You are on page 1of 40

Oral Lichen Planus

Michael J. McCullough, Mohammad S. Alrashdan, and Nicola


Cirillo

Abstract play an essential role in its pathogenesis. The


Lichen planus (LP) is a chronic, inflammatory, diagnosis of OLP is usually made by clinical
mucocutaneous, immune-mediated condition and histological examinations. Lesions similar
with variable clinical presentations. Oral lichen to OLP may develop as a reaction to dental
planus (OLP) is the oral variant and affects restorative materials or systemic medications
about 1–2% of the general adult population or conditions and are called oral lichenoid
with characteristic relapses and remissions. reactions (OLR). OLP and OLR may collec-
OLP is about twice as common in females as tively be referred to as oral lichenoid lesions
in males. The most commonly involved oral (OLLs). Management of symptomatic OLLs
sites are the buccal mucosa, lateral surfaces of varies considerably and ranges from elimina-
the tongue, and gingivae, respectively. Six tion of precipitating factors – local or systemic
clinical patterns of OLP are described in liter- to long-term pharmacological interventions,
ature: reticular, plaque-like, erythematous, ero- mainly with topical immunosuppressants, cor-
sive/ulcerative, papular, and bullous. Helper ticosteroids in particular. In the light of the
and cytotoxic T lymphocytes, in addition to ongoing debate regarding the malignant trans-
antigen-presenting cells, represent the key formation potential of OLLs, a long-term fol-
cells in the inflammatory infiltrate in OLP and low-up protocol is essential.

Keywords
M.J. McCullough (*) Oral lichen planus • Oral lichenoid lesions •
Oral Anatomy, Medicine, and Surgery Section, Melbourne Oral lichenoid reactions • Malignant
Dental School, Faculty of Medicine, Dentistry & Health transformation
Sciences, The University of Melbourne, Carlton, VIC,
Australia
e-mail: m.mccullough@unimelb.edu.au Contents
M.S. Alrashdan Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Faculty of Dentistry, Department of Oral Medicine and Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Oral Surgery, Jordan University of Science and
Technology, Irbid, Jordan Etiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
e-mail: msalrashdan3@just.edu.jo Genetic Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Infectious Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
N. Cirillo Psychological Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
Melbourne Dental School and Oral Health CRC, Faculty of Trauma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
Medicine, Dentistry and Health Sciences, The University Systemic Associations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
of Melbourne, Carlton, VIC, Australia
e-mail: nicola.cirillo@unimelb.edu.au

# Springer International Publishing AG 2017 1


C.S. Farah et al. (eds.), Contemporary Oral Medicine,
DOI 10.1007/978-3-319-28100-1_14-1
2 M.J. McCullough et al.

possible to make a diagnosis based on their clin-


Pathogenesis of OLP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
The Immunology of the Oral Cavity . . . . . . . . . . . . . . . . . . 6 ical appearance alone (Epstein et al. 2003). Addi-
Pathogenic Mechanisms of OLP . . . . . . . . . . . . . . . . . . . . . . 7 tionally, there is a spectrum of oral lichenoid
Clinicopathologic Features . . . . . . . . . . . . . . . . . . . . . . . . . . 13 lesions (OLLs) that may confuse the differential
Clinical Appearance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 diagnosis. These include lichenoid contact
Signs, Symptoms, Clinical Behavior and OLP lesions, lichenoid drug reactions, and lichenoid
Scoring Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 lesions of graft-versus-host disease. Systemic
Histopathology of OLP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Oral Lichenoid Reactions (OLR) . . . . . . . . . . . . . . . . . . . . . 16 medications, such as nonsteroidal anti-
Cutaneous Lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 inflammatory drugs, antihypertensives, and oral
Other Mucosal Lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19 hypoglycemics, can contribute to the develop-
Malignant Potential of OLP . . . . . . . . . . . . . . . . . . . . . . . . . 19 ment of oral lichenoid reactions (OLRs) (Ismail
OLL and Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 et al. 2007). Dental restorative materials, includ-
Tumors Developing in OLP and their ing amalgam, gold, and nickel, may also be
Carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
related to localized OLRs in a number of patients
Patient Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25 (Epstein et al. 2003).
Topical Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26 Treatment is not always necessary, unless
Systemic Drug Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
Miscellaneous Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30 OLLs are symptomatic (Scully et al. 1998). Man-
Follow-up . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32 agement of symptomatic OLLs varies consider-
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
ably and ranges from elimination of precipitating
or provoking local and systemic factors, psycho-
Cross-References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
social interventions, to long-term pharmacologi-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33 cal therapies. Thus, although OLP localizes to the
oral cavity, there are broader implications in terms
of patient management that warrant careful con-
Introduction sideration. The ongoing controversy as to whether
OLP is associated with an increased risk of malig-
The term lichen planus (LP) is derived from the nant transformation adds further complexity to
Greek word leichen meaning tree moss and the this disease.
Latin planus meaning flat. Erasmus Wilson first
described the condition LP in 1869, as a chronic
inflammatory disease affecting the skin, scalp, Epidemiology
nails, and mucosa, with possible rare malignant
transformation (Farhi and Dupin 2010). LP may The estimated prevalence of OLP in general adult
involve the hair follicles (lichen planopilaris, population is 0.5–2% (Scully et al. 1998; Eisen
resulting in scarring alopecia), nails, esophagus, et al. 2005). However, because of a lack of stan-
and, more seldom, the eyes, urinary tract, nasal dardized approaches to the clinical diagnosis of
mucosa, and larynx (Parashar 2011). OLP and OLL, it is difficult to draw accurate
The oral variant, oral lichen planus (OLP), is a figures about the prevalence of OLP. In a demo-
chronic inflammatory disease affecting the oral graphic study from Sweden, 20,333 people aged
mucosa with characteristic relapses and remis- 15 and above were examined for signs of OLP.
sions (Scully et al. 1998; Eisen et al. 2005; Lodi The prevalence reported was 1.9%, 1.6% in men,
et al. 2005a). While cutaneous lesions of LP are and 2.2% in women (Axell and Rundquist 1987).
generally self-limiting and pruritic, oral lesions Another demographic study from India showed a
are commonly chronic, non-remissive, and can prevalence of 2.6% (Murti et al. 1986). A report
be a source of morbidity (Scully et al. 1998). from Malaysia showed a relatively low prevalence
The diagnosis of OLP is usually made by clin- of 0.38% (Zain et al. 1997).
ical and histological examinations. However, in There is a slight female predisposition (female/
classical lesions (bilateral, reticular pattern), it is male sex ratio is 1.5–2:1), and the age of onset is
Oral Lichen Planus 3

generally between 30 and 60 years (Scully et al. clinical presentation of LP. Interferon-gamma
1998; Al-Hashimi et al. 2007; Farhi and Dupin (IFN-γ) UTR 5644 genotype frequencies showed
2010). However, there have been case reports of a significant increase in number of T/T homozy-
OLP occurring in children, with a 17% prevalence gotes in a sample of OLP patients (n = 44) com-
of oral involvement in a population of 100 children pared with controls (n = 140) (40.9% vs. 22.9%;
diagnosed with LP at a mean age of 8.7 years in p = 0.0022) (Carrozzo et al. 2004). It has been
one report (Kanwar and De 2010). suggested that genetic polymorphism of the first
Genital and cutaneous LP are associated with intron of the promoter gene of IFN-gamma may
approximately 20% and 15% of OLP cases, be an important risk factor to develop oral lesions
respectively, while it is estimated that OLP occurs of LP, whereas an increase in the frequency of
in 70–77% of patients with cutaneous LP (Scully 308A TNF-alpha allele may best contribute to
et al. 1998; Farhi and Dupin 2010; Kanwar and De the development of additional skin involvement.
2010). More recently, the concept that a particular
genetic background may be important in a subset
of patients with OLP has been presented
Etiology (Carrozzo et al. 2011). In particular, this study
suggested that those patients who had both OLP
OLP is not a classical autoimmune disorder, and and chronic hepatitis C infection may well have
the precise etiology of this condition is unknown. particular HLA-Cw* alleles (Carrozzo et al.
However, given that cell-mediated immune 2011).
dysregulation targeting the stratified squamous Nevertheless, the overall statement that OLP is
epithelia has been shown to be associated with a genetically determined disease has not been
the pathogenesis of OLP (Epstein et al. 2003; confirmed, and further studies in different geo-
Eisen et al. 2005; Lodi et al. 2005a), it is possible graphic areas are required.
that a LP-specific epidermal antigen is present in
some epithelial cells. The nature of the antigen,
however, remains uncertain. Several predisposing Infectious Agents
factors have been reported to be associated with
OLP and OLLs. Many infectious agents, principally viruses, have
been studied in association with OLP, but the
evidence is generally sparse. Other infectious
Genetic Background agents, such as Helicobacter pylori, were also
suggested to have a link to OLP by some authors
Genetic background seems to play a role in OLP (Shimoyama et al. 2000).
pathogenesis as several familial cases have been The viruses for which an association with OLP
reported (Bermejo-Fenoll and Lopez-Jornet has been suspected can be classified in two
2006); however the association has not been con- groups: (1) viruses for which an association with
sistent. In terms of HLA associations, an increase OLP has been anecdotally suggested and
in HLA-B15, Bw57, B5, B7, BX, DR2 and a (2) viruses for which there is abundant documen-
decrease in the frequency of HLA-DQ1, DR4, tation of an association with OLP, although with
and B18 has been noted for OLP (Roitberg- marked geographic disparities (discussed in sec-
Tambur et al. 1994). On the other hand, in a tion “Systemic Associations”).
comprehensive review of literature, Porter et al. Varicella zoster virus (VZV), Epstein-Barr
reported no significant association with any par- virus (EBV), cytomegalovirus (CMV), human
ticular HLA types in familial LP (Porter et al. herpesvirus 6 (HHV6), human papillomavirus
1997). (HPV), and human immunodeficiency virus
Genetic polymorphisms of several cytokines (HIV) have all been postulated in different studies
have been postulated to be associated with the to have an association with an increased incidence
4 M.J. McCullough et al.

of OLP, yet definitive evidence for the association in turn may contribute to the initiation and clinical
is lacking (Lodi et al. 2005a; Farhi and Dupin expression of OLP (Ivanovski et al. 2005). How-
2010). ever, as a common issue in studies of this type, the
For example, the receptor for EBV (CD21) was authors were unable to determine whether the
shown to be upregulated in OLP, and a signifi- observed psychological alterations constitute a
cantly higher optometric density of EBV anti- direct cause or a consequence of OLP.
earlier antigen (EA) IgG positivity has been When the levels of anxiety and salivary corti-
reported in a sample of OLP patients (n = 22) sol were measured in a group of OLP patients
compared with controls (n = 22), despite no dif- (n = 40), the scores were statistically correlated
ference in the frequency of both EBV IgG and and significantly higher than a control group
IgM for EA and nuclear antigen-1 (EBNA) (mean state anxiety level = 49 vs. 40, cortisol
(Pedersen 1996). levels = 1.46 vs. 0.93 μg/dl) (Koray et al. 2003).
In summary, while association between OLP In conclusion, in spite of the presence of higher
and infectious agents has been reported, at present levels of psychological stress and anxiety among
there is no evidence for a causative role of these OLP patients, the question remains whether the
microorganisms in OLP. psychological factors contribute to the etiology of
OLP or represent a part of resulting morbidity.

Psychological Factors
Trauma
Psychological factors are thought to play a role in
the pathogenesis of OLP. A group of OLP patients Trauma as such has not been reported as an etio-
(n = 9) exhibited higher levels of anxiety, greater logical factor in OLP, although it has been postu-
depression, and increased vulnerability to psycho- lated as a mechanism by which other etiological
logical disorders, as opposed to a group of factors may exert their effects (Scully et al. 1998).
20 healthy controls (Soto Araya et al. 2004). However, no studies were conducted to verify this
In another study that involved 100 OLP hypothesis.
patients, those with erosive LP were shown to The Koebner phenomenon (isomorphic
exhibit higher depression scores than patients response), whereby OLP lesions develop in
with nonerosive LP (Rojo-Moreno et al. 1998). response to mechanical trauma, may partially
In addition to the chronic discomfort that can explain why OLP lesions develop commonly in
result in stress, patients with OLP were shown to sites prone to trauma, i.e., the buccal mucosa or
be concerned about the possibility of malignancy, lateral surfaces of the tongue (Eisen et al. 2005).
the contagious nature of the disease, and the lack
of available patient educational materials
(Burkhart et al. 1997). Systemic Associations
Exacerbations of OLP have been linked to
periods of psychological stress and anxiety in OLP may be associated with some systemic dis-
some studies (Rojo-Moreno et al. 1998). eases; however, few have been thoroughly inves-
In a study that involved 40 OLP patients tigated. The most studied are the associations with
(20 reticular and 20 erosive OLP) assessed against hepatitis C, hypertension, diabetes, thyroid dis-
25 healthy controls using the psychological Min- ease, and graft versus host disease.
nesota Multiphasic Personality Inventory
(MMPI)-202 test, Ivanovski et al. hypothesized Hepatitis C Virus (HCV)
that prolonged emotive stress in many OLP Along with porphyria cutanea tarda and
patients may lead to psychosomatization cryoglobulinemia, OLP is one of the three derma-
(as shown by significantly higher mean scores of tologic diseases that have been most frequently
internalization ratio index in OLP groups) which reported in patients infected with HCV (Farhi and
Oral Lichen Planus 5

Dupin 2010). The first report appeared in 1991 depending on the presence of certain HLA sub-
(Mokni et al. 1991) and suggested an association types (Carrozzo et al. 2001).
between OLP and HCV seropositivity. Since then,
the association has been well documented in some Hypertension and Diabetes Mellitus
Mediterranean populations (Carrozzo et al. 1996; Although OLP patients do not seem to have an
Erkek et al. 2001), in Japan (Nagao et al. 1995), increased risk of diabetes, diabetics who develop
and in a United States metropolitan population OLP were reported to have an increased fre-
(Beaird et al. 2001). Nevertheless, an association quency of atrophic-erosive lesions, especially
between OLP and HCV seropositivity could not affecting the tongue (Bagan et al. 1993). How-
be demonstrated in France (Dupin et al. 1997), the ever, this was not confirmed in any further studies.
United Kingdom (Ingafou et al. 1998), and in An association between OLP, diabetes
countries with high HCV prevalence, such as mellitus, and hypertension was first described by
Egypt (HCV prevalence estimated at 14.7%) Grinspan in a small series of seven patients
(Ibrahim et al. 1999), and Nigeria (HCV preva- (Grinspan et al. 1966). The triad was later named
lence estimated to range from 5% to 20%) as Grinspan’s syndrome. Although Grinspan’s
(Daramola et al. 2002). syndrome may be seen clinically, the association
A systematic review of HCV prevalence in LP between the three conditions may represent a
patients and in matched controls without LP coincidental finding or probably an OLR to med-
yielded 25 relevant studies, including eight with ications used to manage hypertension or diabetes
only OLP (Lodi et al. 2004). A significantly rather than a true syndrome (Scully et al. 1998).
higher proportion of HCV seropositivity was
documented in patients with OLP, with an odds Thyroid Dysfunction
ratio (OR) of 5.7 (95% confidence interval, The association between OLP and thyroid dys-
3.5–9.4). This association was stronger in Medi- function was recently investigated in a retrospec-
terranean countries (OR 6.63, 95% CI, 4.7–9.4) tive Finnish study that confirmed a link between
but disappeared in Northern Europe such as in OLP and hypothyroidism in particular (Siponen
Germany and the United Kingdom (OR 2.14, et al. 2010). This study compared data of 222 OLP
95% CI, 0.6–7.7). Three more recent independent patients with 222 age- and sex-matched controls
meta-analyses (Shengyuan et al. 2009; Lodi et al. and revealed that 10% of OLP patients (n = 15)
2010; Petti et al. 2011) have provided robust evi- versus 5% of the controls (n = 11) had hypothy-
dence that LP and HCV are associated. Overall, roidism (OR 2.39, 1.05–5.61, CI 95%).
there is a significant positive association noted in Other studies suggested a relationship between
studies across the world, although the association OLP and hyperthyroidism. Overall, more well-
is more homogeneous in studies from East and designed, large population-based studies are
Southeast Asia and South America than in studies required to confirm whether an association
from the Middle East and Europe (Baccaglini between OLP and thyroid disorders does exist.
et al. 2013).
In HCV-positive patients, the estimated preva- Graft Versus Host Disease (GVHD)
lence of LP is 1.6–20% (Farhi and Dupin 2010) Nearly 15,000 patients worldwide receive allo-
being higher than expected in the respective geo- genic hematopoietic cell transplants (HSCT)
graphic areas. In a study from Italy, 44 OLP each year, and GVHD will eventually develop in
patients positive for HCV were compared to a about 40–70% and will represent the leading
group of 144 OLP patients negative for HCV. cause of death in such patients (Imanguli et al.
HCV-related OLP group showed a significantly 2008).
higher association with HLA class II allele Acute GVHD occurs within the first 100 days
HLA-DR6 (52% vs. 18%), which may partially of transplantation and comprises dermatitis, enter-
explain the peculiar geographical heterogeneity of itis, and hepatitis with immunosuppression and
the association between HCV and OLP, probably cachexia. Chronic GVHD develops after day
6 M.J. McCullough et al.

100 and comprises an autoimmune-like syndrome chronic inflammatory process. In order to better
comparable to ulcerative colitis, primary biliary understand the pathobiological events that under-
cirrhosis, Sjögren’s syndrome, rheumatoid arthri- lie the development of OLP lesions, it is important
tis, and lupus-like disease with glomerulonephri- to recall some basic concepts of oral immunology.
tis. The skin is a primary target in chronic GVHD
and exhibits either a lichenoid eruption or
sclerodermatous changes (Lodi et al. 2005a). The Immunology of the Oral Cavity
Oral involvement occurs in 33–75% of patients
with acute GVHD and up to 80% of patients with The oral cavity immune system consists of innate
chronic GVHD (Imanguli et al. 2008). Oral muco- and adaptive immune system components. The
sal GVHD resembles OLP both clinically and innate immune system consists of all the immune
histologically. defenses that lack an immunologic memory
Squamous cell carcinoma (SCC) may develop (Delves and Roitt 2000). In the oral cavity the
in oral and cutaneous chronic GVHD, as reported oral mucosa serves as a mechanical barrier to
in few case reports (Lodi et al. 2005b). Most microbial colonization. Loss of tissue integrity
patients who undergo allogeneic HSCT receive places the oral cavity at risk of opportunistic
stem cells from MHC-identical donors. In these microbial infection.
patients, GVHD is initiated by donor T cells that Saliva makes up another portion of the oral
recognize a subset of host peptides called minor innate immune system. Saliva is rich in water
histocompatibility antigens (miHAs). Although and mucin that acts to provide a moist barrier
the antigen specificity of LP and mucocutaneous that helps to limit microbial colonization. It also
GVHD is probably distinct, it is likely that they consists of numerous ions, such as bicarbonate,
share similar immunological effector mechanisms phosphate, etc., that neutralize pH and keep the
resulting in T-cell infiltration, epithelial basement saliva supersaturated to prevent tooth deminerali-
membrane disruption, basal keratinocyte apopto- zation. Salivary antibodies, specifically secretory
sis, and clinical disease (Lodi et al. 2005a). IgA, exist in saliva and act as a first line of defense
The role of TNF-alpha as a major effector by limiting colonization and invasion of microor-
molecule in GVHD has been confirmed in a num- ganisms into the epithelium (Brandtzaeg 2007;
ber of experimental systems. Importantly, neutral- Feller et al. 2013). Saliva contains numerous non-
izing anti-TNF-alpha antibodies have been shown immune innate factors such as salivary peroxi-
to alleviate cutaneous and intestinal GVHD in dase, myeloperoxidase, lysozyme, cystatins,
both mice and humans (Herve et al. 1992; proline-rich proteins, mucin, peroxidase,
Brown et al. 1999). lactoferrin, and statherin which also work to pre-
vent microbial colonization through antifungal,
antibacterial, antiviral, and antiparasitic properties
Pathogenesis of OLP (Tenovuo 1998).
The oral innate immune response also consists
Much is still not known about the of phagocytic cells such as macrophages and neu-
etiopathogenesis of OLP. OLP is characterized trophils. Macrophages are phagocytic cells
by a chronic T-cell inflammatory infiltrate with derived from monocytes, while neutrophils are
basal cell degeneration including vacuolar degen- polymorphonuclear granulocytes. Both work to
eration, hyperkeratosis or parakeratosis, and saw- detect infections organisms and destroy infectious
tooth rete ridges (Sugerman et al. 1993, 2002; organisms through the process of phagocytosis
Zhou et al. 2002). The mechanism that irrevers- (Aderem 2003). Eosinophils, basophils, and
ibly switches on this process resulting in the mast cells also constitute part of the oral innate
chronic disease state is currently unknown. response working to protect against parasites
There are multiple theories on OLP pathogenesis (eosinophils) while also being involved in the
and the mechanisms that underpin the ongoing host allergic reactions. Mast cells especially are
Oral Lichen Planus 7

thought to play an important immunological role T cells are the major effector cell in cell-
in OLP (Zhou et al. 2002). Natural killer mediated immunity. They consist of helper T
(NK) cells target and destroy tumor cells as well cells (CD4+) which recognize MHC class II mol-
as viruses as part of the oral innate immune ecules, cytotoxic T cells (CD8+) which recognize
response by recognizing peptides presented in MHC class I molecules present on all cells, regu-
the context of major histocompatibility complex latory T cells which are essential for preventing
(MHC) class I molecules. The complement sys- autoimmune diseases while also limiting chronic
tem is a series of 20 serum glycoproteins that form inflammation, NK cells, and memory T cells
membrane attack complexes; opsonization and (Delves and Roitt 2000; Vignali et al. 2008).
chemoattractants work to upregulate phagocytosis Helper T cells play an important role in coordinat-
and mobilize the host immune system to clear ing the adaptive immune response by promoting
pathogens (Delves and Roitt 2000). cellular (cytotoxic T-cell-mediated, via activation
The adaptive immune response in the oral cav- of TH1) or humoral (B-cell-mediated, via TH2
ity consists of antigen-presenting cells (APC), T activation) types of response. Cytotoxic T cells
cells and B cells. The main difference between the on recognition of a foreign antigen induce apo-
adaptive and innate immune system is the adap- ptosis in the infected cell. OLP is characterized by
tive immune system’s ability to tailor the response T-cell accumulation, specifically cytotoxic T-cell
to suit different pathogens as well as remember accumulation, within the superficial lamina pro-
past microbial and viral challenges. The adaptive pria that is directed at the basal cell layer
immune system also uses MHC molecules, spe- (Sugerman et al. 2002; Roopashree et al. 2010).
cifically MHC I and II, to distinguish self from It is this chronic cytotoxic T-cell inflammatory
nonself to allow for a targeted immune response process which defines the chronicity of OLP.
(Delves and Roitt 2000). The APCs of the adap-
tive immune system recognize foreign, nonhost
proteins, known as antigens. APC of the adaptive Pathogenic Mechanisms of OLP
immune system includes macrophages, B cells,
dendritic cells, and Langerhans cells. Langerhans The work of Sugerman and colleagues in the
cells reside in the epidermis with the highest 1990s and early 2000s in the field of OLP patho-
counts found in the nonkeratinized oral mucosal genesis and disease mechanisms established the
tissues including the ventral tongue, soft palate, basis of most research conducted thereafter
lip, and floor of mouth (Daniels 1984). (Sugerman et al. 1994, 1995, 1996, 2000, 2000,
Langerhans cells are thought to potentially play 2002; Khan et al. 2003). In general these mecha-
a role in the pathogenesis of OLP (Sugerman et al. nisms can be divided into specific and nonspecific
2002; Roopashree et al. 2010; Gueiros et al. ones, which involve T cells and dendritic cells
2012). activated by specific (yet unknown) antigens and
The major task of plasma cells (which are MMPs, cytokines and other immune cells,
terminally differentiated B lymphocytes), is to respectively.
produce immunoglobulins, specifically IgA, IgG, A number of biochemical changes, including
IgM, IgE, and IgD. Secretory IgA is the primary altered keratinocyte antigen expression and
antibody of the oral cavity found in present in altered keratinocyte function, have been previ-
saliva which acts as the first line of defense ously suggested to be early events in OLP patho-
(Brandtzaeg 2007; Feller et al. 2013). The other genesis (Holmstrup and Dabelsteen 1979). It was
major task of B cells is the production of memory originally proposed that following altered
B cells whose function is to circulate through the keratinocyte antigen expression, a CD8+ T cell
body and mount a rapid response against previ- on routine surveillance in the epithelium may
ously recognized antigens (Delves and Roitt encounter the keratinocyte antigen by chance
2000). (“chance encounter” hypothesis). Alternatively,
the CD8+ T cell may be attracted to the epithelium
8 M.J. McCullough et al.

by keratinocyte-derived chemokines (“directed inflammatory cytokines, it could be partially


migration” hypothesis) (Sugerman et al. 2002). responsible for the characteristic, chronic course
The migration of activated T cells in the OLP of OLP similar to other chronic inflammatory
infiltrate to oral epithelium can be mediated by diseases such as psoriasis and rheumatoid arthritis
intercellular and vascular adhesion molecules (Eisen et al. 2005).
(ICAM-1 and VCAM) (Eisen et al. 1990). The cellular and molecular constituents of
Upregulation of ELAM-1, ICAM-1, and these pathogenic events will be discussed in detail
VCAM-1, especially by endothelial cells in the in the following paragraphs.
subepithelial vascular plexus in OLP, was demon-
strated in an immunohistochemical study that Putative OLP Antigens
assessed various endothelial-associated adhesion While OLP is not considered to be a typical auto-
molecules in frozen sections from 12 OLP cases immune disease, it is believed that one or more
and nine normal controls (Regezi et al. 1996). epithelial antigens are present in basal
Biopsy specimens from patients with OLP con- keratinocytes. Antigens that are presented by
sistently showed significantly higher levels of MHC class II receptor are processed through an
expression of the three molecules. The prolonged endosomal cellular pathway, while antigens that
overexpression of adhesion molecules on endo- are presented by MHC class I are processed
thelial cells may represent the molecular basis for through a cytosolic cellular pathway (Sugerman
the so-called reactive isomorphism seen in OLP et al. 2002). Hence, the putative antigen presented
patients. Previous studies have shown that in OLP by MHC class II to CD4+ helper T cells in OLP
there is an upregulation of epithelial basement may differ from that presented by MHC class I to
membrane extracellular matrix (ECM) proteins, CD8+ cytotoxic T cells. Alternatively, a single
including collagen types IV and VII, laminin and antigen may gain access to both the endosomal
certain integrins – possibly serving as pathways and cytosolic cellular pathways of antigen presen-
for T-cell migration (Eversole 1997). Finally, tation (Sugerman et al. 2002; Roopashree et al.
cytokines (IL-1, IL-8, IL-10, IL-12, and TNF-α), 2010).
secreted by keratinocytes in OLP, are also chemo- Whether one or more different antigens are
tactic for lymphocytes ultimately leading to tissue involved in disease pathogenesis, it has been
destruction in OLP (Sugermann et al. 1996). suggested that simultaneous antigen presentation
The cell-mediated immunological response to CD8+ and CD4+ T cells in the context of MHC
seen in OLP, possibly initiated by endogenous or classes I and II, respectively, is required to
exogenous factors, is thought to result in the pro- develop persistent T-cell infiltration and CD8+
duction of TNF-α and IFN-γ and keratinocyte/T- cytotoxic T-cell activity in OLP (Sugerman et al.
cell/antigen-presenting dendritic cell associations 2002).
(Eisen et al. 2005; Lodi et al. 2005; Payeras et al. A unifying hypothesis of the specific mecha-
2013). The increased production of TH1 cyto- nisms thought to play a role in the pathogenesis of
kines is a key and early event in LP. This even OLP was introduced by Sugerman et al. (2002).
is, at least in part, genetically controlled, and The hypothesis is based on a theoretical interac-
genetic polymorphism of cytokines seems to gov- tion between CD8+ T cells and CD4+ T cells
ern whether lesions develop in the mouth alone through a “request cytotoxic activity” (RCA) cell
(IFN-γ associated) or in the mouth and skin surface molecule expressed by CD8+ T cells and a
(TNF-α associated) (Carrozzo et al. 2004, Scully RCA receptor expressed by the CD4+ T cells to
and Carrozzo 2008). allow confirmation and initiation of cytotoxic
TNF-α may stimulate the activation of nuclear activity by CD8+ T cells. The hypothesis stresses
factor kappa B (NF-κB) whose increased expres- that this interaction can only take place after each
sion has been seen in OLP (Santoro et al. 2003). type of the T-cell antigen receptors is engaged
Because NF-κB translocation in keratinocytes with a related foreign antigen (Ag), i.e., Ag1 in
may induce the production of several the context of MHC class I engaged by CD8+
Oral Lichen Planus 9

T-cell antigen receptors and Ag2 in the context of been suggested by studies that found significantly
MHC class II engaged by CD4+ T-cell antigen higher chemokine production within OLP lesions
receptors. To date, sound experimental evidence when compared to normal or chronically inflamed
to support this theory is lacking. gingival tissues (Yamamoto and Osaki 1995).

Cell-Mediated Immunity CD4+ T Cells


The majority of intraepithelial lymphocytes in
CD8+ T Cells OLP have been shown to be CD8+ cytotoxic T
An early immunohistochemical study assessing cells (Sugerman et al. 2002), while studies have
the lymphocytic infiltrate in OLP showed this to shown that most lymphocytes in the lamina pro-
be composed almost exclusively of T cells, and pria are CD4+ helper T cells (Ishii 1987; Kilpi
the majority of T cells within the epithelium and 1988). A mixed helper and suppressor activity
adjacent to damaged basal keratinocytes were among OLP lesional T-cell clones in vitro was
activated CD8+ lymphocytes (Kilpi 1988). CD8 identified, suggesting that the balance between
+ T cells were also co-localized with apoptotic immunological help and suppression may deter-
keratinocytes in OLP lesions (Sugerman et al. mine the clinical behavior of the disease
2000). The dominant role of CD8+ T cells and (Sugerman et al. 1994). In this interesting report,
CD4+ T cells in OLP pathogenesis was further Sugerman et al. reported that the majority of T
confirmed by the expression of the chemokines lymphocyte lines extracted from six biopsy spec-
CCR5 and CXCR3, known to be selectively imens of OLP (n = 13) expressed the α/β T-cell
expressed by TH1, in the infiltrating lymphocytes, receptor of which 11 were CD8+ and two were
in addition to the CD8+ T-cell respective ligand CD4+ (Sugerman et al. 1994).
RANTES/CCL5 and IP-10/CXCL10 (Iijima et al. Hence, an early event in OLP lesion formation
2003). may be a presentation of a MHC class II antigen to
Analysis of these previous data suggests that CD4+ helper T cells, followed by keratinocyte
CD8+ T cells are involved in disease pathogenesis apoptosis triggered by CD8+ cytotoxic T cells.
and that activated CD8+ T cells may trigger MHC class II antigen presentation in OLP may
keratinocyte apoptosis in OLP. T-cell lines and be mediated by Langerhans cells (LCs) or
clones isolated in vitro from LP lesions were keratinocytes. Furthermore, increased numbers
more cytotoxic against autologous lesional of LCs have been reported in OLP lesions with
keratinocytes than T cells from the clinically nor- upregulated MHC class II expression (Villarroel
mal skin of LP patients, and the majority of non- Dorrego et al. 2002).
cytotoxic clones were CD4+ (helper/inducer Keratinocytes in OLP have also been shown to
clones) (Sugerman et al. 2000). express MHC class II (Ichimura et al. 2006). High
Furthermore, the cytotoxic activity of CD8+ levels of antigen expression, CD40 and CD80
lesional T-cell clones was partially blocked with expression, and IL-12 secretion by MHC class II
anti-MHC class I monoclonal antibody + antigen-presenting cells (APC) in OLP are
(Sugerman et al. 2000). Hence, early in OLP thought to promote a TH1 CD4+ T-cell response
lesion formation, CD8+ lesional T cells may rec- with IL-2 and IFN-γ secretion (Sugerman et al.
ognize an antigen associated with MHC class I on 2002).
lesional keratinocytes. Following antigen recog- Analysis of these data together suggests that
nition and activation, CD8+ cytotoxic T cells are LCs or keratinocytes in OLP may present antigen
thought to trigger keratinocyte apoptosis associated with MHC class II to CD4+ helper
(Sugerman et al. 2002). T cells that are stimulated to secrete the TH1
Activated CD8+ T cells (and possibly cytokines IL-2 and IFN-γ. Subsequently, CD8+
keratinocytes) may release chemokines that cytotoxic T cells may be activated by the
attract additional lymphocytes and other immune combination of (i) antigen associated with MHC
cells into the developing OLP lesion. This has class I on basal keratinocytes and (ii) TH1 CD4+
10 M.J. McCullough et al.

T-cell-derived IL-2 and IFN-γ. Activated CD8+ memory and will induce a secondary immune
cytotoxic T cells may then trigger basal response (Barrett and Raja 1997).
keratinocyte apoptosis in OLP (Sugerman et al. In the OLP lesions, a large number of LCs are
2002). However, it is essential to note that this present in the basal layer of the epithelium
hypothesis has never been proven by robust (Villarroel Dorrego et al. 2002). It has been pos-
evidence. tulated that in these lesions the LCs play an impor-
A recently discovered subgroup of CD4+ T tant role in presenting antigen to the T lymphocyte
cells, namely, TH17 CD4+ subgroup, has been through class II MHC molecules, introducing not
shown to produce IL-26 and IL-22, in addition only an initial sensitivity to the antigen (primary
to IL-17, which are known to be, when immune response) but also a subsequent second-
uncontrolled, inducers of the inflammatory ary immune response and thus the appearance of
response in different autoimmune conditions, the clinical signs of the disease (Payeras et al.
such as multiple sclerosis, psoriasis, and lupus 2013).
(Payeras et al. 2013).
The proportion of lesional Th1 and Th17 cells Mast Cells
and serum IL-17 levels in patients with OLP Mast cells are preferentially located in the lamina
(n = 40) were shown to be significantly greater propria, near blood vessels and nerves. They are
than controls (n = 15), especially in the atrophic- derived from the CD34+ hematopoietic progeni-
erosive OLP patients compared with those pre- tor that has the ability to activate T lymphocyte,
senting with reticular OLP, suggesting that Th17 undergo degranulation, and release a series of
cells and their cytokine Th17 might play an mediators that modulate the inflammatory
important role in OLP pathogenesis (Xie et al. response (Payeras et al. 2013). Studies have
2012). shown an increased mast cell density in OLP
with approximately 60% of them being
Dendritic Cells (DCs) degranulated, compared with 20% in normal buc-
DCs have an important role in the immunological cal mucosa (Sugerman et al. 2002).
response as they activate T cells through antigenic Utilizing IHC, mast cell density in OLP was
stimulation (Banchereau et al. 2000). Studies have found to be markedly higher in the basement
revealed an increase in the number of DCs in OLP, membrane rupture sites as compared to intact
indicating that they may be associated with its sites, suggesting that this cell might play a direct
pathogenesis (Santoro et al. 2005; Gueiros et al. role in the basement membrane destruction, as
2012). According to Santoro et al., the increase of well as in the T CD8+ lymphocyte migration to
different subsets of DCs, such as Langerhans cells the intraepithelial region (Zhou et al. 2002). Thus,
(LCs), stromal DCs, and plasmacytoid dendritic mast cells have been proposed to be involved in
cells (PDCs), may promote the inflammatory the pathogenesis of OLP. Mast cell degranulation
response in OLP (Santoro et al. 2005). in OLP releases a range of pro-inflammatory
Among these, LCs have been the most studied mediators such as TNF-α, chymase, and tryptase.
DCs. These cells reside in the supra-basal layers TNF-α has been shown to upregulate endothelial
of the stratified epithelium of the skin and oral cell adhesion molecule (CD62E, CD54, and
mucosa and have the principle function of captur- CD106) expression in OLP that is required for
ing and presenting antigens. When LCs capture lymphocyte adhesion to the luminal surfaces of
antigen, they are activated, migrate to the regional blood vessels and subsequent extravasation (Zhao
lymph nodes, and are introduced to the T lympho- et al. 1997). Chymase, a mast cell protease, is a
cytes, producing a primary immune response known activator of MMP-9 (Fang et al. 1997).
(Payeras et al. 2013). Subsequently, when LCs It has been proposed that basement membrane
recapture antigen, this antigen will be presented disruption in OLP may be mediated by mast cell
and recognized by T lymphocytes circulating proteases directly or indirectly via activation of T-
cell-secreted MMP-9 (Sugerman et al. 2002).
Oral Lichen Planus 11

Both TNF-α and chymase stimulate secretion of seen in some pathologies, including cancer, arthri-
RANTES by T lymphocytes which in turn stimu- tis, and cardiovascular diseases (Bode 2003).
late mast cells to release TNF-α and chymase. The primary source of MMPs in OLP is prob-
This cyclical activity has been suggested to con- ably the immune infiltrate. In one study, culture
tribute to the chronicity of OLP (Roopashree et al. supernatants from OLP lesional T cells contained
2010). a higher concentration of MMP-9 and TIMP-1
than those obtained from peripheral blood T cells
Macrophages in both the same OLP patients group and the
Macrophages are phagocyte cells derived from healthy controls, suggesting the presence of addi-
blood monocytes, recruited in the tissues in the tional MMP-9 activators in the OLP lesional
presence of chemotactic signals. They are present T-cell supernatants (Zhou et al. 2001).
in the healthy oral mucosa and in larger numbers MMP-9 activators released from the OLP T cell
during pathological processes (Merry et al. 2012). are believed to help in activating pro-MMP-9,
Macrophages are classified as M1 resulting in basement membrane disruption
(pro-inflammatory) or M2 (anti-inflammatory) (Roopashree et al. 2010). Rubaci et al. (2012)
according to the functions of their effectors showed that the expression of MMP-2 (1.3
(Mantovani et al. 2004). The M1 macrophages vs. 0.7) and MMP-7 (1.7 vs. 0.6) in epithelium
might exacerbate OLP manifestation through the and connective tissues from OLP lesions was
production of pro-inflammatory agents such as greater than normal oral mucosa (P < 0.05). This
TNF-α or IL-1b. These agents, in turn, regulate IHC study was undertaken on a cohort of 29 OLP
the presence of adhesion molecules on the endo- patients and ten healthy controls. Likewise, the
thelial cell surface inducing the production of study revealed that MMP-2/TIMP-1 and MMP-7/
chemokines (RANTES) by the keratinocytes and TIMP-1 ratios were higher in the OLP patient
resulting in an increase in the lesional inflamma- group than in the control group (P < 0.05). These
tory cells recruitment. Furthermore, it has been results support the view that increased MMP
shown that the production of TNF-α by the mac- expression and imbalance between MMPs and
rophages can initiate the basal keratinocyte apo- TIMPs may play a role in the pathology of OLP.
ptosis and indirectly increase the disruption rate of
the basement membrane by MMP-9, produced by Chemokines
T cells (Merry et al. 2012). Chemokines are a family of small cytokines (pro-
teins that are involved in cell signaling and medi-
Soluble Factors ating immune reactions), which were initially
identified by their modulator action on the inflam-
Matrix Metalloproteinases (MMPs) matory response (Payeras et al. 2013). More atten-
MMPs are a family of zinc-containing endo-pro- tion has been given to these proteins recently,
teinases with at least 20 members with the princi- especially due to their potential function on endo-
pal function of proteolytic degradation of thelial cells and possible involvement in chronic
connective tissue matrix proteins. MMPs share inflammation and tumor progression (Kiefer and
biochemical properties but retain distinct sub- Siekmann 2011). Moreover, evidence has shown
strate specificities (Zhou et al. 2001). MMP pro- the role chemokines play in different autoimmune
teolysis is regulated by the action of endogenous diseases, such as rheumatoid arthritis and multiple
inhibitors, including the tissue inhibitors of meta- sclerosis, through lymphocyte recruitment and
lloproteinases (TIMPs), which are thought to form establishment of ectopic lymphoid structures in
stable inactive enzyme-inhibitor complexes with the target organs in affected individuals
MMPs or pro-MMPs (Sugerman et al. 2002). An (Godessart and Kunkel 2001).
imbalance between MMPs and TIMPs can be RANTES (regulated on activation, normal T
associated with the process of tissue destruction cell expressed and secreted) is a member of the
CC chemokine family and is produced by various
12 M.J. McCullough et al.

cells, including activated T lymphocytes, bron- in the etiopathogenesis of OLP or a byproduct of


chial epithelial cells, rheumatoid synovial fibro- the disease.
blasts, oral keratinocytes, and mast cells.
RANTES plays a critical role in the recruitment Epithelial Components
of lymphocytes, monocytes, natural killer cells,
eosinophils, basophils, and mast cells in OLP The Epithelial Basement Membrane
(Roopashree et al. 2010). This chemokine has a Keratinocytes contribute to the structure of the
number of cell surface receptors (CCR1, CCR3, epithelial basement membrane by secreting colla-
CCR4, CCR5, CCR9, and CCR10) that have been gen IV and laminin V into the basement mem-
identified in OLP (Sugerman et al. 2002). brane zone (Marinkovich et al. 1993). Evidence
The role that RANTES may play in OLP path- from mouse models suggests that keratinocytes
ogenesis was hypothesized by Sugerman and col- require a basement membrane-derived cell sur-
leagues (2002) who suggested that RANTES vival signal to prevent the onset of apoptosis
secreted by OLP lesional T cells may attract (Pullan et al. 1996). Similarly, lack of appropriate
mast cells into the developing OLP lesion and ECM anchorage results in a form of cell death
subsequently stimulate mast cell degranulation. called “anoikis.” Thus, an intact basement mem-
Degranulating mast cells in OLP would release brane is required for keratinocytes survival, and
TNF-α and chymase which in turn upregulates reciprocally healthy keratinocytes are necessary
OLP lesional T-cell RANTES secretion. for normal basement membrane integrity.
According to this hypothesis, such a cyclical Keratinocytes undergoing cell death are no
mechanism may underlie OLP chronicity. longer able to perform this reciprocal function,
Ichimura et al. (2006) analyzed the and as such keratinocyte apoptosis triggered by
chemokines and their receptors expressed in the intraepithelial CD8+ cytotoxic T cells may result
epithelium of OLP by means of DNA microarray. in epithelial basement membrane disruption in
The study found that high levels of MIP-3a/ OLP, thus fueling anoikis and allowing for non-
LARC/CCL20, and its receptor CCR6, are specific T lymphocytes present in the sub-
expressed on the lesional epithelium. Further- epithelial zone to migrate into the epithelium
more, DC-CK1/CCL18, ELC/CCL19, SDF-1/ (Chainani-Wu et al. 2001). Both keratinocyte apo-
CXCL12, and CXCR4 expressions were also ptosis and basement membrane disruption may be
increased. Immunohistological analysis showed involved in the pathogenesis of OLP, and such a
that high numbers of LCs were present in the cyclical mechanism has been postulated to under-
epithelium of OLP. Lesional epithelium also lie disease chronicity (Sugerman et al. 2002;
expressed high levels of the ligands specific for Lodi et al. 2005a).
CXCR3 and CCR5 (e.g., RANTES/CCL5).
Based on these results, it was suggested that infil- Keratinocyte Apoptosis
tration of LCs may be regulated by CCR6 Keratinocyte apoptosis is a common and key fea-
(as these receptors are expressed by LCs) and ture in the histopathological findings of OLP
that LCs residing in the lesional epithelia may lesions and is considered a major diagnostic cri-
represent a mature phenotype. Moreover, infiltra- terion (Ismail et al. 2007; Eisen et al. 2005; Scully
tion of T cells in OLP could be mediated by et al. 1998). Therefore, keratinocyte apoptosis has
signaling pathways through CXCR3 and CCR5. been postulated to be intentionally involved in the
Thus, it would appear that there is a etiopathogenesis of OLP (Sugerman et al. 2002).
dysregulation of specific chemokines, and chemo- A number of mechanisms have been suggested
kine receptors, and that this would appear to be by which CD8+ cytotoxic T cells can trigger
related to expression of chemokine receptors on keratinocyte apoptosis in OLP. These include
LCs. Similar to the observed dysregulation of (i) T-cell-secreted TNF-α binding to TNF receptor
MMPs/TIMPs and keratinocyte/basement mem- 1 (TNFR1) on the keratinocyte surface, (ii) T-cell
brane noted above, this may be either involved surface CD95L (Fas ligand) binding CD95 (Fas)
Oral Lichen Planus 13

Fig. 1 Clinical presentation of patients with OLP: (a) reticular, (b) erosive/ulcerative, (c) atrophic and plaque-like, and
(d) desquamative gingivitis (atrophic and erosive forms)

on the keratinocyte surface, or (iii) T-cell-secreted buccal mucosa, tongue, alveolar ridge, and gingi-
granzyme B entering the keratinocyte via val tissues (Axell and Rundquist 1987).
perforin-induced membrane pores (Sugerman
et al. 2002). All of these mechanisms may activate
the keratinocyte caspase cascade, resulting in Clinical Appearance
keratinocyte apoptosis. Elevated levels of TNF-α
in the serum of OLP patients were identified OLP classically presents in a bilateral, symmetri-
(Sugermann et al. 1996). cal pattern with the buccal mucosa (bilaterally)
In summary, while it is well established that being the most typical site of involvement
apoptosis of basal keratinocytes is a key feature of (Fig. 1); however, any other oral mucosal sites
OLP, the exact pathogenic mechanisms have not and the lips can also be involved (Al-Hashimi
yet been elucidated. et al. 2007; Ismail et al. 2007; Scully and Carrozzo
2008). Other common sites of involvement
include the tongue (Fig. 1c), gingival (Fig. 1d),
Clinicopathologic Features labial mucosa, and vermilion of the lower lip
(Eisen et al. 2005; Al-Hashimi et al. 2007; Scully
OLP is the most common mucocutaneous condi- and Carrozzo 2008; Parashar 2011). Patients with
tion in the mouth and affects approximately 1–2% isolated lip lesions and tongue lesions have been
of the population (Axell and Rundquist 1987). described although many patients who present
Areas which are typically affected include the with isolated lesions eventually develop more
14 M.J. McCullough et al.

widespread disease (Eisen et al. 2005). Lesions of found OLP to be the most common cause of
OLP affecting the palate, floor of the mouth, and desquamative gingivitis (71% and 75%), while
upper lip are not common. pemphigoid and pemphigus were next in fre-
The oral manifestations of OLP have been quency (Leao et al. 2008; Lo Russo et al. 2009).
described in many large studies. Clinically, there OLP isolated to a single oral site other than the
are six clinical subtypes of OLP that can be seen gingiva is uncommon.
individually or in combination: reticular, plaque- In general, bullous and papular forms are rare
like, atrophic, erosive, papular, and bullous in the oral mucosa (Parashar 2011). The erosive
(Ismail et al. 2007; Scully and Carrozzo 2008; lesions hardly ever remit spontaneously and may
Farhi and Dupin 2010). The most common of lead to confusion with other vesiculobullous dis-
these are the reticular, erosive, and plaque-like eases, which share similar clinical features (Eisen
subtypes, and these variants can coexist within et al. 2005).
the same patient (Fig. 1). The plaque form of OLP mimics leukoplakia in
The reticular lesions, the most recognized form that it appears as a white, homogeneous, slightly
of OLP, encompass white lesions, which appear as elevated, multifocal, smooth lesion. The plaque
a network of connecting and overlapping lines form of OLP commonly affects the dorsum of the
referred to as Wickham striae, papules, or plaques tongue and buccal mucosa (Ismail et al. 2007).
(Eisen et al. 2005). Although some patients may OLL and OLR have similar features, clinically
display an impressive array of diffuse and wide- and histologically, to OLP, but have a less charac-
spread reticulated lesions, they rarely complain of teristic morphology or have a distinct cause,
symptoms and often are unaware of their pres- unlike OLP. OLL therefore needs to be distin-
ence. In the absence of the classic reticular pattern guished because treatment modalities are different
on oral mucosal surfaces, it is challenging to clin- from those for OLP (Al-Hashimi et al. 2007).
ically diagnose non-reticular types (Fig. 1b, d) To better define the criteria for diagnosis of
(Scully and Carrozzo 2008). Histologic confirma- OLP, the World Health Organization (WHO)
tion of the diagnosis is thus required. The erosive devised a set of clinicopathologic criteria in
form of OLP (Fig. 1b) may present with erythema 1978 (Table 1) (Kramer et al. 1978). However,
caused by inflammation or epithelial thinning, or these criteria lacked consensus regarding a clini-
both, and ulceration/pseudomembrane formation cal and histologic diagnosis of OLP, and so mod-
with periphery of the lesion surrounded by retic- ifications were proposed to the WHO criteria in
ular keratotic striae (Ismail et al. 2007). Atrophic 2003 (Table 2), which resulted in a substantial
and erosive OLP lesions result in varying degrees increase in consensus and clinicopathologic cor-
of discomfort. The number of ulcerations is vari- relation (Epstein et al. 2003; van der Meij and van
able as are their size and location; rarely, bullae der Waal 2003). In addition, knowledge about
that rupture easily may be observed in the erosive history of systemic diseases, history of drug use,
form of OLP (Eisen et al. 2005). If the erosive and cutaneous lesions can be helpful in arriving at
subtype of OLP only affects the gingival tissue, a definite diagnosis.
the descriptive clinical term desquamative gingi-
vitis is often used (Fig. 1d). OLP is confined to the
gingiva in about 10% of patients (Mignogna et al. Signs, Symptoms, Clinical Behavior
2005; Scully and Carrozzo 2008). Erosive lesions and OLP Scoring Systems
resembling those observed in other
vesiculobullous diseases including pemphigoid, The clinical signs and symptoms of OLP vary. In
pemphigus, linear IgA disease, and foreign body many patients, the onset of OLP is insidious, and
gingivitis can also produce desquamative gingivi- patients are unaware of their oral condition. Some
tis not easily identified as OLP unless there are patients report roughness of the lining of the
coexistent reticular lesions on the gingiva or else- mouth, sensitivity of the oral mucosa to hot or
where in the oral cavity. Two recent cohort studies spicy foods, painful oral mucosa, red or white
Oral Lichen Planus 15

Table 1 Original WHO diagnostic criteria of OLP (Kramer et al. 1978)


Clinical criteria
Presence of white papule, reticular, annular, plaque-type lesions, gray-white lines radiating from the papules
Presence of lacelike network of slightly raised gray-white lines (reticular pattern)
Presence of atrophic lesions, with or without erosion, and possibly also bullae
Histopathological criteria
Presence of thickened ortho- or parakeratinized layer in sites that are normally keratinized, and if site is normally
nonkeratinized, this layer may be thin
Presence of Civatte bodies in basal layer, epithelium, and superficial part of connective tissue
Presence 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
Signs of liquefaction degeneration in the basal cell layer

Table 2 Modified WHO diagnostic criteria of OLP and OLL (van der Meij and van der Waal 2003)
Clinical criteria
Presence of bilateral, more or less symmetrical lesions
Presence of a lacelike network of slightly raised gray-white lines (reticular pattern)
Erosive, atrophic, bullous, and plaque-like lesions are only accepted as a subtype in the presence of reticular lesions
elsewhere in the oral mucosa
In all other lesions that resemble OLP but not complete with the aforementioned criteria, the term “clinically compatible
with” should be used
Histopathological criteria
Presence of well-defined band-like zone of cellular infiltration that is confined to the superficial part of the connective
tissue, consisting mainly of lymphocytes
Signs of “liquefaction degeneration” in the basal cell layer
Absence of epithelial dysplasia
When the histopathological features are less obvious, the term “histopathologically compatible with” should be used
Final diagnosis of OLP or OLL
To achieve a final diagnosis, clinical as well as histopathological criteria should be included
OLP: a diagnosis of OLP requires fulfillment of both clinical and histopathological criteria
OLL: the term OLL will be used under the following conditions
(1) Clinically typical of OLP but histopathologically only “compatible with” OLP
(2) Histopathologically typical of OLP but clinically only “compatible with” OLP
(3) Clinically “compatible with” OLP and histopathologically “compatible with” OLP

patches on the oral mucosa, or oral ulcerations signs of OLP are diminished (Ismail et al. 2007).
(Ismail et al. 2007). Factors such as stress may aggravate the clinical
Symptoms and signs can range from patients presentation of the disease. Precipitating factors
being unaware of the disease, with lesions that are similar to the Koebner phenomenon, which is
completely asymptomatic as in reticular OLP, to characteristic of cutaneous LP whereby lesions
those experiencing mucosal sensitivity and burning develop in response to trauma, can also affect
and debilitating pain. Approximately two-thirds of the oral cavity where sharp cusps and ill-fitting
the patients affected with OLP experience some dental prosthesis may be the triggers.
degree of oral discomfort (Parashar 2011). Accumulation of plaque and calculus can also
OLP has periods of relapses and remissions. exacerbate OLP, probably because of the Koebner
During a period of exacerbation there will be an phenomenon (Mignogna et al. 2005). Gingival
increase in symptoms and clinical signs, while OLP can eventually lead to gingival recession,
during periods of quiescence, symptoms and advanced periodontal disease, and so forth, and
16 M.J. McCullough et al.

therapeutic periodontal procedures may aggravate Histopathology of OLP


these conditions (Eisen et al. 2005; Scully and
Carrozzo 2008). The histologic features of OLP were first
Oral post-inflammatory pigmentation (OPP) described by Dubreuill in 1906 and later revised
has been described in patients with OLP and by Shklar in 1972 (Parashar 2011). The WHO
OLR as diffuse brown or black pigmentation fol- developed a set of histopathologic criteria for
lowing the lichenoid lesions distribution OLP in 1978, which was most recently modified
(Mergoni et al. 2011). in 2003 (Tables 1 and 2). Definite diagnostic his-
Currently there is no universally accepted scor- tologic findings include liquefactive degeneration
ing system for OLP; however many have been of the basal cells, colloid bodies (Civatte, hyaline,
proposed to objectively and even subjectively cytoid), homogeneous infiltrate of lymphocytes
measure OLP disease severity. Several studies and histiocytes in a dense, band-like pattern
have proposed using scoring systems with the along the epithelium-connective tissue interface
earlier systems based upon a three- or five-point in the superficial dermis, cytologically normal
scale from no disease to severe disease (Eisen et al. maturation of the epithelium, sawtooth rete ridges,
1990; Thongprasom et al. 1992). Other scoring and hyperkeratosis (orthokeratosis or para-
systems base severity on the level of site involve- keratosis) (Fig. 2) (Eisenberg 2000; Eisen et al.
ment (<> 50% site) (Malhotra et al. 2008). 2005; Ismail et al. 2007; Parashar 2011). In addi-
Some of the more detailed scoring systems tion, the surface epithelium may show signs of
have worked to split OLP into three clinical sub- ulceration, typically seen in erosive LP.
types, reticular, atrophic/erythematous, and ero- Several histologic criteria are considered as
sive/ulcerative (Escudier et al. 2007). For some exclusionary in diagnosing OLP, including the
of these studies the oral cavity is divided into absence of basal cell liquefaction degeneration,
different subsites so each site is given an activ- polyclonal inflammatory infiltrate, abnormal
ity/severity score which can be totaled to give an cytology suggestive of dysplasia, abnormal kera-
entire oral cavity score (Chainani-Wu et al. 2001; tinization, flat rete ridges, and absence of colloid
Piboonniyom et al. 2005; Escudier et al. 2007). In bodies (Eisenberg 2000; Ismail et al. 2007).
some of these scoring systems, presence of ery-
thema, erosions, or ulceration is graded higher
than the presence of reticular lesions alone Oral Lichenoid Reactions (OLR)
(Piboonniyom et al. 2005; Escudier et al. 2007).
One of the main advantages of using a detailed There are various lesions that may resemble OLP
scoring system is the ability to objectively mea- both clinically and histopathologically. These
sure disease activity baseline and changes in dis- lesions are collectively termed OLR and encom-
ease activity for the entire oral cavity and specific pass lichenoid drug reactions (LDR) and OLR to
sites. Recently attempts have been made to vali- dental materials, most notably allergic contact
date some of the more detailed scoring systems sensitivity to amalgam (Koch and Bahmer 1995;
(Chainani-Wu et al. 2012). Time has also been Ostman et al. 1996; Thornhill et al. 2003; Issa
taken with these scoring systems to add subjective et al. 2004; Eisen et al. 2005; Ismail et al. 2007).
measures of the patient’s pain and symptoms mak- As with OLP, OLRs present clinically with a
ing this a complete disease scoring system range of different features ranging from asymp-
(Chainani-Wu et al. 2012). tomatic striae and plaque-like lesions to painful
Currently there are many available disease erythematous and ulcerative lesions. Histopatho-
scoring systems which systems in place to mea- logically OLRs align with OLP showing a band-
sure changes in subjective and objective disease like lymphohistiocytic infiltrate (mainly T cell)
activity in OLP. At this stage no one system has within the lamina propria as well as liquefactive
been universally accepted for use. degeneration of the basal cell layer.
Oral Lichen Planus 17

Fig. 2 Histopathology of
OLP. (a) Low power
photomicrograph showing
parakeratosis, band-like
subepithelial chronic
inflammatory infiltrate, and
sawtooth rete ridges (H&E,
original magnification
X100). (b) High-power
photomicrograph showing
several colloid bodies
(arrows) and liquefactive
degeneration of basal
keratinocytes (arrowheads)
(H&E, original
magnification X200)

Cases of OLRs related to restorative materials (Epstein et al. 2003; Bagan et al. 2004;
will typically present in a direct topographic rela- Al-Hashimi et al. 2007; Ismail et al. 2007).
tionship with the restorative material. The aller- No specific criteria exist for the diagnosis of
gens thought to be responsible for amalgam- LDR; however withdrawal and reexposure to the
contact sensitivity include mercury, amalgam drug resulting in resolution and recurrence can be
alloying metal zinc, copper, silver, gold, or palla- considered diagnostic. Patch testing may be a
dium (Koch and Bahmer 1995; Suter and valuable diagnostic tool for OLR as a result of a
Warnakulasuriya 2016). There has been some evi- contact sensitivity to dental materials (Suter and
dence to suggest that some patients with Warnakulasuriya 2016). In cases of a suspected
amalgam-associated OLRs represent a true OLR as a result of amalgam dental restorations,
delayed hypersensitivity reaction as a result of there is evidence to suggest that replacement of
metal haptens released from dental restorative amalgam can result in resolution or improvement
materials (Laine et al. 1999). LDRs will typically of OLR; in some cases this is irrespective of a
arise with a direct temporal association when tak- positive patch testing result (Laine et al. 1992;
ing certain medications including nonsteroidal Ostman et al. 1996; Thornhill et al. 2003; Issa
anti-inflammatory medications (NSAIDs), some et al. 2004). Thus in cases of suspected OLRs, it
antihypertensive medications, oral hypoglyce- is essential that proper testing including histopa-
mics, penicillamine, and antimalarial medications thology and patch testing be undertaken to ensure
18 M.J. McCullough et al.

a correct diagnosis is made as OLP and OLR individuals who have been sensitized through
benefit from different management protocols. long exposure (Ismail et al. 2007).

Dental Materials Systemic Medications


OLR as an allergic reaction to dental materials has Medications, such as antihypertensives ( in par-
been widely reported, with many studies ticular beta-blockers and ACE inhibitors), dap-
documenting contact hypersensitivity to dental sone, oral hypoglycemics, NSAIDs,
materials such as amalgam (Lind et al. 1986; penicillamine, phenothiazines, antimalarials, sul-
Thornhill et al. 2003), composite (Lind 1988), fonylureas, and gold salts, have been associated
dental acrylics (van Loon et al. 1992), cobalt with OLRs (Scully et al. 1998; Al-Hashimi et al.
(Torresani et al. 1994), and nickel (Ismail et al. 2007; Ismail et al. 2007).
2007) presenting as OLR. Some studies also More recently, OLR induced by antiretroviral
showed resolution of OLR following replacement medications for treatment of human immunodefi-
of causative restorations. Laine et al. reported a ciency virus (HIV) has been reported (Scully and
complete remission of OLR lesions associated Diz Dios 2001). Clinical identification of LDR
with amalgam in a small cohort of seven patients, has been based largely on subjective criteria
a marked remission in six patients, and no change although there may sometimes be a tendency for
in two patients (total n = 15) after amalgam the oral lesions to be unilateral and erosive (Eisen
replacement and a mean follow-up of 3.2 years et al. 2005). Histology may be beneficial as
(Laine et al. 1992). In another trial, Thornhill et al. lichenoid lesions may have a more diffuse lym-
reported that amalgam replacement resulted in phocytic infiltrate and contain eosinophils and
improvement in 93% of patients (28 out of 30) plasma cells, and there may be more colloid bod-
who had amalgam-contact hypersensitivity ies than in classical OLP (Eisen et al. 2005; Scully
lesions (Thornhill et al. 2003). Gingival OLR and Carrozzo 2008).
lesions, in particular, were reported to be non- The most reliable method to diagnose LDRs is
responsive to amalgam replacement for unknown to note if the reaction resolves after the offending
reasons (Henriksson et al. 1995). drug is withdrawn and if it returns when the
Diagnosis of OLR will commonly depend on patient is challenged again. As this is both imprac-
the topography and distribution of the lesions as in tical and potentially unsafe, empiric withdrawal of
most cases, OLR are indistinguishable from idio- a potentially offending drug and substitution with
pathic OLP, clinically or histologically (Thornhill another agent may not be warranted. After the
et al. 2003). Cutaneous patch testing may also offending drug is withdrawn, it may be months
play a role in differentiating these lesions (Scully before the LDR resolves. Interestingly, it was
and Carrozzo 2008; Ismail et al. 2007). Thornhill reported that LDR may develop months or even
et al. found that 70% of amalgam-contact hyper- years after a patient takes a drug (Eisen et al. 2005).
sensitivity lesions (presented as lichenoid reac-
tions) were patch test positive for amalgam or
mercury compared with only 3.9% of OLP cases Cutaneous Lesions
(Thornhill et al. 2003).
To date, OLP and OLR lack internationally LP may affect the hair follicles, nails, esophagus,
accepted distinguishing features, and the diagno- and, less frequently, the eyes, urinary tract, geni-
sis of OLR can be challenging as the pathogno- tals, nasal mucosa, and larynx. Scalp involvement
monic features of OLR are yet to be identified causes pruritic, follicular and perifollicular, scaly,
(Ismail et al. 2007). violaceous papules, referred to as lichen
Recent findings have suggested that OLR planopilaris, and can also lead to permanent
appears to be the result of cell-mediated contact patchy hair loss known as scarring alopecia.
hypersensitivity to dental materials, in susceptible When LP affects nails, it causes pitting, subungual
Oral Lichen Planus 19

hyperkeratosis, and permanent nail loss (Farhi and much less common in males than females, recog-
Dupin 2010). nition and treatment of the disease are important
Skin lesions characteristically present as flat- as malignant transformation of penile LP has also
topped, polygonal, violaceous papules regularly been reported (Bain and Geronemus 1989).
covered by a network of fine lines affecting the The clinical features of esophageal LP have
wrists, ankles, and genitalia (Bornstein et al. been well documented, and the disease appears
2006). Cutaneous LP may also appear in several to develop most commonly in patients with OLP
atypical forms that are not easily recognizable. As (Evans et al. 2000), while conjunctival, laryngeal,
previously mentioned, approximately 15% of or other mucosal involvement is rarely reported
patients with OLP develop cutaneous lesions, (Eisen et al. 2005).
while OLP occurs in 70–77% of patients with
cutaneous LP (Eisen 1999; Farhi and Dupin 2010).
Typically, cutaneous lesions develop within Malignant Potential of OLP
several months after the appearance of the oral
lesions, and the severity of the oral lesions does Since the first report of malignant transformation
not seem to correlate with the extent of cutaneous of OLP (Hallopeau 1910), numerous studies have
involvement, i.e., while OLP is chronic and rep- attempted to address this issue. Malignant trans-
resents a common cause of morbidity, cutaneous formation rates ranging from 0% to 12.5% were
lesions are commonly pruritic but self-limiting reported (Gonzalez-Moles et al. 2008). Although
(Eisen et al. 2005). these findings appear to support the potentially
malignant character of OLP, it remains a contro-
versial topic.
Other Mucosal Lesions Several authors agreed to a frequency of malig-
nant transformation between 0.4% and 5%, over
Undoubtedly, the most frequent extraoral site of periods of observation from 0.5 to over 20 years
involvement in female patients with OLP is the with an annual rate between 0.2% and 0.5%. (van
genital mucosa with lesions developing in 20% of der Meij et al. 2003; Al-Hashimi et al. 2007;
women with OLP compared with only 2–4% of Scully and Carrozzo 2008). However, reviews
men with OLP (Ismail et al. 2007; Scully and limited to selected studies on malignant potential
Carrozzo 2008; Farhi and Dupin 2010). The asso- of OLP with a follow-up of more than 2 years
ciation of LP of the vulva, vagina, and gingiva is showed that when strict criteria were applied, the
recognized as the vulvovaginal-gingival syn- malignant transformation rate is 0–2% (Ismail
drome (Pelisse 1989), and the male counterpart et al. 2007).
is known as penogingival syndrome (Bain and It has been thought that the increased risk of
Geronemus 1989). oral cancer appears to be independent of the clin-
When LP affects the genital mucosa, the ero- ical type of OLP and therapy administered
sive form of the disease is the predominant type (Gandolfo et al. 2004).
although asymptomatic reticular lesions can be The first critical review regarding OLP malig-
identified in about a quarter of all patients (Eisen nant transformation appeared in the Journal of
et al. 2005). Various symptoms including burning, Oral Pathology about four decades ago
pain, vaginal discharge, and dyspareunia are fre- (Krutchkoff et al. 1978), included data published
quent and are noted in patients with erythematous up to 1977, and the authors recommended strict
and erosive disease. criteria (Table 3) to be adopted to definitively
Reports of malignant transformation of genital accept the malignant transformation in OLP.
LP in women (Dwyer et al. 1995) underscore the After applying these new criteria, they concluded
need for an early diagnosis and the institution of that only 15 of the 223 cases reported in the
prompt treatment for these patients. Although the literature should be unquestionably accepted as
concomitant involvement of oral and genital LP is malignant transformation in OLP. The remaining
20 M.J. McCullough et al.

Table 3 Criteria for malignant transformation of OLP were reported, of which 33 (34%) met the pro-
(Krutchkoff et al. 1978) posed criteria. According to the authors, the high
A. Original diagnosis must have been properly verified, incidence of malignant transformation described
with histological evidence demonstrating at least the last in many studies may be due to the misdiagnosis of
two of these four features
some lesions as OLP or to the analysis of a highly
Hyperkeratosis or parakeratosis
selected study population (e.g., predominance of
Sawtoothed rete ridges
Superficial infiltrate of lymphocytes
patients referred to specialists). This investigation
Basal cell liquefaction concluded that nearly all of the case reports lacked
B. History and follow-up precise documentation and that still no consensus
Clinical and histological features of the alleged had been established about the criteria for the
transformation must have been adequately described histopathologic diagnosis of OLP (van der Meij
(information on age and sex of patient and on the precise et al. 1999a).
location and clinical description of the lesion)
van der Meij et al. further emphasized the need
The reported transformation should have had proper
follow-up (minimum of 2 years) with all changes in
for standard criteria for a firm diagnosis of OLP to
clinical features properly recorded be universally adopted (van der Meij et al. 1999a, b).
C. Tobacco exposure The designation OLL was later proposed for
Tobacco habits should have been properly documented cases that are clinically characteristic and histo-
to help distinguish between true malignant logically compatible, clinically compatible and
transformation and conventional carcinomas occurring in histologically characteristic, or clinically and his-
the mouths of patients who happened to have OLP
tologically compatible with OLP (Table 2) (van
der Meij and van der Waal 2003). It is currently
proposed by some key authors that OLL rather
cases were excluded for at least one of the follow- than OLP is at high risk of developing cancer
ing reasons: (1) insufficient data to support the (Bornstein et al. 2006; van der Meij et al. 2007;
OLP diagnosis, (2) appearance of oral cancer in Gonzalez-Moles et al. 2008).
an area anatomically distant from the OLP, and On the other hand, a review by Mattsson et al.
(3) inadequate historical data on previous expo- largely based on follow-up studies reported a
sure to carcinogens. The authors commented: “If higher incidence of oral cancer in OLP patients
OLP prevalence is accepted to be 1–2% of general and concluded that OLP should be considered a
population over 15 years, and if malignant trans- potentially malignant condition with a transfor-
formation rate is 1% in a mean period of 5 years, mation rate of 0.5–2% (Mattsson et al. 2002).
then, from 10 to 20 patients per 100 000 inhabi- Many other studies using strict diagnostic criteria
tants should develop oral cancer in a mean period have shown a significant risk of malignant trans-
of 5 years. This would indicate that in many parts formation of OLP to squamous cell carcinoma
of the world all oral carcinomas should develop on (SCC) (Holmstrup et al. 1988; Gandolfo et al.
an OLP, which is rather improbable” (Krutchkoff 2004; Rodstrom et al. 2004).
et al. 1978). They further drew the conclusion that Finally, the World Health Organization, in its
there was insufficient evidence to accept an inher- latest volume on the Pathology and Genetics of
ent biological potential of OLP to progress to Head and Neck Tumours (Gale et al. 2005), has
cancer, but they acknowledged that OLP patients recommended the development of diagnostic
have a slightly higher tendency to develop carci- criteria to differentiate between OLP and OLL
nomas compared to individuals without OLP but declared that both lesions should be consid-
(Krutchkoff et al. 1978). ered at risk of malignant transformation until such
Almost 20 years later, van der Meij et al. criteria become available.
(1999a, b) reviewed studies on the malignant In 1985, Krutchkoff and Eisenberg (1985)
transformation of OLP published from 1977 to coined the term lichenoid dysplasia (LD) to
1999, applying the Krutchkoff criteria. During describe lesions that resemble OLP histologically
this period, 98 new malignant transformations and also show features of dysplasia. They
Oral Lichen Planus 21

suggested that OLP has no inherent predisposition marrow transplantation patients found that oral
to become malignant and that reported cases of cancer had the highest risk among cancers, being
malignant transformation in OLP lesions were due 11.1 times more frequent than expected (Curtis
to lack of discrimination between OLP and LD or et al. 1997). The significant risk factors for OSCC
failure to identify a concomitant exposure to known were chronic GVHD, limited-field irradiation, and
carcinogens (Krutchkoff and Eisenberg 1985). male sex. In one of these studies, head and neck
The assumption for the proposal of malignant cancer was the only solid cancer observed in a
potential in LD rather than OLP was that any group of 78 patients undergoing bone marrow
departure from normal epithelial maturation and transplantation for Fanconi anemia, with the fre-
growth altogether excludes a diagnosis of OLP quency of such tumors was 167 times higher than
(Eisenberg 2000), although consensus on such expected (Deeg et al. 1996).
criteria has never been reached, and some authors Clearly, bone marrow transplantation patients
consider dysplasia a very common feature of OLP have numerous risk factors that may enhance
(Lodi et al. 2005b). their likelihood of developing malignancies (pri-
The entity LD might correspond to two groups mary immunodeficiencies, immunosuppressive
of conditions: those with clinical features similar to treatments, viral infections, and probably genetic
OLP but dysplastic at the histological level and predisposition to cancer) that do not allow a com-
those with lichenoid microscopic features (band- parison with OLP patients; however, the similarity
like lymphocytic infiltration in particular) and clin- of the oral conditions and the apparent common
ical features which do not resemble classic OLP tendency to transform are worthy of careful con-
(unilateral distribution, absence of reticular lesions) sideration (Lodi et al. 2005b). Other lichenoid
(Lodi et al. 2005b). The former could represent an lesions that can undergo malignant transformation
early phase in the malignant transformation of OLP, include discoid lupus erythematosus, in particular
while the latter could represent an OLL with under- of the lip (Voigtlander and Boonen 1990) and
lying various clinical conditions that may have amalgam-associated OLR (Ostman et al. 1996).
lichenoid histopathology including lichenoid reac- Interestingly, in a prospective study on prema-
tions, lupus erythematosus, leukoplakia, erythroleu- lignant potential of OLP, all cases of malignant
koplakia, and proliferative verrucous leukoplakia transformation involved lesions that the authors
(PVL). PVL, particularly in the early stages, can included in the group of OLL because they did not
have features, both clinical and histologic, that can fulfill both clinical and histologic criteria for OLP
be confused with OLP, frequently shows dysplastic (van der Meij et al. 2003). In 2007, van der Meij
changes, and is characterized by a high malignant et al. (2007) also studied the number of expected
transformation rate (Lodi et al. 2005b). oral carcinomas in 67 patients with OLP and
125 patients with OLL. All malignant transforma-
tions (4 of 192, 2.1%) appeared in OLL patients,
OLL and Cancer i.e., an annual OLL malignant transformation rate
of 0.71%. Hence, there was no increase in oral
A very interesting, and potentially informative, cancer risk for patients with OLP but a 142-fold
group of patients regarding OLL and oral malig- increase for patients with OLL (P = 0.04).
nancies are those who underwent allogeneic bone There may be considerable overlap between the
marrow transplantation and developed oral clinical and microscopic features of OLR and OLP.
GVHD. Oral GVHD is clinically and histologi- Although no association has been established
cally indistinguishable from OLP. Case reports between OLR and malignant transformation
(Abdelsayed et al. 2002) and large studies (Deeg (Mattsson et al. 2002), Larsson and Warfvinge
et al. 1996; Curtis et al. 1997) describe numerous proposed that there may be a similar rate of malig-
episodes of oral cancers (mainly SCC) in patients nant transformation in OLR to that observed in
with oral GVHD. A large study that investigated OLP, especially in lesions at the lateral border of
the incidence of solid tumors in 20,000 bone the tongue, a frequent site for OLR due to the close
22 M.J. McCullough et al.

Fig. 3 Oral squamous cell carcinoma in OLP. Clinical carcinomas in an OLP patient whose right buccal mucosa
presentation of patients with OLP and who subsequently had previously shown histopathological evidence of OLP.
developed OSCC: (a) An exophytic squamous cell carci- Subsequently developed a more florid erythematous
noma that developed on the right lateral tongue in a patient mucosa in the same area (pictured) that was biopsied and
who previously had histopathologically diagnosed OLP in diagnosed as OSCC. (c) Eight years later, the same patient
the same area. (b) An example of metachronous developed a second OSCC, controlaterally

contact with silver amalgam restorations. They of transformation – rather they considered the loss
reported that the cancer had developed on an of lesion homogeneity at a specific site to be most
OLR in 4 of 724 patients with tongue cancer relevant. This clinical sign is especially useful
(Larsson and Warfvinge 2005). when only a small area is involved, as OLP usu-
ally affects various areas or a large area.
An important reported feature of the presenta-
Tumors Developing in OLP and their tion and clinical course of carcinomas that arise on
Carcinogenesis OLP is their tendency of multiplicity. Mignogna
et al. (2002) found that 29% of patients develop-
Clinically, carcinomas that develop in previously ing carcinomas in OLP had two or more indepen-
diagnosed OLP lesions have been reported to be dent neoplastic lesions (19% with a second tumor,
exophytic keratotic lesions (Lo Muzio et al. 1998; 10% with >2 metachronous tumors) (Fig. 3b, c).
Fatahzadeh et al. 2004) (Fig. 3a), but some may This finding confirmed previous reports by
also show endophytic growth patterns (Lo Muzio Duffey et al. (1996) (20% of patients with second
et al. 1998). Markopoulos et al. (1997) suggested primary tumors) and Lo Muzio et al. (1998)
that rapid expansion of OLP lesion should raise (35.7% of patients with second primary tumors).
suspicion of malignant transformation, but The relatively high frequency of multiple
Mignogna et al. (2001) found neither the exten- intraoral localizations of second primary tumors
sion nor severity of symptoms a useful indicator in previously diagnosed OLP may be attributed to
Oral Lichen Planus 23

the field cancerization phenomenon and indicates tobacco and that secondary to OLP. For this rea-
that OLP may have an intrinsic predisposition to son, some authors recommend the exclusion of
tumor development (Mignogna et al. 2007). smokers with OLP from studies (van der Meij
The metastatic capacity of carcinomas devel- et al. 1999a, b, Lozada-Nur 2000). However,
oping in OLP has been addressed by an earlier according to Lodi et al. (2005b), although some
report by Mignogna et al. (2002) who showed that cases described may be mainly related to tobacco
24% of these patients had detectable lymph node consumption, the exclusion of one putative risk
metastases at the time of diagnosis. In their more factor based on the presence of another appears
recent work, the same authors (Mignogna et al. inappropriate and could prevent the identification
2007) reported that 94% of 97 neoplastic events of new risk factors. Thus, for example, this
observed were TisN0M0 or T1N0M0 approach would have impeded identification of
(intraepithelial neoplasia or microinvasive carci- the super-multiplicative risk of combined tobacco
noma <1 mm) and 6% were stage III (three and alcohol consumption for oral and oropharyn-
tumors) or IV (three tumors). geal cancer development.
Histopathologically, most tumors detected in Therefore, the putative role in OLP transfor-
OLP are well-differentiated SCCs [70% in the mation of well-known risk factors for oral cancer
study by Lo Muzio et al. (1998); 100% in the (tobacco and alcohol) has not been properly eval-
study by Markopoulos et al. (1997)]. Finally, uated in most studies (Lodi et al. 2005b). In a
there are conflicting results on the prognosis of single study, in which this interaction was
patients with neoplasia in OLP, some indicating a addressed, it was suggested that alcohol and
poor prognosis (Hietanen et al. 1999; Mignogna tobacco, or their interaction, cannot explain the
et al. 2001; Mignogna et al. 2002), but Mignogna excess risk for oral cancer found in OLP
et al. (2007) reported 100% 3-year and 97% (Gandolfo et al. 2004).
5-year survival, although there may have been a However, many authors (Murti et al. 1986;
bias in this study as the neoplastic events Barnard et al. 1993; Eisen 2002; van der Meij
corresponded to severe dysplasias ⁄carcinomas in et al. 2003; Gandolfo et al. 2004) found no rela-
situ in most patients, due to a meticulous follow- tionship between tobacco and alcohol consump-
up program. tion on one side and malignant transformation on
Numerous studies (Silverman et al. 1985; the other side in OLP patients.
Markopoulos et al. 1997; Hietanen et al. 1999; Results published by Rajentheran et al. (1999)
van der Meij et al. 2003; Mignogna et al. 2006) indicated that tobacco and alcohol consumption
have been unable to identify risk factors for cancer may even be lower in these patients than in
development in patients with OLP. It has therefore patients developing oral cancer in the absence of
been proposed by some authors that carcinoma- OLP. In a cohort of 24 OLP patients with subse-
tous transformation is part of the natural history of quent OSCC, Mignogna et al. (Mignogna et al.
the disease or is attributable to unknown risk 2001) found only three patients to be smokers and
factors (van der Meij et al. 2003). none to be alcoholic. Munoz et al. (2007) reported
Several risk factors for malignant transforma- that only two out of ten OLP patients who devel-
tions in OLP have been proposed. These include oped OSCC were smokers, none was an alcoholic
erosive forms, tongue lesions, women more than abuser.
men, and sixth to seventh decades of life, but none With respect to the clinical form of OLP,
of them gained significant agreement among numerous authors (Silverman et al. 1985; Barnard
researchers (Gonzalez-Moles et al. 2008). et al. 1993; Markopoulos et al. 1997; Hietanen
Another relevant issue in the context of malignant et al. 1999; Rajentheran et al. 1999; Eisen 2002;
transformation of OLP is whether to include van der Meij et al. 2003) found that atrophic-
patients with chronic oral exposure to carcino- erosive forms predisposed to cancer development,
gens, as it will probably be impossible to differ- but this remains a controversial issue. In some
entiate between the transformation caused by series (Hietanen et al. 1999; Mignogna et al.
24 M.J. McCullough et al.

2001, 2007), plaque-like OLP lesions were also et al. 1999; Gandolfo et al. 2004; Mignogna et al.
relevant, both when they appeared alone and 2007) and might even reduce it. Thus, it has been
when associated with atrophic-erosive lesions. proposed that a microenvironment rich in
Analyses of malignant transformation risk fac- pro-inflammatory cytokines may be especially
tors have also considered the different intraoral favorable for neoplastic promotion, suggesting
sites of OLP. The tongue appears to be the pre- that more aggressive immunosuppressant treat-
ferred site for the emergence of cancer (Holmstrup ments against the inflammatory response in OLP
et al. 1988; Barnard et al. 1993; Markopoulos might restore normal immune surveillance and
et al. 1997; Munoz et al. 2007). interrupt neoplastic progression (Eisen 2002). In
Other studies, however, have reported sites a study of OLP patients treated mostly with topi-
other than the tongue to have a higher risk of cal and/or systemic steroids, therapeutic modali-
malignant transformation in OLP. Mignogna ties did not affect the risk of malignant
et al. (2001) found a significantly higher fre- transformation (Gandolfo et al. 2004).
quency of carcinomas at the midline of the palate, Malignant transformation of OLP has been
gingivae, and lips in a cohort of 502 OLP patients suggested to be related to, or dependent upon, a
who were followed up for periods from 4 months series of molecular stimuli originating in the
to 12 years and an overall malignant transforma- inflammatory infiltrate (Mignogna et al. 2004).
tion rate of 3.7%. Chronic inflammation has been associated with
Regarding gender and age, there appears to be various types of cancer (Clevers 2004), and it
a general consensus that the risk is higher in has been widely reported that the inflammatory
women than in men (Gonzalez-Moles et al. infiltrate can be a strong risk factor for cancer
2008). Some authors reported that an oral cancer development in ulcerative colitis, atrophic gastri-
most frequently develops on an OLP between the tis, and Barret’s esophagitis, among other diseases
sixth and seventh decade of life (Barnard et al. (Balkwill and Mantovani 2001). In fact, it was
1993; Hietanen et al. 1999). proposed by some authors that OLP could be
The mean interval between OLP diagnosis and included in this group of diseases (Mignogna
cancer diagnosis ranges widely from 20.8 months et al. 2004). Some molecules and radicals gener-
to 10.1 years, although the maximum risk is ated by inflammatory cells can act as mutagenic
reportedly between 3 and 6 years after OLP diag- agents for epithelial cells or influence important
nosis (Gonzalez-Moles et al. 2008). The risk of cell cycle regulation mechanisms (Gonzalez-
malignant progression may also increase with the Moles et al. 2008).
use of immunosuppressive agents. Although no Most studies on cell proliferation in OLP have
specific data are available in OLP patients, it is reported a marked increase in the proliferation rate
well known that immunosuppressive treatment, of basal epithelial cells (Valente et al. 2001;
which generally includes corticosteroids, cyclo- Gonzalez-Moles et al. 2006), and some authors
sporine, and tacrolimus, is a risk factor for cancer, have proposed that this might be an important
including OSCC (Kruse and Gratz 2009). A grow- event in the development of cancer in OLP
ing body of evidence now suggests that glucocor- (Taniguchi et al. 2002).
ticoids can act as antiapoptotic agents in epithelial Mignogna et al. (2004) presented the possible
cells to promote cancer progression (Azher et al. role of each type of inflammatory cells in the
2016). The discovery that corticosteroids can malignant transformation of OLP. According to
directly target the oral mucosa via GR expressed their hypothesis, macrophages, mast cells, lym-
by oral keratinocytes (Cirillo et al. 2012) may phocytes, and fibroblasts can contribute to the
have salient clinical implications in the under- process of carcinogenesis in OLP by secreting
standing of the malignant potential of OLP. cytokines, chemokines, MMPs, and RANTES
However, other authors consider that immuno- molecules which have the ability to cause DNA
suppressant therapy does not increase the risk of damage, bypass p53 tumor suppression function,
transformation (Hietanen et al. 1999; Rajentheran
Oral Lichen Planus 25

induce immortalization, and influence growth, which simply favors colonization and overgrowth
survival, angiogenesis, and invasion. of Candida spp. (McCullough et al. 2002).
In addition to the proposal that OLP-related
inflammatory microenvironment is able to initiate
tumorigenesis in normal epithelium, this microen- Patient Management
vironment has also been suggested to represent a
predisposing and enhancing factor toward the The characteristic clinical aspects of OLP (sym-
molecular changes caused by conventional envi- metry, bilateral distribution) are thought by some
ronmental carcinogens, such as tobacco and alco- researchers to be sufficient to make a correct diag-
hol (Mignogna et al. 2004). Interestingly, nosis especially if there are classic skin lesions
smoking was recently shown to alter the inflam- present (Eisen et al. 2005).
matory infiltrate in OLL, reducing the expression However, an oral biopsy with histopathologic
of macrophages, which may in turn affect the study is usually recommended to confirm the clin-
immune surveillance and theoretically the mech- ical diagnosis and mainly to exclude dysplasia and
anisms of malignant transformation (Alrashdan malignancy (Eisen et al. 2005; Al-Hashimi et al.
et al. 2016). 2007; Ismail et al. 2007; Scully and Carrozzo
The role of Candida spp. in the symptomatol- 2008).
ogy and malignant potential of OLP is unclear. The histopathologic assessment of OLP has
Candida spp. is frequently identified in patients been described as a subjective and insufficiently
with intraoral lesions of OLP and other oral poten- reproducible process (van der Meij et al. 1999a),
tially premalignant or malignant lesions including and in about 50% of OLP cases, there is a lack of
oral leukoplakia and OSCC (Zeng et al. 2009; clinicopathologic correlation in the diagnostic
Masaki et al. 2011). Candida spp. has been assessment of OLP (van der Meij and van der
shown to present higher frequency and coloniza- Waal 2003). Gingival LP may be more difficult
tion in those with OSCC compared to controls to diagnose, and direct immunofluorescence of
( p = 0.001 and 0.033, respectively) (Alnuaimi perilesional mucosa may facilitate the diagnosis
et al. 2015). With reference to OLP, studies have and exclude other causes such as vesiculobullous
shown the presence of oral yeast to be anywhere diseases (Eisen et al. 2005). The value of direct
between 40% and 80% for patients with OLP and immunofluorescence for confirmation of the dis-
around 20% and 40% for controls (Jainkittivong ease is well accepted, especially with non-
et al. 2007; Lodi et al. 2007; Masaki et al. 2011). diagnostic histopathologic features and for the
In relation to the association of Candida spp. desquamative gingivitis form of OLP (Eisen
and OLP, the evidence is conflicting. Specifically et al. 2005; Scully and Carrozzo 2008).
the evidence is conflicting in terms of whether Direct immunofluorescence studies of OLP
there is a significant relationship between Can- have shown a linear pattern and intense positive
dida spp. and erosive OLP, specifically whether fluorescence with antifibrogen outlining the base-
this relationship plays a role which enhances ment membrane zone and cytoid-like bodies with
inflammation to aggravate the pathogenic condi- positive immunoglobulin M labeling (Eisen et al.
tion (Zeng et al. 2009). 2005; Ismail et al. 2007; Scully and Carrozzo
Similarly, there is conflicting evidence with 2008). Indirect immunofluorescence studies are
regard to Candida invasion in OLP. At this stage not routinely used in the clinical diagnosis of OLP.
it is not certain if the presence of Candida in OLP It has been proposed that allergy to dental
and other premalignant and malignant conditions materials is common in patients with OLR. Cuta-
is simply a coincidental finding and whether neous patch testing is a recognized and accepted
changes in the local environment as a result of method to identify allergens responsible for type I
OLP, such as roughness of the mucosal surface and IV allergic reactions with Dental Series
and hyperkeratosis, create an ideal environment Epicutaneous Test Battery (Trolab) of patch test
allergens being commonly used. The test
26 M.J. McCullough et al.

substances are applied to normal skin of the back Many therapeutic modalities have been
and read after 72 h. The patient is considered a suggested in the treatment of OLP, with the most
patch test positive to an allergen if they develop currently accepted and reliable modality being the
erythematous vesicular or ulcerative reaction at use of topical steroids with consideration to be
the site of contact (Ismail et al. 2007). given for use of systemic steroids in cases of
Skin patch testing to investigate contact sensi- severe widespread disease and/or refractory
tivity responses to mercury and amalgam pro- cases of OLP (Vincent et al. 1990; Al-Hashimi
duced conflicting results with variable numbers et al. 2007).
of patients being positive in different studies. In
one study, Laine et al. (1992) studied 118 patients
with OLR topographically associated with dental Topical Agents
fillings, 68% of such patients were patch test
positive to metals of fillings materials, particularly Glucocorticoids
mercury and silver nitrate. In another trial, Wong The most commonly employed and useful agents
et al. reported a positive patch test in 39% of a total for the treatment of OLP are topical corticoste-
of 84 patients who presented with OLR related to roids. A response to treatment with midpotency
amalgam fillings (Wong and Freeman 2003). corticosteroids such as triamcinolone, potent fluo-
In a systematic review that analyzed data from rinated corticosteroids such as fluocinolone
14 cohort and five case-controlled studies with a acetonide and fluocinonide, and superpotent halo-
total of 1,158 patients (27% males and 73% genated corticosteroids such as clobetasol has
females, age range 23–79 years) with OLR asso- been reported in 30–100% of treated patients
ciated with amalgam, 16–91% of patients were (Carbone et al. 1999; Thongprasom et al. 2003;
patch test positive for at least one mercury com- Eisen et al. 2005).
pound (Issa et al. 2004). Al-Hashimi et al. (2007) reviewed 12 clinical
Treatment of OLP should be directed at achiev- trials in the context of corticosteroid use for OLP
ing specific goals after considering the degree of (four were placebo controlled, one assessed sys-
clinical involvement, the predominant clinical temic corticosteroids). Most studies were not
type of lesions, the patient’s symptoms, and age. focused on investigating the value of corticoste-
Reticular lesions that are asymptomatic generally roids in the treatment of OLP per se but compared
require no therapy but only observation for the effectiveness of different formulations, differ-
change. In general, all treatment should be aimed ent classes of corticosteroid, different strengths of
at managing atrophic and ulcerative lesions, alle- topical steroids, and different frequency of appli-
viating symptoms, and potentially decreasing the cation. The specific medications included in the
risk of malignant transformation (Eisen et al. review were fluocinonide, fluocinolone acetonide,
2005; Lodi et al. 2005; Al-Hashimi et al. 2007; triamcinolone acetonide, clobetasol propionate,
Scully and Carrozzo 2008). fluticasone propionate, and betamethasone valer-
Mechanical trauma or irritants such as sharp ate/sodium phosphate for topical therapy, with
filling margins, rough surfaces, or badly fitting dosages ranging from 0.025%, 0.1%, to 0.5%
dentures should receive attention. A drug history and the frequency of application varying from
should be obtained to identify reversible causes of two, three, to four times a day. The average dura-
lichenoid eruptions as discontinuation of the tion of the studies was between 4 and 8 weeks,
offending agent can be curative (Eisen et al. 2005). except for one, which was for 6 months, and the
Hypersensitivity reactions should be suspected overall conclusion suggests that corticosteroids
when OLLs are confined to oral mucosal sites in are effective in the management of OLP and are
close proximity to dental restorations. An optimal unlikely to cause serious side effects (Al-Hashimi
oral hygiene program should be instituted in et al. 2007). There were no studies determining if
patients with gingival disease (Eisen et al. 2005). adhesive vehicles are better than mouth rinses.
However, empirical evidence seems to suggest
Oral Lichen Planus 27

that mouth rinses are of value in patients with antifungal therapy before the patient begins using
widespread symptomatic OLP where the lesions topical steroids. Many of these patients can be
are not easily accessible to the placement of identified prior to commencing corticosteroids
ointments or gels. The evidence also suggests and preventive treatment initiated coincident or
that higher potency corticosteroids, such as immediately prior to the initial applications. Com-
clobetasol, are probably more effective mon conditions that have been proposed to predis-
(Al-Hashimi et al. 2007). There was insufficient pose to candidal overgrowth include xerostomia;
evidence regarding different dosages, formula- systemic and/or topical use of antibiotics, cortico-
tions, or modes of delivery of topical steroids steroid asthma inhalants, prostheses, and cigarette
(e.g., paste, spray, mouthwash) to make an smoking (Savage and McCullough 2005).
evidence-based recommendation (Al-Hashimi Development of candidosis often leads to
et al. 2007). immediate interruption to treatment, prolonged
The greatest obstacle in using topical cortico- and amplified morbidity, additional treatment for
steroids in the mouth is the lack of adherence to the infection, delayed management of the original
the mucosa for a sufficient length of time. For this condition, and clouding of the baseline pathosis
reason, some investigators prefer using topical present. Wherever possible, anticipation and pre-
corticosteroids in adhesive pastes although there vention are preferable to a reactive response.
is no data that topical steroids in adhesive bases A refractory response, described by some as
are more effective than as base preparations tachyphylaxis, is characterized by decreasing effi-
(Lo Muzio et al. 2001). Elixir forms of corticoste- cacy of corticosteroids during continued treatment
roids, such as dexamethasone, triamcinolone, and (Hengge et al. 2006) and may result from a num-
clobetasol, have been used as an oral rinse for ber of areas including poor patient compliance;
patients with diffuse oral involvement or for inappropriate instruction and patient use; inappro-
elderly patients who may find it technically diffi- priate application, for example, a carrier may be
cult to apply medication to various active loca- helpful for the gingiva; agent of insufficient
tions of the oral cavity. Careful consideration potency; incorrect diagnosis; and failure to
should be given to the vehicle as unlike skin remove any local cause, for example, a corroded
compounds, which have been well studied, clini- amalgam restoration causing a OLR (Savage and
cal trials that have compared the strength of corti- McCullough 2005).
costeroids in various bases in the oral cavity are Systemic absorption has been reported, and it
generally lacking (Eisen et al. 2005). is thought that absorption of small amounts
Few serious side effects arise with topical cor- through the oral mucosa can take place, but clin-
ticosteroids as they are generally well tolerated. ical experience and laboratory studies have shown
Side effects reported include secondary this not to be of clinical significance in almost all
candidosis, nausea, oral use not tolerated, refrac- cases (Savage and McCullough 2005).
tory response, mucosal atrophy, oral dryness, sore Interestingly, although systemic absorption
throat, bad taste, delayed healing, and systemic and adrenal suppression were reported with
absorption (Savage and McCullough 2005; long-term use of superpotent corticosteroids for
Thongprasom and Dhanuthai 2008). chronic skin diseases (Levin and Maibach 2002),
Given the high rate of commensal oral yeast this does not seem to be the case with oral corti-
carriage in the community, it is expected that some costeroids used for OLP (Thongprasom and
patients will develop a secondary erythematous Dhanuthai 2008). Exceptions arise, and this is an
candidosis or pseudomembranous candidosis issue that should receive consideration with par-
(thrush) (Thongprasom and Dhanuthai 2008). As ticular patient groups along with the occasional
many as one-third of OLP patients treated with idiosyncratic response. Patients with medical con-
topical corticosteroids have been reported to ditions that are of particular concern include dia-
develop secondary candidosis (Vincent et al. betes, hypertension, and tuberculosis. Steroid
1990) which necessitates treatment or instituting mouth rinses in patients with extensive areas of
28 M.J. McCullough et al.

disease and excessive and unmonitored usage are focused on patients with symptomatic OLP that
also a concern. had not responded to topical corticosteroids or
Therefore, careful and frequent follow-up who were at risk of adverse side effects from
examinations are necessary especially with corticosteroids. In their review, Al-Hashimi et al.
chronic use of topical corticosteroids. Temporary (2007) assessed four studies regarding cyclospor-
burning or stinging at the site of application has ine use for OLP (three used 500 mg mouthwash
also been reported with triamcinolone acetonide and one used adhesive gel form). In all of the
0.1% ointment (Laeijendecker et al. 2006). studies, the side effects were minimal and mainly
In general, it has been recommended that ther- consisted of a transient burning sensation, bad
apy should be initiated with a potent preparation taste, and high cost. In OLP patients, systemic
to achieve a rapid response, particularly in erosive absorption is probably low, and most studies did
OLP lesions, and then lowering the strength with not detect cyclosporin in peripheral blood. The
healing, and eventually once the disease becomes results of all of the studies showed a marked
inactive and there is either an absence of lesions or improvement in the oral symptoms. However,
the presence of only white reticular lesions, therapy cyclosporine mouth rinse was not significantly
may be temporarily discontinued (Eisen et al. 2005). better than 1% triamcinolone paste in a controlled,
For intractable erosive OLP lesions, randomized prospective trial that involved
intralesional injections of hydrocortisone, dexa- 13 OLP patients randomly assigned to treatment
methasone, triamcinolone acetonide, and methyl- with cyclosporine rinse or triamcinolone paste for
prednisolone have been used (Eisen et al. 2005). 6 weeks (Sieg et al. 1995).
Frequent injections of steroids, however, are pain- In general, cyclosporin can be an alternative to
ful, not invariably effective, and may result in an conventional treatments for initial control of OLP.
unwanted systemic dose (Eisen et al. 2005). However, it should not be considered as a first
drug of choice because of the high cost of long-
Calcineurin Inhibitors term treatment and the availability of effective
Calcineurin inhibitors are microbially derived alternatives. Severe side effects of systemic cyclo-
immunosuppressive agents that have been primar- sporin, such as hypertension and nephrotoxicity,
ily used in transplant medicine and in the treat- preclude its long-term use for OLP (Lodi
ment of immune-mediated diseases with the et al. 2005).
principle agents being tacrolimus, pimecrolimus, Tacrolimus is a macrolide immunosuppressant
and cyclosporine (Al Johani et al. 2009). derived from Streptomyces tsukubaensis. It is a
Calcineurin inhibitors bind to different cyto- relatively selective inhibitor of calcineurin and
plasmic proteins of T lymphocytes (cyclosporine was initially developed as a systemic agent to
to cyclophilin, tacrolimus, and pimecrolimus to lessen allograft rejection. Tacrolimus has the abil-
FK506-binding protein) to form complexes that ity to inhibit T-cell activation at 10–100 times
in turn inhibit calcineurin leading to suppression lower concentration than cyclosporin. Notably,
of transcription and production of many cyto- topical tacrolimus seems to penetrate skin better
kines. Calcineurin inhibitors have been suggested than topical cyclosporin (Lodi et al. 2005; Al
to be of clinical benefit in the management of Johani et al. 2009). Formulated for topical appli-
some immunologically mediated oral mucosal cation in the management of atopic dermatitis
disorders including OLP (Al Johani et al. 2009). (AD), it was approved in 2000 by the United
There are now numerous reports of the efficacy States Food and Drug Administration (FDA) to
of calcineurin inhibitors in the management of be used in moderate to severe AD for patients
OLP, and effectiveness has been assessed via older than 2 years. Topical tacrolimus has proven
open-label prospective studies, randomized trials, to be of benefit in the treatment of other disorders
retrospective studies, case series, and described in including cutaneous psoriasis, contact allergy,
several case reports (Lodi et al. 2005; Al-Hashimi corticosteroid-induced rosacea, pyoderma
et al. 2007; Al Johani et al. 2009). Initial studies gangrenosum, alopecia areata, mucocutaneous
Oral Lichen Planus 29

LP, and GVHD (Al Johani et al. 2009). Topical randomized, double-blind controlled trial,
tacrolimus is available in different concentrations followed by a 6-month follow-up period in
(0.03%, 0.1%). 30 unresponsive OLP patients. Both agents were
Systemic tacrolimus is substantially less effective in inducing clinical improvement; how-
expensive and 10–100 times more potent than ever, pimecrolimus showed better stability in ther-
cyclosporine, even though relative potency of apeutic effectiveness with statistically significant
topical preparations has never been evaluated. higher number of patients (10 vs. 4) not requiring
Tacrolimus used topically can control symptoms any further treatment at the end of the follow-up
and significantly improve refractory erosive OLP period.
(Hodgson et al. 2003). Local irritation is the most The theoretical increased risk of developing
common adverse effect reported (Lodi et al. 2005; malignancy with use of either tacrolimus or
Al Johani et al. 2009). Other side effects include pimecrolimus has been raised with the FDA; in
transient taste disturbance and sore throat. the USA “Black Box” warning attached to these
Tacrolimus ointment 0.1% was shown to be well agents is based on theoretical increased risk of
tolerated and appeared to be effective in erosive malignancy (SCC and lymphoma) in patients
OLP that did not respond to topical steroids in a using these agents for cutaneous psoriasis. In a
small cohort of six erosive OLP (Morrison et al. recent case report of a patient with OLP, the top-
2002). Although topical tacrolimus is effective ical use of tacrolimus 0.1% was suggested to be
and well tolerated, some OLP patients have the cause of the development of an SCC of the
noted flare-ups soon after stopping the treatment. tongue (Becker et al. 2006). Therefore, some
The treatment of chronic erosive oral LP with low authors believe that the use of these agents should
concentrations of tacrolimus was found to yield a be restricted and patients should be made aware of
rapid and important palliative effect in an open- these concerns (Al-Hashimi et al. 2007). On the
label prospective study that included eight erosive other hand, other researchers find that although
OLP with 6 months of treatment; however, all systemic tacrolimus may increase the risk of
patients relapsed after 12-month follow-up (Oliv- malignancy, there is no strong evidence that top-
ier et al. 2002). Currently, there remains little ical application of tacrolimus is associated with
evidence demonstrating that tacrolimus is notably such an increased risk (Al Johani et al. 2009). In
superior to topical corticosteroids for the treat- support of this view, a recent case-control study
ment of OLP (Al Johani et al. 2009). that involved 294 patients did not find any
Pimecrolimus is derived from the macrolide increased risk of lymphoma in patients with AD
ascomycin and shares the same cellular binding treated with topical calcineurin inhibitors
protein (FK506-binding protein-12) as tacrolimus (Arellano et al. 2007).
and blocks the transcription of cytokines by Overall, there remains little information of the
inhibiting the calcineurin pathway. Topical carcinogenic potential of tacrolimus or
pimecrolimus is a cream developed specifically pimecrolimus, and the new recommendations from
for the treatment of AD and approved for the the European Medicines Agency state that the ben-
treatment of patients with mild to moderate AD efits of these calcineurin inhibitors outweigh the
disease in patients older than 2 years. There is risks. The European Medicines Agency, however,
currently limited data on the potential use of topical recommends intermittent use of topical tacrolimus
pimecrolimus for the treatment of oral mucosal with the lowest strength possible and only for short
disease, with few reports suggesting it to be effec- periods of time (Al Johani et al. 2009).
tive in the management of symptoms and erosions/
ulcerations of OLP (Dissemond et al. 2004). Retinoids
In a recent study, Arduino et al. (2014) found Systemic and topical forms of retinoids have been
no difference between pimecrolimus 1% cream used in the treatment of OLP (Lodi et al. 2005;
and tacrolimus 0.1% ointment in managing recal- Al-Hashimi et al. 2007). Topical tretinoin or isotret-
citrant atrophic-erosive OLP in an 8-week inoin has been used to treat OLP, particularly
30 M.J. McCullough et al.

atrophic-erosive forms, with considerable improve- (n = 23) after a mean follow-up period of
ment (Scardina et al. 2006), but retinoids often cause 36 months (Carbone et al. 2003). Complete remis-
adverse effects and are less effective than topical sion was achieved in 68% of the prednisone group
corticosteroids (Scully and Carrozzo 2008). and 705 of the topical clobetasol group
(P = 0.94).
Topical Antifungal Therapy in OLP Systemic corticosteroids are, therefore, usually
The role Candida plays in the symptomatology of reserved for cases where topical approaches have
OLP is currently unknown. Based on the current failed, where there is recalcitrant, erosive, or ery-
evidence, a question therefore arises as to whether thematous OLP, or for widespread OLP when the
antifungals are required to treat symptomatic OLP skin, genitals, esophagus, or scalp is also involved
along with corticosteroids, not only prevent sec- (Al-Hashimi et al. 2007; Scully and Carrozzo
ondary candidosis and the symptoms associated 2008). Prednisolone 40–80 mg daily is usually
with this but also as a prophylactic treatment to sufficient to achieve a response: its toxicity
reduce Candida spp. overgrowth and the associ- requires that it should be used only when neces-
ated factors which may encourage a carcinogenic sary, at the lowest dose, and for the shortest time
event. A randomized double-blinded clinical trial possible with recommendation of (5–7 days) and
undertaken by Lodi et al. (2007) with 35 patients then withdrawn abruptly, or the dose should be
reviewed the efficacy of topically applied reduced by 5–10 mg/day gradually over
clobetasol propionate with and without topical 2–4 weeks (Scully and Carrozzo 2008). Adverse
miconazole in patients with symptomatic OLP. effects are possible even with short courses but
Results showed significant improvement in both may be minimized if patients can tolerate the same
the test ( p = 0.0020) and control ( p < 0.001) total dose on alternate days.
groups, and it was concluded while miconazole Systemic calcineurin inhibitors are associated
prevented secondary candidosis, which occurred with significant adverse effects including hyper-
in 30% of control subjects; however adjunctive tension, nephrotoxicity, and infections secondary
topical antifungal treatment did not affect the effi- to immunosuppressive status of the patients which
cacy of treatment or improve outcomes such as correlated with the dosage, blood levels, and dura-
pain or lesion extension (Lodi et al. 2007). Due to tion of therapy (Al Johani et al. 2009). Moreover,
the current lack of controlled studies and literature systemic tacrolimus can increase the risk of malig-
in this particular area, recent systemic review by nancy (e.g., oropharyngeal and skin cancers) by
Lodi et al. (2012) and Cochrane review by suppressing immune surveillance and inhibiting
Thongprasom et al. (2011) concluded that there DNA repair and apoptosis (Yarosh et al. 2005).
is currently insufficient evidence to determine Therefore, systemic calcineurin inhibitors are not
whether adjunctive antifungal therapy is effective recommended for OLP treatment.
in the treatment of oral lichen planus
(Thongprasom et al. 2011; Lodi et al. 2012).
Miscellaneous Agents

Systemic Drug Treatment Azathioprine


Azathioprine has been reportedly successful as a
Several studies have reported that systemic corti- “steroid-sparing agent” for cutaneous LP, and
costeroids are the most effective treatment for there is limited published evidence suggesting it
OLP; however, a comparative study that involved may have a similar role in recalcitrant OLP
a total of 49 OLP patients did not find differences (Silverman et al. 1991). In general, the results
in response between systemic prednisone (1 mg/ are no better than systemic steroids alone or sys-
kg/day) with topical clobetasol in an adhesive temic steroids in conjunction with topical steroids
base (n = 26) and topical clobetasol alone (Lodi et al. 2005b).
Oral Lichen Planus 31

Dapsone lesions after 2 months from the beginning of treat-


Dapsone has been used in the treatment of erosive ment (Mansourian et al. 2011).
OLP with some benefit (Beck and Brandrup According to a recent Cochrane review
1986). It should be considered in resistant cases, (Thongprasom et al. 2011), there is weak evidence
particularly when severe erosive lesions are pre- from two placebo-controlled RCTs, using differ-
sent. Significant adverse effects such as hemolysis ent formulations, that AV may be associated with
have been reported, and thus the use of dapsone in a reduction in pain in OLP (Choonhakarn et al.
the treatment OLP is generally not recommended 2008; Salazar-Sanchez et al. 2010). It should be
(Matthews et al. 1989). emphasized that the above trials used different AV
formulations and the amount of active drug sub-
Mycophenolate Mofetil stance in AV varies depending also on the age of
Mycophenolate has shown promise as an alterna- the plant, the growing and harvesting conditions,
tive to azathioprine as an immunomodulatory the parts of the plant, and the extraction methods
agent with a better safety profile in the manage- used. The great level of variability could have
ment of graft rejection in organ transplant recipi- affected the results of the published studies and
ents, and GVHD, and therefore may be a represents a challenge for future research
candidate for use in recalcitrant OLP (Eisen (Thongprasom et al. 2013).
2003). In a recent retrospective review of clinical
responses of ten patients with severe ulcerative LP Biologics
(vulvovaginal with gingival involvement, n = 8; The management of various immune-mediated
penogingival, n = 1; oral, n = 1) treated with disorders has changed dramatically by the advent
mycophenolate, Wee et al. (2012) showed remis- of biologic therapies. Biologics are designed by
sion in six patients, well-controlled disease in one, recombinant biotechnology to target particular
and partially controlled in the other three. The steps in the pathogenesis of immunoinflammatory
mean duration for mycophenolate treatment was diseases. Structurally, biologics include receptor
3.7 years, and the mean follow-up was 4.2 years. fusion proteins, monoclonal antibodies, and
Mycophenolate was well tolerated in all patients recombinant cytokines. Functionally, biologics
except in two who reported mild headaches and can be divided into T-cell or cytokine modulators
tiredness (Wee et al. 2012). (Zhang et al. 2011). Various immunobiologics
have been recently applied in the treatment of
Aloe vera psoriasis and rheumatic diseases as biologic
Aloe vera (AV) is a cactus-like plant that belongs immunomodulators may represent a more effec-
to the Liliaceae family. The reported pharmaco- tive and targeted approach than conventional
logical actions of AV include anti-inflammatory, treatment modalities (Shirota et al. 2008).
antibacterial, antiviral, and antifungal properties Various diseases including Behcet’s disease,
and hypoglycemic effects (Yagi et al. 2002). Top- recurrent aphthous stomatitis, benign mucous
ical Aloe vera is an emerging new modality in the membrane pemphigoid, and LP are considered
treatment of OLP that has been recently investi- potential candidates for the use of biologics
gated. In a randomized double-blind study, AV (O’Neill 2008).
mouthwash significantly improved the oral qual- After the pathogenesis of OLP has been thor-
ity of life for 32 OLP patients over a period of oughly investigated, several biologic agents,
3 months as compared to a placebo group especially those targeting T cells such as TNF-α
(Salazar-Sanchez et al. 2010). In another study, inhibitors, are suggested as a future alternative to
comparable effects of AV mouthwash were steroid therapy. However, possible reactions and
reported when compared to 0.1% triamcinolone side effects will always be a major concern (Zhang
acetonide paste in a group of OLP patients (n = 23 et al. 2011).
each) in terms of VAS of pain, burning sensation, TNF-α inhibitors currently available are
clinical presentation, and healing of the OLP etanercept, infliximab, and adalimumab, all of
32 M.J. McCullough et al.

which are FDA approved for psoriasis. The clin- corticosteroids with placebo in patients with symp-
ical feasibility has been demonstrated not only by tomatic OLP. From the 28 trials included in this
a successful treatment of LP with etanercept systematic review, the wide range of interventions
(Yarom 2007) but also by the efficacy of compared means there is insufficient evidence to
adalimumab for the treatment of cutaneous and support the superior effectiveness of any specific
OLP (Chao 2009). Moreover, there are several treatment (Lodi et al. 2012).
ongoing clinical trials to evaluate the safety and
effectiveness of etanercept in treating oral and
cutaneous LP. According to a recent review by Follow-up
O’Neill and Scully (2013), the best data, although
limited, are for the use of the T-cell modulator Patient follow-up ranging from every 2 months to
alefacept in OLP. Limitations on future studies annually is accepted as part of long-term care for
with such agents include the need for continuous patients with OLP largely to screen for changes
or intermittent use of anti-TNF-α therapy for long- that may indicate malignant transformation
term control of OLP and the fact that TNF-α (Mignogna et al. 2006). At a minimum, annual
blockers have the potential for initiating lichenoid monitoring is recommended (Al-Hashimi et al.
eruptions (Asarch et al. 2009).This possibly 2007; Parashar 2011) and favorably two to four
accounts for the rarity of reports of use of TNF reviews (Scully et al. 1998; Mattsson et al. 2002;
antagonists in LP to date and may be a barrier to van der Meij et al. 2007). More frequent exami-
further use. Additional concerns center around the nations are recommended for patients with OLL
potential for reactivation of viral infections with with dysplasia.
concern about those patients with LP who have If changes are noted in a lesion at follow-up
concomitant HCV infection (Lodi et al. 2010). visits, then an additional biopsy or biopsies should
The potential risk of malignancy in LP and those be performed and the follow-up intervals short-
reports of the development of oral cancer in ened (Ismail et al. 2007).
patients receiving TNF-α blockers may be an For those OLP patients who develop OSSC,
additional reason to use these agents with caution Mignogna et al. (2002) proposed the strict follow-
(O’Neill and Scully 2013). up of patients with oral and neck examinations
every 2 months during the 5- to 9-month period
Other Agents and Treatment Modalities after the diagnosis of oral carcinoma, when the
Other agents have been suggested for OLP treat- risk of metastasis or second primary tumor is
ment but with weaker evidence. These include maximum. The same authors subsequently
levamisole, amitriptyline, amlexanox, hyaluronic reported that a program of three follow-up exam-
acid, thalidomide, ignatia, curcuminoids, lyco- inations a year enables detection of malignant
pene, phototherapy, lasers, surgery, and cryosur- transformation in early or microinvasive
gery (Lodi et al. 2005; Al-Hashimi et al. 2007; intraepithelial states, which generally have a
Thongprasom et al. 2013). very good prognosis (Mignogna et al. 2006).
In conclusion, it appears that although a wide
range of treatment options is available for patients
with OLP, the level of clinical efficacy is incon- Conclusion
sistent, and individualized protocols are required
in cases with recalcitrant OLP. LP is a common chronic inflammatory disease of
Recently, Lodi et al. (2012) reviewed 28 ran- the skin and oral mucosa with an estimated prev-
domized controlled trials (RCTs) in the context of alence of 0.5–2%. Although the cause of OLP is
interventions for OLP. Although topical corticoste- unknown, it is thought to result from the complex
roids are considered to be the first-line treatment, interplay of host, lifestyle, and environmental fac-
no RCTs were identified that compared topical tors resulting in cell-mediated immune
Oral Lichen Planus 33

dysregulation. The oral appearance of OLP has References


been described as six clinical subtypes seen indi-
vidually or in combination (reticular, plaque-like, Abdelsayed RA, Sumner T, Allen CM, Treadway A, Ness
GM, Penza SL. Oral precancerous and malignant
atrophic, erosive, papular, and bullous), with the
lesions associated with graft-versus-host disease: report
most common being reticular occurring bilaterally of 2 cases. Oral Surg Oral Med Oral Pathol Oral Radiol
in the posterior buccal mucosa. Endod. 2002;93(1):75–80.
Symptoms and signs can range from patients Aderem A. Phagocytosis and the inflammatory response.
J Infect Dis. 2003;187(Suppl 2):S340–5.
being unaware of the disease, with lesions that are
Al Johani KA, Hegarty AM, Porter SR, Fedele
completely asymptomatic as in reticular OLP, to S. Calcineurin inhibitors in oral medicine. J Am Acad
those experiencing mucosal sensitivity and burn- Dermatol. 2009;61(5):829–40.
ing and debilitating pain. Treatment generally Al-Hashimi I, Schifter M, Lockhart PB, Wray D,
Brennan M, Migliorati CA, Axell T, Bruce AJ,
consists of the topical use of anti-inflammatory
Carpenter W, Eisenberg E, Epstein JB, Holmstrup P,
agents for symptomatic relief. Malignant transfor- Jontell M, Lozada-Nur F, Nair R, Silverman B,
mation of OLP remains a controversy and vari- Thongprasom K, Thornhill M, Warnakulasuriya S,
ably reported between 0% and 5%. Obviously van der Waal I. Oral lichen planus and oral lichenoid
lesions: diagnostic and therapeutic considerations. Oral
there is much that remains unknown regarding
Surg Oral Med Oral Pathol Oral Radiol Endod.
oral mucosal lichen planus, such as the exact 2007;103(Suppl):S25 e21–12.
cause, pathogenic mechanism involved, and pro- Alnuaimi AD, Wiesenfeld D, O’Brien-Simpson NM,
cess by which malignant transformation occurs. Reynolds EC, McCullough MJ. Oral Candida coloni-
zation in oral cancer patients and its relationship with
Further directed research is required so that we
traditional risk factors of oral cancer: a matched case-
may be able to best advise and treat our patients. control study. Oral Oncol. 2015;51(2):139–45.
Alrashdan MS, Angel C, Cirillo N, McCullough
M. Smoking habits and clinical patterns can alter the
inflammatory infiltrate in oral lichenoid lesions. Oral
Cross-References Surg Oral Med Oral Pathol Oral Radiol. 2016;121
(1):49–57.
▶ Clinical Evaluation of Oral Diseases Arduino PG, Carbone M, Della Ferrera F, Elia A,
▶ Clinical Immunology in Diagnoses of Maxillo- Conrotto D, Gambino A, Comba A, Calogiuri PL,
Broccoletti R. Pimecrolimus vs. tacrolimus for the top-
facial Disease
ical treatment of unresponsive oral erosive lichen
▶ Cutaneous Pathology of the Head and Neck planus: a 8 week randomized double-blind controlled
Region study. J Eur Acad Dermatol Venereol. 2014;28
▶ Gingival Pathology (4):475–82.
Arellano FM, Wentworth CE, Arana A, Fernandez C, Paul
▶ Interface Between Oral and Systemic Disease
CF. Risk of lymphoma following exposure to
▶ Laboratory Medicine and Diagnostic Pathology calcineurin inhibitors and topical steroids in patients
▶ Normal Variation in the Anatomy, Biology and with atopic dermatitis. J Invest Dermatol. 2007;127
Histology of the Maxillofacial Region (4):808–16.
Asarch A, Gottlieb AB, Lee J, Masterpol KS, Scheinman
▶ Oral and Maxillofacial Fungal Infections
PL, Stadecker MJ, Massarotti EM, Bush ML. Lichen
▶ Oral Manifestations of Systemic Diseases and planus-like eruptions: an emerging side effect of tumor
their Treatments necrosis factor-alpha antagonists. J Am Acad
▶ Oral Mucosal Malignancies Dermatol. 2009;61(1):104–11.
Axell T, Rundquist L. Oral lichen planus – a demographic
▶ Oral Ulcerative Lesions
study. Community Dent Oral Epidemiol. 1987;15
▶ Oral Vesicular and Bullous Lesions (1):52–6.
▶ Orofacial Pain in Patients with Cancer and Azher S, Azami O, Amato C, McCullough M,
Mucosal Diseases Celentano A, Cirillo N. The non-conventional effects
of glucocorticoids in cancer. J Cell Physiol.
▶ Pharmaco-Therapeutic Approaches in Oral
2016;231:2368–73.
Medicine Baccaglini L, Thongprasom K, Carrozzo M, Bigby
▶ White and Red Lesions of the Oral Mucosa M. Urban legends series: lichen planus. Oral Dis.
2013;19(2):128–43.
34 M.J. McCullough et al.

Bagan JV, Donat JS, Penarrocha M, Milian MA, Sanchis Carbone M, Goss E, Carrozzo M, Castellano S,
JM. Oral lichen planus and diabetes mellitus. a clinico- Conrotto D, Broccoletti R, Gandolfo S. Systemic and
pathological study. Bull Group Int Rech Sci Stomatol topical corticosteroid treatment of oral lichen planus: a
Odontol. 1993;36(1–2):3–6. comparative study with long-term follow-up. J Oral
Bagan JV, Thongprasom K, Scully C. Adverse oral reac- Pathol Med. 2003;32(6):323–9.
tions associated with the COX-2 inhibitor rofecoxib. Carrozzo M, Gandolfo S, Carbone M, Colombatto P,
Oral Dis. 2004;10(6):401–3. Broccoletti R, Garzino-Demo P, Ghisetti V. Hepatitis
Bain L, Geronemus R. The association of lichen planus of C virus infection in Italian patients with oral lichen
the penis with squamous cell carcinoma in situ and with planus: a prospective case-control study. J Oral Pathol
verrucous squamous carcinoma. J Dermatol Surg Med. 1996;25(10):527–33.
Oncol. 1989;15(4):413–7. Carrozzo M, Francia Di Celle P, Gandolfo S, Carbone M,
Balkwill F, Mantovani A. Inflammation and cancer: back Conrotto D, Fasano ME, Roggero S, Rendine S,
to Virchow? Lancet. 2001;357(9255):539–45. Ghisetti V. Increased frequency of HLA-DR6 allele in
Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Italian patients with hepatitis C virus-associated oral
Liu YJ, Pulendran B, Palucka K. Immunobiology of lichen planus. Br J Dermatol. 2001;144(4):803–8.
dendritic cells. Annu Rev Immunol. 2000;18:767–811. Carrozzo M, Uboldi de Capei M, Dametto E, Fasano ME,
Barnard NA, Scully C, Eveson JW, Cunningham S, Porter Arduino P, Broccoletti R, Vezza D, Rendine S, Curtoni
SR. Oral cancer development in patients with oral ES, Gandolfo S. Tumor necrosis factor-alpha and
lichen planus. J Oral Pathol Med. 1993;22(9):421–4. interferon-gamma polymorphisms contribute to sus-
Barrett AW, Raja AM. The immunohistochemical identifi- ceptibility to oral lichen planus. J Invest Dermatol.
cation of human oral mucosal melanocytes. Arch Oral 2004;122(1):87–94.
Biol. 1997;42(1):77–81. Carrozzo M, Elia A, Mereu V, Dametto E, Fasano M,
Beaird LM, Kahloon N, Franco J, Fairley JA. Incidence of Broccoletti R, Rendine S, Amoroso A. HLA-C/KIR
hepatitis C in lichen planus. J Am Acad Dermatol. genotypes in oral lichen planus patients infected or
2001;44(2):311–2. non-infected with hepatitis C virus. Oral Dis. 2011;17
Beck HI, Brandrup F. Treatment of erosive lichen planus (3):309–13.
with dapsone. Acta Derm Venereol. 1986;66(4):366–7. Chainani-Wu N, Silverman Jr SOL, Lozada-Nur F, May-
Becker JC, Houben R, Vetter CS, Brocker EB. The carci- er P, Watson JJ. Oral lichen planus: patient profile,
nogenic potential of tacrolimus ointment beyond disease progression and treatment responses. J Am
immune suppression: a hypothesis creating case report. Dent Assoc. 2001;132(7):901–9.
BMC Cancer. 2006;6:7. Chainani-Wu N, Madden E, Lozada-Nur F, Silverman Jr
Bermejo-Fenoll A, Lopez-Jornet P. Familial oral lichen S. High-dose curcuminoids are efficacious in the reduc-
planus: presentation of six families. Oral Surg Oral tion in symptoms and signs of oral lichen planus. J Am
Med Oral Pathol Oral Radiol Endod. 2006;102(2): Acad Dermatol. 2012;66(5):752–60.
e12–5. Chao TJ. Adalimumab in the management of cutaneous
Bode W. Structural basis of matrix metalloproteinase func- and oral lichen planus. Cutis. 2009;84(6):325–8.
tion. Biochem Soc Symp. 2003;70:1–14. Choonhakarn C, Busaracome P, Sripanidkulchai B,
Bornstein MM, Kalas L, Lemp S, Altermatt HJ, Rees TD, Sarakarn P. The efficacy of Aloe vera gel in the treat-
Buser D. Oral lichen planus and malignant transforma- ment of oral lichen planus: a randomized controlled
tion: a retrospective follow-up study of clinical and trial. Br J Dermatol. 2008;158(3):573–7.
histopathologic data. Quintessence Int. 2006;37 Cirillo N, Hassona Y, Pignatelli M, Gasparoto TH, Morgan
(4):261–71. DJ, Prime SS. Characterization of a novel oral gluco-
Brandtzaeg P. Do salivary antibodies reliably reflect both corticoid system and its possible role in disease. J Dent
mucosal and systemic immunity? Ann N Y Acad Sci. Res. 2012;91(1):97–103.
2007;1098:288–311. Clevers H. At the crossroads of inflammation and cancer.
Brown GR, Lindberg G, Meddings J, Silva M, Beutler B, Cell. 2004;118(6):671–4.
Thiele D. Tumor necrosis factor inhibitor ameliorates Curtis RE, Rowlings PA, Deeg HJ, Shriner DA, Socie G,
murine intestinal graft-versus-host disease. Gastroen- Travis LB, Horowitz MM, Witherspoon RP, Hoover
terology. 1999;116(3):593–601. RN, Sobocinski KA, Fraumeni Jr JF, Boice Jr
Burkhart NW, Burkes EJ, Burker EJ. Meeting the educa- JD. Solid cancers after bone marrow transplantation.
tional needs of patients with oral lichen planus. Gen N Engl J Med. 1997;336(13):897–904.
Dent. 1997;45(2):126–32; quiz 143–24. Daniels TE. Human mucosal Langerhans cells: postmor-
Carbone M, Conrotto D, Carrozzo M, Broccoletti R, tem identification of regional variations in oral mucosa.
Gandolfo S, Scully C. Topical corticosteroids in asso- J Invest Dermatol. 1984;82(1):21–4.
ciation with miconazole and chlorhexidine in the long- Daramola OO, George AO, Ogunbiyi AO. Hepatitis C
term management of atrophic-erosive oral lichen virus and lichen planus in Nigerians: any relationship?
planus: a placebo-controlled and comparative study Int J Dermatol. 2002;41(4):217–9.
between clobetasol and fluocinonide. Oral Dis. 1999;5 Deeg HJ, Socie G, Schoch G, Henry-Amar M,
(1):44–9. Witherspoon RP, Devergie A, Sullivan KM,
Oral Lichen Planus 35

Gluckman E, Storb R. Malignancies after marrow Eversole LR. Immunopathogenesis of oral lichen planus
transplantation for aplastic anemia and fanconi anemia: and recurrent aphthous stomatitis. Semin Cutan Med
a joint Seattle and Paris analysis of results in Surg. 1997;16(4):284–94.
700 patients. Blood. 1996;87(1):386–92. Fang KC, Raymond WW, Blount JL, Caughey GH. Dog
Delves PJ, Roitt IM. The immune system. First of two mast cell alpha-chymase activates progelatinase B by
parts. N Engl J Med. 2000;343(1):37–49. cleaving the Phe88-Gln89 and Phe91-Glu92 bonds of
Dissemond J, Schroter S, Franckson T, Herbig S, Goos the catalytic domain. J Biol Chem. 1997;272
M. Pimecrolimus in an adhesive ointment as a new (41):25628–35.
treatment option for oral lichen planus. Br J Dermatol. Farhi D, Dupin N. Pathophysiology, etiologic factors, and
2004;150(4):782–4. clinical management of oral lichen planus, part I: facts
Duffey DC, Eversole LR, Abemayor E. Oral lichen planus and controversies. Clin Dermatol. 2010;28(1):100–8.
and its association with squamous cell carcinoma: an Fatahzadeh M, Rinaggio J, Chiodo T. Squamous cell car-
update on pathogenesis and treatment implications. cinoma arising in an oral lichenoid lesion. J Am Dent
Laryngoscope. 1996;106(3 Pt 1):357–62. Assoc. 2004;135(6):754–9; quiz 796.
Dupin N, Chosidow O, Lunel F, Fretz C, Szpirglas H, Feller L, Altini M, Khammissa RA, Chandran R,
Frances C. Oral lichen planus and hepatitis C virus Bouckaert M, Lemmer J. Oral mucosal immunity.
infection: a fortuitous association? Arch Dermatol. Oral Surg Oral Med Oral Pathol Oral Radiol.
1997;133(8):1052–3. 2013;116(5):576–83.
Dwyer CM, Kerr RE, Millan DW. Squamous carcinoma Gale N, Pilch BZ, Sidransky D, Westra WH, Califano
following lichen planus of the vulva. Clin Exp J. Epithelial precursor lesions. In: Barnes L, Evenson
Dermatol. 1995;20(2):171–2. JW, Reichart P, Sidransky D, editors. Pathology &
Eisen D. The evaluation of cutaneous, genital, scalp, nail, genetics. Head and neck tumours. Lyon: IARC; 2005.
esophageal, and ocular involvement in patients with p. 177–81.
oral lichen planus. Oral Surg Oral Med Oral Pathol Gandolfo S, Richiardi L, Carrozzo M, Broccoletti R,
Oral Radiol Endod. 1999;88(4):431–6. Carbone M, Pagano M, Vestita C, Rosso S, Merletti
Eisen D. The clinical features, malignant potential, and F. Risk of oral squamous cell carcinoma in 402 patients
systemic associations of oral lichen planus: a study of with oral lichen planus: a follow-up study in an Italian
723 patients. J Am Acad Dermatol. 2002;46 population. Oral Oncol. 2004;40(1):77–83.
(2):207–14. Godessart N, Kunkel SL. Chemokines in autoimmune
Eisen D. The clinical manifestations and treatment of oral disease. Curr Opin Immunol. 2001;13(6):670–5.
lichen planus. Dermatol Clin. 2003;21(1):79–89. Gonzalez-Moles MA, Bascones-Ilundain C, Gil Montoya
Eisen D, Ellis CN, Duell EA, Griffiths CE, Voorhees JA, Ruiz-Avila I, Delgado-Rodriguez M, Bascones-
JJ. Effect of topical cyclosporine rinse on oral lichen Martinez A. Cell cycle regulating mechanisms in oral
planus. A double-blind analysis. N Engl J Med. lichen planus: molecular bases in epithelium pre-
1990a;323(5):290–4. disposed to malignant transformation. Arch Oral Biol.
Eisen D, Griffiths CE, Ellis CN, Nickoloff BJ, Voorhees 2006;51(12):1093–103.
JJ. Cyclosporin wash for oral lichen planus. Lancet. Gonzalez-Moles MA, Scully C, Gil-Montoya JA. Oral
1990b;335(8688):535–6. lichen planus: controversies surrounding malignant
Eisen D, Carrozzo M, Bagan Sebastian JV, Thongprasom transformation. Oral Dis. 2008;14(3):229–43.
K. Number V Oral lichen planus: clinical features and Grinspan D, Diaz J, Abulafia J, Villapol L,
management. Oral Dis. 2005;11(6):338–49. Schneiderman J, Palese D, Berdichesky R. Our experi-
Eisenberg E. Oral lichen planus: a benign lesion. J Oral ence with lichen ruber planus of the buccal mucosa.
Maxillofac Surg. 2000;58(11):1278–85. Ann Dermatol Syphiligr (Paris). 1966;93(5):531–42.
Epstein JB, Wan LS, Gorsky M, Zhang L. Oral lichen Gueiros LA, Gondak R, Jorge Junior J, Coletta RD,
planus: progress in understanding its malignant poten- Carvalho Ade A, Leao JC, de Almeida OP, Vargas
tial and the implications for clinical management. Oral PA. Increased number of Langerhans cells in oral lichen
Surg Oral Med Oral Pathol Oral Radiol Endod. 2003;96 planus and oral lichenoid lesions. Oral Surg Oral Med
(1):32–7. Oral Pathol Oral Radiol. 2012;113(5):661–6.
Erkek E, Bozdogan O, Olut AI. Hepatitis C virus infection Hengge UR, Ruzicka T, Schwartz RA, Cork MJ. Adverse
prevalence in lichen planus: examination of lesional effects of topical glucocorticosteroids. J Am Acad
and normal skin of hepatitis C virus-infected patients Dermatol. 2006;54(1):1–15; quiz 16–18.
with lichen planus for the presence of hepatitis C virus Henriksson E, Mattsson U, Hakansson J. Healing of
RNA. Clin Exp Dermatol. 2001;26(6):540–4. lichenoid reactions following removal of amalgam. A
Escudier M, Ahmed N, Shirlaw P, Setterfield J, Tappuni A, clinical follow-up. J Clin Periodontol. 1995;22
Black MM, Challacombe SJ. A scoring system for (4):287–94.
mucosal disease severity with special reference to oral Herve P, Flesch M, Tiberghien P, Wijdenes J, Racadot E,
lichen planus. Br J Dermatol. 2007;157(4):765–70. Bordigoni P, Plouvier E, Stephan JL, Bourdeau H,
Evans AV, Fletcher CL, Owen WJ, Hay RJ. Oesophageal Holler E, et al. Phase I-II trial of a monoclonal anti-
lichen planus. Clin Exp Dermatol. 2000;25(1):36–7. tumor necrosis factor alpha antibody for the treatment
36 M.J. McCullough et al.

of refractory severe acute graft-versus-host disease. undergoing topical steroid therapy. Oral Surg Oral
Blood. 1992;79(12):3362–8. Med Oral Pathol Oral Radiol Endod. 2007;104
Hietanen J, Paasonen MR, Kuhlefelt M, Malmstrom M. A (1):61–6.
retrospective study of oral lichen planus patients with Kanwar AJ, De D. Lichen planus in childhood: report of
concurrent or subsequent development of malignancy. 100 cases. Clin Exp Dermatol. 2010;35(3):257–62.
Oral Oncol. 1999;35(3):278–82. Khan A, Farah CS, Savage NW, Walsh LJ, Harbrow DJ,
Hodgson TA, Sahni N, Kaliakatsou F, Buchanan JA, Porter Sugerman PB. Th1 cytokines in oral lichen planus.
SR. Long-term efficacy and safety of topical tacrolimus J Oral Pathol Med. 2003;32(2):77–83.
in the management of ulcerative/erosive oral lichen Kiefer F, Siekmann AF. The role of chemokines and their
planus. Eur J Dermatol. 2003;13(5):466–70. receptors in angiogenesis. Cell Mol Life Sci. 2011;68
Holmstrup P, Dabelsteen E. Changes in carbohydrate (17):2811–30.
expression of lichen planus affected oral epithelial cell Kilpi A. Characterization of mononuclear cells of inflam-
membranes. J Invest Dermatol. 1979;73(5):364–7. matory infiltrates in oral tissues. A histochemical and
Holmstrup P, Thorn JJ, Rindum J, Pindborg JJ. Malignant immunohistochemical study of labial salivary glands in
development of lichen planus-affected oral mucosa. J Sjogren’s syndrome and of oral lesions in systemic
Oral Pathol. 1988;17(5):219–25. lupus erythematosus and in lichen planus. Proc Finn
Hallopeau H. Sur um cãs de lichen de Wilson gingival avec Dent Soc. 1988;84(Suppl 3):1–93.
neoplasicvoisinedans la region maxillaire. Bull Soc Fr Koch P, Bahmer FA. Oral lichenoid lesions, mercury
DermatolSyphiligr. 1910;17: 33. hypersensitivity and combined hypersensitivity to mer-
Ibrahim HA, Baddour MM, Morsi MG, Abdelkader cury and other metals: histologically-proven reproduc-
AA. Should we routinely check for hepatitis B and C tion of the reaction by patch testing with metal salts.
in patients with lichen planus or cutaneous vasculitis? Contact Dermatitis. 1995;33(5):323–8.
East Mediterr Health J. 1999;5(1):71–8. Koray M, Dulger O, Ak G, Horasanli S, Ucok A,
Ichimura M, Hiratsuka K, Ogura N, Utsunomiya T, Tanyeri H, Badur S. The evaluation of anxiety and
Sakamaki H, Kondoh T, Abiko Y, Otake S, Yamamoto salivary cortisol levels in patients with oral lichen
M. Expression profile of chemokines and chemokine planus. Oral Dis. 2003;9(6):298–301.
receptors in epithelial cell layers of oral lichen planus. J Kramer IR, Lucas RB, Pindborg JJ, Sobin LH. Definition
Oral Pathol Med. 2006;35(3):167–74. of leukoplakia and related lesions: an aid to studies on
Iijima W, Ohtani H, Nakayama T, Sugawara Y, Sato E, oral precancer. Oral Surg Oral Med Oral Pathol.
Nagura H, Yoshie O, Sasano T. Infiltrating CD8+ T 1978;46(4):518–39.
cells in oral lichen planus predominantly express CCR5 Kruse AL, Gratz KW. Oral carcinoma after hematopoietic
and CXCR3 and carry respective chemokine ligands stem cell transplantation – a new classification based on
RANTES/CCL5 and IP-10/CXCL10 in their cytolytic a literature review over 30 years. Head Neck Oncol.
granules: a potential self-recruiting mechanism. Am J 2009;1:29.
Pathol. 2003;163(1):261–8. Krutchkoff DJ, Eisenberg E. Lichenoid dysplasia: a dis-
Imanguli MM, Alevizos I, Brown R, Pavletic SZ, Atkinson tinct histopathologic entity. Oral Surg Oral Med Oral
JC. Oral graft-versus-host disease. Oral Dis. 2008;14 Pathol. 1985;60(3):308–15.
(5):396–412. Krutchkoff DJ, Cutler L, Laskowski S. Oral lichen planus:
Ingafou M, Porter SR, Scully C, Teo CG. No evidence of the evidence regarding potential malignant transforma-
HCV infection or liver disease in British patients with tion. J Oral Pathol. 1978;7(1):1–7.
oral lichen planus. Int J Oral Maxillofac Surg. 1998;27 Laeijendecker R, Tank B, Dekker SK, Neumann HA. A
(1):65–6. comparison of treatment of oral lichen planus with
Ishii T. Immunohistochemical demonstration of T cell sub- topical tacrolimus and triamcinolone acetonide oint-
sets and accessory cells in oral lichen planus. J Oral ment. Acta Derm Venereol. 2006;86(3):227–9.
Pathol. 1987;16(7):356–61. Laine J, Kalimo K, Forssell H, Happonen RP. Resolution
Ismail SB, Kumar SK, Zain RB. Oral lichen planus and of oral lichenoid lesions after replacement of amalgam
lichenoid reactions: etiopathogenesis, diagnosis, man- restorations in patients allergic to mercury compounds.
agement and malignant transformation. J Oral Sci. Br J Dermatol. 1992;126(1):10–5.
2007;49(2):89–106. Laine J, Konttinen YT, Beliaev N, Happonen
Issa Y, Brunton PA, Glenny AM, Duxbury AJ. Healing of RP. Immunocompetent cells in amalgam-associated
oral lichenoid lesions after replacing amalgam restora- oral lichenoid contact lesions. J Oral Pathol Med.
tions: a systematic review. Oral Surg Oral Med Oral 1999;28(3):117–21.
Pathol Oral Radiol Endod. 2004;98(5):553–65. Larsson A, Warfvinge G. Oral lichenoid contact reactions
Ivanovski K, Nakova M, Warburton G, Pesevska S, may occasionally transform into malignancy. Eur J
Filipovska A, Nares S, Nunn ME, Angelova D, Cancer Prev. 2005;14(6):525–9.
Angelov N. Psychological profile in oral lichen planus. Leao JC, Ingafou M, Khan A, Scully C, Porter
J Clin Periodontol. 2005;32(10):1034–40. S. Desquamative gingivitis: retrospective analysis of
Jainkittivong A, Kuvatanasuchati J, Pipattanagovit P, disease associations of a large cohort. Oral Dis.
Sinheng W. Candida in oral lichen planus patients 2008;14(6):556–60.
Oral Lichen Planus 37

Levin C, Maibach HI. Topical corticosteroid-induced adre- oral lichen planus: a randomized comparative study. J
nocortical insufficiency: clinical implications. Am J Am Acad Dermatol. 2008;58(4):596–602.
Clin Dermatol. 2002;3(3):141–7. Mansourian A, Momen-Heravi F, Saheb-Jamee M,
Lind PO. Oral lichenoid reactions related to composite Esfehani M, Khalilzadeh O, Momen-Beitollahi
restorations. Preliminary report. Acta Odontol Scand. J. Comparison of Aloe vera mouthwash with triamcin-
1988;46(1):63–5. olone acetonide 0.1% on oral lichen planus: a random-
Lind PO, Hurlen B, Lyberg T, Aas E. Amalgam-related ized double-blinded clinical trial. Am J Med Sci.
oral lichenoid reaction. Scand J Dent Res. 1986;94 2011;342(6):447–51.
(5):448–51. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A,
Lo Muzio L, Mignogna MD, Favia G, Procaccini M, Testa Locati M. The chemokine system in diverse forms of
NF, Bucci E. The possible association between oral macrophage activation and polarization. Trends
lichen planus and oral squamous cell carcinoma: a Immunol. 2004;25(12):677–86.
clinical evaluation on 14 cases and a review of the Marinkovich MP, Keene DR, Rimberg CS, Burgeson
literature. Oral Oncol. 1998;34(4):239–46. RE. Cellular origin of the dermal-epidermal basement
Lo Muzio L, della Valle A, Mignogna MD, Pannone G, membrane. Dev Dyn. 1993;197(4):255–67.
Bucci P, Bucci E, Sciubba J. The treatment of oral Markopoulos AK, Antoniades D, Papanayotou P, Tri-
aphthous ulceration or erosive lichen planus with top- gonidis G. Malignant potential of oral lichen planus; a
ical clobetasol propionate in three preparations: a clin- follow-up study of 326 patients. Oral Oncol. 1997;33
ical and pilot study on 54 patients. J Oral Pathol Med. (4):263–9.
2001;30(10):611–7. Masaki M, Sato T, Sugawara Y, Sasano T, Takahashi
Lo Russo L, Fierro G, Guiglia R, Compilato D, Testa NF, N. Detection and identification of non-Candida
Lo Muzio L, Campisi G. Epidemiology of albicans species in human oral lichen planus. Micro-
desquamative gingivitis: evaluation of 125 patients biol Immunol. 2011;55(1):66–70.
and review of the literature. Int J Dermatol. 2009;48 Matthews RW, Pinkney RC, Scully C. The management of
(10):1049–52. intransigent desquamative gingivitis with Dapsone.
Lodi G, Giuliani M, Majorana A, Sardella A, Bez C, Ann Dent. 1989;48(1):41–3.
Demarosi F, Carrassi A. Lichen planus and hepatitis C Mattsson U, Jontell M, Holmstrup P. Oral lichen planus
virus: a multicentre study of patients with oral lesions and malignant transformation: is a recall of patients
and a systematic review. Br J Dermatol. 2004;151 justified? Crit Rev Oral Biol Med. 2002;13(5):390–6.
(6):1172–81. McCullough M, Jaber M, Barrett AW, Bain L, Speight PM,
Lodi G, Scully C, Carrozzo M, Griffiths M, Sugerman PB, Porter SR. Oral yeast carriage correlates with presence
Thongprasom K. Current controversies in oral lichen of oral epithelial dysplasia. Oral Oncol. 2002;38
planus: report of an international consensus meeting. (4):391–3.
Part 1. Viral infections and etiopathogenesis. Oral Surg Mergoni G, Ergun S, Vescovi P, Mete O, Tanyeri H, Meleti
Oral Med Oral Pathol Oral Radiol Endod. 2005a;100 M. Oral postinflammatory pigmentation: an analysis of
(1):40–51. 7 cases. Med Oral Patol Oral Cir Bucal. 2011;16(1):
Lodi G, Scully C, Carrozzo M, Griffiths M, Sugerman PB, e11–4.
Thongprasom K. Current controversies in oral lichen Merry R, Belfield L, McArdle P, McLennan A, Crean S,
planus: report of an international consensus meeting. Foey A. Oral health and pathology: a macrophage
Part 2. Clinical management and malignant transforma- account. Br J Oral Maxillofac Surg. 2012;50(1):2–7.
tion. Oral Surg Oral Med Oral Pathol Oral Radiol Mignogna MD, Lo Muzio L, Lo Russo L, Fedele S,
Endod. 2005b;100(2):164–78. Ruoppo E, Bucci E. Clinical guidelines in early detec-
Lodi G, Tarozzi M, Sardella A, Demarosi F, Canegallo L, tion of oral squamous cell carcinoma arising in oral
Di Benedetto D, Carrassi A. Miconazole as adjuvant lichen planus: a 5-year experience. Oral Oncol.
therapy for oral lichen planus: a double-blind random- 2001;37(3):262–7.
ized controlled trial. Br J Dermatol. 2007;156 Mignogna MD, Lo Russo L, Fedele S, Ruoppo E,
(6):1336–41. Califano L, Lo Muzio L. Clinical behaviour of malig-
Lodi G, Pellicano R, Carrozzo M. Hepatitis C virus infec- nant transforming oral lichen planus. Eur J Surg Oncol.
tion and lichen planus: a systematic review with meta- 2002;28(8):838–43.
analysis. Oral Dis. 2010;16(7):601–12. Mignogna MD, Fedele S, Lo Russo L, Lo Muzio L, Bucci
Lodi G, Carrozzo M, Furness S, Thongprasom E. Immune activation and chronic inflammation as the
K. Interventions for treating oral lichen planus: a sys- cause of malignancy in oral lichen planus: is there any
tematic review. Br J Dermatol. 2012;166(5):938–47. evidence ? Oral Oncol. 2004;40(2):120–30.
Lozada-Nur F. Oral lichen planus and oral cancer: is there Mignogna MD, Lo Russo L, Fedele S. Gingival involve-
enough epidemiologic evidence? Oral Surg Oral Med ment of oral lichen planus in a series of 700 patients.
Oral Pathol Oral Radiol Endod. 2000;89(3):265–6. J Clin Periodontol. 2005;32(10):1029–33.
Malhotra AK, Khaitan BK, Sethuraman G, Sharma Mignogna MD, Fedele S, Lo Russo L. Dysplasia/neoplasia
VK. Betamethasone oral mini-pulse therapy compared surveillance in oral lichen planus patients: a description
with topical triamcinolone acetonide (0.1%) paste in
38 M.J. McCullough et al.

of clinical criteria adopted at a single centre and their Oral Surg Oral Med Oral Pathol Oral Radiol Endod.
impact on prognosis. Oral Oncol. 2006;42(8):819–24. 1997;83(3):358–66.
Mignogna MD, Fedele S, Lo Russo L, Mignogna C, de Pullan S, Wilson J, Metcalfe A, Edwards GM,
Rosa G, Porter SR. Field cancerization in oral lichen Goberdhan N, Tilly J, Hickman JA, Dive C, Streuli
planus. Eur J Surg Oncol. 2007;33(3):383–9. CH. Requirement of basement membrane for the sup-
Mokni M, Rybojad M, Puppin Jr D, Catala S, Venezia F, pression of programmed cell death in mammary epi-
Djian R, Morel P. Lichen planus and hepatitis C virus. J thelium. J Cell Sci. 1996;109(Pt 3):631–42.
Am Acad Dermatol. 1991;24(5 Pt 1):792. Rajentheran R, McLean NR, Kelly CG, Reed MF, Nolan
Morrison L, Kratochvil 3rd FJ, Gorman A. An open trial of A. Malignant transformation of oral lichen planus. Eur
topical tacrolimus for erosive oral lichen planus. J Am J Surg Oncol. 1999;25(5):520–3.
Acad Dermatol. 2002;47(4):617–20. Regezi JA, Dekker NP, MacPhail LA, Lozada-Nur F,
Munoz AA, Haddad RI, Woo SB, Bhattacharyya McCalmont TH. Vascular adhesion molecules in oral
N. Behavior of oral squamous cell carcinoma in sub- lichen planus. Oral Surg Oral Med Oral Pathol Oral
jects with prior lichen planus. Otolaryngol Head Neck Radiol Endod. 1996;81(6):682–90.
Surg. 2007;136(3):401–4. Rodstrom PO, Jontell M, Mattsson U, Holmberg E. Cancer
Murti PR, Daftary DK, Bhonsle RB, Gupta PC, Mehta FS, and oral lichen planus in a Swedish population. Oral
Pindborg JJ. Malignant potential of oral lichen planus: Oncol. 2004;40(2):131–8.
observations in 722 patients from India. J Oral Pathol. Roitberg-Tambur A, Friedmann A, Korn S, Markitziu A,
1986;15(2):71–7. Pisanti S, Safirman C, Nelken D, Brautbar C. Serologic
Nagao Y, Sata M, Tanikawa K, Itoh K, Kameyama and molecular analysis of the HLA system in Israeli
T. Lichen planus and hepatitis C virus in the northern Jewish patients with oral erosive lichen planus. Tissue
Kyushu region of Japan. Eur J Clin Investig. 1995;25 Antigens. 1994;43(4):219–23.
(12):910–4. Rojo-Moreno JL, Bagan JV, Rojo-Moreno J, Donat JS,
O’Neill ID. Off-label use of biologicals in the management Milian MA, Jimenez Y. Psychologic factors and oral
of inflammatory oral mucosal disease. J Oral Pathol lichen planus. A psychometric evaluation of 100 cases.
Med. 2008;37(10):575–81. Oral Surg Oral Med Oral Pathol Oral Radiol Endod.
O’Neill ID, Scully C. Biologics in oral medicine: ulcerative 1998;86(6):687–91.
disorders. Oral Dis. 2013;19(1):37–45. Roopashree MR, Gondhalekar RV, Shashikanth MC,
Olivier V, Lacour JP, Mousnier A, Garraffo R, Monteil RA, George J, Thippeswamy SH, Shukla A. Pathogenesis
Ortonne JP. Treatment of chronic erosive oral lichen of oral lichen planus – a review. J Oral Pathol Med.
planus with low concentrations of topical tacrolimus: 2010;39(10):729–34.
an open prospective study. Arch Dermatol. 2002;138 Rubaci AH, Kazancioglu HO, Olgac V, Ak G. The roles of
(10):1335–8. matrix metalloproteinases-2, -7, -10 and tissue inhibitor
Ostman PO, Anneroth G, Skoglund A. Amalgam- of metalloproteinase-1 in the pathogenesis of oral
associated oral lichenoid reactions. Clinical and histo- lichen planus. J Oral Pathol Med. 2012;41(9):689–96.
logic changes after removal of amalgam fillings. Oral Salazar-Sanchez N, Lopez-Jornet P, Camacho-Alonso F,
Surg Oral Med Oral Pathol Oral Radiol Endod. 1996;81 Sanchez-Siles M. Efficacy of topical Aloe vera in
(4):459–65. patients with oral lichen planus: a randomized double-
Parashar P. Oral lichen planus. Otolaryngol Clin N blind study. J Oral Pathol Med. 2010;39(10):735–40.
Am. 2011;44(1): 89–107, vi. Santoro A, Majorana A, Bardellini E, Festa S, Sapelli P,
Payeras MR, Cherubini K, Figueiredo MA, Salum Facchetti F. NF-kappaB expression in oral and cutane-
FG. Oral lichen planus: focus on etiopathogenesis. ous lichen planus. J Pathol. 2003;201(3):466–72.
Arch Oral Biol. 2013;58(9):1057–69. Santoro A, Majorana A, Roversi L, Gentili F, Marrelli S,
Pedersen A. Abnormal EBV immune status in oral lichen Vermi W, Bardellini E, Sapelli P, Facchetti
planus. Oral Dis. 1996;2(2):125–8. F. Recruitment of dendritic cells in oral lichen planus.
Pelisse M. The vulvo-vaginal-gingival syndrome. A new J Pathol. 2005;205(4):426–34.
form of erosive lichen planus. Int J Dermatol. 1989;28 Savage NW, McCullough MJ. Topical corticosteroids in
(6):381–4. dental practice. Aust Dent J. 2005;50(4 Suppl 2):
Petti S, Rabiei M, De Luca M, Scully C. The magnitude of S40–4.
the association between hepatitis C virus infection and Scardina GA, Messina P, Carini F, Maresi E. A randomized
oral lichen planus: meta-analysis and case control trial assessing the effectiveness of different concentra-
study. Odontology. 2011;99(2):168–78. tions of isotretinoin in the management of lichen
Piboonniyom SO, Treister N, Pitiphat W, Woo SB. Scoring planus. Int J Oral Maxillofac Surg. 2006;35(1):67–71.
system for monitoring oral lichenoid lesions: a prelim- Scully C, Carrozzo M. Oral mucosal disease: lichen planus.
inary study. Oral Surg Oral Med Oral Pathol Oral Br J Oral Maxillofac Surg. 2008;46(1):15–21.
Radiol Endod. 2005;99(6):696–703. Scully C, Diz Dios P. Orofacial effects of antiretroviral
Porter SR, Kirby A, Olsen I, Barrett W. Immunologic therapies. Oral Dis. 2001;7(4):205–10.
aspects of dermal and oral lichen planus: a review. Scully C, Beyli M, Ferreiro MC, Ficarra G, Gill Y,
Griffiths M, Holmstrup P, Mutlu S, Porter S, Wray
Oral Lichen Planus 39

D. Update on oral lichen planus: etiopathogenesis and Suter VG, Warnakulasuriya S. The role of patch testing in
management. Crit Rev Oral Biol Med. 1998;9 the management of oral lichenoid reactions. J Oral
(1):86–122. Pathol Med. 2016;45(1):48-57.
Shengyuan L, Songpo Y, Wen W, Wenjing T, Haitao Z, Taniguchi Y, Nagao T, Maeda H, Kameyama Y,
Binyou W. Hepatitis C virus and lichen planus: a recip- Warnakulasuriya KA. Epithelial cell proliferation in
rocal association determined by a meta-analysis. Arch oral lichen planus. Cell Prolif. 2002;35(Suppl
Dermatol. 2009;145(9):1040–7. 1):103–9.
Shimoyama T, Horie N, Kato T, Kaneko T, Komiyama Tenovuo J. Antimicrobial function of human saliva – how
K. Helicobacter pylori in oral ulcerations. J Oral Sci. important is it for oral health? Acta Odontol Scand.
2000;42(4):225–9. 1998;56(5):250–6.
Shirota Y, Illei GG, Nikolov NP. Biologic treatments for Thongprasom K, Dhanuthai K. Steriods in the treatment of
systemic rheumatic diseases. Oral Dis. 2008;14 lichen planus: a review. J Oral Sci. 2008;50(4):377–85.
(3):206–16. Thongprasom K, Luangjarmekorn L, Sererat T, Taweesap
Sieg P, Von Domarus H, Von Zitzewitz V, Iven H, Farber W. Relative efficacy of fluocinolone acetonide com-
L. Topical cyclosporin in oral lichen planus: a con- pared with triamcinolone acetonide in treatment of
trolled, randomized, prospective trial. Br J Dermatol. oral lichen planus. J Oral Pathol Med. 1992;21
1995;132(5):790–4. (10):456–8.
Silverman Jr S, Gorsky M, Lozada-Nur F. A prospective Thongprasom K, Luengvisut P, Wongwatanakij A,
follow-up study of 570 patients with oral lichen planus: Boonjatturus C. Clinical evaluation in treatment of
persistence, remission, and malignant association. Oral oral lichen planus with topical fluocinolone acetonide:
Surg Oral Med Oral Pathol. 1985;60(1):30–4. a 2-year follow-up. J Oral Pathol Med. 2003;32
Silverman Jr S, Gorsky M, Lozada-Nur F, Giannotti K. A (6):315–22.
prospective study of findings and management in Thongprasom K, Carrozzo M, Furness S, Lodi
214 patients with oral lichen planus. Oral Surg Oral G. Interventions for treating oral lichen planus.
Med Oral Pathol. 1991;72(6):665–70. Cochrane Database Syst Rev. 2011;(7):Cd001168.
Siponen M, Huuskonen L, Laara E, Salo T. Association of Thongprasom K, Prapinjumrune C, Carrozzo M. Novel
oral lichen planus with thyroid disease in a Finnish therapies for oral lichen planus. J Oral Pathol Med.
population: a retrospective case-control study. Oral 2013;42:721–7.
Surg Oral Med Oral Pathol Oral Radiol Endod. Thornhill MH, Pemberton MN, Simmons RK, Theaker
2010;110(3):319–24. ED. Amalgam-contact hypersensitivity lesions and
Soto Araya M, Rojas Alcayaga G, Esguep A. Association oral lichen planus. Oral Surg Oral Med Oral Pathol
between psychological disorders and the presence of Oral Radiol Endod. 2003;95(3):291–9.
oral lichen planus, burning mouth syndrome and recur- Torresani C, Nannini R, Bondi A, Guadagni M, Manara
rent aphthous stomatitis. Med Oral. 2004;9(1):1–7. GC. Erosive oral lichen planus due to sensitization to
Sugerman PB, Savage NW, Walsh LJ, Seymour cobalt chloride. Clin Exp Dermatol. 1994;19(6):535–6.
GJ. Disease mechanisms in oral lichen planus. A pos- Valente G, Pagano M, Carrozzo M, Carbone M, Bobba V,
sible role for autoimmunity. Australas J Dermatol. Palestro G, Gandolfo S. Sequential immunohistochem-
1993;34(2):63–9. ical p53 expression in biopsies of oral lichen planus
Sugerman PB, Savage NW, Seymour GJ. Phenotype and undergoing malignant evolution. J Oral Pathol Med.
suppressor activity of T-lymphocyte clones extracted 2001;30(3):135–40.
from lesions of oral lichen planus. Br J Dermatol. van der Meij EH, van der Waal I. Lack of clinicopathologic
1994;131(3):319–24. correlation in the diagnosis of oral lichen planus based
Sugerman PB, Savage NW, Xu LJ, Walsh LJ, Seymour on the presently available diagnostic criteria and sug-
GJ. Heat shock protein expression in oral lichen planus. gestions for modifications. J Oral Pathol Med. 2003;32
J Oral Pathol Med. 1995;24(1):1–8. (9):507–12.
Sugerman PB, Satterwhite K, Bigby M. Autocytotoxic van der Meij EH, Reibel J, Slootweg PJ, van der Wal JE, de
T-cell clones in lichen planus. Br J Dermatol. Jong WF, van der Waal I. Interobserver and
2000a;142(3):449–56. intraobserver variability in the histologic assessment
Sugerman PB, Savage NW, Zhou X, Walsh LJ, Bigby of oral lichen planus. J Oral Pathol Med. 1999a;28
M. Oral lichen planus. Clin Dermatol. 2000b;18 (6):274–7.
(5):533–9. van der Meij EH, Schepman KP, Smeele LE, van der Wal
Sugerman PB, Savage NW, Walsh LJ, Zhao ZZ, Zhou XJ, JE, Bezemer PD, van der Waal I. A review of the recent
Khan A, Seymour GJ, Bigby M. The pathogenesis of literature regarding malignant transformation of oral
oral lichen planus. Crit Rev Oral Biol Med. 2002;13 lichen planus. Oral Surg Oral Med Oral Pathol Oral
(4):350–65. Radiol Endod. 1999b;88(3):307–10.
Sugermann PB, Savage NW, Seymour GJ, Walsh LJ. Is van der Meij EH, Schepman KP, van der Waal I. The
there a role for tumor necrosis factor-alpha possible premalignant character of oral lichen planus
(TNF-alpha) in oral lichen planus? J Oral Pathol Med. and oral lichenoid lesions: a prospective study. Oral
1996;25(5):219–24.
40 M.J. McCullough et al.

Surg Oral Med Oral Pathol Oral Radiol Endod. 2003;96 and anti-inflammatory effects of aloesin derivatives in
(2):164–71. Aloe vera. Planta Med. 2002;68(11):957–60.
van der Meij EH, Mast H, van der Waal I. The possible Yamamoto T, Osaki T. Characteristic cytokines generated
premalignant character of oral lichen planus and oral by keratinocytes and mononuclear infiltrates in oral
lichenoid lesions: a prospective five-year follow-up lichen planus. J Invest Dermatol. 1995;104(5):784–8.
study of 192 patients. Oral Oncol. 2007;43(8):742–8. Yarom N. Etanercept for the management of oral lichen
van Loon LA, Bos JD, Davidson CL. Clinical evaluation of planus. Am J Clin Dermatol. 2007;8(2):121.
fifty-six patients referred with symptoms tentatively Yarosh DB, Pena AV, Nay SL, Canning MT, Brown
related to allergic contact stomatitis. Oral Surg Oral DA. Calcineurin inhibitors decrease DNA repair and
Med Oral Pathol. 1992;74(5):572–5. apoptosis in human keratinocytes following ultraviolet
Vignali DA, Collison LW, Workman CJ. How regulatory T B irradiation. J Invest Dermatol. 2005;125(5):1020–5.
cells work. Nat Rev Immunol. 2008;8(7):523–32. Zain RB, Ikeda N, Razak IA, Axell T, Majid ZA, Gupta
Villarroel Dorrego M, Correnti M, Delgado R, Tapia PC, Yaacob M. A national epidemiological survey of
FJ. Oral lichen planus: immunohistology of mucosal oral mucosal lesions in Malaysia. Community Dent
lesions. J Oral Pathol Med. 2002;31(7):410–4. Oral Epidemiol. 1997;25(5):377–83.
Vincent SD, Fotos PG, Baker KA, Williams TP. Oral lichen Zeng X, Hou X, Wang Z, Jiang L, Xiong C, Zhou M, Chen
planus: the clinical, historical, and therapeutic features Q. Carriage rate and virulence attributes of oral Can-
of 100 cases. Oral Surg Oral Med Oral Pathol. 1990;70 dida albicans isolates from patients with oral lichen
(2):165–71. planus: a study in an ethnic Chinese cohort. Mycoses.
Voigtlander V, Boonen H. Squamous cell carcinoma of the 2009;52(2):161–5.
lower lip in discoid lupus erythematosus associated Zhang J, Zhou G, Du GF, Xu XY, Zhou HM. Biologics, an
with hereditary deficiency of complement 2. Z Hautkr. alternative therapeutic approach for oral lichen planus.
1990;65(9):836–7. J Oral Pathol Med. 2011;40(7):521–4.
Wee JS, Shirlaw PJ, Challacombe SJ, Setterfield Zhao ZZ, Savage NW, Pujic Z, Walsh
JF. Efficacy of mycophenolate mofetil in severe muco- LJ. Immunohistochemical localization of mast cells
cutaneous lichen planus: a retrospective review of and mast cell-nerve interactions in oral lichen planus.
10 patients. Br J Dermatol. 2012;167(1):36–43. Oral Dis. 1997;3(2):71–6.
Wong L, Freeman S. Oral lichenoid lesions (OLL) and Zhou XJ, Sugerman PB, Savage NW, Walsh LJ. Matrix
mercury in amalgam fillings. Contact Dermatitis. metalloproteinases and their inhibitors in oral lichen
2003;48(2):74–9. planus. J Cutan Pathol. 2001;28(2):72–82.
Xie S, Ding L, Xiong Z, Zhu S. Implications of Th1 and Zhou XJ, Sugerman PB, Savage NW, Walsh LJ, Seymour
Th17 cells in pathogenesis of oral lichen planus. J GJ. Intra-epithelial CD8+ T cells and basement mem-
Huazhong Univ Sci Technolog Med Sci. 2012;32 brane disruption in oral lichen planus. J Oral Pathol
(3):451–7. Med. 2002;31(1):23–7.
Yagi A, Kabash A, Okamura N, Haraguchi H, Moustafa
SM, Khalifa TI. Antioxidant, free radical scavenging

You might also like