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Benhatchi K, et al. CTLA4 exon1 A49G polymorphism in Slovak patients with rheumatoid arthritis and Hashimoto thyroiditis-results and the review of the literature. Clin Rheumatol. 2011;30(10):1319–24.CrossRefPubMed Benhatchi K, et al. CTLA4 exon1 A49G polymorphism in Slovak patients with rheumatoid arthritis and Hashimoto thyroiditis-results and the review of the literature. Clin Rheumatol. 2011;30(10):1319–24.CrossRefPubMed
35.
Zurück zum Zitat Repnik K, Potocnik U. CTLA4 CT60 single-nucleotide polymorphism is associated with Slovenian inflammatory bowel disease patients and regulates expression of CTLA4 isoforms. DNA Cell Biol. 2010;29(10):603–10.CrossRefPubMed Repnik K, Potocnik U. CTLA4 CT60 single-nucleotide polymorphism is associated with Slovenian inflammatory bowel disease patients and regulates expression of CTLA4 isoforms. DNA Cell Biol. 2010;29(10):603–10.CrossRefPubMed
36.
Zurück zum Zitat Megiorni F, et al. Cytotoxic T-lymphocyte antigen 4 (CTLA4) +49AG and CT60 gene polymorphisms in alopecia areata: a case-control association study in the Italian population. Arch Dermatol Res. 2013;305(7):665–70.CrossRefPubMed Megiorni F, et al. Cytotoxic T-lymphocyte antigen 4 (CTLA4) +49AG and CT60 gene polymorphisms in alopecia areata: a case-control association study in the Italian population. Arch Dermatol Res. 2013;305(7):665–70.CrossRefPubMed
37.
Zurück zum Zitat Nusslein HG, et al. Efficacy and prognostic factors of treatment retention with intravenous abatacept for rheumatoid arthritis: 24-month results from an international, prospective, real-world study. Clin Exp Rheumatol. 2016;34(3):489–99.PubMed Nusslein HG, et al. Efficacy and prognostic factors of treatment retention with intravenous abatacept for rheumatoid arthritis: 24-month results from an international, prospective, real-world study. Clin Exp Rheumatol. 2016;34(3):489–99.PubMed
38.
Zurück zum Zitat Petukhova L, et al. The genetics of alopecia areata: what’s new and how will it help our patients? Dermatol Ther. 2011;24(3):326–36.CrossRefPubMed Petukhova L, et al. The genetics of alopecia areata: what’s new and how will it help our patients? Dermatol Ther. 2011;24(3):326–36.CrossRefPubMed
39.
Zurück zum Zitat Ali A, Martin JMT. Hair growth in patients alopecia areata totalis after treatment with simvastatin and ezetimibe. J Drugs Dermatol. 2010;9(1):62–4.PubMed Ali A, Martin JMT. Hair growth in patients alopecia areata totalis after treatment with simvastatin and ezetimibe. J Drugs Dermatol. 2010;9(1):62–4.PubMed
40.
Zurück zum Zitat Lattouf C, et al. Treatment of alopecia areata with simvastatin/ezetimibe. J Am Acad Dermatol. 2015;72(2):359–61.CrossRefPubMed Lattouf C, et al. Treatment of alopecia areata with simvastatin/ezetimibe. J Am Acad Dermatol. 2015;72(2):359–61.CrossRefPubMed
41.
Zurück zum Zitat Loi C, Starace M, Piraccini BM. Alopecia areata (AA) and treatment with simvastatin/ezetimibe: experience of 20 patients. J Am Acad Dermatol. 2016;74(5):e99–100.CrossRefPubMed Loi C, Starace M, Piraccini BM. Alopecia areata (AA) and treatment with simvastatin/ezetimibe: experience of 20 patients. J Am Acad Dermatol. 2016;74(5):e99–100.CrossRefPubMed
42.
Zurück zum Zitat Moreira FT, et al. Effects of two lipid lowering therapies on immune responses in hyperlipidemic subjects. Life Sci. 2014;98(2):83–7.CrossRefPubMed Moreira FT, et al. Effects of two lipid lowering therapies on immune responses in hyperlipidemic subjects. Life Sci. 2014;98(2):83–7.CrossRefPubMed
43.
Zurück zum Zitat Bolduc C, Bissonnette R. Safety and efficacy of adalimumab for the treatment of severe alopecia areata: case series of three patients. J Cutan Med Surg. 2012;16(4):257–60.CrossRefPubMed Bolduc C, Bissonnette R. Safety and efficacy of adalimumab for the treatment of severe alopecia areata: case series of three patients. J Cutan Med Surg. 2012;16(4):257–60.CrossRefPubMed
44.
Zurück zum Zitat Hernandez MV, et al. Development of alopecia areata after biological therapy with TNF-alpha blockers: description of a case and review of the literature. Clin Exp Rheumatol. 2009;27(5):892–3.PubMed Hernandez MV, et al. Development of alopecia areata after biological therapy with TNF-alpha blockers: description of a case and review of the literature. Clin Exp Rheumatol. 2009;27(5):892–3.PubMed
46.
Zurück zum Zitat Freyschmidt-Paul P, et al. Interferon-gamma-deficient mice are resistant to the development of alopecia areata. Br J Dermatol. 2006;155(3):515–21.CrossRefPubMed Freyschmidt-Paul P, et al. Interferon-gamma-deficient mice are resistant to the development of alopecia areata. Br J Dermatol. 2006;155(3):515–21.CrossRefPubMed
47.
Zurück zum Zitat Freyschmidt-Paul P, et al. The functional relevance of the type 1 cytokines IFN-gamma and IL-2 in alopecia areata of C3H/HeJ mice. J Investig Dermatol Symp Proc. 2005;10(3):282–3.CrossRefPubMed Freyschmidt-Paul P, et al. The functional relevance of the type 1 cytokines IFN-gamma and IL-2 in alopecia areata of C3H/HeJ mice. J Investig Dermatol Symp Proc. 2005;10(3):282–3.CrossRefPubMed
48.
Zurück zum Zitat Pham MN, von Herrath MG, Vela JL. Antigen-specific regulatory T cells and low dose of IL-2 in treatment of type 1 diabetes. Front Immunol. 2015;6:651.PubMed Pham MN, von Herrath MG, Vela JL. Antigen-specific regulatory T cells and low dose of IL-2 in treatment of type 1 diabetes. Front Immunol. 2015;6:651.PubMed
49.
Zurück zum Zitat Castela E, et al. Effects of low-dose recombinant interleukin 2 to promote T-regulatory cells in alopecia areata. JAMA Dermatol. 2014;150(7):748–51.CrossRefPubMed Castela E, et al. Effects of low-dose recombinant interleukin 2 to promote T-regulatory cells in alopecia areata. JAMA Dermatol. 2014;150(7):748–51.CrossRefPubMed
50.
Zurück zum Zitat Guttman-Yassky E, et al. Extensive alopecia areata is reversed by IL-12/IL-23p40 cytokine antagonism. J Allergy Clin Immunol. 2016;137(1):301–4.CrossRefPubMed Guttman-Yassky E, et al. Extensive alopecia areata is reversed by IL-12/IL-23p40 cytokine antagonism. J Allergy Clin Immunol. 2016;137(1):301–4.CrossRefPubMed
Metadaten
Titel
The Changing Landscape of Alopecia Areata: The Translational Landscape
verfasst von
Etienne C. E. Wang
Angela M. Christiano
Publikationsdatum
23.06.2017
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 7/2017
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-017-0540-9

Weitere Artikel der Ausgabe 7/2017

Advances in Therapy 7/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.