key: cord-0069943-6zj2qxlo authors: Boch, Katharina; Langan, Ewan A.; Kridin, Khalaf; Zillikens, Detlef; Ludwig, Ralf J.; Bieber, Katja title: Lichen Planus date: 2021-11-01 journal: Front Med (Lausanne) DOI: 10.3389/fmed.2021.737813 sha: 6079167f24a280abd366a91c829aa3c4651b2183 doc_id: 69943 cord_uid: 6zj2qxlo Lichen planus (LP) is a T cell-mediated disease affecting the stratified squamous epithelia of the skin and/or mucus membrane. Histologically, the disease is characterized by a lichenoid inflammatory infiltrate and vacuolar degeneration of the basal layer of the epidermis. LP has three major subtypes: Cutaneous, mucosal and appendageal LP. Rarely, it may affect the nails in the absence of skin and/or mucosal changes. LP may also be induced by several drugs, typically anti-hypertensive medication or be associated with infections, particularly viral hepatitis. The diagnosis is based on the clinical presentation and characteristic histological findings. Although the disease is often self-limiting, the intractable pruritus and painful mucosal erosions result in significant morbidity. The current first-line treatment are topical and/or systemic corticosteroids. In addition, immunosuppressants may be used as corticosteroid-sparing agents. These, however are often not sufficient to control disease. Janus kinase inhibitors and biologics (anti-IL-12/23, anti-IL17) have emerged as novel future treatment options. Thus, one may expect a dramatic change of the treatment landscape of LP in the near future. The term lichen planus (LP) stems from the Greek word "leichen, " which means "tree moss, " and the Latin word "planus, " which means "flat, " which aptly describes the surface of the cutaneous lesion (1) . LP is a group of chronic inflammatory diseases affecting stratified squamous epithelia. Recently, LP is perceived as a T cell-mediated autoimmune disease, in which cytotoxic CD8+ Tcells are recruited into the skin and subsequently lead to an interface dermatitis (2) (3) (4) (5) (6) (7) (8) . Viruses, drugs and contact allergens have all been reported to be possibly associated with development of LP (9) (10) (11) (12) (13) (14) (15) (16) (17) (18) (19) . Clinically, LP is hallmarked by characteristic lesions, affecting the skin, hair, nails and/or mucous membranes. The classical skin changes are pruritic, purple, polygonal, flat-topped (planar) papules crossed by fine white lines, while erosions are seen on the mucous membranes (Figure 1) . The latter may be associated with pain and/oral burning sensation (1) . An overview of clinical subtypes and rare variants are listed in Table 1 . LP preferentially affects middle-aged adults, with no known gender pre-disposition (1, 14) . Whilst the clinical features are relatively characteristic, histological confirmation of the diagnosis is recommended to exclude potential differential diagnoses. The typical band-like lymphocytic infiltrate and interface dermatitis are the characteristic findings-irrespective of skin location or disease subtype. In addition to routine histology, direct immunofluorescence (IF) microscopy may demonstrate C3 and/or IgG at the dermal-epidermal junction and deposition of IgM as so-called colloid bodies (20) . The overall goal of treatment is symptom control and resolution of the skin lesions. Selection of treatment should be based on the severity of the disease, the extent of the subjective symptoms, as well as taking into (14) . Cutaneous LP is usually self-limiting and resolves within 6 months in over 50% of patients and within 18 months in up to 85% of patients (14, 21) . By contrast, mucosal LP is often chronic and may be refractory to treatment (22, 23) . LP with hypertrophic cutaneous lesions and isolated nail or scalp involvement is also often chronic in nature. Persistent cutaneous and mucosal lesions are considered as a premalignant condition. Thus, patients should be followed up regularly for both, adjustment of treatment, and screening for the development of malignancies. The prevalence of LP is 0.89% in the general population and 0.98% in patients seeking dermatological care according to a recent meta-analysis of 46 studies (24) . The prevalence of cutaneous LP was reported to range between 0.2 and 1.0% of the adult population, and it is outnumbered by oral LP in most study populations (1, 9) . The incidence of LP is less well-characterized and displays considerable geographical heterogeneity as it ranges between 14 and 250 cases/100,000 person-years (25) (26) (27) (28) (29) . This variability more likely mirrors methodological differences in the sampled populations rather than the existence of an ethnic pre-disposition. Moreover, the aforementioned studies adopted various eligibility criteria and pooled patients with oral and cutaneous LP together. While oral LP affects females more frequently than males (24, 30) , cutaneous LP does not demonstrate a prominent sex predilection (21) . Cutaneous LP tends to manifest during the fifth and sixth decades of life, with almost two-thirds of patients presenting with the disease between the ages of 30 and 60 years (9, 31, 32) . Oral LP tends to develop 10 years later than cutaneous LP (33) . While no ethnic predilection is renowned in LP, a recent meta-analysis revealed that the pooled prevalence of oral LP was lower among patients of Asian ancestry (24) . The epidemiology of LP remains to be fully delineated as the current knowledge stems mainly from scattered small-scale retrospective studies. Given that the care of patients with LP spreads across different medical specialties, in both primary and specialized healthcare, precise estimation of its incidence and prevalence is methodologically challenging. The observation of familial LP (34) , the occurrence of LP in monozygotic twins (35) and HLA-based susceptibility association studies all point toward a genetic pre-disposition for LP. Several HLA alleles are associated with LP, for example between HLA-B27, HLA-B51, HLA-Bw57 (oral LP in English patients), HLA DR1 (cutaneous/oral LP), HLA-DR9 (oral LP in Japanese and Chinese patients), HLA DR6 (HCV-associated oral LP), and HLA DRB1 * 11 and DQB1 * 03 alleles (lichen planopilaris) (17, (36) (37) (38) (39) (40) . So far, only one genome-wide association study (GWAS) has been published in LP. In total, 261 patients with hepatitis C infection with (n = 71) or without (n = 190) LP were genotyped. The findings were validated in a small group of patients (n = 45), of which only 7 were affected by LP. In addition to the association with the HLA, single-nucleotide polymorphisms (SNP) in loci encoding for NRP2 and IGFBP4 that increase or reduce risk of LP association, respectively, were found (37) . Recently, a phenomewide association study confirmed the HLA association in LP and additionally found two additional SNPs to be associated with LP. These SNP encode for three genes: TSBP1, HCG23, and BTNL2 (41). Further gene associations had been described for several cytokines (IFN-γ, TNF, TNFαR, IL-4, IL-6, IL-18) and others (NFκB, PGE2, Prothrombin) (40). Several environmental factors have been implemented to trigger LP. Systemic viral infection, such as hepatitis C, may modify self-antigens on the surface of basal keratinocytes, or alter the immune balance, promoting a lichenoid inflammation (15) (16) (17) (18) 42) . The association between LP and hepatitis C has recently been substantiated in a large cohort study. Here, the prevalence of chronic inflammatory skin disease, including LP, was contrasted in over 23,000 patients with hepatitis C and a 3-fold greater number of non-hepatitis controls. In this study, the adjusted hazard ratio (HR) for subjects with hepatitis C to develop LP was 13.14 (95% CI: 7.10-24.31), indicating a significantly higher risk to develop LP for patients with hepatitis C. Among all evaluated chronic inflammatory skin diseases, the HR to develop LP for patients with hepatitis C was the second highest (43) . Other viruses that are associated with triggering LP are members of the human herpesvirus (HHV) family, specifically (HHV)-6 and HHV-7 (44) . However, studies relating to HHV-6 were not validated in other studies (45) . Moreover, localized skin disease due to herpes simplex, varicella zoster, or human papilloma virus 16 (46-52) may cause LP. There are also reports that vaccine administration, including influenza and hepatitis B virus vaccines, may be associated with the development of LP (53) . Additional environmental factors have been implicated in the development of oral LP. These include changes in the oral microbiome (e.g., Candida sp., various other bacterial infections) and dental metals precipitating allergic contact reaction (54) (55) (56) (57) . In line with this observation, the diversity of the skin bacterial communities may be involved in LP pathogenesis. Under steady-state conditions, a low diversity of bacterial communities on the skin are associated with an increased expression of proinflammatory cytokines (TNFα and CXCL1) and CD11c, pointing toward an increased infiltration with macrophages (58) . These cytokines, as well as macrophages are also found in lesional LP skin (59) (60) (61) . Thus, a low diversity of cutaneous bacterial communities may generate a pro-inflammatory state, even under steady-state conditions, that shares features of LP ( Figure 2) ; thereby, potentially lowering the threshold for LP to develop. Among metals that may be associated with oral LP include amalgam (mercury), copper, and gold. Drugs may also elicit lichenoid-like reactions, which may be both clinically and histologically indistinguishable from classic LP. The most commonly implicated drugs ( Table 2 ) are angiotensinconverting enzyme inhibitors, thiazide diuretics, antimalarials, anti-inflammatory drugs, antimicrobials, antihypertensives, psychiatric drugs, antidiabetics, PD-1-inhibitors, quinidine, penicillamine, and metals (62) (63) (64) (65) . Another peculiar potential environmental trigger for LP is UV-filters in sunscreens and hair-care products that have been noted to be associated with frontal fibrosing alopecia and lichen planopilaris (66, 67) . Most of the findings on LP pathogenesis are based on morphology. Only a limited number of studies also demonstrated a functional impact of cells and/or molecules on LP pathogenesis. A cell-mediated immune response is at the core of LP pathogenesis, with cytotoxic, CD8+ T-cells in the center (Figure 2 ). Yet, both CD4+ and CD8+ T-cells accumulate in the dermis and oral mucosa, whilst a CD8+ T-cell-dominant infiltrate is seen within the epidermis (3, 4) . Other groups have reported that CD8+ and CD45RO+ T-cells are the major cell type in the inflammatory infiltrate and that the T cell receptor (TCR) αβ, and to a lesser extent TCR γδ, are expressed (68) . The functional contribution of T-cells to LP pathogenesis is further supported by a recent study that showed granule exocytosis with the release of perforin and granzyme B. In this context, to a lesser extent, the Fas/Fas-ligand system appears to be involved, the main pathway of cytotoxicity by CD4+ and CD8+ T-cells in humans (5) . In addition to T-cells, mast cells may contribute to LP pathogenesis given that they are often found in the inflammatory infiltrate and show signs of activation (69) (70) (71) (72) . Immunohistochemistry of oral LP also demonstrated the presence of dendritic cells (73) . The fact that CD8+ Tcells and mast cells are detected in lesions of LP patients led to the conclusion that non-specific mechanisms like mast cell degranulation and protease activation are involved in the pathogenesis of LP. These mechanisms may combine to cause Tcell accumulation in lesions and induce keratinocyte apoptosis (74) . In line, an increased protease expression has been described in LP lesions that potentially contributes to the disruption of the basement membrane gelatinases (e.g., MMP-2, MMP-7, and−9), chymase, tryptase, capthepsins and caspase-3 (74) (75) (76) (77) (78) (79) . Several alterations in the expression of cytokines and chemokines in lesions or serum of patients with LP have been described. Serum levels of interleukin (IL)-5, IL-6, IL-8, IL-9, IL-10, IL-12 IL-17, IL-22, tumor necrosis factor-α, transforming growth factor-β, interferon (IFN)-γ, CXCR-3, CXCR-4, CXCL-10, CXCL-12, CCR1, CCR3, CCR4, CCL5-CCR5, and CCL17-CCR4) have been found elevated (80) (81) (82) (83) (84) (85) (86) (87) (88) (89) . In addition, an increased expression IFN-γ and IL-17 in the skin of LP lesions has been described (81, 90)-albeit some other studies refuted these observations (91) . Case reports indicated that off-label treatment of LP with Janus kinase (JAK) inhibitors (JAKi), such as tofacitinib, led to marked improvement of the disease (92) (93) (94) . As IFN-γ-induced signaling centers on the activation of JAK (95), IFN-γ and JAK are likely to be central to the pathogenesis of LP. Functional evidence for a pathogenic contribution of IL-17, including the IL-17 pathway, stems from the observation of increased IL-17 and IL-23 expression in LP (84) , as well as the clinical improvement following off-label treatment of LP patients with the anti-IL-17 antibody secukinumab, or the IL-12/23-targeting ustekinumab or the IL-23 inhibitor guselkumab. Of note, clinical improvement of LP following IL-17 or IL-23 blockade was accompanied by a strong reduction of the Th1 and Th17/Tc17 cellular mucosal and cutaneous infiltrates (96) . This supports the previously mentioned notion that these T-cell subsets may be key effector cells in LP. The relative lack of functional insights into LP pathogenesis may be due to the limited number of pre-clinical model systems. So far, only one mouse model has been established that resembles aspects of LP pathogenesis. This model is based on the intradermal transfer of autoreactive CD4+ T-cells producing IFN-γ and TNF into syngeneic mice, inducing cellular infiltrates Frontiers in Medicine | www.frontiersin.org FIGURE 2 | Schematic overview of lichen planus pathogenesis. Based on the increasing evidence of the autoimmune nature of lichen planus (LP), its pathogenesis may be divided in two distinct phases: Afferent phase, where tolerance to autoantigens is lost, and the efferent phase that is characterized by a T cell-driven skin inflammation. (1) Under steady state conditions the diversity of the cutaneous microbiome can alter the "inflammatory state" of the skin. If the diversity of the bacterial communities on the skin is low, this is associated with an increased expression of pro-inflammatory cytokines such as TNFα and CXCL1, as well as an increase of CD11c, suggesting an increased presence of macrophages. (2, 3) Following this presumed injury, the loss of tolerance in LP occurs in the context of an association with the MHC that may additionally be shaped by associated viral infections and other environmental factors. The site of the APC/T-cell interaction is so far unknown. It may occur locally and/or in skin draining lymph nodes. (4) Next, autoreactive T cells reach the skin by extravasation from the blood vessels. (5) Within the skin, T cells become activated by binding to the specific autoantigens. Effector functions of cytotoxic T cells is mediated by (6) proteases, such as granzyme B, as well as perforin. In addition, (7) mast cells become activated and may further aggravate inflammation in LP. However, the precise sequence of events and their interactions are only incompletely understood. with epidermotropism with basal vacuolar degeneration and colloid bodies (2) . Furthermore, desmoglein (Dsg) 3-specific Tcells are also capable of inducing histologically LP-like changes (97) . Transfer of Dsg3-specific T-cells into immunodeficient mice induced an interface dermatitis (a distinct form of T-cellmediated autoimmunity) in mice. The induction of the interface Besides systemic viral infection, several other diseases were shown to be associated with LP. A high prevalence of thyroid disease is found amongst patients with oral LP (98) , whereas the association between LP and diabetes mellitus is less wellestablished (99) . In addition to the association with chronic inflammatory diseases, patients with LP present a higher risk for dyslipidaemia, which could be explained by the cytokines involved in the pathogenesis of the disease, such as TNF-α, IL-6, IL-10, and IL-4 (100, 101) . Autoimmune diseases such as alopecia areata, ulcerative colitis, vitiligo, morphea, lichen sclerosus and myasthenia gravis are over-represented in patients with LP (14) . Over 20 different clinical manifestations of LP are described ( Table 1) . Herein, we focus on the most common variants, as well as LP pemphigoides, lichenoid GVHD and lichenoid drug eruptions. The hallmark of cutaneous LP are purple or violet, polygonal, shiny, flat-topped, firm, papules, and plaques with white streaks (Wickham striae) (40) . Wickham striae are best visualized by dermoscopy (102, 103) . The cutaneous lesions may vary in size from several millimeters to more than one centimeter. The lesions may be clustered or disseminated and whilst the typical locations are the wrists, lower back, and ankles, a distribution in photo-exposed areas is also well-recognized (Figures 1a-c) . Skin conditions may also appear following the lines of trauma (isomorphic response, Figure 1f ). The dominant subjective symptom is pruritus, which may be severe and refractory to standard anti-pruritic therapies. The typical lesions of mucosal LP are painful and persistent erosions (erosive LP) or diffuse erythema and peeling of the mucosa (desquamative LP) (40) . In addition, Wickham striae may be present in a lacy or fern-like pattern. Mucosal LP can be further subclassified into oral LP, affecting the buccal mucosa, the tongue, and to a lesser extend the gums and lips (Figures 1m,n) or genital LP, affecting the glans penis, labia majora, labia minora and vaginal introitus (Figures 1p-r) . Chronic disease may result in scarring, with the formation of adhesions, resorption of labia minora and ultimately introital stenosis. Penile LP usually presents with papules around the glans penis, white streaks and erosions. In rare cases, mucosal LP may also affect the lacrimal glands, eyelids, external ear canal, esophagus, larynx, bladder, and anus. Lichen planopilaris (LPP) presents as tiny red spiny follicular papules and extending smooth areas on the scalp or less often, elsewhere on the hair-bearing regions body areas (104, 105) . Destruction of the hair follicles leads to permanently bald patches characterized by sparse "lonely hairs" (Figures 1i,j) . Frontal fibrosing alopecia is a variant of LPP that affects the anterior scalp, forehead and eyebrows. Another subtype of LPP is Graham-Little-Piccardi-Lasseur Syndrome with the following characteristics: multifocal, patchy, cicatricial alopecia present on the scalp, non-cicatricial alopecia of the axillae, non-cicatricial alopecia of the perineum, and follicular hyperkeratosis of the trunk and extremities (106) . LP may affect one or more nails (Figure 1l) , sometimes in the absence of skin involvement. LP thins the nail plate, which may become grooved and ridged. The nail may darken, thicken or lift off the nail bed (onycholysis). Sometimes, the cuticle is destroyed and forms a scar (pterygium). The nails may shed or stop growing altogether, and they may rarely, completely disappear (anonychia). An important clinical feature of nail LP is the occurrence of a dorsal pterygium. LP pemphigoides is clinically characterized by the simultaneous occurrence of lichenoid and bullous skin lesions. By some, LP pemphigoides is considered as an autoimmune dermatosis with autoimmunity toward type XVII collagen (COL17). By contrast, others consider LP pemphigoides the co-occurrence of 2 independent skin diseases, or as a variant of LP (14, 107, 108) . Graft-vs.-host disease (GVHD) is the primary complication of allogeneic bone marrow transplantation and the skin is the most commonly involved organ. The clinical picture varies and often is similar to autoimmune or inflammatory diseases. Cutaneous GVDH can imitate classical lichen planus with purple, polygonal, pruritic papules (but without Wickham striae) or lichen planus pigmentosus (109, 110) . GVHD refers to the inflammatory manifestations, when immunocompetent T-cells from a donor recognize and react against "foreign" tissue antigens in an immunocompromised host, this autoreactive pre-condition leads to a Th2 immune response induced interface dermatitis (109) . Lichenoid drug eruptions often mimic idiopathic lichen planus although there can be features that may help to distinguish them, which may include: symmetrical rash on the trunk and limbs, predominantly in sun-exposed areas. Skin features do normally not show Wickham striae, nail and mucous membrane involvement is missing. Medications reported to trigger a lichenoid drug eruptions are, exemplary (14) : ACE inhibitors, beta-blockers, nifedipine, methyldopa, hydrochlorothiazide, frusemide, spironolactone, non-steroidal anti-inflammatory drugs (NSAIDs), carbamazepine, phenytoin, ketoconazole, 5-fluorouracil, imatinib, hydroxychloroquine, sulfonylurea, dapsone, mesalazine, sulfasalazine, allopurinol, iodides and radiocontrast media, interferon-α, omeprazole, penicillamine, tetracycline, infliximab, etanercept, adalimumab, imatinib, misoprostol, sildenafil, and herpes zoster/influenza vaccines. Contact allergies also may mimic lichen planus: Oral lichenoid lesions may be associated to type-IV-sensitization to mercury or dental amalgam (111, 112) ; lichenoid skin lesions usually result from contact with rubber, chemicals used in clothing dyes or chemicals in wine industries (113) . Histopathology A skin/mucosal biopsy is recommended to confirm the diagnosis of LP. The typical histological findings are acanthosis and hyperkeratosis, wedge-shaped hypergranulosis, vacuolic degeneration of the basal layer, alteration or loss of rete ridges resulting in a sawtooth appearance and a dense, band-like lymphocytic infiltrate in the upper dermis along the dermalepidermal junction (Figures 1h,k,o,s) . Apoptotic keratinocytes are often seen near the basal layer and are termed colloid bodies. For LP affecting the scalp, for example LPP, shows beside the penitent LP features often the destruction of hair follicle root sheaths and follicular plugging as well as the loss of sebaceous glands as well (114) (Figure 1k) . Additionally, a lesional biopsy for direct IF microscopy can be a useful, especially when trying to differentiate between LP and other autoimmune diseases, such as pemphigus vulgaris, mucous membrane pemphigoid, or lupus erythematosus (LE) (115, 116) . In LP, direct IF microscopy (Figure 1g ) may reveal globular deposits of IgA, IgM, IgG, C3, or fibrinogen mixed with apoptotic keratinocytes (117, 118) . The differential diagnosis of cutaneous LP is broad and includes graft-vs. host-disease, psoriasis vulgaris, guttate psoriasis, secondary syphilis, pityriasis lichenoides, pityriasis rosea, lichen nitidus, lichen simplex chronicus, lichen sclerosus, lichen striatus, linear epidermal naevus, eczema, prurigo nodularis, erythema dyschromicum perstans, eczematid-like purpura, drug eruption, granuloma annulare, lichen amyloidosus, Kaposi sarcoma and lupus erythematosus. In most cases, histology permits a reasonable differentiation between these diseases and inflammatory disorders. Similarly, an extensive list of differential diagnoses should be considered when diagnosing LP of the oral cavity including pemphigus vulgaris, mucous membrane pemphigoid, lupus erythematosus, secondary syphilis, traumatic patches, and candidiasis. Vulval/penile LP can be difficult to distinguish from lichen sclerosis, mucous membrane pemphigoid, psoriasis, intraepithelial neoplasia, graft-vs.-host disease, erosive dermatitis, and intertrigo. Histology and direct IF microscopy should allow a definite diagnosis of LP and the exclusion of other diseases. Various diseases may appear similar to LPP, especially when the destruction of the hair follicles leads to permanently bald or even scarring patches without inflammation or tiny red spiny follicular papules, such as patchy alopecia in systemic LE, alopecia areata, diffuse alopecia due to secondary syphilis or severe folliculitis. Brunsting-Perry cicatricial pemphigoid is rare variant of mucous membrane pemphigoid associated with scarring on the head and neck region. Differentiating them can be difficult, besides punch biopsy trichometric analysis, fungal culture, blood tests are recommended to find the underlying medical condition. Nail LP can be challenging to differentiate from psoriasis, atopical dermatitis, alopecia areata and onychomycosis. For the later, appropriate laboratory testing for presence of fungi is recommended. The ultimate aim of treatment is the resolution of the skin lesions and their associated symptoms. This is particularly important in oral LP where painful erosions can result in significant malnutrition and weight loss. Drug-induced LP should always be considered and excluded prior to commencing immunosuppressive therapy (14) and the responsible drug discontinued or substituted. An LP-associated diseases should be checked in each patient. Hypertrophic and mucosal LP lesions are potentially premalignant and regular follow-up and biopsies should be considered to exclude malignant transformation ( Table 3) . The first-line treatments for limited LP are (super)potent topical steroids, with intralesional steroid injection reserved for hypertrophic and/or unresponsive lesions (14, 119, 120) . For disseminated disease, systemic corticosteroids can be considered, either as oral therapy or intravenous "pulse" therapy, to achieve disease control. Thereafter, the oral dose can be tapered or the interval between intravenous administrations extended (14, 121, 122) . Other first-line therapies include systemic retinoids (acitretin/isotretinoin) or cyclosporine (14, (123) (124) (125) (126) . If diffuse cutaneous LP remains unresponsive, second-line therapy should be considered. These include sulphasalazine, and phototherapy such as broadband/narrowband UVB or psoralen and UVA (PUVA), and the combination of UV/PUVA with retinoids (14, 121, (127) (128) (129) (130) . For topical treatment of limited and diffuse cutaneous LP calcineurin inhibitors can be used to reduce side-effects of topical steroids (14, 131) . Third-line treatments include hydroxychloroquine, azathioprine, methotrexate, mycophenolate mofetil, or biologics targeting IL-12/23 (14, 121, 130, (132) (133) (134) (135) (136) . Based on the fact that in LP proinflammatory signaling pathways result in T-cell-dependent immune response, oral JAKi may represent a future treatment option (137) . Oral antihistamines may be helpful to minimize the itch (14) . Topical antipruritic agents such as menthol, camphor, or polidocanol can be prescribed as an adjuvant to the main treatment (14) . The majority of patients with cutaneous lesions spontaneously clear within 12-24 months (21); however, relapses are common. Healing may also be complicated by the development of post-inflammatory hyperpigmentation (1, 9) . Mucosal LP is often difficult to treat, particularly when extensive erosions are present. Long-term follow-up is necessary to monitor disease activity and to exclude malignant transformation of erosive lesions (14) . The mainstay of treatment of mucosal LP are topical corticosteroids (14, 138) . Superpotent steroids can be applied topically (in the form of an adhesive paste) twice daily for 1-2 months, and then administered as required (14) . Intralesional steroid injections are worth considering when lesions are particularly painful and fail to respond to topical therapy (14, 138, 139) . Systemic corticosteroids are reserved for patients with severe erosive mucosal LP (recalcitrant, multi-site, ulcers) and to more rapidly induce a remission (14) . Further systemic first-line treatments are retinoids (acitretin/isotretinoin) and cyclosporine (14) . Second-line treatments include sulphasalasine, azathioprine, hydroxychloroquine, methotrexate, mycophenolate mofetil, and/or use of topical calcineurin inhibitors (14, 133, (140) (141) (142) (143) (144) (145) (146) (147) . Third-line treatment may include cyclophosphamide, thalidomide, metronidazole, trimethoprim-sulphomethoxazole, antibiotic treatment, itraconazole, griseofulvin, dapsone and extracorporeal photochemotherapy (ECP) (14, (148) (149) (150) (151) (152) (153) (154) (155) (156) . In the management of oral LP, lidocain solution as mouthwash may be helpful to reduce pain. Amphotericin B solution as mouthwash several times daily (after food consumption) may prevent secondary candida infection. Patch tests may be recommended for patients with oral lichen planus affecting the gums and who have fillings with amalgam, to assess for contact allergy to thiomersal, a mercurial compound (14) . Mucosal LP may clear spontaneously within 5 years, but typically it is a chronic disease with a remitting and relapsing course (22, 23) . The general principles of the management of genital LP are similar to those of LP confined to the oral mucosa (14) . Most cases of papulosquamous genital LP are self-limited, and treatment with emollients and mid-potency steroids for a few weeks leads to complete remission. First-line treatment for erosive LP of the vulval or penile mucosa are superpotent topical corticosteroids (14, 157) , which can be gradually tapered (14) . Calcineurin inhibitors (tacrolimus/pimecrolimus) are a further topical treatment option (14) . The aim for the treatment of erosive genital lesions is the prevention or limitation of scarring. In women, synechia formation with vaginal stenosis may be prevented by the use of vaginal dilators and application of intra-vaginal steroids to treat mucosal inflammation (14) . In uncircumcised men, circumcision is usually recommended to avoid phimosis (14) . Local anesthetic gel, low-dose tricyclic antidepressants or anticonvulsants may relieve itch and ease discomfort and nystatin cream can prevent secondary fungal infections (14) . The aim of the treatment is disease control to prevent permanent hair loss due to scarring (14) . Furthermore, treatment can reduce itching and burning of the scalp. Topical steroids are treatment of first choice (97, (158) (159) (160) . Intralesional steroid injections may improve response rates (161) . Topical calcineurin inhibitors may be used as monotherapy or as an adjuvant to systemic therapy proved effective (162) . Systemic steroids are the mainstay of treatment for rapidly progressive disease to prevent scarring (163) , while introducing cyclosporine, methotrexate, or hydroxychloroquine as steroid sparing agents (160, (164) (165) (166) (167) (168) . Suggested second-line options are retinoids (acitretin/isotretinoin), tetracycline/doxycycline, mycophenolate mofetil, adalimumab, pioglitazone, thalidomide, or rituximab (160, 162, 165, 167, (169) (170) (171) (172) (173) (174) (175) . LP of the nails is generally difficult to treat and the prognosis is poor (14) . LP affecting the nails frequently leads to permanent destruction of the nail matrix and bed with functional limitations. Therefore, early treatment is essential, even in mild cases of nail LP (176) . Potent topical steroids under occlusive dressings are the preferred, first-line topical treatment (14) . Due to the poor short-term efficacy of topical steroids and long-term side effects, triamcinolone acetonide injections (intralesional) should be considered as further first-line therapies (176, 177) . Oral prednisone 0.5 mg/kg for 3 weeks demonstrated a marked improvement and is useful when multiple nails are affected (14) . Oral retinoids are second-line choices (178) (179) (180) , and immunosuppressive agents may also be considered (14, 181, 182) . In a case series, topical tacrolimus ointment 0.1% was successfully used in treatment of nail LP (183) . Corticosteroids are the backbone the treatment of cutaneous lichenoid GVHD, but ∼30% need additional immunosuppressant such as cyclosporine. cyclophosphamide, methotrexate, azathioprine, mycophenolate mofetil, pentostatin, or high-dose thalidomide and hydroxychloroquine (184) . Another option for skin involvement might be phototherapy, while extracorporeal photochemotherapy may improve cutaneous as well as systemic involvement (184) . The triggering agent should be stopped (14) ; improvement of the skin lesions can take weeks to months. Commonly flat pigmented freckles persist and fade more slowly. Steroids (topical/systemically) may be supportive to give relief or rapid resolution. In LP, new therapeutic options currently stem from case reports and/or case report series. These have set the rationale for the planning of current clinical trials in LP ( Table 4 ). The molecular targets currently persued for LP can be categorized into biologics targeting cytokines and small molecules blocking intracellular signaling. In addition, photodynamic therapy has consistently been reported to have favorable outcomes in LP patients (185) . Currently licensed (for other indications than LP) biologics targeting IL-17 or the IL-17R are secukinumab, ixekizumab and brodalumab. In 2017, occurrence of oral LP was noted in a psoriasis patient treated with secukinumab. As concurrently oral candidiasis, a relatively common adverse event under anti-IL-17 treatment, was present, the causality of IL-17 inhibition and induction of oral LP remained ambiguous (186) . In addition to this case, 3 more cases of cutaneous/oral lichenoid eruptions associated with IL-17 inhibition were noted, albeit (oral) LP was not formally diagnosed (187) (188) (189) . By contrast, response to IL-17 inhibition has been reported in a total of 5 LP patients (96, 190, 191) . Grounded on the latter observations, as well as the increased serum and tissue IL-17 expression in both oral and cutaneous LP (83, 192, 193) , two clinical studies currently evaluate the impact of IL-17 inhibition using secukinumab or ixekizumab in patients with LP (NCT04300296, NCT05030415). Biologics targeting either IL-12 and/or IL-23 (ustekinumab) or IL-23 alone (risankizumab, tildrakizumab and guselkumab) have so far not been associated with the induction of LP. However, one report noted a failure of LP to respond to ustekinumab (194) . Later observations noted a response of IL-12/23 inhibition on one A search on clinicaltrials.gov was conducted on September 25th, 2021 with the key word "lichen planus". A total of 116 clinical studies were identified. We focused on studies with a molecular target. A total of 14 completed interventional studies with molecular targets were updated since 2020. In addition, 2 studies with a defined molecular target (status: not recruiting/recruiting, enrolling by invitation/active, not recruiting) were found. We also included completed (with updates since 2020) and ongoing studies using photodynamic therapy. Number in brackets in the Status column indicate the last year, the study was updated in clincaltrials.gov. CIU, Chronic idiopathic urticarial; LP, Lichen planus; PDT, Photodynamic therapy. patient with LP pemphigoides (195) , and three patients with LP (96, 196) . Use of IL-12/23 was grounded on the observation of increased IL-23 expression in oral LP (84) , as well as an increased serum concentration of IL-23 in LP patients (197) . Of note, we are not aware of any study addressing the impact of IL-12/23 inhibition in LP. By contrast to IL-17 and IL-23, blockade of TNF-α has not emerged as a promising therapeutic target in LP. There are several reports on lichenoid drug eruptions following TNFα inhibition (198, 199) , and only one report on a successful treatment of lichen planus with the anti-TNF-α antibody adalimumab (135) . On the expression level, increased TNFα expression has been noted in the skin and serum of LP patients (60, 90) . In line, a study evaluating the impact of the TNF-α inhibitor etanercept of LP was terminated due to slow recruitment in 2018 (NCT00285779). Recent work showed that the inflammation in LP is dominated by an IFN-γ and an IL-21 signature, along with an increased expression of phospho-STAT1 in the dermal infiltrate (200) . In another T-cell mediated inflammatory skin disease, namely alopecia areata, the identification of an IFN gene signature in affected skin identified JAK inhibitors as potential new treatments for alopecia areata, which showed efficacy in phase 2 clinical trials (201, 202) . Based on these morphological observations and considerations, the authors concluded that use of JAK inhibitors may be beneficial in LP (200) . Successful treatment of a treatment-refractory LP patient with the JAK1/3-selective JAKi tofacitinib supports this notion. Furthermore, in 2 independent case series, tofacitinib used as either monotherapy or adjunctive therapy led to clinical improvement in 11/13 patients (95, 96) . In line with these observations, a clinical trial currently investigated the impact of topical ruxolitinib in LP patients. The trial was completed in 2020. Results are shown at clinicaltrial.gov: 12 patients were enrolled, 3 were lost to follow-up, most likely related to the Covid-19 pandemic, and no serious adverse events occurred (NCT03697460). Grounded on the broad anti-inflammatory activity of the PDE4 inhibitor apremilast, the safety and efficacy of the drug in LP patient's refractory to topical corticosteroid treatment was evaluated in an investigator-initiated, single-center, nonrandomized, open-label, pilot study in 2013. Patients were treated with 2 × 20 mg apremilast per day for 12 weeks. The primary endpoint was achieving a 2-grade or more in Physician Global Assessment (PGA) at 12 weeks. While all patients demonstrated a significant clinical improvement, 3/10 met the primary endpoint (137) . Subsequently, a total of 5 LP patients, mostly with treatment-refractory disease, were reported to improve when treated with apremilast (203) (204) (205) . Based on these reports, apremilast is currently evaluated in a randomized placebo-controlled clinical trial in women with genital erosive lichen planus (206) . Currently, patients are recruited to this study (NCT03656666). Other clinical trials are evaluating the impact of topical mechlorethamine, a topical chemotherapy used for the treatment of cutaneous T cell lymphoma (207) , in LP. The study was completed in 2019, but so far results have not been published (NCT03417141). A study treating LP patients with the opiate opioid receptor antagonist naltrexone was recently completed (NCT04409041). Again, results are pending to be published. In this line, a small case series reported on the beneficial outcome of naltrexone in LPP (208) . Most of non-pharmacological interventions for LP that are currently evaluated in clinical trails focus on the use of photodynamic therapy (PDT) (NCT04976673, NCT01282515). Both studies are completed, while the results have not been published so far. Up to date, a total of 5 controlled studies has addressed the impact of PDT in oral LP (209) (210) (211) (212) (213) . In most studies, topical steroids were used as an active comparator. In 3/5 studies, no difference between the 2 treatment modalities (that both led to a reduced severity of LP) was observed (209, 211, 212) , whilst in 2/5 studies, a superior effect of PDT was noted (211, 213) . In an open study using PDT in oral LP, a significant change of molecular disease markers (reduced numbers of CD4+ and CD8+ T-cells in the lesions, reduced numbers of activated T cells in the circulation) were observed in parallel to the clinical improvement (214) . In addition, a recent retrospective investigation and review of the impact of narrowband UVB phototherapy and psoralen plus UVA (PUVA) photochemotherapy as second-line treatment of LP showed a relatively good response (complete responses in a little over 70% for both narrow-bad UVB and PUVA), whereby adverse events were only observed in patients treated with oral PUVA (215) . Previous reports also indicated a good response of recalcitrant LP to extracorporeal photochemotherapy (ECP) (155, 216, 217) . In a larger case series, 9/12 patients showed complete remission and 3/12 a partial response. In follow-up, relapse occurred frequently when ECP sessions were less frequent or stopped (216) . Since 2010, no more reports on the use of ECP in LP were published. Hence, ECP may be used in LP cases refractory of several previous therapies, and additional treatments should be administrated to maintain the (presumed) initial good response to ECP. Overall, LP is an under-recognized dermatosis, whose epidemiology and pathogenesis is only partially understood, the disease is associated with significant morbidity, and current treatment options are limited in their success. Given the lack of double-blind randomized control trials, treatment is often based on clinical experience and the results of retrospective metaanalyses (121, 218) . Biological treatments (93) and JAKi (96) hold significant promise as future therapeutic options. The lack of animal models underscores the importance of a comprehensive understanding of the pathogenesis of LP elucidating human phenotype-genotype correlations facilitating renewed efforts to unravel the cellular and molecular changes underlying the disease (219) . Still, with the emergence of biological treatment options and of JAKi that both derived from careful clinical observations, the treatment landscape of LP will hopefully improve in the near future. KBo, EL, KK, RL, and KBi wrote the manuscript. All authors read, commented, and approved the final version of the manuscript. Lichen planus Lichenoid tissue reaction (LTR) induced by local transfer of Ia-reactive T-cell clones. II LTR by epidermal invasion of cytotoxic lymphokine-producing autoreactive T cells Autocytotoxic T-cell clones in lichen planus Induction and control of lichenoid tissue reactions Granule exocytosis, and not the fas/fas ligand system, is the main pathway of cytotoxicity mediated by alloantigenspecific CD4(+) as well as CD8(+) cytotoxic T lymphocytes in humans Lichenoid tissue reaction/interface dermatitis: clinical and histological perspectives Indoleamine 2,3-dioxygenase (IDO): the antagonist of type I interferon-driven skin inflammation? T-cell-mediated injury to keratinocytes: insights from animal models of the lichenoid tissue reaction Clinical practice. Lichen planus Shugaa-Addin B. Hepatitis C virus infections in oral lichen planus: a systematic review and meta-analysis Oral Lichen planus Lichenoid drug eruptions Lichen planus-like eruptions: an emerging side effect of tumor necrosis factor-alpha antagonists European S1 guidelines on the management of lichen planus: a cooperation of the European Dermatology Forum with the European Academy of Dermatology and Venereology Hepatitis C virus and lichen planus: a case-control study of 340 patients Hepatitis C virus RNA detection in oral lichen planus tissue Increased frequency of HLA-DR6 allele in Italian patients with hepatitis C virus-associated oral lichen planus Oral lichen planus pathogenesis: a role for the HCV-specific cellular immune response Direct Immunofluorescence Studies in Lichen Planus Long-term follow-up of lichen planus Oral lichen planus: different clinical features in HCVpositive and HCV-negative patients Retrospective analysis of the clinical characteristics and patient-reported outcomes in vulval lichen planus: results from a single-center study Global prevalence and incidence estimates of oral lichen planus: a systematic review and meta-analysis Incidence rate of oral lichen planus among Indian villagers Incidence rates for oral leukoplakia and lichen planus in a Japanese population The incidence of molluscum contagiosum, scabies and lichen planus Epidemiology, clinical features, and associated factors in 78 cases of lichen planus on black skin Incidence of Lichen planus and subsequent mortality in finnish women Oral lichen planus-a demographic study Clinical epidemiology and treatment of lichen planus: a retrospective review of 2 tertiary care centers Clinical variants of lichen planus Course of oral lichen planus: a retrospective study of 808 northern Italian patients Familial lichen planus Lichen planus in monozygotic twins Lichen planopilaris is associated with HLA DRB1 * 11 and DQB1 * 03 alleles Genome-wide association study identifies risk variants for lichen planus in patients with hepatitis C virus infection The association between HLA DR, DQ antigens, and vulval lichen sclerosus in the UK: HLA DRB112 and its associated DRB112/DQB10301/04/09/010 haplotype confers susceptibility to vulval lichen sclerosus, and HLA DRB10301/04 and its associated DRB10301/04/DQB10201/02/03 haplotype protects from vulval lichen sclerosus The vulvovaginal gingival syndrome: a severe subgroup of lichen planus with characteristic clinical features and a novel association with the class II HLA DQB1 * 0201 allele Cutaneous and mucosal lichen planus: a comprehensive review of clinical subtypes, risk factors, diagnosis, and prognosis Phenomewide association study maps new diseases to the human major histocompatibility complex region Hepatitis C virus and lichen planus: a reciprocal association determined by a meta-analysis Association between hepatitis C virus infection and subsequent chronic inflammatory skin disease Lichen planus remission is associated with a decrease of human herpes virus type 7 protein expression in plasmacytoid dendritic cells Association of human herpes virus 6 infection with lichen planopilaris Cutaneous reactions at sites of herpes zoster scars: an expanded spectrum Detection of varicella-zoster virus antigens in lesional skin of zosteriform lichen planus but not in that of linear lichen planus Peripheral and local human papillomavirus 16-specific CD8+ T-cell expansions characterize erosive oral lichen planus Pathomechanisms of lichen planus autoimmunity elicited by cross-reactive T cells Oral lichen planus and HPV lesions Association of human papillomavirus with oral lichen planus and oral leukoplakia: a meta-analysis Association of herpes zoster and chronic inflammatory skin disease in US inpatients Lichen planus occurring after hepatitis B vaccination: a new case Oral Candida colonization and oral lichen planus Candida albicans induces TLR2/MyD88/NF-kappaB signaling and inflammation in oral lichen planus-derived keratinocytes Correlation between the sap gene of Candida albicans and oral lichen planus The presence of bacteria within tissue provides insights into the pathogenesis of oral lichen planus Skin microbiota-associated inflammation precedes autoantibody induced tissue damage in experimental epidermolysis bullosa acquisita Gene expression based evidence of innate immune response activation in the epithelium with oral lichen planus The expression of TNF-alpha and ICAM-1 in lesions of lichen planus and its implication Increased infiltration of CD11 c +/CD123 + dendritic cell subsets and upregulation of TLR/IFN-α signaling participate in pathogenesis of oral lichen planus. Oral Surg Oral Med Oral Pathol Oral Radiol Three cases of linear lichen planus caused by dental metal compounds Lichen planus and lichenoid reactions of the oral mucosa Drug-induced lichen planus Ultraviolet filters in hair-care products: a possible link with frontal fibrosing alopecia and lichen planopilaris Male frontal fibrosing alopecia: study of 35 cases and association with sunscreens, facial skin and hair care products An IFN-associated cytotoxic cellular immune response against viral, self-, or tumor antigens is a common pathogenetic feature in "interface dermatitis Role of mast cells in oral lichen planus and oral lichenoid reactions Mast cell degranulation and the role of T cell RANTES in oral lichen planus Immunohistochemical localization of mast cells and mast cell-nerve interactions in oral lichen planus Benign mast cell hyperplasia and atypical mast cell infiltrates in penile lichen planus in adult men Recruitment of dendritic cells in oral lichen planus Pathogenesis of oral lichen planus-a review The association and potentially destructive role of Th9/IL-9 is synergistic with Th17 cells by elevating MMP9 production in local lesions of oral lichen planus The roles of matrix metalloproteinases-2,−7,−10 and tissue inhibitor of metalloproteinase-1 in the pathogenesis of oral lichen planus The expression of Cathepsin L in oral lichen planus Role of cathepsin B as a marker of malignant transformation in oral lichen planus: an immunohistochemical study Cathepsin K expression is increased in oral lichen planus Etiology and pathogenesis of oral lichen planus: an overview. Oral Surg Oral Med Oral Pathol Oral Radiol Distinct interferon-gamma and interleukin-9 expression in cutaneous and oral lichen planus Immunoexpression of interleukin-22 and interleukin-23 in oral and cutaneous lichen planus lesions: a preliminary study Increased serum level and high tissue immunoexpression of interleukin 17 in cutaneous lichen planus: a novel therapeutic target for recalcitrant cases? Overexpression and selectively regulatory roles of IL-23/IL-17 axis in the lesions of oral lichen planus Oral lichen planus interactome reveals CXCR4 and CXCL12 as candidate therapeutic targets Expression and biological functions of the CCL5-CCR5 axis in oral lichen planus The expression of CXCL10/CXCR3 and effect of the axis on the function of T lymphocyte involved in oral lichen planus Potential roles of the CCL17-CCR4 axis in immunopathogenesis of oral lichen planus Infiltrating CD8+ T cells in oral lichen planus predominantly express CCR5 and CXCR3 and carry respective chemokine ligands RANTES/CCL5 and IP-10/CXCL10 in their cytolytic granules: a potential self-recruiting mechanism Inflammation-related cytokines in oral lichen planus: an overview Cytokine profiles in serum of patients with oral lichen planus Tofacitinib for the treatment of lichen planopilaris: a case series Treatment of severe lichen planus with the JAK inhibitor tofacitinib Tofacitinib in Hypertrophic Lichen Planus Interferon target-gene expression and epigenomic signatures in health and disease Therapeutic Targeting of Th17/Tc17 Cells Leads to Clinical Improvement of Lichen Planus Desmoglein 3-specific CD4+ T cells induce pemphigus vulgaris and interface dermatitis in mice The association of thyroid disease and oral lichen planus: a literature review and meta-analysis. Front Endocrinol Lichen planus and diabetes mellitus: systematic review and meta-analysis Lipid levels in patients with lichen planus: a case-control study Cardiovascular risk factors in patients with lichen planus Dermoscopy of active lichen planus Accuracy of dermoscopic criteria for the diagnosis of psoriasis, dermatitis, lichen planus and pityriasis rosea Lichen planopilaris epidemiology: a retrospective study of 80 cases Therapeutic management of classic lichen planopilaris: a systematic review Piccardi-Lassueur-Graham-Little syndrome associated with frontal fibrosing alopecia Lichen planus pemphigoides: from lichenoid inflammation to autoantibody-mediated blistering Lichen planus pemphigoides Cutaneous graft-versus-host disease Chronic graft versus host disease presenting as lichen planus pigmentosus Amalgam contact allergy in oral lichenoid lesions Oral lichenoid contact lesions to mercury and dental amalgam-a review Lichenoid tissue reaction/interface dermatitis: recognition, classification, etiology, and clinicopathological overtones Lichen planopilaris: retrospective study on the characteristics and treatment of 291 patients Lupus erythematosus/lichen planus overlap syndrome: successful treatment with acitretin Histopathological discriminant criteria between lichenoid drug eruption and idiopathic lichen planus: retrospective study on selected samples Lichen planus and cicatricial conjunctivitis: disease course and response to therapy of 11 patients Successful treatment of chronic skin diseases with clobetasol propionate and a hydrocolloid occlusive dressing Treatments for cutaneous lichen planus: a systematic review and meta-analysis Betamethasone oral mini-pulse therapy compared with topical triamcinolone acetonide (01%) paste in oral lichen planus: a randomized comparative study Treatment of lichen planus with acitretin. A double-blind, placebocontrolled study in 65 patients Isotretinoin for oral lichen planus Systemic isotretinoin treatment of oral and cutaneous lichen planus Treatment of severe lichen planus with cyclosporine Comparison of the narrow band UVB versus systemic corticosteroids in the treatment of lichen planus: a randomized clinical trial Lichen planus of the lower limbs: successful treatment with psoralen cream plus ultraviolet A photochemotherapy Successful treatment of lichen planus with sulfasalazine in 20 patients Treatment of lichen planus. An evidencebased medicine analysis of efficacy Efficacy of topical calcineurin inhibitors in lichen planus Individualizing treatment and choice of medication in lichen planus: a step by step approach Azathioprine for the treatment of severe erosive oral and generalized lichen planus Methotrexate for the treatment of generalized lichen planus Successful treatment of lichen planus with adalimumab Lichen planus pemphigoides treated with ustekinumab An openlabel pilot study of apremilast for the treatment of moderate to severe lichen planus: a case series Systemic and topical corticosteroid treatment of oral lichen planus: a comparative study with long-term follow-up A prospective study of findings and management in 214 patients with oral lichen planus. Oral Surg Oral Med Oral Pathol Efficacy of sulfasalazine in the treatment of generalized lichen planus: randomized double-blinded clinical trial on 52 patients Generalized severe lichen planus treated with azathioprine Hydroxychloroquine sulfate (Plaquenil) improves oral lichen planus: an open trial Oral lichen planus: a case series with emphasis on therapy Successful treatment of oral erosive lichen planus with mycophenolate mofetil Alefacept for erosive lichen planus: a case series Adalimumab in the management of cutaneous and oral lichen planus Off-label use of biologicals in the management of inflammatory oral mucosal disease Sustained remission of generalized lichen planus induced by cyclophosphamide randomized double-blind, positive-control trial of topical thalidomide in erosive oral lichen planus. Oral Surg Oral Med Oral Pathol Oral Radiol Endod Efficacy of oral metronidazole in treatment of cutaneous and mucosal lichen planus The efficacy of tetracycline antibiotics for treatment of lichen planus: an open-label clinical trial Pulsed itraconazole therapy in eruptive lichen planus Treating erosive lichen planus with griseofulvin: a report of four cases Treatment of erosive lichen planus with dapsone Extracorporeal photochemotherapy for chronic erosive lichen planus Psoralen plus UVA vs. UVB-311 nm for the treatment of lichen planus Influence of treatment of erosive lichen planus of the vulva on its prognosis Comparison of systemic mycophenolate mofetil with topical clobetasol in lichen planopilaris: a parallel-group, assessor-and analyst-blinded, randomized controlled trial Lichen planopilaris: report of 30 cases and review of the literature A case series of 46 patients with lichen planopilaris: demographics, clinical evaluation, and treatment experience A case-series of 29 patients with lichen planopilaris: the Cleveland Clinic Foundation experience on evaluation, diagnosis, and treatment Lichen planopilaris: clinical and pathologic study of forty-five patients Treatment of frontal fibrosing alopecia and lichen planopilaris: a systematic review Lichen planopilaris: update on diagnosis and treatment Lichen planopilaris: retrospective study and stepwise therapeutic approach Hydroxychloroquine and lichen planopilaris: efficacy and introduction of Lichen Planopilaris Activity Index scoring system Clinical efficacy and safety of methotrexate versus hydroxychloroquine in preventing lichen planopilaris progress: a randomized clinical trial Efficacy and safety of mycophenolate mofetil for lichen planopilaris Therapy-resistant folliculitis decalvans and lichen planopilaris successfully treated with adalimumab Lichen planopilaris treated with a peroxisome proliferator-activated receptor gamma agonist Use of pioglitazone in patients with lichen planopilaris Lichen planopilaris treated with thalidomide Thalidomide is ineffective in the treatment of lichen planopilaris Rapid and complete resolution of lichen planopilaris in juvenile chronic arthritis treated with rituximab Isolated nail lichen planus: An expert consensus on treatment of the classical form Intralesional steroid injection alleviates nail lichen planus Successful treatment of nail lichen planus with alitretinoin: report of 2 cases and review of the literature Nail lichen planus -a possible new indication for oral alitretinoin Lichen planus with associated myasthenia gravis-successful treatment with acitretin Nail lichen planus: successful treatment with etanercept Successful treatment of palmoplantar nail lichen planus with cyclosporine Successful treatment of nail lichen planus with topical tacrolimus Lichenoid variant of chronic cutaneous graft versus host reaction post blood transfusion: a rare event post blood transfusion Photodynamic therapy of oral lichen planus Oral lichen planus associated with candidiasis during secukinumab treatment Oral lichenoid reaction in a psoriatic patient treated with secukinumab: a drug-related rather than a class-related adverse event? Lichenoid drug eruption after treatment with ixekizumab for plaque psoriasis Severe, ulcerative, lichenoid mucositis associated with secukinumab Lichen planopilaris coexisting with plaque psoriasis effectively treated with brodalumab Successful treatment of recalcitrant genital lichen planus with secukinumab Immunoexpression of Th17-related cytokines in oral lichen planus Serum level of interleukin 17 in patients with erosive and non erosive oral lichen planus Failure of lichen planopilaris to respond to ustekinumab Lichen planus pemphigoides treated with ustekinumab Clinical healing of erosive oral lichen planus with tildrakizumab implicates the interleukin-23/interleukin-17 pathway in the pathogenesis of lichen planus Elevated serum interleukin-23 levels in patients with oral and cutaneous lichen planus Lichenoid drug eruption caused by adalimumab: a case report and literature review Lichen planus under anti TNF therapy for ankylosing spondylitis The inflammation in cutaneous lichen planus is dominated by IFN-and IL-21-A basis for therapeutic JAK1 inhibition Alopecia areata is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition A phase 2a randomized, placebo-controlled study to evaluate the efficacy and safety of the oral Janus kinase inhibitors ritlecitinib and brepocitinib in alopecia areata: 24-week results Oral lichen planus treated with apremilast Apremilast is effective in lichen planus mucosae-associated stenotic esophagitis Treatment of recalcitrant erosive oral lichen planus and desquamative gingivitis with oral apremilast Apremilast for genital erosive lichen planus in women (the AP-GELP Study): study protocol for a randomised placebo-controlled clinical trial Topical chemotherapy in cutaneous T-cell lymphoma: positive results of a randomized, controlled, multicenter trial testing the efficacy and safety of a novel mechlorethamine, 002%, gel in mycosis fungoides Novel treatment using low-dose naltrexone for lichen planopilaris comparative study of toluidine blue-mediated photodynamic therapy versus topical corticosteroids in the treatment of erosive-atrophic oral lichen planus: a randomized clinical controlled trial Vulvovaginal photodynamic therapy vs. topical corticosteroids in genital erosive lichen planus: a randomized controlled trial Evaluation of photodynamic therapy in treatment of oral erosive lichen planus in comparison with topically applied corticosteroids Comparing clinical effects of photodynamic therapy as a novel method with topical corticosteroid for treatment of Oral Lichen Planus Efficacy of photodynamic therapy or low level laser therapy against steroid therapy in the treatment of erosive-atrophic oral lichen planus Photodynamic therapy in oral lichen planus: a prospective casecontrolled pilot study Effectiveness of narrowband UVB phototherapy and psoralen plus UVA photochemotherapy in the treatment of generalized lichen planus: results from a large retrospective analysis and an update of the literature Treatment of refractory erosive oral lichen planus with extracorporeal photochemotherapy: 12 cases Effectiveness of extracorporeal photochemotherapy in the treatment of a case of refractory erosive lichen planus Interventions for treating oral lichen planus: corticosteroid therapies Phenotyping of adaptive immune responses in inflammatory diseases The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.Publisher's Note: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.Copyright © 2021 Boch, Langan, Kridin, Zillikens, Ludwig and Bieber. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.