Lichen planus

 

Salient features


 

·       Idiopathic inflammatory disease of the skin, hair, nails and mucous membranes, seen most commonly in middle-aged adults

 

·       Flat-topped violaceous papules and plaques favoring the wrists, forearms, genitalia, distal lower extremities, and presacral area

 

·       Clinical variants include actinic, annular, atrophic, bullous, hypertrophic, inverse, linear, ulcerative, vulvovaginal–gingival, lichen planopilaris, lichen planuspigmentosus, and drug-induced

 

·       Some lichenoid drug eruptions have a photo distribution, while others are clinically and histologically indistinguishable from idiopathic lichen planus

 

·       The most commonly incriminated drugs include angiotensin-converting enzyme (ACE) inhibitors, thiazide diuretics, antimalarials, quinidine, and gold

 

·       Histologically, there is a dense, band-like lymphocytic infiltrate and keratinocyte apoptosis with destruction of the epidermal basal cell layer

 

·       In this T-cell-mediated autoimmune disorder, basal keratinocytes express altered self-antigens on their surface


 

Definition

 

Lichenoid disorders are inflammatory dermatoses characterized clinically by flattopped, papular lesions and histologically by lichenoid tissue reaction, characterized by vacuolar alteration of epidermal basal cells intimately associated with a band-like infiltrate of primarily lymphocytes in the papillary dermis. Lichen planus is the prototype of lichenoid dermatoses.

 

 


Lichenoid tissue reaction (LTR) 

During the initiation of LTR, pDCs are activated by circulating immune complexes, viruses or by self-DNA/LL37 complexes induced during skin injury, leading to the abundant production of IFN-α. The IFN-α signal may represent an early event in the initiation of lichenoid skin reaction. IFN-α induces the production of CXCR3 ligands in structural cells of the skin and activates cytotoxic T cells secreting TH1 cytokines either directly or indirectly through activation of mDCs. Activated cytotoxic TH1 cells and pDCs amplify perpetuate the production of CXCR3 ligands through secretion of IFNs leading to additional recruitment of pDC and pathogenic T cells, and development of a LTR phenotype.

 

Introduction

 

Lichen planus is a common disorder of stratified squamous epithelia in which auto-cytoxic T lymphocytes trigger apoptosis of epithelial cells leading to chronic inflammation.

Lichen planus (LP) is an idiopathic inflammatory disease of the skin, hair, nails and mucous membranes, often with a chronic course with relapses and periods of remission. Classic LP is characterized by pruritic, violaceous papules that favor the extremities. Histologically, a dense, band-like lymphocytic infiltrate is seen underlying an acanthotic epidermis with hypergranulosis, apoptosis, and destruction of the basal cell layer. It has a multifactorial etiology with complex interactions of innate and adaptive immune responses and the disorder has been associated with multiple environmental exposures, including viral infections, medications, vaccinations and dental restorative materials. LP is an autoimmune reaction against epitopes on lesional keratinocytes that have been modified by viral or drug antigens.



Epidemiology

 

Cutaneous LP affects 1% of the adult population, whereas oral lesions affects up to 4% of the population.  The onset of LP occurs most commonly between the ages of 30 and 60 years, women are affected approximately twice as often as men.

 

Pathogenesis

 

LP represents T-cell-mediated autoimmune damage to basal keratinocytes that express altered self-antigens on their surface, which can be triggered by a variety of exogenous antigens, including drugs, metals (gold or mercury) or infection (hepatitis C virus). A critical component for the generation of effector T cells with cytotoxic potential is due to presentation of these exogenous antigens in the context of antigen-presenting cells.

 

Genetics

 

There could be HLA-associated genetic susceptibility that would explain a predisposition in certain persons. Familial cases are reported. LP occurs in up to 10% of first-degree relatives of affected patients. Cases of familial LP tend to have an earlier age of onset (affecting children and young adults), a higher relapse rate, and more frequent oral mucosal involvement. Different HLA haplotypes are reported in familial lichen planus, including HLA-B7, -Aw19, -B18, and -Cw8. In nonfamilial lichen planus, HLA-A3, -A5, -A28, -B8, -B16, and -Bw35 are more common. HLA-B8 is more common in patients with oral lichen planus as a sole manifestation, and HLA-Bw35 is more strongly associated with cutaneous lichen planus.

 

Environmental factors

 

Environmental factors including viral infections, drugs, and contact allergens have been potentially associated with LP.


 Viruses


Of the many potential exogenous antigens, greater attention has been focused on the possible role of viruses, particularly hepatitis C virus (HCV). Of the various types of LP, it is the oral form that is most commonly viewed as a manifestation of HCV infection. The PCR technique detected HCV RNA in 93% of oral LP lesions, suggesting HCV replication within LP lesions.

Other viruses have been implicated in the pathogenesis of LP, including hepatitis B virus (HBV), human herpes virus 6 (HHV6) and HHV7 and varicella zoster virus. Typically, hepatitis B vaccines have been shown in some patients to trigger LP, often after the second injection of vaccine.


Contact allergens


Contact allergy to a variety of metals within dental restorations causes exacerbation or induction of oral LP, positive patch test results, and then regression or complete clearing after removal of the sensitizing metal and replacement with other materials. Because involved allergens are dissolved and spread via saliva, mucosal reactions may extend beyond the contact areas. The metals that aggravate oral LP include amalgam (mercury), copper and gold. Although approximately 95% of patients had improvement after removal of the sensitizing metal, 75% of patients with negative patch test results also reported clearing of oral LP after removal of the metal and replacement with other materials. One possible explanation for this finding is that mercury served as an irritant and induced lesions via the Koebner phenomenon.

Of note, the development of contact allergy to metals within dental restorations in patients with LP could be explained by easy penetration of the metal via damaged mucosa.

 

Drugs


Cutaneous eruptions identical to LP (both clinically and histologically) have been linked to a variety of drugs. The terms “lichen planus-like” and “lichenoid” are often used to describe this phenomenon. A wide variety of drugs have been associated with lichenoid drug eruptions. The mechanism by which certain drugs can induce a lichenoid tissue reaction in susceptible hosts is unknown, but appears to be similar to LP. The most commonly incriminated drugs include angiotensin-converting enzyme (ACE) inhibitors, thiazide diuretics, antimalarials, quinidine and gold.


Betel nut


Social use of the betel nut is relatively common in India and SouthEast Asia. The product that is chewed, betel quid, is a mixture of substances, including the areca nut and betel leaf, and is associated with oral LP.


Lichen planuslike contact dermatitis


An LPlike eruption occurs in up to 25% of persons exposed to chemicals found in color developer such as paraphenylenediamine.

 

Immunopathology

 

 

 


Phases of lichen planus


A. In the induction phase, keratinocytes and plasmacytoid dendritic cells (pDCs), upon stimulation of their Toll-like receptors (TLRs) by pathogens or endogenous ligands, can release type 1 IFNs (e.g. IFN-α); this represents an early event in the cascade leading to T-cell-mediated epidermal damage. Activated keratinocytes, via production of IL-1β and TNF-α, can induce activation and migration of DCs. Chemokines, such as IP-10/CXCL10, released locally by pDCs, serve to attract CXCR3-expressing CD8+ or CD4+ effector memory T cells (which have differentiated from naive T cells within lymph nodes following presentation by DCs of self-peptides modified by exogenous antigens [viruses, medications and contact allergens]). Additional chemokine and chemokine receptor pairs have also been implicated in this process and the precise mix of chemokines and cytokines released into the tissue plays an important role in determining the composition of the inflammatory infiltrates.

 

B. In the evolution phase, effector T cells (Te) that come to express skin homing receptors (E-selectin ligands) migrate into the inflammatory site and upon recognition of antigens, are activated and release proinflammatory cytokines and cytotoxic granules, which in turn cause epidermal injury. In addition, Fas/FasL interactions can trigger cell death of both lymphocytes and keratinocytes with possible elimination of potentially harmful auto aggressive T cells. “Inflammatory” and “homeostatic” chemokines produced by keratinocytes direct the traffic of not only “pathogenic” T cells (Te) but also ‘immune surveillance” T cells (Ts) or regulatory T cells (Treg) into the sites; the relative balance of chemokines produced may determine the outcome of the T-cell mediate immune response.

 

 


 


 

Clinical features

 

The seven Ps—(1) purple, (2) polygonal, (3) planar, (4) pruritic, (5) papule, (6) plaque and (7) persistent describe LP. The primary lesion is a firm, 1 to 3-mm flat-topped papule with irregular angulated border (polygonal-shaped). WICKHAM 1895 described the characteristic appearance of whitish striae and punctuations that develop atop the flat surfaced papules. The surface is slightly shiny or transparent and close inspection of the surface shows a lacy, reticular pattern of crisscrossed, thin white lines (Wickham’s striae), seen best with hand lens after application of a drop of mineral oil. The latter correspond histologically to focal thickening of the granular layer. Wickham striae are readily apparent by dermoscopy. Newly evolving lesions are pink-white, but over time they assume a distinctive violaceous or purple hue with a peculiar waxy luster. Lesions that persist for months may become thicker and dark red (hypertrophic lichen planus). Papules can be isolated or they may cluster or coalesce into larger plaques, or annular or diffusely papular (guttate), or linear, appearing in response to a scratch or trauma (Koebner phenomenon). Rarely, a line of papules may extend the length of the extremity. Vesicles or bullae may appear on preexisting lesions or on normal skin. Typically, a greyish brown pigmentation can be observed in lesions that have resolved due to deposition of melanin in the superficial dermis. Annular lesions are especially common on the penis and rarely may be the predominant type of lesion present, later leading to atrophy. A variant in which groups of ‘spiny’ lesions resembling keratosis pilaris develop around hair follicles called lichen planopilaris.   Colocalization of LP and vitiligo has been reported. LP can affect any part of the body surface, but the most frequently involved sites are the flexor surfaces of the wrists and forearms, the dorsal surface of the hands, anterior aspect of the lower legs, neck, and the lumbar region and around the ankles. The ankles and shins are the commonest sites for hypertrophic lesions. When the palms and soles are affected, the lesions tend to be firm and rough (hyperkeratosis) with a yellowish hue. Isolated lesions of LP have been reported to involve one or both eyelids or the lips.

Pruritus is a fairly consistent feature in LP; occasionally, pruritus is completely absent. Hypertrophic lesions usually itch severely. Paradoxically, there is seldom evidence of scratching, as the patient tends to rub to gain relief. Itching at sites without visible skin lesions can occur. In the mouth, the patient may complain of discomfort, stinging or pain; ulcerated lesions are especially painful. Great discomfort may be caused by hot foods and drinks.

 

Natural history and morphology of lichen planus


Acute eruptive lichen planus usually begins as erythematous papules which progress and turn violaceous. Few of these lesions may further coalesce to form plaques that may be violaceous or even hypertrophic hyper pigmented type. Lesions resolve with classic post inflammatory hyper pigmentation.

 

 


 


Clinical variants

 

Many variations in the clinical presentation have been described and are generally categorized according to (1) the configuration of lesions, (2) the morphologic appearance, or (3) the site of involvement. These variations are patterned by subtle or unknown properties of the disease. The prototypic papule can be altered or modified in configuration, morphology, or anatomic distribution.

 

The most common sites of involvement in LP based on subtypes.


Subtypes

Most common sites of involvement


CLP

Actinic

Sun-exposed areas such as face, V-chest, hands

Annular

Male genitalia (penis, scrotum), axilla, groin folds

Atrophic

All parts of body especially lower extremities

Erosive

Soles of feet

Follicular

Scalp

Guttate

Trunk

Hypertrophic

Anterior leg, ankles, and interphalangeal joints

Linear

Leg-excoriated area

Papular

Flexor surfaces (the main initial presentation)

Bullous

Feet

Pigmentosus

Sun exposed areas such as face, V-chest, hands

Pigmentosus-inversus

Intertriginous and flexural areas

Nail involvement

Fingernails and toenails

Palmoplantar involvement

(1)  Malleoli

(2)  Palms

(3)  Soles (internal plantar arch)

Lichen planopilaris

(1)  Vertex of scalp (classic type)

(2)  Frontal of scalp (FFA type)

(3)  Graham Little-Piccardi-Lassueur syndrome 


Mucosal LP

Oral

 Reticular

(1) Buccal mucosa and mucobuccal folds
(2) Lateral and dorsal tongue
(3) Gingiva and lips

 Atrophic

Attached gingiva

 Hypertrophic

Buccal mucosa

 Erosive

(1) Lateral and dorsal portions of tongue
(2) Buccal mucosa

 Bullous

Posterior and inferior areas of buccal mucosa

 Plaque-like

Dorsum of the tongue and buccal mucosa


Vulvovaginal

All subtypes

Vaginal introitus, clitoris, clitoral hood, labia minora, and majora, vagina

Esophageal

 

-Proximal esophagus
-Proximal and distal esophagus
-Distal esophagus, strong possibility of concomitant mucosal involvement (80%)

                                               Special forms

Drug induced              Sun-exposed areas, lacks oral involvement

LP/LE overlap                              Distal extremities, sun exposed

Lichen planus pemphigoides      Extremities

 


 


Configuration of Lesions

 

Lichen planus, annular


Annular lesions occur in approximately 10% of patients with LP and are usually scattered among more typical lesions, but the former may represent the predominant finding. Annular lesions commonly develop as arcuate groupings of individual papules that develop rings or peripheral extension of clustered papules with central clearing. The annular edge is slightly raised and typically purple to white in color, while the central portion is depressed, slightly atrophic, hyper pigmented or skin-colored. The most common site of involvement is the axilla, followed by the penis, extremities, and groin. Most patients are asymptomatic, while some have pruritus. Actinic lichen planus seen on sun-exposed, dark-skinned young adults and children is frequently annular in shape.

 

Linear Lichen Planus


Although linear lesions frequently occur in sites of scratching or trauma in patients with LP as a result of the Koebner phenomenon, the term linear LP is usually reserved for lesions that appear spontaneously within the lines of Blaschko.

 

Morphology of Lesions

 

Lichen planus, hypertrophic

 

Hypertrophic lichen planus (lichen planus verrucosus) usually occurs on the extremities, especially the shins, dorsal aspect of the foot or around the ankles, and interphalangeal joints. Additionally, they tend to be highly pruritic, refractory to treatment, and associated with relapse. After a long time, papules lose their characteristic features and become confluent as reddish brown or purplish, thickened, elevated, round-to-elongated (band like) plaques and nodules with a rough or hyperkeratotic surface and may be covered by a fine adherent scale. Occasionally, verrucous plaques develop. The lesions are usually symmetric and tend to be chronic because of repetitive scratching. Chronic venous stasis frequently contributes to the development of this condition. When such lesions eventually clear, an area of pigmentation and scarring and some degree of atrophy may remain. Squamous cell carcinoma may develop within a background of longstanding hypertrophic LP.

 

Atrophic Lichen Planus

 

Atrophic LP may represent a resolving phase of LP, given the history of the lesions: papules coalesce to form larger plaques that over time become atrophic centrally, with residual hyper pigmentation. They are most common on the proximal lower extremities. Other reported sites included the axillae, glans penis, and trunk. The clinical appearance of atrophic LP is likely a result of marked thinning of the epidermis rather than degeneration of elastic fibers, and the epidermal atrophy may be accentuated by the use of potent topical corticosteroids. The lesions often resemble lichen sclerosus et atrophicus. However, the histologic features are diagnostic.

 

Lichen Planus Pigmentosus


This is a pigmentary disorder seen in India or in the Middle East, which may or may not be associated with typical LP papules and favors young to middle-aged adults with skin photo types III–V. It presents as irregularly shaped or oval, slate grey to brownish black macules and patches in either sun-exposed areas (especially the forehead, temples, neck and upper limbs) or intertriginous zones, especially the axillae, although it can be more widespread. It is usually asymptomatic, but some patients describe mild pruritus or burning. While the distribution is symmetric in the vast majority of patients, a unilateral linear variant following the lines of Blaschko has also been described. It is mostly diffuse, but reticular, blotchy and perifollicular forms are seen. The palms, soles, nails and oral mucosa are usually not involved. LPP is a chronic disorder with exacerbations and remissions. Histologic findings include an atrophic epidermis, a vacuolar alteration of the basal cell layer with a scarce lymphohistiocytic lichenoid infiltrate in the upper dermis, and dermal melanophages.

The etiology of LPP is unknown. The photo distribution in some patients suggests that UV can play a pathogenic role, and topical application of mustard oil (which contains allylisothiocyanate, a potential photosensitizer) and amla oil have been proposed as possible inciting agents. In one uncontrolled study, lightening of the pigmentation occurred in 54% of patients treated with topical tacrolimus for 12–16 weeks.

 

Lichen planus, actinic


Most patients are children or young adults. The onset of this variant is during the spring and summer, and the lesions primarily involve sun-exposed skin of the face, followed by the nape of the neck, the dorsal surfaces of the hands and arms. The lesions usually consist of red–brown plaques with an annular configuration with a thread, rolled edge or appear as discoid patches, which have a deeply hyper pigmented center surrounded by a striking hypo pigmented zone. Sunlight exposure appears to be central to the pathogenesis of actinic LP.  Melasma-like hyper pigmented patches have been observed in actinic LP.

 

Lichen planus, guttate

 

Lesions are widely scattered and remain discrete, may be all small (1 mm across) or larger (up to 1 cm), and individual lesions seldom become chronic. Guttate psoriasis must be excluded.

 

Follicular Lichen Planus/lichen planopilaris


In lichen planopilaris, involvement of the hair follicle is observed, both clinically and histologically. This variant is also called follicular LP.

Follicular lichen planus may occur alone or in association with other forms of cutaneous or mucosal lichen planus. Individual keratotic follicular papules and studded plaques are seen. Sites of predilection include the trunk and medial aspects of the proximal extremities. Follicular lichen planus may affect the scalp is likely to lead to a scarring alopecia.

 


Site of Involvement


Lichen Planus of the Scalp


Lichen planopilaris (LPP) or follicular lichen planus may affect the scalp in a distinctive clinical and histologic pattern that affects women more than men and this form may occur alone or with typical LP lesions elsewhere.

LPP can be subdivided into three variants: (1) classic LPP, (2) frontal fibrosing alopecia, and (3) Lassueur–Graham–Little–Piccardi syndrome.

In classic LPP, individual keratotic follicular papules surrounded by a narrow violaceous rim that coalesce to form plaques with subsequent hair loss resulting in patchy scarring alopecia of scalp are usually seen.  The inflammatory process may result in scarring and loss of follicular structure, i.e. a permanent alopecia. Perifollicular erythema and acuminate keratotic plugs are characteristic features. Lichen planopilaris resulting in atrophic, scarred, porcelain-colored area centrally with most active lesions consisting of acuminate keratotic follicular plugs and perifollicular scaling along with perifollicular erythema, are found at the margins of alopecia patches and follicular convergence resulting in doll's hair formation. Follicular-centered lesions are usually observed under close inspection of the scalp and orifices, particularly at the margins of the alopecic area or within patches still bearing hair. Dermatoscopic features of LPP include absence of follicular opening, cicatricial white patches, perifollicular casts and perifollicular scale, blue gray dots, perifollicular erythema and polytrichia (two or three hairs).

A variant of lichen planopilaris known as Graham Little–Piccardi–Lassueur syndrome is characterized by the triad of:  progressive patchy cicatricial alopecia of the scalp, non scarring alopecia of axillae, pubic area, and eyebrows as well as grouped spinous follicular papules resembling lichen spinulosus or keratosis pilaris on the trunk and extremities.

Frontal fibrosing alopecia is an uncommon condition characterized by progressive frontotemporal recession due to inflammatory destruction of hair follicles. Up to 75% of women with FFA report concomitant loss of the eyebrows, which tends to be non-inflammatory. It is more common in postmenopausal women. It may be associated with mucocutaneous lichen planus.

 

Pseudopelade of Brocq is a rare clinical syndrome of scarring alopecia and fibrosis, in which distinct pathologic features are absent. It is generally accepted that pseudopelade of Brocq is the end stage of follicular fibrosis caused by a primary inflammatory dermatosis such as lichen planus, lupus erythematosus, pustular-scarring forms of folliculitis.

 

 

Mucosal Lichen Planus


Lichen planus can affect the mucosal surfaces of mouth, vagina, esophagus, conjunctiva, urethra, anus, nose, and larynx.

 

ORAL LP


OLP is probably about eight times more common than cutaneous lichen planus. The disease develops in women more than twice as often as in men and most commonly occurs in the fifth to sixth decade of life, although the disease can sometimes affect children. Oral involvement is observed in up to 75% of patients with cutaneous LP, but the former can be the only manifestation of the disease, and oral lesions may precede, accompany or follow lesions elsewhere.

In patients whose initial presentation is oral LP will eventually develop cutaneous LP in approximately 15% and vulvar and/or vaginal involvement in 25%. However, scalp, nail, esophageal and ocular involvement are uncommon. Whereas cutaneous lesions of lichen planus (LP) are self-limiting and pruritic, oral lesions are chronic, rarely undergo spontaneous remission, potentially premalignant, often a source of morbidity and difficult to palliate.

 

Etiology


Most oral lichen planus is idiopathic but significantly greater anxiety and depression are observed among patients with oral lichen planus compared with controls. Lesions clinically and histologically similar, termed ‘lichenoid lesions’, are sometimes caused by dental restorative materials, (e.g. chromate, gold and thimerosal), chronic GVHD, drug use (e.g. nonsteroidal antiinflammatory agents), diabetes or chronic liver disease, especially chronic active hepatitis due to HCV infection.

 

Clinical features


OLP may manifest in one of three clinical forms: 1) reticular, including raised hyperkeratotic papules and plaques; 2) erythematosus, including atrophic lesions; and 3) erosive, including ulcerated and bullous lesions. Reticular lesions, the most recognized form of OLP, often occur as isolated lesions and may be the only clinical manifestation of the disease. They are often symptomless but may cause soreness. Erythematous lesions are accompanied by reticular lesions, and erosive lesions are accompanied by both reticular and erythematous lesions in almost all cases. This feature helps clinically differentiate OLP from other diseases such as pemphigus and pemphigoid, which are characterized by isolated areas of erythema and/or erosions. Erythematous and erosive lesions result in varying degrees of pain as well as burning, swelling, irritation, and bleeding with brushing.

The diagnosis of OLP can be made from the clinical features if they are sufficiently characteristic, particularly if typical skin lesions are also present, but biopsy is recommended to confirm the diagnosis.

OLP develops most frequently on the posterior buccal mucosa followed by the tongue, gingiva, labial mucosa, and vermilion of the lower lip. Lesions on the palate, floor of the mouth, and upper lip are uncommonly observed. The reticular pattern is characterized by slightly raised whitish linear lines in a lace-like pattern or in rings with short radiating spines (wickham striae). The striae often display a peripheral erythematous zone, which reflects sub epithelial inflammation. Lines are wavy and parallel. The most common site of involvement is the posterior buccal mucosa; lesions are often bilateral and symmetric. On the tongue, the lesions are usually in the form of fixed, irregular white plaques, often slightly depressed below the surrounding normal mucous membrane, especially on the upper surface and edges. Reticular OLP can sometimes be observed at the vermillion border of the lip. There may be striking pigmentation of oral LP in darkly pigmented races.

There is a higher incidence of plaque-like lesions among tobacco smokers called smoker’s patches’, which characteristically involve the palate.

Approximately 10% of patients with OLP have the disease confined to the gingiva and present as raised white lacy lesions or more commonly as desquamative gingivitis, typically with atrophic and erosive lesions. On the other hand, lichen planus is the most common cause for desquamative gingivitis. Direct immunofluorescence is an effective method of excluding other causes of desquamative gingivitis that share similar clinical features with OLP, including pemphigus, pemphigoid, linear IgA disease and foreign body gingivitis. Diabetes is a possible associated disease of oral LP, and candidiasis may coexist with LP in some patients.

Erosive lichen planus, which frequently affects the dorsum and lateral borders of the tongue or the buccal mucosae on both sides, is uncommon. The erosions are often large, slightly depressed or raised with a yellow slough, and have an irregular outline, but they are not always as painful as might be imagined. The surrounding mucosa is often erythematous and glazed in appearance, with loss of filiform papillae of the tongue, and there are often pathognomonic whitish striae. It has been reported that the erosive oral LP is less frequently associated with cutaneous LP than are all other types of oral LP. Such mucous membrane lesions are more therapy-resistant and less likely to spontaneously remit than cutaneous lesions. Malignant transformation of longstanding, non-healing ulcerative lesions in the mouth has been reported, but it must be distinguished from pseudoepitheliomatous hyperplasia. The WHO has defined oral LP as a “premalignant condition” because of this risk.

 

Oral lichenoid reaction is similar clinically and histologically to oral lichen planus, however, with identifiable etiology. Differentiating these two entities is often difficult. Oral lichenoid reactions may be asymmetrical on the buccal mucosa and occur adjacent to dental amalgam restorations, especially fillings that are worn and cracked. If patch testing reveals mercury allergy, changing to another type of filling may prove beneficial.

 

Very occasionally, LP lesions extend to the larynx or esophagus. Esophageal lichen planus is rare and affects the proximal esophagus. It manifests as progressive dysphagia and odynophagia. Endoscopic findings can include lacy white papules, pinpoint erosions, desquamation, pseudo membranes, and stenosis. Histologically, it shows parakeratosis, epithelial atrophy, and lack of hypergranulosis. Squamous cell carcinoma of esophagus may develop after longstanding disease.

 

Several studies have reported a relationship between oral LP and chronic liver disease, particularly that due to HCV infection. Some authors found an association of HCV with the reticular and plaque forms, whereas other authors reported an association of HCV with the erosive type. In the HCV-positive oral LP group, oral lesions are more frequently located on the tongue, labial mucosa and gingivae.

 

Modified World Health Organization diagnostic criteria of OLP

 

CLINICAL CRITERIA

 

ØPresence of bilateral, more or less symmetrical lesions

ØPresence of a lacelike network of slightly raised gray- white lines (reticular pattern)

ØErosive, atrophic, bullous and plaque-type lesions are accepted only as a subtype in the presence of reticular lesion elsewhere in the oral mucosa

ØIn all other lesions that resemble OLP but do not complete the aforementioned criteria, the term “clinically compatible with” should be used

 

HISOPATHOLOGIC CRITERIA

 

ØPresence of a well-defined band like zone of cellular infiltration that is confined to the superficial part of the connective tissue, consisting mainly of lymphocytes

ØSigns of liquefaction degeneration in the basal cell layer

ØAbsence of epithelial dysplasia

ØWhen the histopathologic features are less obvious, the term “histopathologically compatible with” should be used

 


Male genitalia


Male genitalia are involved in 25% of cases, and the glans penis is most commonly affected, with annular lesions frequently present. Other sites commonly affected are shaft of the penis, prepuce and scrotum. Anal lesions of mucosal lichen planus present with leukokeratosis, hyperkeratosis, fissuring, and erosions.

 

Vulvovaginal LP


Lesions on the female genitalia may occur alone, be combined with lesions in the mouth only, or be part of widespread involvement. The clinical presentation of LP of the vulva spans a spectrum from subtle, fine, reticulate papules to severe erosive disease accompanied by dyspareunia, scarring and loss of the normal vulvar architecture. Vaginal involvement occurs in up to 70% of women with erosive vulvar LP.

Diagnostic criteria have been proposed recently and include:

·       Welldemarcated erosions or glazed erythematous areas at the vaginal introitus

·       The presence of a hyperkeratotic white border to erythematous areas/ erosions and/or Wickham's striae in the surrounding skin

·       Symptoms of pain/burning

·       Scarring/loss of normal architecture

·       Presence of vaginal inflammation and

·       Involvement of other mucosal sites.

The association of erosive LP of the vulva and vagina with desquamative gingivitis has been termed the vulvovaginal–gingival syndrome.

 

Lichen planus of the palms and soles

 

Lichen planus of the palms and soles generally occurs as an isolated phenomenon, but may appear simultaneously with disease in other areas. Lesions on the palms and soles are uncommon, lack the characteristic shape and color of lesions elsewhere. The lesions differ from classic lesions of lichen planus in that the papules are larger and aggregate into semi translucent plaques with a globular waxy surface and are firm to the touch and yellow in hue. Itching may be absent.

Ulcerations can occur within palmoplantar lesions of LP, particularly those on the soles of one or both feet, often recalcitrant to conventional therapy.  Although palmoplantar LP is more common in men than in women, ulcerative LP prevails in female patients. The ulcers are large and intensely painful with gradual, permanent loss of the toenails; there may be secondary webbing of the toes.  Chronic ulcerative lesions are at risk of developing squamous cell carcinoma.

 

Lichen Planus of the nails

 

The nails are affected in ~10% of patients with LP and usually several nails are affected. Fingernails are more frequently affected than toenails, with initial involvement of two or three fingernails before subsequent involvement of the remaining digits. There are three major forms of nail LP:  classic nail lichen planus, Trachyonychia (twenty-nail dystrophy) and Idiopathic atrophy of the nails. The characteristic nail abnormalities include lateral thinning, longitudinal ridging, and fissuring. These changes are manifestations of matrix damage, which can lead to scarring and dorsal pterygium formation if left untreated. Nonspecific changes in the nail bed include yellow discoloration, onycholysis, and subungual hyperkeratosis. Details are described in the chapter of Nail disorders.

 

Inverse Lichen Planus


In this unusual variant, an inverse distribution pattern is observed. Pink to violaceous papules and plaques appear in intertriginous zones (axillae > inguinal and inframammary folds) and less often in the popliteal and antecubital fossae. Occasionally, there are LP lesions elsewhere on the body. Hyper pigmentation is usually present as well and it may be the sole manifestation, leading to overlap with LP pigmentosus.

 


Special Forms of Lichen Planus or Lichenoid Eruptions

 

Lichenoid drug eruption (drug-induced LP)


Lichen planus-like or lichenoid eruptions describe a group of cutaneous reactions identical or similar to lichen planus. Lichenoid drug eruptions have been reported after ingestion, contact, or inhalation of certain chemicals. Despite the significant overlap between LP and lichenoid drug eruption, there are both clinical and histologic clues that favor one diagnosis over the other. Both sexes are equally affected, but lichenoid drug eruptions tend to occur in adults approximately 10 years older than those with idiopathic LP.

 

FEATURES FOR DISTINGUISHING LICHENOID DRUG ERUPTION FROM LICHEN PLANUS

 

Lichenoid drug eruption

Idiopathic lichen planus

Mean age

66 years

50 years

Location

More generalized; often spares the “classic” sites of LP

Wrists, flexor forearms, presacral, lower legs, genitalia

Morphology

More eczematous, psoriasiform or pityriasis rosea-like

Shiny, flat-topped, polygonal, violaceous papules

Wickham's striae

Uncommon

Present

Hyper pigmentation

Very common, sometimes persistent

Common

Photo distribution

Frequent*

Unusual

Mucous membranes

Usually spared

Often involved

Histology

Varying degree of eosinophilic and/or plasma cell infiltrates

Eosinophils and plasma cells uncommon

Deep infiltrate may be present

Dense band-like infiltrate of lymphocytes

Parakeratosis

No parakeratosis

 

*

Especially with medications such as hydrochlorothiazide.

 

Mixed’ lichen planus/discoid lupus erythematosus disease patterns

 

This is a rare variant characterized by patients whose lesions have overlapping features of both LP and lupus erythematosus (LE).  Atrophic plaques and patches with hypo pigmentation and a livid red to blue–violet color with telangiectasia and minimal scaling are characteristic. Transient bullae may develop. Classic lesions of lichen planus, photosensitivity, pruritus, and follicular plugging are not common. Lesions may develop anywhere, but are preferentially located in acral sites. This disease variant is characterized by a prolonged course and lack of response to treatment. Histologic and direct immunofluorescence (DIF) microscopic findings show features of both LP and LE. Histologically, a lichenoid reaction typical for lichen planus and histologic features of lupus erythematosus are usually present in the same biopsy. By direct immunofluorescence, cytoid bodies staining with IgG, IgM, and C3 intraepidermally or at the dermal–epidermal junction, as seen in classic lichen planus, are most common. Linear to granular deposition of IgM and C3 (as seen in lupus erythematosus, but not in lichen planus) and shaggy deposition of fibrinogen at the basement membrane zone, typical of lichen planus are observed occasionally. Whether systemic immunologic abnormalities such as high titers of ANA are present in these patients is controversial, but case reports suggest that some patients have disease at the chronic cutaneous LE end of the clinical spectrum while others meet the criteria for systemic LE.

 

Lichen planus, bullous and pemphigoides

 

In bullous LP, vesicles and bullae develop within pre-existing lesions of LP as a result of intense lichenoid inflammation and severe liquefaction degeneration of the basal cell layer. This leads to epidermal–dermal separation, i.e. exaggerated Max-Joseph spaces. Histologically, there is sub epidermal bulla formation with typical changes of LP, and direct and indirect IMF is negative. The eruption is usually only of short duration. In contrast, patients with LP pemphigoides have circulating IgG autoantibodies directed against the 180 kDa BP antigen (BP180; BPAG2), as in idiopathic BP. In this variant, the LP tends to be acute and generalized and is followed by the sudden appearance of large tense bullae both within LP lesions and previously uninvolved skin, but the diagnosis of LP usually precedes the LP pemphigoides. LP pemphigoides has been precipitated by psoralen and UVA (PUVA) and has evolved into pemphigoid nodularis. In LP pemphigoides, the histology shows a subepidermal bulla with no evidence of associated LP. Direct IMF shows linear basement membrane zone deposition of IgG and C3 in perilesional skin. Immunoelectron microscopic studies reveal deposition of IgG and C3 in the base of the bulla and not in the roof as found in bullous pemphigoid.

Immunoblotting data have revealed that circulating autoantibodies in LP pemphigoides react with an epitope within the Cterminal NC16A domain of bullous pemphigoid 180 kDa antigen. To date, no reactivity against the 230 kDa BP antigen, type VII collagen, or the laminin-5 subunits has been detected. It seems that damage to the basal layer by a lichenoid infiltrate may expose hidden basement membrane antigens to the autoreactive T cells, leading to the formation of auto antibodies and subepidermal bullae. The mean age of patients with LP pemphigoides is lower than that of those with classic bullous pemphigoid, and the course of the disease also tends to be less severe.

 

Acute and subacute lichen planus with a confluence of lesions

 

Lichen planus may occur abruptly as a generalized, intensely pruritic eruption. Initially, the papules are pinpoint, numerous, and isolated. The papules may remain discrete or become confluent as large, red, eczematous-like, thin plaques. A highly characteristic, diffuse, dark brown, post inflammatory pigmentation remains as the disease subsides. Lichenoid drug eruptions are frequently of this diffuse type. Low-grade fever may be present in the first few days, and lesions appear on the trunk and extremities. The disease is seldom seen on the face or scalp and is rare on the palms and soles.  Successive crops may occur.  The clinical course is usually self-limited, and in general lesions resolve within 3 to 9 months.

 

Lichen Planus and Malignant Transformation


It is currently believed that the risk of malignant transformation is fairly low. Risk factors that increase the likelihood of developing oral cancer are long-standing disease, erosive or atrophic types, tobacco use, and possibly esophageal involvement. It is generally accepted that about 2% of patients with oral lichen planus develop SCC. The majority of these cases are in situ carcinoma or with micro invasive pattern. The most common site for cancer is the tongue, followed by buccal mucosa, gingiva, and, rarely, the lip. Clinically, the lesions appear as indurated, nonhealing ulcers, or exophytic lesions with a keratotic surface. Red atrophic plaques could also be seen and often correlate with SCC in situ. Advanced cases could result in nodal metastases and occasionally death.

The risk of skin malignancy in cutaneous lichen planus is extremely low. Most patients developing SCC in cutaneous lichen planus had a history of either arsenic or X-ray exposure.

 

Laboratory tests to consider for LP

 

·       Complete blood count

·       Patch testing, particularly for oral LP

·       Lipid panel

·       Thyroid function tests for lichen planopilaris

·       Antithyroid peroxidase antibodies

·       Antithyroglobulin antibodies

·       Hepatitis c virus testing for oral LP

 

Patch tests in patients with oral lichen planus usually reveal positive results in a majority of patients. Sensitivity to mercury and gold is often positive in one-half of the cases.  Avoidance of these clinically relevant sensitizers results in amelioration of disease. Dyslipidemia is more common in patients with lichen planus.

 


Histopathology


 





Microscopic features:  Orthokeratoic hyperkeratosis, wedge shaped hypergranulosis and saw-tooth appearance of the epidermis.

Effacement of the dermoepidermal junction by band-like mononuclear inflammatory cell infiltrate.

Hydropic degeneration of basal cells; Colloid body formation; Pigment incontinence.


Despite its different clinical manifestations, the histopathology of LP is fairly uniform and is the most useful investigation to confirm a diagnosis of LP.  The primary features are hyperkeratosis without parakeratosis, focal increases in the granular cell layer, irregular acanthosis with a “sawtooth” appearance, liquefactive degeneration of the basal cell layer, and a band-like lymphocytic infiltrate at the dermal–epidermal junction. A focal increase in thickness of the granular layer and infiltrate corresponds to the presence of Wickham's striae. Degenerating basal epidermal cells are transformed into colloid bodies (also referred to as Civatte, hyaline or cytoid bodies), which appear singly or in clumps and are usually present in the lower levels of the epidermis and the superficial dermis. Vacuolar changes within the basal cell layer may become confluent and result in small separations between the epidermis and the dermis (called “Max-Joseph spaces”). There is often incontinence of pigment with multiple dermal melanophages.

The histopathology of oral LP is similar to that of cutaneous lichen planus, although saw tooth rete ridges are rarely seen in oral biopsies. Oral LP lesions often show parakeratosis rather than hyperkeratosis, and the epidermis frequently becomes atrophic.

Lesions of lichen planopilaris are characterized by an inflammatory cell infiltrate around the hair follicles even at an early stage. Most of the inflammation involves the upper half of the follicle, with the isthmus affected in one-third of patients. Destruction of the follicles represents a later stage.

In LP pemphigoides, a primary histologic feature of the bullous lesions is a subepidermal separation with an abundance of eosinophils (occasionally neutrophil-rich infiltrates), whereas the papular lesions have the usual features of LP. DIF microscopy of peribullous skin shows linear deposition of IgG and C3 along the dermal–epidermal junction, similar to BP.

Apoptotic cells, as evidenced by Civatte bodies, tend to remain in the lower levels of the epidermis in idiopathic LP; in contrast, they are found in the cornified or granular layer in lichenoid drug eruptions. Direct immunofluorescence shows globular deposits of immunoglobulin M (IgM) and occasionally IgG and IgA, representing apoptotic keratinocytes, around the DEJ and lower epidermis, with fibrin deposition at the region of the DEJ. Although the band-like inflammatory infiltrate is usually restricted to the papillary dermis in LP and involvement of the deep vascular plexus suggests a lichenoid drug eruption (though observed in <50% of cases). In lichen planopilaris lesions, IgM, IgG and IgA are found in varying combinations along the follicle–dermal interface.

 


DIF of Lichen Planus showing IgM positive cytoid bodies in upper dermis.

 




DIF of Lichen Planus showing shaggy deposits of fibrinogen at DEJ.

  

The pattern of DIF results of potential differential diagnoses of lichen planus.


Differential

DIF pattern


Lichen planus

Globular IgM and fibrin deposition at BMZ

Bullous pemphigoid

Linear C3, IgG at BMZ, less common IgA

 

 

Dermatitis herpetiformis

Granular IgA at BMZ with concentration at the papillary tips

DLE/SLE

Linear band or continuous granular IgG, IgA, IgM, and C3 at BMZ

Epidermolysis bullosa acquisita

Linear IgG and C3 at BMZ

 

 


 


 

Linear IgA bullous dermatosis

Linear IgA at BMZ, less common IgG, IgM, and C3

Mucous membrane pemphigoid

Linear IgG and C3 at BMZ, less common IgA, IgM, and/or fibrin

Paraneoplastic pemphigus

Intercellular IgG and C3 with or without BMZ involvement

Pemphigus vulgaris

Intercellular IgG (IgG1 and IgG4) in “chicken wire pattern,” less common C3 and IgM


BMZ: basement membrane zone; DIF: direct immunofluorescence; Ig: immunoglobulin.

 

Dermatoscopy


The use of dermoscopy is found useful in some cases when Wickham's striae can be visualized better with the devise. Typical features of dermoscopy images of LP include a network of whitish striae with red globules at the periphery.

 

Prognosis and Clinical Course


Lichen planus is an unpredictable disease that typically persists for 1–2 years, but which may follow a chronic, relapsing course over many years. The duration varies according to the extent and site of involvement and morphology of lesions. Generalized eruptions tend to have a rapid course and heal spontaneously faster than limited cutaneous disease. Lichen planopilaris is one of the most chronic and often progressive disease variants with little potential for hair regrowth after follicular inflammation and destruction. Hypertrophic lichen planus typically follows a protracted, unremitting course. Spontaneous regression is also an uncommon feature of oral lichen planus. The mean duration for oral lichen planus is 5 years. The reticular variant has a better prognosis than erosive disease that does not heal spontaneously. Generally, the duration of disease conforms to the following order, from shortest duration to longest duration: (1) generalized cutaneous (2) non-generalized cutaneous, (3) cutaneous + mucous membrane, (4) mucous membrane, (5) hypertrophic, and (6) lichen planopilaris.

Relapse of disease occurs in 15%–20% of cases and tends to occur in the same area as the initial episode. Recurrences are more common in generalized lichen planus and are usually of shorter duration.

 


Management

 

Management of lichen planus can be challenging and discouraging for both the patient and physician. Lichen planus may be associated with only minor symptoms or may cause considerable discomfort and disability. Hence, treatment options should be assessed for attendant risks and benefits and tailored to the extent and severity of disease. Avoidance of exacerbating drugs and minimizing trauma to skin and mucosal tissues, are routinely recommended.

Treatment of LP depends on the localization, clinical form and severity. For cutaneous LP, the aim of treatment is to reduce pruritus and time to resolution of the lesions. No treatment is usually recommended for asymptomatic oral LP. However, painful, erosive LP may need aggressive and longterm treatments. Nail or scalp involvement, which may induce scars, genital LP and esophageal and conjunctival involvement, which may induce strictures and fibrosis, may require rapid treatment to avoid scaring or avoid a fatal course.

The main therapy for LP is corticosteroids, commonly recommended as first line treatment for LP.

 

Cutaneous lichen planus

 

TREATMENT

DOSAGE

COMMENTS

High-potency topical corticosteroids

Administered twice daily

First-line therapy

 

Oral corticosteroids (prednisone)

30 to 60 mg daily for three to six weeks, then dose is tapered over the next four to six weeks

For severe, widespread lichen planus

 

Phototherapy

30- to 40-minute treatments, two or three times weekly

For severe disease; narrow-band ultraviolet B is preferred over psoralen plus ultraviolet A


noteAcitretin is used in more severe cases of cutaneous lichen planus that do not respond to topical treatment; this is an off-label use.

 

 

Therapeutic ladder

 

First line

·        Limited cutaneous LP: very potent corticosteroids (clobetasol propionate ointment 0.05%); potent topical corticosteroids can be used in less severe forms of LP or during maintenance therapy

·        Widespread cutaneous LP: prednisolone 0.5–1 mg/kg per day until improvement


Second line

·        Prednisolone 0.5–1 mg/kg per day until improvement

·        Acitretin 30 mg per day for 8 weeks

·        PUVA or UVB: two to three times a week, 12 times alone or associated with systemic retinoids

 

First line

 

Topical corticosteroids are the first line treatment for limited cutaneous LP. The use of very potent corticosteroids is required (clobetasol propionate ointment 0.05%) once daily at night until remission. If no response is observed after 6 weeks, second line therapy has to be considered. Topical corticosteroids are particularly popular for children. For hypertrophic LP, very potent corticosteroids need to be applied under an occlusive bandage.

Intralesional triamcinolone acetonide (5–10 mg/mL) is effective in treating resistant cutaneous lichen planus. This may also be used in lichen planus of the nails with injection of the proximal nail fold every 4 weeks. Regression of lesions occurs within 3–4 months. For hypertrophic lichen planus, higher concentrations of intralesional triamcinolone acetonide (10–20 mg/mL) may be required. Regular and close observation should be performed to monitor for any signs of atrophy or localized hypo pigmentation that should prompt cessation of therapy. Intralesional injection is of special value in lichen planopilaris.

Oral corticosteroids are the first line treatment for widespread cutaneous LP: the minimal effective daily dose of prednisone for treating LP is greater than 20 mg/day (e.g., 30–80 mg prednisone) for 2–6 weeks with subsequent taper over 4–6 weeks. Rebound and relapses may occur, but long-term maintenance therapy with systemic corticosteroids should be avoided.

 

Second line

 

Retinoids


Acitretin is the only systemic retinoid that has a relatively good level of evidence regarding its efficacy in the treatment of cutaneous LP. Acitretin 1 mg/kg per day is helpful as adjunctive measure in severe (oral, hypertrophic) cases, but usually additional topical treatment is required. A therapeutic regimen consisting of acitretin 30 mg/day for 8 weeks resulted in significant improvement and marked remission is achieved in the treatment group. Retinoids tend to be used for recalcitrant cases, and, therefore, relapses may occur after discontinuation of the drug; as a result, long-term maintenance therapy may be required. Considering the side effects of teratogenicity and mucocutaneous dryness, acitretin is used as second line therapy in cutaneous LP. In case reports, alitretinoin, up to 25 mg/day for 4 weeks, was reported to clear oral, esophageal and cutaneous LP.

 

Phototherapy


In patients with resistant longstanding LP, significant improvement has been observed after systemic or bath PUVA. However, the risk of promoting carcinogenesis, especially in patients with skin types I and II, has to be balanced against the benefits. Psoralen and ultraviolet A (PUVA) light photo chemotherapy is usually successful in generalized cutaneous lichen planus. PUVA phototherapy may also be combined with acitretin. Trioxsalen, 50 mg added to 150 L of water, then exposing the patients to UVA after 10 minutes of bathing gave good results. For generalized disease, clearance within a mean of 3 months was observed in 70% of patients treated with narrowband UVB therapy. Phototherapy can increase the risk of residual hyper pigmentation in darkskinned patients. Phototherapy has been used in conjunction with oral glucocorticoids to hasten the response.

 

 

Third line

 

Immunosuppressive Agents


Oral cyclosporine has been used successfully in inducing remission in severe cases of LP resistant to systemic retinoids and corticosteroids. Complete responses were observed with doses of cyclosporine ranging from 1 to 6 mg/kg/day. Pruritus usually disappears after 1–2 weeks. Clearance of the rash is seen in 4–6 weeks. Adverse effects as well as relapse after discontinuation of the drug, limit its use to severe cases.

Azathioprine is useful in recalcitrant, generalized cutaneous lichen planus and in lichen planus pemphigoides.

Based upon a case series, weekly low-dose methotrexate is of value in generalized LP.  A complete response was achieved in 10 of 11 patients within 1 month and the medication was well tolerated without adverse effects.

Mycophenolate mofetil, an immunosuppressive agent which specifically and reversibly inhibits the proliferation of activated T cells, is reported to be effective in the management of disseminated, erosive, hypertrophic and bullous variants of LP. It is also effective at reducing the signs and symptoms of active lichen planopilaris in 83% of patients who had failed multiple prior treatments. It may be preferable to other immunosuppressive drugs such as cyclosporine because of its safer side-effect profile.

 

Miscellaneous


Hydroxychloroquine is effective in decreasing symptoms and signs in Lichen planopilaris and its variant frontal fibrosing alopecia in 69% and 83% of patients after 6 and 12 months of treatment, respectively. It is also the main treatment in actinic lichen planus at 200–400 mg/day in addition to sun protection.

Oral metronidazole 500 mg twice daily for 1–2 months also reportedly clears the majority of cases of generalized lichen planus.

Based on the benefit in bullous pemphigoid, combination therapy with tetracycline or doxycycline and nicotinamide has been reported as useful in the treatment of lichen planus pemphigoides. Tetracycline should be considered in lichen planopilaris.

An open-label pilot study of apremilast, a novel oral phosphodiesterase IV inhibitor, in 10 patients with moderate to severe LP demonstrated statistically significant clinical improvement in all of the patients after 12 weeks.

 

 

Oral lichen planus

 

 

 


 

Treatment ladder


First line

·        Symptomatic oral LP: very potent corticosteroids (clobetasol propionate ointment 0.05%) three times daily until remission, then maintenance therapy

 

·        Potent topical corticosteroids can be used in less severe forms of LP or during maintenance therapy: soluble prednisolone tablets, 5 mg in 15 mL water for mouthwash three times daily for widespread oral LP.  An alternative to prednisolone tablets is betamethasone soluble tablets (Betnesol) 0.5 mg

 

·        Severe erosive LP: prednisolone 0.5–1 mg/kg per day until improvement

 

Second line

·        Papular and plaquelike white forms and in absence of erosive lesions: topical retinoids (retinoic acid or isotretinoin lotion/gel 0.1%) twice daily

 

·        Resistant to topical corticosteroids: prednisolone 0.5–1 mg/kg per day until improvement

 

Third line

·        Corticodependent or corticoresistant erosive LP: systemic immunosuppressive agents (e.g. azathioprine, mycophenolate mofetil, methotrexate, oral and topical ciclosporin)

 


General Measures


Treatment is not always necessary, unless there are symptoms. Unfortunately, although the natural history of cutaneous lichen planus is one of remission in most cases, oral lichen planus rarely remits, and thus treatment is indicated for symptomatic oral lichen planus.

The aims of treatment are to heal erosive lesions in order to reduce pain and permit normal food intake. Education of the patient should emphasize that oral LP frequently has a chronic course marked by treatmentinduced remission followed by relapse. Considering the potential higher risk of squamous cell carcinoma, the need for regular clinical surveillance on a longterm basis has also to be explained.

Predisposing factors should be corrected. Replacement of amalgam or gold dental restorations with composite material is frequently of considerable benefit in patients with oral lichenoid reactions, even in the apparent absence of relevant patch test results. Chronic lesions on the buccal mucosa in contact with metal or other contact sensitizers frequently heal promptly after replacement. Occasionally, lesions at sites distant to oral lesions may also clear after removal of metal restorations. Gingival lesions may respond less favorably. If drugs are implicated, the physician should be consulted as to possible changes in therapy. If there is diabetes or HCV infection, this should be treated by a physician. Good oral hygiene and regular personal and professional dental care need to be encouraged. Alcohol and tobacco as well as spicy or acidic foods and drinks should be avoided.

 

First line

 

Topical Steroids


Topical corticosteroids are the first line treatment for symptomatic oral LP. Use of occlusive materials suitable for mucous membranes, such as Orabase, may provide protection, sustained tissue contact with the medication, and alleviate the discomfort associated with erosive lesions. In the absence of specific available formulation for oral lesions, super potent corticosteroids (0.05% clobetasol propionate ointment) can be applied three times daily with a gloved finger on erosive lesions. It is recommended to not drink or eat for 1 h after the application. Alternatively, for widespread, less severe lesions or maintenance therapy, a patient can use a soluble prednisolone tablet 5 mg dissolved in 15 ml water for a mouthwash swish and rinse three times daily. Oral candidiasis is the most frequent complication of this treatment. The use of chlorhexidine gluconate mouthwashes and topical anticandidal medications is recommended during therapy.

 

Topical retinoids


For papular and plaquelike forms of oral LP, topical retinoids (tretinoin gel) can be recommended as first line treatment. Isotretinoin gel is also effective, especially in nonerosive oral lesions. However, recurrence is common after discontinuation of therapy. Topical retinoids are often used in conjunction with topical glucocorticoids.

 

Second line

 

If no or insufficient improvement is observed after 6 weeks of topical corticosteroids and for severe case of erosive LP associated with eating difficulties and eventual weight loss, oral corticosteroids can be used alone, or, more commonly, in conjunction with topical glucocorticoids. Prednisone dose 0.5 -1mg/kg day, until remission, generally 4–6 weeks and then tapered over the next 3–6 weeks, or Betamethasone 5 mg on two consecutive days per week show benefit. Relapses are common after dose reduction or discontinuation. Slow reductions in dose are needed because of the risk of relapse.

Two-thirds of patients showed marked improvement or complete remission within 8 weeks of instituting acitretin, 30 mg/day in erosive OLP.

 

Third line

 

Tacrolimus and Pimecrolimus


Tacrolimus, effective in erosive mucosal disease, provides rapid relief from pain and burning with minimal side effects. It is at least equally effective to clobetasol propionate 0.05% ointment and triamcinolone acetonide 0.1% paste in the treatment of oral lichen planus.

Pimecrolimus 1% cream was shown to be as effective as triamcinolone acetonide 0.1% paste in treating oral lichen planus, and proved to be effective in treating vulvar disease.

Current US Food and Drug Administration (FDA) labelling states that topical pimecrolimus and tacrolimus should not be used to treat premalignant conditions. They have to be used with caution in oral LP due to the inherent risk of malignant transformation.

 

Mild lichen planus

 

Topical corticosteroids are the mainstay of therapy (e.g. triamcinolone acetate, betamethasone or fluocinolone). Erosive and gingival lesions are often recalcitrant. Next, highpotency corticosteroids such as clobetasol, fluocinonide or fluticasone may be employed initially and then changed to a lower potency drug.

 

Moderate lichen planus

 

If there is severe or extensive oral involvement, topical ciclosporin or tacrolimus may be of significant benefit, often being used with a highpotency or superpotent topical steroid such as clobetasol, fluticasone or mometasone. Topical creams or pastes can be applied in a suitable customized tray or veneer to be worn at night. This regimen is useful in the management of lichen planusrelated desquamative gingivitis recalcitrant to other therapies.

 

Severe lichen planus

 

In severe lichen planus in multiple sites, patients may require systemic corticosteroids, azathioprine, cyclophosphamide, hydroxychloroquine, acitretin, thalidomide or ciclosporin. Dapsone is occasionally effective.

 

Nonreticular lichen planus


Patients with nonreticular lichen planus should be monitored to exclude development of carcinoma; tobacco and alcohol use should be minimized.

 

Anogenital lichen planus

 

Early superpotent corticosteroid treatment (0.05% clobetasol propionate ointment once daily) aims to resolve symptoms and prevent synechiae and scarring of the genital area. Potent corticosteroids can be used daily during the initial phase until symptom resolution. Foreskin retraction or removal surgery in uncircumcised men and vaginal dilators in women are used to prevent synechiae formation. Surgery is required after complete resolution of the active lesions if adhesion occurs.

 

Lichen planopilaris

 

In order to lessen pain and pruritus, and to avoid irreversible alopecia, first line treatment is potent corticosteroids (e.g. 0.05% clobetasol propionate ointment twice daily). In severe cases or in cases of insufficient improvement after corticosteroid ointment, second line treatment is monthly intralesional injection of triamcinolone acetonide (0.5–10 mg/mL) or oral corticosteroids (prednisolone 1 mg/kg/day).

 

 


Proposed treatment strategy for classic lichen planopilaris.

Notes: *Topical/intralesional steroids may be added to systemic therapies in the case of persistence of limited active areas. §A short course of systemic steroids should be considered only to halt the progression and to improve symptoms in rapidly progressive and severe cases.

 

Frontal fibrosing alopecia


Treatments can be of limited efficacy and includes topical and intralesional corticosteroids, antibiotics, hydroxychloroquine, topical and oral immunomodulators and 5αreductase inhibitors.

 

Nail lichen planus

 

The aims of treatment are to reduce pain and avoid irreversible nail scars. Intramuscular injection or oral systemic glucocorticoids and/or intralesional injection or topical glucocorticoids are commonly used.

When three or less than three nails are involved, the recommended treatment is a twice daily application of superpotent corticosteroids (e.g. 0.05% clobetasol propionate ointment), or if lesions are more severe, monthly intralesional injection of triamcinolone acetonide (0.5–10 mg/mL) in the periungual sites.

When more than three nails are involved, systemic corticosteroids are required. Treatment with oral prednisolone (0.5–1 mg/kg/day) for 4–66 weeks or intramuscular triamcinolone acetonide injection has been reported.

 

Severe erosive lichen planus

 

Systemic corticosteroids are first line treatment. Systemic immunosuppressive agents (e.g. azathioprine, mycophenolate mofetil and methotrexate) are used as corticosteroidsparing agents for patients with erosive or potentially scarring LP requiring prolonged systemic corticosteroids or in disease unresponsive to corticosteroids.

 

Mixed’ lichen planus/discoid lupus erythematosus disease patterns

 

Both ciclosporin and acitretin can be of benefit in treating LP/DLE overlap syndrome.

 

Actinic lichen planus

 

Actinic LP has been treated with acitretin and topical corticosteroids and with ciclosporin.

 

Bullous lichen planus and lichen planus pemphigoides

 

Some cases require treatment with systemic steroids or azathioprine. A combination of corticosteroids and acitretin has been used.

 

Popular Posts