Plaque psoriasis
Overview
Psoriasis encompasses a group of
related immune‐mediated
inflammatory skin diseases that affect up to 3% of the population. The
commonest presentation of psoriasis is plaque psoriasis. Psoriasis is associated
with co‐morbidities
including obesity, diabetes, vascular disease, depression and inflammatory
bowel disease, which contribute to the considerable morbidity and mortality.
A spectrum of treatments
is available. Patients with mild disease are usually treated with topical
corticosteroids and vitamin D derivatives. Other topical therapies include
tar-based preparations, anthralin, calcineurin inhibitors and tazarotene. In
patients with more widespread disease, traditional approaches such as photo (chemo)
therapy (UVB or PUVA), methotrexate, cyclosporine, or acitretin may be
required. Alternatively, targeted immunomodulators (“biologic” therapies) that
are highly specific (e.g. targeting TNF-α, IL-12/23, IL-17) are increasingly
being used, albeit at a greater cost than conventional approaches.
Because psoriasis is a
chronic disease, it requires a long-term treatment strategy, which takes into
account potential medication side effects and individual needs. Treatment
strategies are often divided into a clearing phase and a maintenance phase.
Salient features
· Psoriasis is a chronic, immune-mediated disorder that results
from a polygenic predisposition combined with environmental triggers, e.g.
trauma, infections, medications, psychological stress
· The underlying pathophysiology involves various classes of T
cells and their interactions with dendritic cells and cells involved in innate
immunity, including neutrophils and keratinocytes
· Identification of susceptibility genes has pointed to a major
role for the innate and adaptive immune systems as well as altered epidermal
differentiation
· Sharply demarcated, scaly, erythematous plaques characterize the
most common form of psoriasis; occasionally, sterile pustules are seen
· The most common sites of involvement are the scalp, elbows and
knees, followed by the nails, hands, feet and trunk (including the intergluteal
fold)
· Typical histologic findings include acanthosis with elongated
rete ridges, hypogranulosis, hyper- and parakeratosis, dilated blood vessels,
and a perivascular infiltrate of lymphocytes with neutrophils singly or within
aggregates in the epidermis
· Psoriatic arthritis is the major associated systemic
manifestation and the most common presentation is asymmetric oligoarthritis of
the small joints of the hands and feet; other comorbidities include depression,
metabolic syndrome, and cardiovascular disease in patients with moderate to
severe disease
· Topical therapy is used to treat limited disease, whereas
phototherapy, methotrexate, cyclosporine and targeted immuno-modulators
(“biologic” therapies) that target key immune effector cells and cytokines lead
to significant clinical improvement of moderate to severe psoriasis
Introduction
A common, chronic, disfiguring, inflammatory and
proliferative condition of the skin,
associated with systemic manifestations in many organ systems, in
which both genetic and environmental play a
critical role in the etiology and pathogenesis. The most
characteristic lesions consist of red (salmon pink), scaly (large adherent
silvery centrally attached scales), sharply demarcated, indurated plaques,
present particularly over extensor surfaces and scalp. Various environmental triggering
factors, e.g. trauma, infections or medications, may elicit disease in
predisposed individuals. The disease is life-long and its course is
unpredictable but is usually chronic with exacerbations and remissions that are
emotionally and physically debilitating.
Psoriasis is a systemic disease process in
which up to 20–30% of patients has or will develop psoriatic arthritis. In
addition, in patients with moderate to severe psoriasis, there is an increased
relative risk for metabolic syndrome and atherosclerotic cardiovascular
disease.
Provisional working classification of
psoriasis
Clinical forms of psoriasis
(based on morphology or natural history)
·
Plaque
psoriasis (psoriasis vulgaris)
·
Acute
guttate psoriasis
·
‘Unstable’
psoriasis
·
Erythrodermic
psoriasis
·
Pustular
psoriasis
·
Atypical
forms of psoriasis
Other specified forms of
psoriasis (based on age or precipitants)
·
Linear
and segmental psoriasis
·
Psoriasis
in childhood and old age
·
Photo
aggravated psoriasis
·
Drug‐induced or
exacerbated psoriasis
·
HIV‐induced or exacerbated
psoriasis
Psoriasis affecting specific
sites
·
Scalp
psoriasis
·
Follicular
psoriasis
·
Seborrheic
psoriasis (sebopsoriasis)
·
Flexural
psoriasis (inverse psoriasis)
·
Genital
psoriasis
·
Non‐pustular palmoplantar
psoriasis
·
Nail
psoriasis
·
Mucosal
lesions
·
Ocular
lesions
PSORIASIS, PLAQUE
Epidemiological
aspects
Incidence and prevalence
1-3 per cent of world’s populations
have psoriasis.
Age of onset
Psoriasis can first appear at any age, but two peaks age of onset:
one at 20–30 years and a second peak at 50–60 years. In approximately 75% of
patients, the onset is before the age of 40 years. Although the age of onset is
earlier in women than in men, the natural history is similar – chronic with
intermittent remissions. Plaque psoriasis is the most frequent form of the
disease in children, followed by guttate psoriasis.
Sex
Males and females are equally affected
by psoriasis vulgaris.
Genetic epidemiology
Based
on population data, lifetime risks of psoriasis is 28 % if one parent is affected, 65%
if both parents are affected, and 6% if one sibling affected, compared to 4%
when no parent or sibling is affected. Genomic imprinting may explain why
psoriatic fathers are more likely to pass on the disease to their children than
are psoriatic mothers. HLA-Cw6 is strongly linked to the age of onset of
psoriasis. This finding led Henseler and Christophers to propose two different forms
of psoriasis exist: type 1 psoriasis, with
age of onset before 40 years, is hereditary with a positive family history, strongly
HLA associated (particularly HLA-Cw6), and more likely to be severe and type II psoriasis, with age of onset
after 40 years, is sporadic with no
family history, lacking HLA
associated, and usually mild.
Guttate psoriasis is almost invariably
HLA associated and thought to be closely linked pathogenetically to type 1
plaque psoriasis.
Environmental risk (Precipitating)
factors
Triggering factors, both external (directly interacting with
the skin) and systemic, can elicit psoriasis in genetically predisposed
individuals. These factors not only initiate the disease process but also cause
exacerbation of pre-existing disease.
The interplay between innate and adaptive immune cells in the
pathogenesis of Psoriasis
Psoriasis can be triggered by several
factors, including genetic variants, infection, wound, obesity, stress, and
drugs. Early upstream events in psoriasis involve the innate immune activation
of skin resident keratinocytes or fibroblasts or recruited plasmacytoid
dendritic cells (pDCs) or neutrophils. Cytokines derived from these innate
immune cells promote myeloid dendritic cell maturation, with consequent Th17 T
cell development and the beginning of the adaptive immune phase. T cell infiltrate
promotes inflammatory amplification of innate immune cells, leading to the
formation of an autoimmune self-amplifying loop that drives pathogenic hyper
proliferation of keratinocytes and manifestations of psoriasis.
External triggering factors
Trauma - Koebner
and reverse Koebner phenomena
Psoriasis is one of several conditions in which various
types of trauma may elicit the disease in previously uninvolved skin (Koebner
reaction). A wide range of injurious local stimuli, including physical,
chemical, electrical, surgical, infective and inflammatory insults, has been
recognized to elicit psoriatic lesions. The lag time
between the trauma and the appearance of skin lesions is usually 2–6 weeks.
Clearing of existing psoriasis following injury has been observed and termed the
reverse Koebner reaction. The Koebner reaction is often thought to be more
frequent in actively spreading, severe psoriasis. The Koebner phenomenon suggests that psoriasis is a
systemic disease that can be triggered locally in the skin.
Trauma can initiate innate immune activation with subsequent
specific immune activation and keratinocyte hyper proliferation and
angiogenesis. Up to 25% of patients report the development of psoriasis in
sites of skin trauma and this is commoner in patients who are HLA‐C:06:02 positive.
Systemic triggering factors
Infection
Acute
guttate psoriasis is strongly associated with preceding or concurrent
streptococcal infection, particularly of the throat (tonsillitis). There is evidence that streptococcal
infection may be important in chronic plaque psoriasis also. Oligoclonal expansion of T cells occurs in the tonsils in
response to streptococcal colonization, and the same T‐cell repertoire is found in the
peripheral blood and skin of patients with guttate psoriasis. It has been
proposed that psoriasis is an autoimmune disease that is fueled by persistent
intracellular streptococcal infection. Streptococci are facultative
intracellular bacteria and are not eliminated by standard antibiotic therapy,
which may explain the lack of efficacy of antibiotics in treating guttate
psoriasis. Tonsillectomy appears to be effective in some patients because there
is a reduction in skin homing T cells.
Further, acute episodes of guttate
psoriasis are much more common in individuals with a family history of plaque
psoriasis and one-third of cases of guttate psoriasis progress to the chronic
plaque form.
Drugs
Drugs may be associated
with the precipitation or exacerbation of psoriasis. A drug can affect the
patient with psoriasis in several ways: (1) exacerbation of pre-existing
psoriasis; (2) induction of lesions of psoriasis in clinically normal skin in a
person with psoriasis; (3) precipitation of psoriasis de novo; and (4) development of treatment resistance.
In addition, in sites of involvement by other cutaneous drug reactions (e.g.
exanthematous), psoriatic lesions may appear secondary to the Koebner
phenomenon, and plaques may develop at sites of injections of interferon. The clinical
manifestations of drug-induced psoriasis span the spectrum of psoriasis, from
limited or generalized plaques to erythroderma and pustulosis of the palms and
soles. Nail changes and scalp involvement may also be seen.
There are many drugs reported to be
responsible for the onset or exacerbation of psoriasis. Chief amongst are lithium salts, synthetic antimalarials, interferon α and TNF‐α inhibitors. Given the known co‐morbidities of psoriasis, β‐blockers, ACE inhibitors and NSAIDS are widely prescribed in
patients with psoriasis and yet, in most, do not appear to have a major
detrimental effect.
Lesions of drug-induced
psoriasis usually regress within weeks to a few months of discontinuing the
inciting drug. However, psoriasiform eruptions, including palmoplantar
pustulosis, may be more persistent in patients receiving TNF-α inhibitor.
Histologic findings are
not always characteristic. They may suggest psoriasis or be more compatible
with a lichenoid drug eruption.
Sunlight
Although sunlight is generally
beneficial, in 5–20% of patients psoriasis may be
provoked by strong sunlight and cause summer exacerbations in exposed skin.
Some
of these patients gave a history of polymorphic light eruption (PLE) with
psoriasis appearing as a Koebner phenomenon within PLE lesions. Recent work has
indicated that severely photosensitive psoriasis is predominantly female,
distinct from PLE, and strongly associated with HLA-Cw6, family history and
very early age of onset. Photo-chemotherapy (PUVA) can be helpful in these
patients.
Metabolic factors
The early onset of psoriasis in women,
with a peak around puberty, changes during pregnancy and provocation of
psoriasis by high dose estrogen therapy potentially indicates a role for
hormonal factors in the disease.
Hypocalcemia
following
accidental parathyroidectomy has been
reported to be a triggering factor for generalized pustular psoriasis. If psoriasis changes
in pregnancy, it is more likely to improve than worsen and about 50% of the patients may show improvement in pregnancy, while in the postpartum
period it is more likely to deteriorate. However,
pregnant women may develop pustular psoriasis, also referred to as impetigo
herpetiformis, sometimes in association with hypocalcemia.
Psychogenic stress
Emotional upset is a well-established systemic triggering
factor in psoriasis, with a heightened cortisol response to stress having been
demonstrated in affected patients. It
has been associated with initial presentations of the disease and up to 80% of
psoriasis patients report that psychological distress results in flares of
their pre-existing disease. Moderate to severe psoriasis is associated with
increased levels of depression, anxiety and worry compared to the general
population.
Alcohol consumption,
smoking and obesity
Obesity, increased alcohol
consumption, and smoking have all been associated with psoriasis.
It has long been suspected that both
cigarettes and alcohol have a detrimental effect on psoriasis. Alcohol may
exacerbate pre-existing disease but does not appear to induce psoriasis. This
effect seems greater in men than in women. Psoriasis
appears to be more prevalent (15%) in patients with alcoholic liver disease. Heavy drinkers tend
to have more extensive and inflamed disease. Excess drinking is undoubtedly
also a consequence of the disease and leads to treatment resistance and reduced
therapeutic compliance. Abstinence has been reported to induce remission.
In one analysis, smoking
appeared to have a role in the onset of psoriasis, while obesity appeared to be
a consequence of psoriasis.
Cigarette smoking is increased
amongst patients with plaque psoriasis compared to the normal population,
particularly in women. Patients who smoke are at increased risk of developing
psoriasis, of having more severe disease and of developing psoriatic arthritis.
Smoking has multiple immunological effects that may contribute to the
initiation and persistence of psoriasis. There is a particularly strong
association between cigarette smoking and palmoplantar pustulosis in women. Some
studies have found that the prevalence of psoriasis in a population of
individuals who stop smoking or who lose weight eventually reverts to
background levels.
HIV and acquired immune deficiency
syndrome
The association between severe
psoriasis, psoriatic arthropathy and human immunodeficiency virus (HIV)
infection is well recognized.
Pathogenesis
The cardinal features of lesional
psoriatic skin are: (i) epidermal hyper- proliferation with loss of
differentiation; (ii) increased angiogenesis within the
dermis leading to dilatation
and proliferation of dermal blood vessels and (iii) a marked dermal and epidermal accumulation of inflammatory cells,
particularly neutrophils and T lymphocytes.
Epidermal
proliferation
Normal
basal keratinocytes proliferate, differentiate and ascend through the layers of
the epidermis before undergoing senescence and entering the keratin layer. The epidermis of
psoriasis replicates too quickly. Keratinocytes proliferate out of control, and
an excessive number of germinative cells enter the cell cycle as consequence of
an increase in the proliferating cell compartment in the basal and supra-basal
levels of the epidermis with subsequent rapid progression
through the epidermal layers and decreased differentiation. The growth fraction
of epidermal basal cells is greatly increased to almost 100% compared with 30%
in normal skin. This ‘out of control’ proliferation is rather like a car going
too fast because the accelerator is stuck, and cannot be stopped by putting a
foot on the brake. The epidermal turnover time is greatly shortened, to less
than 10 days compared with 30 to 60 days in normal skin. This epidermal
hyperproliferation accounts for many of the metabolic abnormalities associated
with psoriasis. It is not confined to obvious plaques: similar but less marked
changes also occur in the apparently normal skin of psoriatics.
Vascular
changes
Vertical dermal capillary loops in
lesional skin are dilated, elongated and twisted. It has been demonstrated that
epidermal keratinocytes are the primary source of angiogenic activity. These
cells produce an array of soluble mediators with angiogenic activity including
vascular endothelial growth factor (VEGF). It is over-expressed in psoriatic
epidermis as are its receptors on lesional psoriatic microvasculature.
In addition to vascular growth, dermal
capillaries contribute to the inflammatory process actively through surface
expression of molecules involved in leukocyte homing, induced by inflammatory
mediators such as histamine, neuropeptides, interleukin-1 (IL-1) and tumor
necrosis factor-alpha (TNF-α). Importantly, E selectin is induced and
intercellular adhesion molecule-1 (ICAM- 1) up-regulated on dermal vessels in
lesional tissue, thus providing a mechanism for skin homing T lymphocytes to
accumulate within lesional dermis and epidermis.
Immunology
and inflammation
It is increasingly clear that the
innate immune system, which provides an early response against harm to the
host, is dysregulated in psoriasis. Further, in psoriatic stratum corneum,
there is an abundance of antimicrobial peptides such as defensins and
cathelicidins, which have the capacity to activate innate immunity.
Innate immune mechanisms in turn lead
to antigen driven T cell expansion and activation. A subset of T cell, thought
to play a key role in autoimmunity, termed Th17, and is the primary pathogenic
subset. Evidence includes genetic association with IL-23 being a key cytokine
in generation of Th17 cells, abundance of IL-23 in psoriatic tissue.
Based on the above information, a
pathogenic model has been suggested in which aberrations in the stratum corneum
trigger activation of innate immune mechanisms, which in turn lead to
recruitment and activation of Th17 cells, which provide effector cytokines
leading to epidermal hyper proliferation and a proinflammatory state.
Pathogenesis of psoriasis – Exposure to microbial
or mechanical injury damage associated molecular patterns/ pathogen associated
molecular patterns leads to activation of antigen presenting cells like
macrophages and dermal dendritic cells; failure to maintain skin barrier due to
late cornified envelope proteins 3C/3B deletion leads to continuous exposure to
such antigens. Interaction of APC and T cells leads to activation of Th1 and
Th17 cells mediated by IL-23. Liberation of IL-17 and IL-22 by Th17 cells, and
tumor necrosis factor-α and IFN-γ by Th1 cells further perpetuates the
keratinocyte injury creating a vicious positive feedback cycle.
Genetic and
environmental factors act in conjunction to produce immune dysregulation in the
presence of a defective skin barrier. Interaction of damage associated
molecular patterns (DAMP) and the pathogen associated molecular patterns (PAMP)
with their receptors, such as TLR and NOD like receptors, causes the activation
of keratinocytes and the epidermal innate immune system and thus, increased
secretion of antimicrobial proteins. This interaction between DAMP/PAMP with
TLR/NOD like receptors is also followed by liberation of inflammatory cytokines
such as TNF-α, IL-8 and IL-1β, all of which are potent chemo attractants. In
patients who carry the psoriasis susceptibility genes, such as HLA-C*06,
LCE3B/LCE3C-del or defensin genes, exposure to PAMP leads to a heightened
inflammatory response and defective skin barrier repair with increased
expression of keratins 6 and 17, and the LCE3 family. Aberrant skin repair
allows a sustained exposure to PAMPs which are engulfed by Langerhans cells and
dendritic cells.
Immunopathogenesis
The underlying pathophysiology involves T cells and their
interactions with dendritic cells and cells involved in innate immunity,
including keratinocytes.
Immunopathogenesis of psoriasis
The occurrence of triggering
environmental factors in genetically predisposed individuals, carrying
susceptibility alleles of psoriasis-associated genes, results in disease
development. During the initiation phase, stressed keratinocytes can release
self DNA and RNA, which form complexes with the cathelicidin LL37 that then
induce interferon-α (IFN-α) production by plasmacytoid dendritic cells (pDCs;
recruited into the skin via fibroblast-released chemerin), thereby activating
dermal DCs (dDCs). Keratinocyte-derived interleukin-1β (IL-1β), IL-6 and tumor
necrosis factor-α (TNF-α) also contribute to the activation of dDCs. Activated
dDCs then migrate to the skin-draining lymph nodes to present an as-yet-unknown
antigen (either of self or of microbial origin) to naive T cells and (via
secretion of different types of cytokines by DCs) promote their differentiation
into T helper 1 (Th1), Th17 and Th22 cells. Th1 cells (expressing cutaneous
lymphocyte antigen [CLA], CXC-chemokine receptor 3 [CXCR3] and CC-chemokine
receptor 4 [CCR4]), Th17 cells (expressing CLA, CCR4 and CCR6) and Th22 cells
(expressing CCR4 and CCR10) migrate via lymphatic and blood vessels into
psoriatic dermis, attracted by the keratinocyte-derived chemokines CCL20,
CXCL9–11 and CCL17; this ultimately leads to the formation of a psoriatic
plaque. Th1 cells release IFN-γ and TNF-α, which amplify the inflammatory
cascade, acting on keratinocytes and dDCs. Th17 cells secrete IL-17A and IL-17F
(and also IFN-γ and IL-22), which stimulate keratinocyte proliferation and the
release of β-defensin 1/2, S100A7/8/9 and the neutrophil-recruiting chemokines
CXCL1, CXCL3, CXCL5 and CXCL8. Neutrophils (N) infiltrate the stratum corneum
and produce reactive oxygen species (ROS) and α-defensin with antimicrobial activity,
as well as CXCL8, IL-6 and CCL20. Th22 cells secrete IL-22, which induces
further release of keratinocyte-derived T cell-recruiting chemokines. Moreover,
inflammatory DCs (iDCs) produce IL-23, nitric oxide (NO) radicals and TNF-α,
while natural killer T (NKT) cells release TNF-α and IFN-γ. Keratinocytes also
release vascular endothelial growth factor (VEGF), basic fibroblast growth
factor (bFGF), and angiopoietin (Ang), thereby promoting neoangiogenesis.
Macrophage (M)-derived chemokine CCL19 promotes clustering of Th cells
expressing chemokine receptor CCR7 with DC in the proximity of blood vessels,
with further T-cell activation. At the dermal–epidermal junction, memory CD8+
cytotoxic T cells (Tc1) expressing very-late antigen-1 (VLA-1) bind to collagen
IV, allowing entry into the epidermis and contributing to disease pathogenesis
by releasing both Th1 and Th17 cytokines. Cross-talk between keratinocytes,
producing TNF-α, IL-1β and transforming growth factor-β (TGF-β), and
fibroblasts, which in turn release keratinocyte growth factor (KGF), epidermal
growth factor (EGF) and TGF-β, and possibly Th22 cells releasing FGFs,
contribute to tissue reorganization and deposition of extracellular matrix
(e.g. collagen, proteoglycans).
Histopathology
The main changes are the following.
1 Parakeratosis (nuclei retained in the horny layer).
2 Irregular thickening of the epidermis over the rete
ridges, but thinning over dermal papillae. Bleeding may occur when scale is
scratched off (Auspitz’s sign).
3 Epidermal polymorphonuclear leucocyte infiltrates and
micro-abscesses (described originally by Munro).
4 Dilated and tortuous capillary loops in the dermal
papillae.
5 T-lymphocyte infiltrate in upper dermis.
Typical
histologic findings include acanthosis with uniform elongated rete ridges, thinning
of the suprapapillary plate, hypogranulosis, hyper- and parakeratosis, dilated and
tortuous capillary loops in the dermal papillae and a perivascular infiltrate of lymphocytes with
neutrophils singly or within aggregates in the epidermis.
Initial lesion
In the initial lesion, i.e. a smooth surface pinhead-sized
papule, the histopathologic features are not yet diagnostic with a preponderance
of dermal changes. A superficial perivascular infiltrate of lymphocytes and
macrophages is seen in the dermis along with papillary edema and a dilation of
capillaries appears to precede epidermal changes in early developing lesions. In acute eruptive guttate lesions, mast cell degranulation
is a constant feature.
There is mild epidermal acanthosis without
parakeratosis. Macrophages and lymphocytes appear in the lower half of the
epidermis and some spongiosis of the epidermis is seen at these sites. Per a
large body of evidence, neutrophils are not yet detected in this early phase.
Active lesion
A fully developed guttate lesion or
the marginal zone of an enlarging psoriatic plaque is designated as an “active
lesion”. The histopathologic findings in an active lesion are diagnostic for
psoriasis.
In the
papillary dermis, the capillaries are increased in
number and length and they have a dilated tortuous appearance. Marked edema is
seen, especially at the tops of the papillae. There is a mixed perivascular
infiltrate of lymphocytes, macrophages and neutrophils, and lymphocytes and
neutrophils have migrated into the epidermis.
The epidermis
is acanthotic with rete ridges of even length with focal accumulations of
neutrophils and lymphocytes. There is spongiosis in the malpighian
layer. Above these foci, the granular
layer is absent and the stratum corneum still contains flattened nuclei
(parakeratosis). The accumulation of neutrophils within a spongiotic malpighian
layer to form the characteristic “spongiform pustules of Kogoj” and the accumulation of neutrophil
remnants in the stratum corneum, surrounded by parakeratosis, as a
“microabscess of Munro” are the two findings that are pathognomonic for
psoriasis and AGEP.
Stable lesion
In the papillary dermis,
the capillaries are elongated and tortuous, extending upward into elongated
club-shaped dermal papillae and almost touch the undersurface of
the thinning suprapapillary epidermis;
only a small suprapapillary plate of epidermal cells covers the tip of these
dermal papillae and are surrounded by a mixed mononuclear and
neutrophil infiltrate, as well as extravasated erythrocytes. Invasion of the
epidermis with leukocytes takes place particularly in the suprapapillary
region. This micromorphology explains the
“Auspitz” phenomenon.
The hyperproliferation of the epidermis now has reached its
characteristic pattern. The rete ridges are elongated and have a squared-off
appearance. Some rete ridges coalesce at the base. In some lesions,
micropustules of Kogoj and microabscesses of Munro may be seen.
Clinical features
History
Pruritus
is often the dominant symptom and, although not as severe as in atopic eczema,
is experienced by the majority of patients. Skin tightness and burning are
frequent in unstable, erythrodermic or pustular psoriasis and pain may be
experienced in areas of fissure formation, particularly in palmoplantar or
flexural disease. Shedding of scale can be a significant symptom, for instance
in scalp psoriasis, and contributes to feelings of embarrassment.
The
first manifestation of psoriasis may develop at any age and in general those
with earlier onset disease are more likely to have a family history of
psoriasis. The course of the disease including the frequency of relapses and
remissions varies greatly between individuals. Exacerbating factors should be enquired
after, and responses to previous treatments noted. It is important to ask
concerning the involvement by psoriasis of specific sites that may not be
volunteered by the patient, for instance the ano‐genital
region. A detailed medical history should be taken including symptoms of common
co‐morbidities,
particularly of articular symptoms.
Presentation
Plaque psoriasis is
the most common type of psoriasis, accounting for about 80–90% of all cases and
is characterized by a relatively
symmetric distribution of sharply demarcated erythematous papules and plaques
with micaceous scales. The degree of body surface area involvement can vary,
from limited to extensive. The scalp, elbows, knees and presacrum are sites of
predilection, as are the hands and feet. The genitalia are involved in up to
45% of patients. Plaques may persist for months to years at the same locations.
Although the course of this disease is chronic, periods of complete remission
do occur and remissions of 5 years have been reported in approximately 15% of
patients.
At any one point in time, different
variants may coexist in a particular individual, but the skin lesions all share
the same important hallmarks: erythema, thickening and scale. The
color of the plaques, a full rich red (sometimes referred to as ‘salmon pink’),
has a particular depth of hue not normally seen in eczema or lichen simplex.
This quality of color is of particular diagnostic value in lesions on the
palms, soles and scalp. In fair‐skinned individuals, the color
is less rich and almost magenta pink. In dark‐skinned
races, the quality of the color is lost. On the legs, a bluish tint is often
present, but this differs from the violaceous hue of lichen planus. The lesions
are well defined, with a sharply delineated edge. When they merge, annular and
gyrate figures may be produced. This definition is of special diagnostic value on
the scalp and penis, when other evidence of psoriasis is absent, and in the
flexures. The epidermal thickening characteristic of the psoriatic process
causes the lesions to be raised from the adjoining skin, and easily palpable.
There may be any number of lesions or only a single one. When multiple, lesions
are usually monomorphic and distributed relatively symmetrically. They vary in
diameter from one to several centimeters and are oval or irregular in shape.
Large plaques form by their coalescence and are commonly seen on the legs and
sacral region. When plaques occur across the line of joint movement, fissuring
may occur. In addition to their highly
characteristic sharp demarcation, psoriatic lesions are sometimes surrounded by
a pale blanching ring, which is referred to as Woronoff's ring. Linear
and geometric configurations may arise at the sites of trauma as an isomorphic
(Koebner) phenomenon. Erythema may persist at the site of a previously treated
plaque for many months. Post inflammatory hypo pigmentation or hyper
pigmentation is also both frequent and occasionally lentigines may persist
following clearance of a plaque.
Most psoriasis
lesions are surmounted by silvery white scales, which vary considerably in
thickness. The amount of scaling may be minimal in partially treated disease,
and in the flexures. When scaling is not evident it can often be induced by
light scratching, a useful sign in diagnostically uncertain lesions.
The classic findings of erythema, thickening and scale are
reflections of the histologic findings of elongated dilated capillaries that
are close to the skin surface, epidermal acanthosis plus cellular infiltrates,
and abnormal keratinization, respectively. If the superficial silvery white
scales are removed via curettage (grattage method), a characteristic coherence
is observed, as if one has scratched on a wax candle (“signe de la tache de
bougie”). Subsequently, a smooth, glossy red surface
membrane is seen, which will also come off as a whole. If the latter is
removed,
where the thin suprapapillary epidermis is torn off, then a wet surface is seen with characteristic pinpoint
bleeding. This finding, called Auspitz sign is the clinical reflection of
elongated vessels in the dermal papillae together with thinning of the
suprapapillary epidermis.
During exacerbations, psoriatic lesions often itch. Pinpoint
papules surrounding existing psoriatic plaques indicate that the patient is in
an unstable phase of the disease. In addition, expanding psoriatic lesions are
characterized by an active edge with a more intense erythema. Inflamed lesions
may be slightly tender. The involution of a lesion usually starts in its
center, resulting in annular psoriatic lesions.
Because
the percentage of body surface area involved does not reflect the severity of
the individual lesions with respect to erythema, induration and scaling, the
Psoriasis Area and Severity Index (PASI) was formulated. This is a single
calculated score that is based on the body surface area involved (in each of
four anatomic areas – head, upper extremities, trunk and lower extremities) and
clinical grading of lesional erythema, induration and scaling. The PASI is a
cumbersome calculation and is more commonly utilized for clinical trials than
for the routine management of patients with psoriasis.
CALCULATION OF THE PSORIASIS
AREA AND SEVERITY INDEX-PASI |
||||
Severity of psoriatic lesions |
||||
[0, none; 1, slight; 2, moderate; 3, severe;
4, very severe] |
||||
Head |
Trunk |
Upper limbs |
Lower limbs |
|
Erythema |
0 to 4 |
0 to 4 |
0 to 4 |
0 to 4 |
Induration |
0 to 4 |
0 to 4 |
0 to 4 |
0 to 4 |
Scaling |
0 to 4 |
0 to 4 |
0 to 4 |
0 to 4 |
Total score = |
Sum of the above |
Sum of the above |
Sum of the above |
Sum of the above |
Area of psoriatic involvement |
||||
[0, none; 1, <10%; 2, 10 to <30%; 3,
30 to <50%; 4, 50 to <70%; 5, 70 to <90%; 6, 90–100%] |
||||
Degree of involvement = |
0 to 6 |
0 to 6 |
0 to 6 |
0 to 6 |
Multiply × |
× |
× |
× |
× |
Correction factor for area of involvement = |
0.10 |
0.30 |
0.20 |
0.40 |
× × |
A |
B |
C |
D |
A + B + C + D = total PASI |
Psoriasis affecting
specific sites
Scalp psoriasis
The
scalp is one of the most common sites for plaque psoriasis and may be the first
and only site affected. The whole scalp may
be diffusely involved, or multiple discrete plaques of varying size may be seen,
in contrast to the less well-defined areas of
involvement in seborrheic dermatitis. At times, however, it is not possible to
distinguish seborrheic dermatitis from psoriasis, and the two disorders may
coexist. Very thick plaques can develop, especially at the occiput.
Plaques tend to be restricted to hair‐bearing areas, extending a short distance beyond the
hairline and around the ears. A morphological entity consisting of plaques of
asbestos-like scaling, firmly adherent to the scalp and for some distance to the scalp hairs has been termed
pityriasis (tinea) amiantacea. It is most common in children and young adults,
and is best regarded as a non-specific reaction pattern.
Although pityriasis amiantacea can also be seen in patients with other
scaling scalp conditions such as seborrheic
dermatitis, secondarily infected atopic dermatitis and tinea capitis, psoriasis
is the most common cause. It may be an early manifestation occurring before the
other stigmata of psoriasis. Hair loss, sometimes cicatricial, is seen in
pityriasis amiantacea. Otherwise, common scalp psoriasis is
not a frequent cause of alopecia, although it may occur.
Follicular psoriasis
Psoriasis
around hair follicle openings on
the trunk and limbs – follicular psoriasis – may occur as an isolated
phenomenon, or in association with plaque psoriasis. The lesions are smaller
than the typical lesions of guttate psoriasis and may be either grouped or
diffuse. In children it may be confused with pityriasis rubra pilaris (PRP).
Seborrhoeic psoriasis
(sebopsoriasis)
There is no reason why a genetically constituted
psoriatic should not develop seborrheic dermatitis. Plaques of thin sharply
demarcated erythema with variable scale may occur in the typical distribution
of seborrheic dermatitis such as scalp, hairline, medial eyebrows, paranasal
areas, external ears, presternal and interscapular area having features of both
diseases.
It
may occur as an isolated phenomenon, or in association with plaque
psoriasis. When it arises as an isolated
phenomenon, it is difficult to distinguish from seborrheic dermatitis, and may
represent a koebnerization of psoriasis within this entity. Involvement of the
face other than in a seborrheic distribution is uncommon in adults, but it
occurs in widespread psoriasis elsewhere on the skin.
Flexural psoriasis (inverse
psoriasis)
Flexural lesions are more common in older adults than children and are characterized by shiny, pink to red, sharply
demarcated thin plaques. Although
the lesions are themselves anhidrotic, the effect of hyperhidrosis of the
surrounding skin, maceration and friction alter the appearance of the
psoriasis, which retains its characteristic color but scaling is greatly
reduced or absent. The surface has a glazed hue and
fissuring at the depth of the fold is common. The most common sites of involvement are the
axillae, inguinal crease, umbilicus, intergluteal cleft, submammary region and retroauricular folds. Flexural areas are involved in association with
plaque psoriasis elsewhere on the skin or less frequently, lesions remain
confined to flexural sites, known as “inverse” psoriasis. Inverse psoriasis may
occur as a primary disorder or as a Koebner phenomenon to localized dermatophyte, candidal or bacterial infections or seborrheic
intertriginous dermatoses that can trigger flexural
psoriasis. Failure
to respond to antibacterial or antifungal preparations should arouse suspicion.
Involvement of the napkin area (psoriatic napkin eruption) may be the first
presentation of psoriasis in infancy.
Genital psoriasis
The
genital skin is often involved in individuals with inverse psoriasis, less
frequently in those with plaque psoriasis and may be the only manifestation of
psoriasis.
Skin
of the scrotum and penile shaft may be affected by psoriasis but the glans
penis is the most frequently affected part. In circumcised men, lesions on the
glans are similar in appearance to plaques at other sites. In the
uncircumcised, the plaques lack scale but the color and well‐defined
edge are usually distinctive.
The
most common vulval presentation is a symmetrical, erythematous, non‐scaly,
well‐demarcated
thin plaque affecting the labia majora with marked pruritus.
Non‐pustular palmoplantar
psoriasis
On
the palms and soles, psoriasis may present as typical scaly plaques on which a
fine silvery scale can be evoked by scratching; as less well‐defined
plaques resembling lichen simplex or hyperkeratotic eczema; or as a pustulosis.
It is often difficult to distinguish between psoriasis and eczema, with which
it may sometimes appear to alternate. A sharply defined edge at the wrist,
forearm or palm and absence of vesiculation are helpful. On the dorsal surface,
the knuckles frequently show a dull‐red thickening of the
skin. Elsewhere on the hands and feet, psoriasis retains its typical character.
There may be a relationship to trauma or occupational irritants.
Nail psoriasis
Nail involvement has been reported in about 40% of psoriatic
patients. The fingernails are more often affected than the toenails. This is seen in
association with all types of psoriasis of the skin, and patients with nail involvement appear to have an increased
incidence of psoriatic arthropathy.
There is no sex predilection, but patients over 40 years of age are affected
twice as often as those under 20 years. Nail disease is more likely to be
severe if psoriasis is early onset and familial.
Psoriasis may affect any part of the nail unit, including the nail matrix, nail bed and hyponychium. Nail pitting is the best known and possibly the most frequent psoriatic nail abnormality. Small parakeratotic foci in the proximal portion of the nail matrix results in loss of parakeratotic cells from the surface of the nail plate, leaving a variable number of tiny, punched-out depressions on the nail plate surface. This is a process analogous to the shedding of psoriatic skin scale. Pitting is the most frequently seen in fingernails; individual pits being uniform in size at about 1 mm diameter and sometimes arranged longitudinally. They emerge from under the cuticle and grow out with the nail. Leukonychia and loss of transparency (less common findings) are due to involvement of the mid portion of the matrix. If the whole nail matrix is involved, a whitish, crumbly, poorly adherent “nail” is seen. Psoriasis of the nail bed may cause localized separation of the nail plate. Cellular debris and serum accumulate in this space. The brown-yellow color observed through the nail plate looks like a spot of oil called “oil spot” or “oil drop” phenomenon (Salmon patches) which is highly specific for psoriasis. Splinter hemorrhages are the result of increased capillary fragility, and subungual hyperkeratosis and distal onycholysis are due to parakeratosis of the distal nail bed.
Mucosal lesions
Migratory annular erythematous lesions with hydrated white
scale (annulus migrans) have been observed in patients with acrodermatitis
continua of Hallopeau and generalized pustular psoriasis. The most common
location is the tongue, and the clinical (and histologic) appearance is similar
to geographic tongue.
Ocular lesions
Psoriasis
may affect ocular structures directly, or by associated immunological
phenomena. Direct involvement of the eyelid margins may cause blepharitis and
its consequences, which are the most frequent ocular complications of
psoriasis. A chronic non‐specific conjunctivitis and xerosis are
also common in psoriasis. Uveitis is an important immunologically mediated
complication seen in patients with more extensive psoriasis. It is more
strongly associated with psoriatic arthritis.
Clinical
variants (based on morphology or natural history)
Acute guttate psoriasis
Guttate psoriasis is more commonly seen in children and adolescents. More than 30% of psoriatic patients have their first episode before age 20; in many instances, an episode of guttate psoriasis is the first indication of the patient’s propensity for the disease. Streptococcal pharyngitis caused by group A streptococci or a viral upper respiratory tract infection may precede the eruption by 1 or 2 weeks. In over half of the patients, an elevated antistreptolysin O, anti-DNase B or streptozyme titer is found, indicating a recent streptococcal infection. Throat cultures should be taken to rule out streptococcal infection. A guttate flare may also occur in adults with plaque psoriasis.
This describes the sudden onset of a shower of bright red, drop shaped discrete small scaly papules and plaques. In the early stages, there may be little scaling and the color is not specific. The lesions are from 2 mm to 1 cm in diameter, round or slightly oval. Lesions increase in diameter with time. They are scattered more or less evenly over the body, particularly on the trunk and proximal part of the limbs, rarely on the palms and soles. It may also affect face, ears and scalp. The lesions on the face are often sparse, difficult to see and disappear quickly. Although guttate lesions are normally profuse, there are occasionally no more than half a dozen present on the body. Pruritus is variable. The diagnosis is made mainly on the nature of the scaling, the general distribution and evidence for preceding infection. Lesions usually resolve spontaneously over about 3 months; it responds more readily to treatment than does chronic plaque psoriasis. A minority of patients with acute guttate psoriasis subsequently develops plaque psoriasis. A guttate flare may also occur in adults with plaque psoriasis and is more common in patients whose psoriasis had its onset in childhood.
Unstable’ psoriasis (syn. active
psoriasis, eruptive inflammatory psoriasis)
In
contrast to the lesions in plaque psoriasis which are static for prolonged
periods, in some individuals and in some phases of the disease there is more
marked activity in the form of enlargement of plaques that can become more
intensely erythematous, and the development of many new smaller plaques.
Patients may complain of more pain or pruritus within the plaques. The Koebner
phenomenon is thought to be more frequent in this phase of the disease. The
immediate outcome is unpredictable; the lesions may return to the inactive state,
or progress to localized pustular or erythrodermic psoriasis. Recognized
precipitants include withdrawal of systemic or potent topical corticosteroids,
treatment with irritants such as tar or dithranol, acute infection,
hypocalcemia and severe emotional upset.
Erythrodermic psoriasis
Erythrodermic
psoriasis, in which most or the entire body surface is affected by psoriasis,
is uncommon, occurring in 1–2% of patients but psoriasis has been found to be
the underlying cause in about 25% of cases of erythroderma. Erythroderma in
psoriasis may be chronic, due to the gradual extension of plaque psoriasis, or
acute as part of the spectrum of ‘unstable’ psoriasis. In the chronic form, the
individual may be systemically well, the clinical characteristics of psoriasis
are retained and there are usually some areas of uninvolved skin. Mild topical treatments
are well tolerated and the overall prognosis is good.
The
acute form is often precipitated by environmental or therapeutic triggers
including systemic illness, alcoholism, antimalarials, irritating topical
treatments, ultraviolet radiation or by withdrawal of systemic corticosteroids,
ciclosporin or methotrexate. The patient may be febrile and systemically ill.
Dependent edema is common. Itching is often severe. The entire skin may be
affected and the clinical characteristics of psoriasis are often lost.
Untreated, the course is prolonged, relapses are frequent and there is an
appreciable mortality. Complications are those of acute skin failure, including
sepsis, temperature dysregulation (hypothermia or hyperthermia),
hypoalbuminemia, anemia, and hypervolemia and high output cardiac failure.
Metabolic complications
of erythroderma
Persistent, universal inflammation of the skin may have
important consequences for thermoregulation, hemodynamics, intestinal
absorption, protein, water and other metabolism. Under normal environmental
conditions, radiant and convective heat loss from the body surface is
increased, occasionally leading to dangerous hypothermia. An increase in
metabolic activity provides compensatory increases in body heat production, but
at the expense of tissue catabolism and muscle wasting if prolonged. At the
same time, the psoriatic or erythrodermic skin is hypohidrotic or anhidrotic,
because of intraepidermal occlusion of the sweat duct. There is attendant risk
of hyperthermia in very high ambient temperatures. Skin blood flow, blood volume
and cardiac output may all be increased, and if these changes persist, they may
lead to failure of a cardiovascular system already compromised by hypertension,
myocardial or valvular heart disease, or anemia (particularly in old people). A
healthy cardiovascular system will tolerate the increased metabolic demand for
years. Malabsorption (dermatogenic enteropathy) may occur, reverting as the
psoriasis remits. The principal loss in the profuse scaling of exfoliative
psoriasis is of protein (keratin) but some iron is also lost. In fulminating
psoriasis, further protein loss may be attributable to enteropathy, as well as
leakage from the circulation into the skin. Eventually, hypoalbuminemia may
contribute to the edema caused by the skin inflammation itself, or cardiac
failure. Mild anemia is prone to develop, because of a combination of iron loss
and possibly impaired absorption and utilization of iron. Serum and red cell
folate and serum B12 may also be low. The barrier efficiency of the skin in
psoriatic erythroderma is impaired, the chief effect being increased
transepidermal water loss. The urine output tends to drop and, if water intake
is inadequate for any reason, dehydration results.
Although there are many causes of
erythroderma, clues to the diagnosis of psoriatic erythroderma include previous
plaques in classic locations, characteristic nail changes, and central facial
sparing.
Atypical forms of psoriasis
Rupioid,
elephantine and ostraceous psoriasis are plaques associated with gross
hyperkeratosis. Rupioid psoriasis refers to limpet‐like
cone‐shaped
lesions. The term elephantine psoriasis is very persistent, thickly scaling,
large plaques that occur on the back, limbs, hips or elsewhere. Ostraceous psoriasis
refers to a ring‐like hyperkeratotic lesion with a
concave surface, resembling an oyster shell.
Psoriasis
in childhood and old age
Psoriasis in children
Psoriasis in children is more common in girls than boys
and it significantly associated with several co‐morbidities
including obesity and diabetes.
All
of the clinical variants of psoriasis described in adults are recognized in
childhood. The napkin area is frequently the first site affected; under the age
of 2 years, napkin psoriasis with or without disseminated lesions is the most
frequent form, presenting with well‐defined erythema
devoid of scale.
Guttate
psoriasis is more frequent in children than adults, particularly under the age
of 12 years. In older children, plaque psoriasis is the most frequent
presentation, and the face and ano‐genital sites appear
to be affected more frequently than in adults. The disease often first appears
in the scalp, where it may present as pityriasis amiantacea. Other flexural
forms also occur. The disease may mimic chronic blepharitis or perleche,
usually unilaterally, with a small plaque of psoriasis on one eyelid extending
to the lid margin, or on the cheek at the angle of the mouth. An indolent
pustular acrodermatitis, sometimes of only one digit, usually eventually proves
to be psoriatic. More extensive, chronic lesions of the hands and feet may
occur with persistent dryness, hyperkeratosis and fissuring. Pitting of the
fingernails may be the only manifestation for months or even years. Follicular
psoriasis occurs on the extensor prominences of elbows and knees. Erythrodermic,
pustular psoriasis and psoriatic arthritis are rare in childhood.
Psoriasis in elderly
In
older age groups, psoriasis that starts for the first time after the age of 65
years tends to be less extensive than early‐onset disease. There
is less often a family history of psoriasis. Plaque psoriasis involving the
scalp is the commonest phenotype. Inverse psoriasis and erythrodermic psoriasis
may be more common than in early‐onset disease whereas
guttate and generalized pustular psoriasis are rare.
HIV‐induced or exacerbated
psoriasis
The
association between severe psoriasis, psoriatic arthropathy and HIV infection
is well recognized. Psoriasis may be the first presentation of HIV infection,
as may the deterioration in previously stable disease. Plaque psoriasis is the
most frequent phenotype, with a predilection for the scalp and palmoplantar
skin. Sebopsoriasis, rupioid psoriasis and psoriatic erythroderma are also
common, as are mixed patterns. Psoriasis tends to be more prevalent in the
later stages of HIV‐related immune dysfunction, but may
occur earlier. The mechanism of worsening of psoriasis in HIV infection is
unclear and represents a paradox, given that helper T cells are the major
target of HIV. Suggested explanations include HIV‐induced
reduction in regulatory T cells, an increased number of memory CD8+ T cells,
effects of HIV on dendritic cell populations, HIV proteins acting as super
antigens or shared genetic variants between psoriasis and HIV responder status.
One suggestion is that HIV affects peripheral blood helper T cells and affects
to a lesser degree cutaneous resident memory T cells which are pathogenic in
psoriasis. Psoriasis tends to improve with a reduced viral load, especially on
treatment with highly active antiretroviral therapy.
Differential diagnosis
In seborrheic dermatitis, the
lesions are lighter in color, less well defined and covered with a dull or
branny scale. Eczema at times develops a psoriasiform appearance, especially on
the legs. Hyperkeratotic eczema of the palms is a common cause of misdiagnosis.
Color, scratch‐evoked
scaling and well‐defined
margins are suggestive of psoriasis, and nail changes may be diagnostic. Lichen
planus could give rise to difficulty when the two diseases alternate or
coexist, especially when present as hypertrophic lesions on the legs, as penile
lesions and on the palms. The violaceous color, Wickham's striae and the
presence of oral changes are usually decisive. Lichen simplex can resemble
psoriasis closely, particularly on the scalp and near the elbow. The
intensified skin markings, rather ill‐defined edge and the marked itching are characteristic, and
the point of the elbow tends to be avoided. Candidiasis shows a glistening deep
red color resembling psoriasis, particularly in the flexures, but scaling tends
to be confined to the edge, and small satellite pustules and papules are
usually evident outside the main area. Tinea cruris has a well‐defined, often polycyclic edge, but Trichophyton
rubrum infections, especially of the palm, may cause difficulty. If
corticosteroids have been applied, scaling may be absent and the diagnosis must
be made by microscopy and culture of skin scrapings.
Complications and co‐morbidities
Patients
with psoriasis experience increased morbidity and mortality from a range of
systemic diseases, affecting most major organ systems.
Immune‐mediated inflammatory diseases
Psoriatic
arthritis is the most frequent inflammatory disease associated with psoriasis. Inflammatory
bowel disease (IBD) such as crohn disease and
ulcerative colitis, and psoriasis share an association with sacroiliitis and
HLA-B27 positivity. The risks of other immune‐mediated
diseases are autoimmune thyroid disease and type 1 diabetes.
Psychological
distress
Psoriasis
is associated with significant psychological distress. Excessive alcohol
consumption is found more commonly in men with severe psoriasis, and could be a
consequence of stress caused by severe skin disease.
Cancer
The
incidence of non‐melanoma skin cancer is more common in
psoriasis patients compared with the general population. It has been shown that psoriatic patients who have had
>200 PUVA treatments are at increased risk for the development of skin
cancers, especially squamous cell carcinomas (SCCs) and basal cell carcinoma, and this may be
compounded by immunosuppressive treatments, including ciclosporin,
methotrexate, TNF‐α inhibitors and possibly high‐dose
UVB.
Association with internal diseases (including
comorbidities)
In
patients with moderate to severe psoriasis, there is an increased relative
risk for atherosclerotic cardiovascular disease and metabolic syndrome. |
Cardiovascular disease
Cardiovascular diseases, e.g.
myocardial infarctions, peripheral arterial disease, cerebrovascular accidents,
are more common in patients with severe psoriasis. The latter is associated
with a threefold increased risk for myocardial infarction and a 3.5–4.4-year
reduction in life expectancy. This is largely due to an increased risk for
having metabolic syndrome. Patients with severe psoriasis die at a
younger age than unaffected people and cardiovascular disease accounts for the
majority of this excess mortality. In
patients with psoriasis, serum levels of C-reactive protein (CRP) have been
reported to be elevated (as compared to healthy controls), and elevated CRP
levels represent a risk factor for the development of cardiovascular disease.
It has also been shown that TNF-α and IL-6 can target adipocytes and induce
dyslipidemia. Recently, evidence has been accumulating that treatment with
methotrexate and/or TNF-α inhibitors may reduce the risk of atherosclerotic
cardiovascular disease. Patients with psoriasis may also
be at increased risk for the development of venous thromboembolism.
The
increased vascular risk also relates to behaviors (such as cigarette smoking,
which is more frequent amongst those with psoriasis).
Metabolic syndrome
The
metabolic syndrome (truncal obesity, hyperlipidemia, hypertension and insulin
resistance) is significantly elevated in patients with psoriasis mainly in
those having more extensive disease. The
strongest of these associations is with obesity, which can be seen in childhood
psoriasis, and tends to predate the onset of psoriasis. Obesity in adults
appears to be a risk factor for the development of psoriasis. Patients who are
obese (BMI >30) have more severe psoriasis than lean patients. Obesity may
decrease treatment responsiveness and is one of the risk factors for the
development of premature cardiovascular disease. Psoriasis severity is
associated with insulin resistance and medications that target insulin
resistance may be effective in psoriasis and also weight loss in obese patients
with psoriasis improve psoriasis severity.
Hepatobiliary disease
Death rates due to
liver disease have been reported to be significantly elevated in patients with
severe psoriasis. Non-alcoholic steatohepatitis, characterized by fatty infiltration, periportal
inflammation and focal necrosis, is more commonly observed in patients with
psoriasis. In patients with psoriasis, non-alcoholic fatty liver disease is correlated
with the presence of obesity, hyperlipidemia, the metabolic syndrome, an AST: ALT
ratio >1, and psoriatic arthritis. Chronic
administration of methotrexate is associated with a significant risk for
hepatic damage in patients with psoriasis, whereas similar methotrexate dosages
in patients with rheumatoid arthritis do not pose such hepatotoxic potential,
possibly because of genetic predisposition, increased alcohol consumption by
psoriasis patients, and a higher incidence of non-alcoholic fatty liver
disease.
Neutrophilic cholangitis is a recognized cause of liver dysfunction in patients
with generalized pustular psoriasis and occasionally in plaque psoriasis.
Others
There
are rare reports of acute respiratory distress syndrome complicating erythrodermic
or generalized pustular psoriasis. Psoriasis is also associated with an
increased risk of hyperuricemia and gout.
Disease course and prognosis
This remains as unpredictable as it was 150 years ago.
‘Psoriasis is at all times and under all forms a very troublesome and, often,
an intractable disease, but it is rarely dangerous to life. It is impossible
to say, in any particular case, how long the disease will last, whether a
relapse will occur, or for what period of time the patient will remain free
from psoriasis. Relapse is the rule, however completely the lesions have been
treated and by whatever method. Guttate attacks carry a better prognosis and
have longer remissions after treatment. At the other extreme, erythrodermic and
pustular forms carry an appreciable mortality and arthropathic forms a
considerable morbidity. Early onset and a family history of the disease appear
to worsen the prognosis. It is now known that severe plaque psoriasis will also
appreciably shorten life expectancy by an average of 4–6 years. There is little
doubt that even severe cases can be maintained in prolonged remission by the
use of systemic therapies, including the biologics.
Investigations
The
diagnosis of psoriasis is usually clinical. A skin biopsy of lesional skin may
occasionally be helpful in atypical cases.
Management
The
diagnosis of psoriasis is usually based on clinical features. In the few cases in
which clinical history and examination are not diagnostic, biopsy is indicated
to establish the correct diagnosis. The majority of psoriasis cases fall into
three major categories: guttate, erythrodermic/pustular, and chronic plaque, of
which the latter is by far the most common. Guttate psoriasis is often a self-limited
disease with spontaneous resolution within 6-12 weeks. In mild cases of guttate
psoriasis, treatment may not be needed, but with widespread disease, UVB
phototherapy in association with topical therapy is very effective. Erythrodermic/pustular
psoriasis is often associated with systemic symptoms and necessitates treatment
with fast-acting systemic medications. The most commonly used drug for
erythrodermic and pustular psoriasis is acetretin. In occasional cases of
pustular psoriasis, systemic steroids may be indicated (asterisk). Dotted arrows indicate that guttate, erythrodermic and pustular forms often evolve
into chronic plaque psoriasis. Therapeutic choices for chronic plaque psoriasis
are typically based on the extent of the disease. Among the main treatment
regimens (topical treatment, phototherapy, day treatment centers, and systemic
treatments), first and second line modalities are indicated by the solid
and dashes lines respectively.
Individuals with conditions that limit their activities, including painful
palmoplanter involvement and psoriatic arthritis, may require more potent treatments
irrespective of the extent of affected body surface area. Likewise, psychological
issues and impact on quality of life should be taken into consideration. Within
each treatment regimen, first-line and second-line choices are grouped.
Cyclosporine A is not considered a first-line long-term systemic treatment
because of its side effects, but short term treatment can be helpful for
induction of remission. If patients have incomplete response to or are unable
to tolerate individual first-line systemic medications, combination regimens,
rotational treatments, or use of biological therapies should be considered.
General
Treatment of
psoriasis should be individualized.
Care of the psoriatic patient needs to focus not only on the skin, but also on
the co-morbidities that exist or might develop. To date, no treatment has been
shown to cure this disease and alters its natural history, so counseling of the patient and family members regarding
its natural history and management strategies is recommended.
Some treatments are better suited for rapid clearing; others are better suited to be maintenance treatment. The optimum management involves the sequential use of therapeutic agents involving three steps, namely, the clearing phase (induction of remission), the transitional phase, and the maintenance phase (maintenance of remission). In inducing remission, 75% reduction in PASI from baseline (PASI 75) has been widely adopted as a minimum response criterion for severe plaque psoriasis in clinical trials. With the availability of higher efficacy treatments, this standard is rising to PASI 90.
Determining degree of inflammation
The most
common form of psoriasis is the localized chronic plaque disease involving the
skin and scalp. It must be determined whether the plaque is inflamed before
instituting therapy. Red, sore plaques can be irritated by tar, calcipotriol,
and anthralin. Irritation can induce further activity. Inflammation should be
suppressed with topical steroids and/or antibiotics before initiation of other
treatments.
Determining the end of treatment
The plaque
is effectively treated when induration has disappeared. Residual erythema, hypo
pigmentation, or brown hyper pigmentation is common when the plaque clears;
patients frequently mistake the residual color for disease and continue
treatment. If the plaque cannot be felt by drawing the finger over the skin
surface, treatment may be stopped.
Duration of remission
Among
topical monotherapy, tazarotene induce longer remissions than calcipotriene and
corticosteroids; among systemic agents, longer remissions occur with acitretin
than cyclosporine or methotrexate, but compared with the remission rate of
phototherapeutic modalities, the remission rates are much less.
Topical
treatment
Topical therapies are the mainstay of treatment for psoriasis of limited extent. The main groups of topical therapies are emollients, corticosteroids, vitamin D and its analogues, coal tar and retinoids
Topical
corticosteroids
Maximal quantities: 50 g/week of a super potent
corticosteroid; 100 g/week of a potent corticosteroid.
INDICATIONS AND CONTRAINDICATIONS FOR TOPICAL CORTICOSTEROIDS |
Indications |
·
Mild
to moderate psoriasis: first-line treatment as monotherapy or in combination ·
Severe
psoriasis: often in combination with a vitamin D3 analogue,
a topical retinoid, anthralin or tar; also as an adjunct to systemic therapy
or photo(chemo)therapy ·
Monotherapy
for flexural and facial psoriasis (usually mild strength) ·
Recalcitrant
plaques often require occlusion (plastic, hydrocolloid) |
Contraindications |
·
Bacterial,
viral, and mycotic infections ·
Atrophy
of the skin ·
Allergic
contact dermatitis due to corticosteroids or constituents of the formulation ·
Pregnancy
or lactation* |
* Can consider limited use of mild- to
moderate-strength corticosteroids.
Corticosteroids are anti-inflammatory,
ant proliferative, immunosuppressive, and vasoconstrictive. Lower potency
corticosteroids are used for limited periods of time on the face, neck,
flexures, genitalia, areas with thin skin, and in infants and are also used in
unstable, erythrodermic and generalized pustular psoriasis. In other areas and
in adults, mid- or high-potency agents are used. Patients with thick, chronic
plaques such as the palms and soles require treatment with the highest potency
corticosteroids, such as clobetasol. For clobetasol and halobetasol, maximal
weekly use should be 50 gm or less. Newer formulations of corticosteroids,
particularly foams, are easier to apply than traditional creams or ointments
and can be used for scalp, truncal or limb psoriasis. Once-daily application has been shown to be as effective as
twice-daily application, and long-term remissions may be maintained by
applications on alternate days.
Topical steroids give
fast but temporary relief. They are most useful for reducing inflammation and
controlling itching. Initially, when the patient is introduced to topical steroids,
the results are most gratifying. However, tachyphylaxis, or tolerance, occurs,
and the medication becomes less effective with continued use. Patients remember
the initial response and continue topical steroids in anticipation of continued
effectiveness. Long-term use of topical steroids results in atrophy and
telangiectasia. Topical steroids are useful for treating inflamed and
intertriginous plaques.
A group I
through V steroid applied one to four times a day in a cream or ointment base
is required for best results. Group V topical steroids applied once or twice a
day should be used in the intertriginous areas and on the face. Some plaques
resolve completely, but most remain only partially reduced with continued
application. Continual application for more than 3 weeks should be discouraged.
Remissions are usually brief and the plaques may return shortly after treatment
is terminated. Topical steroid creams applied under an occlusive plastic
dressing promote more rapid clearing, but remissions are not extended.
At least 80% of patients treated with high-potency topical
corticosteroids experience clearance. In fact, the maximum improvement is
usually achieved within 2 weeks. With maintenance therapy consisting of 12
weeks of intermittent applications of betamethasone dipropionate ointment
(restricted to weekends), 74% of patients remained in remission. Unfortunately,
no efficacy data are available on prolonged treatment for more than 3 months.
As tachyphylaxis and/or rebound can occur fairly rapidly, i.e. within a few
days to weeks, intermittent treatment schedules (e.g. once every 2 or 3 days or
on weekends) are advised for more prolonged treatment courses. Combination
topical therapy can take advantage of the rapid effect of topical
corticosteroids as well as the prolonged benefits of topical vitamin D3
analogues.
Potent topical corticosteroids tend not to induce a lasting remission, unlike
tar which has the potential to do so.
Intralesional steroids
Intralesional
injection of small resistant plaques with triamcinolone acetonide (Kenalog 5 to
10 mg/ml) almost invariably clears the lesion, for instance on the backs of the
hands and knuckles and accords long-term remission. Atrophy may occur with the
10 mg/ml concentration.
Vitamin
D3 analogues
Vitamin D3 analogues should be avoided or limited in
patients with abnormalities in bone or calcium metabolism (e.g. sarcoidosis) or
renal insufficiency; they are pregnancy category C. If used in conjunction with
phototherapy, need to apply after UV irradiation or at least several hours
prior, because they may reduce UV penetration into the skin.
Calcipotriene/calcipotriol |
Calcitriol |
Tacalcitol |
|
Concentration |
50 mcg/g |
3 mcg/g |
2 mcg/g; 4 mcg/g |
Formulations |
Ointment, cream,
lotion, solution, foam |
Ointment |
Ointment, lotion |
Frequency |
BID |
BID |
BID; daily |
Maximum weekly amount* |
100 g |
200 g |
70 g; 70 g† |
Chemical structure |
26,27-cyclo-vitamin D3 |
1α,25-dihydroxy
vitamin D (active natural vitamin D3) |
1α,24-dihydroxy
vitamin D3 |
Inactivating enzymes |
22,23-reductase;
24-oxido-reductase |
24-hydroxylase |
25-hydroxylase |
* To avoid hypercalcemia.
† Maximum weekly amount for 8 weeks of
treatment.
When the epidermis is hyper proliferative, vitamin D3
inhibits epidermal proliferation, and it induces normal differentiation by binding
with vitamin D receptor; it also acts on the immunocytes, shifting the Th1
cytokine profile of plaques towards Th2. Vitamin D3 analogues also inhibits several neutrophil functions. Due to their therapeutic efficacy and limited toxicity,
calcipotriene (calcipotriol) and other vitamin D3 analogues have
become a first-line therapy for psoriasis.
The
naturally occurring active metabolite of vitamin D3, calcitriol, and
synthetic analogues calcipotriol and tacalcitol are all effective when applied
topically in psoriasis. The most widely prescribed analogue in current use,
calcipotriol (50 μg/g) ointment, has been reported to be at least as effective
as 0.1% betamethasone valerate ointment in plaque psoriasis. Vitamin D and its
analogues all have the potential to affect systemic calcium homeostasis with
hypercalciuria and hypercalcemia. It is prudent to restrict the amount of
calcipotriol (50 μg/g) ointment to less than 100 g per week, and to monitor
serum and urinary calcium levels should these doses be exceeded. Hypercalcemia
is reported with excessive quantities of calcipotriene applied over large
surface areas.
Calcipotriene
monotherapy has been shown to result in a ~60% reduction of PASI after 8 weeks
of treatment, but practical use in psoriasis patients usually involves
combination therapy with topical corticosteroids is
commonly employed, both during the clearing phase as well as intermittently
during long-term treatment. Calcipotriene is not as effective as group I
corticosteroids, but regimens using calcipotriene and group I corticosteroids
are superior over either agent alone. Most patients now use the following
regimen: Calcipotriene is applied in the morning and a group I corticosteroid
is applied in the evening for 2 weeks. Then a maintenance regimen is begun
using group I corticosteroids twice daily on weekends and calcipotriene twice
daily on weekdays. Application of
calcipotriene twice a day is much more effective than once a day application. Based
on this observation, a stable ointment formulation of calcipotriol 50 μg/g and
betamethasone dipropionate 0.5 mg/g has been licensed for therapy. This
combination shows superior efficacy to either calcipotriol or betamethasone
alone with better clearance and faster onset of action. When calcipotriene and betamethasone dipropionate are combined, a
~70% reduction in PASI has been observed (ointment formulation) as has
clearing/minimal disease in ~70% of patients with scalp psoriasis (gel
formulation). Calcipotriene treatment can produce a mild irritant contact
dermatitis at the site of application. The face and intertriginous areas are
prone to this side effect.
They are
effective and safe and well tolerated for the short- and long-term treatment of
psoriasis. Tachyphylaxis (tolerance) does not occur with calcipotriene.
Calcipotriene solution is also available for scalp psoriasis.
Calcitriol
is a new ointment formulation. It has similar properties to calcipotriene used
in the same manner. It is limited to 200 gm/week and is a pregnancy category C
drug. Use with caution in patients receiving medications known to increase
serum calcium levels, such as thiazide diuretics. Caution should also be
exercised in patients receiving calcium supplements or high doses of vitamin D.
Transient local skin irritation may occur, but calcitriol 3 μg/g ointment is
better tolerated than calcipotriol ointment for treatment of the face and
flexures.
Tacalcitol
4 μg/g ointment applied once daily is effective for the treatment of plaque
psoriasis but is probably less effective than calcipotriol 50 μg/g ointments.
Calcipotriol
and tacalcitol have been combined with other therapies. Calcipotriol ointment
enhances the efficacy of PUVA and UVB phototherapy. As UVA partly inactivates
calcipotriol and UVB is absorbed by calcipotriol, it is recommended that
calcipotriol is not applied immediately prior to phototherapy exposures.
Calcipotriol 50 μg/g ointment used in combination with methotrexate enables
lower cumulative doses of methotrexate to be used.
Topical
calcineurin inhibitors
The
topical calcineurin inhibitors tacrolimus and pimecrolimus were initially
developed for the treatment of atopic eczema in childhood. They are useful for
thinner skin areas such as the face, neck, flexures and genitalia and may be
effective under occlusion for psoriatic plaques at other sites. Their advantage
is that unlike topical corticosteroids they are selective, unlikely to be
absorbed systemically and do not produce skin atrophy making them more suitable
for long‐term
use. The most common side effect is burning and itching that reduces with
ongoing usage and can be reduced by not applying immediately after bathing.
This side effect is worse with tacrolimus. Both are pregnancy category C, are
found in human milk, and are not recommended for nursing mothers.
Topical retinoids
All-trans-retinoic acid and 13-cis-retinoic acid,
although effective in the treatment of acne, are not effective for psoriasis.
However, topical tazarotene, an acetylene retinoid that selectively binds
retinoic acid receptor (RAR)-β and RAR-γ, can be used to treat psoriasis. Tazarotene has been
shown to decrease epidermal proliferation and it inhibits psoriasis-associated
differentiation (e.g. transglutaminase expression, K16 expression). It is available as cream formulations and is applied once or twice daily.
In view
of its modest efficacy as monotherapy, it is usually prescribed as a
second-line therapy. Irritation of the skin with burning, pruritus, and
erythema can limit the use of tazarotene. For this reason, combination therapy
with topical corticosteroids is useful. The maximal area that can be treated
with tazarotene is 10–20% of the body surface, and safety data are available for
up to 1 year of treatment.
INDICATIONS AND CONTRAINDICATIONS FOR TOPICAL TAZAROTENE |
Indications |
·
Mild to moderate psoriasis: second-line
treatment as monotherapy or in combination |
Contraindications |
·
Unstable plaque psoriasis in a phase of progression ·
Erythrodermic psoriasis ·
Allergic contact dermatitis to tazarotene
or constituents of the formulation ·
Pregnancy or lactation |
Additional topical treatments
If the psoriatic plaques have thick scale, this needs to be
reduced to enhance penetration of topical medications and ultraviolet (UV)
light. Options include salt-water baths, topical salicylic acid and oral
retinoids. Salicylic acid 5–10% has a substantial keratolytic effect and, in
the case of scalp psoriasis, salicylic acid can be formulated in an oil or
ointment base. Application of salicylic acid to localized areas can be done
daily, but, for more widespread areas, two to three times per week is
preferred. This is to prevent systemic intoxication, especially in infants or
those with reduced renal function.
Coal tar
Coal
tar has a range of anti-inflammatory actions and is effective as an
antipruritic. Coal tar suppresses DNA synthesis.
Although crude coal tar may be the most effective tar available for the
treatment of psoriasis, a distilled product, liquor carbonis detergens (LCD),
is also used. In view of its mutagenic potential, tar is contraindicated in
pregnant or lactating women.
The Goeckerman regimen consists
of the combination of crude coal tar along with ultraviolet light. It is very
effective treatment for patients with severe psoriasis. Many formulations of
coal tar are available over-the-counter.
Coal tar formulations are
often poorly tolerated because of staining of clothes and tar odor. Newer
preparations are more cosmetically acceptable. They may be applied daily for
extended periods of time. The response is unpredictable but many patients are
gratified with this safe, inexpensive treatment.
Nonmedicated
topical moisturizers
Nonmedicated topical
moisturizers may be effective treatment. They are applied one to three times
daily. Patients who are not satisfied with irritating, expensive prescription
topical medications often turn to use of just topical moisturizers with a
gratifying effect.
Treatment of scalp psoriasis
The scalp is difficult to treat
because hair interferes with the application of medicine and shields the skin
from ultraviolet light. Symptoms of tenderness and itching vary considerably.
The goal is to provide symptomatic and cosmetic relief. It is unnecessary and
impractical to attempt to keep the scalp constantly clear.
Removing scale
Scale must be removed first to
facilitate penetration of medicine. Superficial scale can be removed with
shampoos that contain tar and salicylic acid. Thicker scale is removed by
applying 10% liquor carbonis detergens (LCD) in Nivea oil to the scalp and
washing 6 to 8 hours later with a shampoo. Combing during the shampoo helps
dislodge scale.
LCD (10%), a tar extract of crude
oil tar, is mixed with Nivea oil by the pharmacist. The unpleasant mixture is
liberally massaged into the scalp at bedtime and washed out each morning. Warming
the mixture before application enhances scale penetration. A shower cap
protects pillows and also encourages scale penetration. An impressive amount of
scale is removed in the first few days. Nightly applications are continued
until the scalp is acceptably clear.
Mild-to-moderate scalp involvement
When used at least every other
day, tar shampoos may be effective in controlling moderate scaling. Few drops of
corticosteroid solutions can cover a wide area. Steroid gels (e.g. clobetasol
gel), which have a keratolytic base and penetrate hair, are effective for localized
plaques. Fluocinolone acetonide 0.01% oil (peanut oil) is an effective topical
steroid that can be applied to the entire scalp and occluded with a shower cap.
The scalp is dampened before application. The oil base penetrates and loosens
scale. Treatment is repeated each night for 1 to 3 weeks until itching and
erythema are controlled.
Clobetasol foam is available in
50-gm container. The foam becomes a liquid upon contact with the skin. This
formulation is effective and pleasant, and easily penetrates through hair.
Temporary stinging may occur during application.
Small plaques are effectively
treated with intralesional steroid injections of triamcinolone acetonide
(Kenalog 10 mg/ml). Remissions following use of intralesional steroids are much
longer than those following topical steroids.
Ketoconazole cream is sometimes
useful. Oral ketoconazole (400 mg daily) may be effective in some cases but the
possibility of drug toxicity limits its usefulness.
Treatment of diffuse and thick scalp psoriasis
Calcipotriene 0.005% and
betamethasone dipropionate 0.064% lotion is a topical suspension for the
treatment of moderate-to-severe psoriasis of the scalp in adults. Apply the
suspension to affected areas once daily for 2 weeks or until cleared. Treatment
may be continued for up to 8 weeks. The maximum weekly dose should not exceed
100 gm. Patients should shake the bottle before using the product.
For patients
who fail topical measures, systemic immunosuppressive agents (eg. methotrexate,
cyclosporine) or biological agents may be administered.
Treatment of
inverse/intertriginous psoriasis and genital psoriasis
Inverse psoriasis can involve the
axillae; inframammary areas; abdominal, inguinal, and gluteal folds; groin;
genitalia; perineum; and perirectal area. It is erythematous, less indurated,
and well demarcated thin plaques with minimal scale. Genital psoriasis can be
distressing. Penetration of medications is facilitated with moist opposing skin
surfaces. The risk of atrophy by potent topical corticosteroids is increased.
These areas respond to weaker topical steroids and calcineurin inhibitors
(topical tacrolimus and topical pimecrolimus). Inflammation in these areas is
often misinterpreted as a yeast infection.
Phototherapy
Photo (chemo) therapy
Photo (chemo) therapy represents
a mainstay in the treatment of moderate to severe psoriasis. Phototherapy with
narrowband ultraviolet B (UVB) and photo-chemotherapy with ultraviolet A (UVA)
following ingestion of or topical application of a psoralen are classic
treatment options. Monochromatic 308 nm light from an excimer laser or
equivalent source can be used to target individual psoriatic plaques.
Narrow‐band UVB
Phototherapy
with narrow‐band
UVB is the recommended first line therapy for most patients with moderate to
severe psoriasis. NB‐UVB has been combined with numerous
therapies with apparent success, including methotrexate, acitretin and
biological therapies. NB‐UVB should be avoided in patients who
are taking ciclosporin due to concerns regarding accelerated cutaneous
carcinogenesis.
NB‐UVB
treatment should be given three times weekly. Thrice‐weekly
NB‐UVB
has been shown to clear psoriasis more quickly than twice‐weekly
NB‐UVB.
NB‐UVB
can cause erythema and pruritus in the acute setting and rarely blisters can
develop in the resolving plaques. NB‐UVB may be less
carcinogenic than PUVA, leading some authors to suggest that it can be used in
patients who have reached the maximum number of lifetime PUVA treatments. Due
to concerns regarding increased risk of SCC, the scrotum and genital skin
should be protected from irradiation.
NB‐UVB
causes cutaneous immunomodulation by inducing apoptosis of T cells and reduces
epidermal hyper proliferation.
Narrow‐band
UVB is a highly effective, remission‐inducing therapy in
psoriasis with a long safety record. It continues to play a significant role in
psoriasis management.
PUVA photo
chemotherapy
PUVA
is the acronym used for the combination of psoralens and long‐wave
ultraviolet radiation (UVA 320–400 nm). Psoralens bind to DNA and when
activated by UVA cause permanent DNA damage with resulting cell death. This
reduces keratinocyte hyper proliferation, decreases antigen‐presenting
cells in the dermis, reduces angiogenesis, and causes T‐cell
apoptosis with associated clearance and remission of psoriasis. The psoralens
that are commonly used are 8‐methoxypsoralen (8‐MOP),
5‐methoxypsoralen
(5‐MOP)
and trioxypsoralen (T‐MOP). Psoralens are used either
topically or orally in combination with UVA.
Oral
PUVA therapy is given in dermatology center according to dosing protocols. 8‐MOP
is usually given at a dose of 0.6 mg/kg 2 h before UVA therapy. PUVA is usually
given as a twice‐weekly treatment and the average number
of treatments to clearance is 18. Twice‐weekly PUVA is as
effective as three times weekly PUVA. The scrotum and genital skin are more
sensitive to the development of PUVA‐induced skin cancers
and should be protected from irradiation.
This
modality is highly efficacious in treating psoriasis with reported clearance
rates of over 90%. The average duration of remission (the time taken for 50% of
a patient's psoriasis to return) is approximately 6 months.
PUVA
can cause both acute and chronic side effects; these include erythema, maximal
at 72–96 h, and blistering that is usually dose and skin photo type dependent.
PUVA may also cause neurogenic pruritus (PUVA itch), commoner in fairer skin
types and usually dose related. Patients describe a crawling under the skin
that affects both psoriasis and normal looking skin. If treatment continues
PUVA itch usually persists. PUVA is associated with an increased incidence of
non‐melanoma
skin cancer. This is true for both basal cell carcinoma and SCC.
Immunosuppression with ciclosporin also appears to increase this risk. The
relative risk of cutaneous SCC starts to increase after 250 treatments in a
lifetime. The British Photo dermatology Group therefore recommends that a
patient should not receive more than 200 treatments of PUVA. PUVA may be
combined effectively with oral retinoids, so‐called re‐PUVA.
This allows an overall dose reduction in UVA dose but remission times are
typically shorter.
PUVA
has been associated with cataract formation, so it is recommended that patients
wear UVA protecting plastic‐lensed glasses on treatment
days. Psoralens are teratogenic and patients should be advised about this risk
even in patients undergoing topical or bath PUVA.
PUVA
remains one of the mainstays of treatments for moderate to severe psoriasis but
its use is declining, and has been largely replaced with NB‐UVB.
Systemic
therapy
In
general, systemic treatment is used for moderate-to-severe psoriasis, variably
defined as patients with 10 % or more involvement of body surface area that is
not responsive to topical therapy or phototherapy. The main systemic treatments
in current clinical practice are methotrexate, cyclosporine and acitretin.
METHOTREXATE
Methotrexate is a folate antagonist that inhibits DNA
synthesis by dihydrofolate reductase inhibition. Although initially thought to
inhibit keratinocyte hyper proliferation, it appears to inhibit lymphocyte proliferation
more profoundly (circulating and cutaneous) and is a likely explanation for its
antipsoriatic activity.
MTX is a first-line systemic therapy for psoriasis as it is highly
efficacious for severe disease and all clinical variants of psoriasis. It is also used in localized severe psoriasis,
generalized pustular psoriasis and erythroderma, although it’s relatively slow
onset of action makes it less suitable for rapid control of severe disease. In chronic plaque psoriasis, initial improvement is observed
between 1 and 7 weeks and maximum improvement can be expected after 8–12 weeks
of treatment. Depending on the dosage schedule, the percentage of patients
reaching PASI 75, i.e. a 75% reduction in the PASI score, ranges from 24% (low
starting dose) to 60% (high starting dose) after 12–16 weeks of treatment.
MTX
is administered weekly, usually as a single oral dose (but occasionally
intramuscularly or subcutaneously) and less often every 12 hours for three
doses per week; the maximum weekly dosage is usually 25 mg. Lower doses of
methotrexate may be effective, particularly in the elderly, which may relate to
reduced renal excretion in this age group. Potential side effects restrict its
use to moderate to severe disease resistant to topical treatments and photo
(chemo) therapy and/or situations in which these are contraindicated. The use
of folic acid supplementation is needed during MTX administration as it reduces side effects, including gastrointestinal.
Methotrexate
therapy for psoriasis can cause a myriad of side effects including nausea, bone
marrow suppression, mucositis and hepatotoxicity. The modern use of low‐dose
once‐weekly
methotrexate for psoriasis is associated with hepatic fibrosis in a minority of
patients. Methotrexate hepatotoxicity nonetheless appears to be commoner in
psoriasis patients than in those with rheumatoid arthritis. This may be due to
increased risk factors for liver injury amongst psoriasis patients such as
obesity, alcohol abuse and diabetes and the fact that higher therapeutic doses
are generally used when treating psoriasis compared to rheumatoid arthritis.
Standard liver function tests are insufficient to monitor patients for the
development of hepatic fibrosis. In a significant minority of patients liver
fibrosis can occur with normal liver enzymes. In the past, liver biopsies have
been recommended in order to detect occult fibrosis but the use of the serum
aminoterminal peptide of pro‐collagen III (PIIINP) as a
biomarker of fibrosis has reduced the need for routine liver biopsies in these
patients. Some studies in rheumatoid arthritis suggest that the use of
concomitant folic acid may reduce abnormalities in liver enzymes. Pulmonary
fibrosis has only been reported rarely in psoriasis patients treated with
methotrexate. This contrasts with rheumatoid arthritis where pulmonary fibrosis
induced by methotrexate is a significant concern. Guidelines do however
recommend a chest X‐ray prior to starting methotrexate
therapy. This is in order to screen for pre‐existent pulmonary
pathology that may be exacerbated by methotrexate.
Indications
Methotrexate
(MTX) has been used for more than 40 years to treat severe psoriasis. It
induces remissions in the majority of treated patients and maintains remissions
for long periods with continued therapy. It is relatively safe and well
tolerated. Methotrexate is used alone or in combination with biologics. Safe
use of methotrexate requires appropriate monitoring.
A. Severe psoriasis
·
Chronic plaque psoriasis
(>10–15% BSA or
that affects certain areas of body so that normal function and employment are
prevented)
·
Pustular psoriasis
(generalized or localized)
·
Erythrodermic psoriasis
·
Psoriatic arthritis
(moderate to severe)
·
Severe nail psoriasis
B. Psoriasis not responding to topical treatments,
photo(chemo)therapy and/or systemic retinoids
Contraindications |
Absolute ·
Severe
anemia, leukopenia, and/or thrombocytopenia ·
Significant
liver function abnormalities, hepatitis (active and/or recent), severe
fibrosis, cirrhosis, excessive alcohol intake ·
Severe
infections ·
Concomitant
medications that increase MTX levels, e.g. trimethoprim–sulfamethoxazole ·
Significantly
reduced pulmonary function · Pregnancy or lactation ·
Peptic
ulcer (active) ·
Hypersensitivity
to MTX ·
Unreliable
patient Relative ·
Impaired
kidney function (creatinine clearance <60 ml/min)† ·
Currently
planning to have children (male and female patients)‡ ·
Active
infections ·
Gastritis ·
Pleural
effusion or ascites† ·
Concomitant
hepatotoxic medications ·
Immunodeficiency
syndromes ·
Concomitant
radiation therapy · Obesity (body mass
index >30) · Diabetes mellitus † Requires significant reduction in dosage. ‡ Because of possible mutagenic risk and
teratogenicity, discontinue MTX 3 months prior to attempts to conceive;
continue contraception during these 3 months. |
SIDE EFFECTS OF
METHOTREXATE |
||
Subjective symptoms |
Hematopoietic† |
Mucocutaneous |
1.
Most
frequent*,† 1.
Nausea 2.
Vomiting 3.
Abdominal
pain 4.
Fatigue 5.
Headache 2.
Occasional 1.
Loss
of libido 2.
Impaired
memory |
1.
Leukopenia‡ 2.
Thrombocytopenia 3.
Anemia |
1.
Oral
erosions† 2.
Alopecia 3.
Delayed
phototoxicity 4.
Tenderness
and/or necrosis of plaques due to overdose 5.
Rarely,
urticaria, angioedema, vasculitis |
Hepatic |
Pulmonary |
Fetal development |
1.
Elevated LFT results 1.
Minor elevations of LFT results are common; if
elevation exceeds 2× normal, must check more frequently; if exceeds 3×
normal, consider dose reduction; if exceeds 5× normal, discontinue Hepatitis† |
Interstitial
pneumonitis (acute-onset cough, dyspnea) |
Birth defects: cranial
and absence of digits |
Carcinogenesis¶ |
Idiosyncratic reaction |
|
1.
In
rheumatoid arthritis patients, increased risk of lymphoma 2.
Risk
factor for cutaneous SCC in PUVA-treated patients |
1.
Early
in course with full dosage 2.
Severe
pneumonia 3.
Gastrointestinal
hemorrhage 4.
Pancytopenia |
Opportunistic infections |
Additional/unusual |
1.
Pneumocystis
jiroveci pneumonia 2.
Cryptococcosis 3.
Disseminated
histoplasmosis 4.
Disseminated
herpes zoster |
1.
Osteopathy
(pain, osteoporosis, compression fractures) 2.
Ventricular
cardiac arrhythmias 3.
Lowered
seizure threshold |
* Same day
or 2–3 days following MTX administration; may respond to folic acid, dose
reduction or antiemetic drug.
† May be
reduced by folic acid (1–5 mg/day except on day MTX administered).
‡ Up to
10% of patients on oral regimens.
§ Up to
25% of patients receiving long-term therapy.
¶ No
increased risk of malignancy was observed in large series of psoriasis patients.
Mechanism
of Action
MTX suppresses psoriatic epidermal cell reproduction and
has anti-inflammatory and immunomodulatory effects.
Dihydrofolate
reductase (DHFR) converts dihydrofolate to tetrahydrofolate (fully reduced
folic acid), which is a necessary cofactor in the synthesis of thymidylate and
purine nucleotides, which, in turn, are required for DNA/RNA synthesis. MTX
competitively inhibits DHFR, although this inhibition can be at least partially
reduced by concomitant folic acid administration. MTX also exerts partially
reversible inhibition downstream on thymidylate synthetase, inhibiting cell
division during the S phase.
Although
originally believed to suppress keratinocyte proliferation, it is more likely
that MTX inhibits DNA synthesis in immunologically active cells. MTX decreases
inflammation through other mechanisms as well. For example, by inhibiting
aminoimidocarboxyamido-ribonucleotide transformylase, MTX increases local
tissue concentrations of the potent anti-inflammatory mediator adenosine. By
inhibiting methionine synthase, MTX reduces production of the proinflammatory
mediator S-adenyl methionine. Methotrexate is immunosuppressive; use should be avoided
in patients with active infections.
Dosages
MTX is
administered as a once-weekly dose of up to 25 mg for dermatologic and
rheumatologic diseases. Historically, MTX was administered in three doses over
24 hours (8 a.m. and 8 p.m. day 1, and 8 a.m. day 2). Dividing the dose can
decrease minor gastrointestinal side effects in some patients and also have theoretical advantages for the divided dosing regimen
from a cell cycle kinetics standpoint. However, since the clinical result is the
same, a single dose, which is easier and less confusing (for the patient and
the pharmacist), is currently recommended. The patient should be admonished to
adhere to the dosing schedule religiously, as more frequent dosing is much more
likely to produce major hematologic complications and potentially an increased
risk of liver fibrosis. Parenteral administration (intramuscular or
subcutaneous) is available for patients who cannot tolerate oral MTX and has
also been advocated by some clinicians for use in erythrodermic patients who
may have decreased gastrointestinal absorption or when compliance is an issue.
Available
in 2.5 mg tablets, therapy should begin with a test dose of 2.5–5 mg and repeat laboratory
tests with a complete blood count (CBC) including
differential and platelet count and LFT in approximately 7 days. This
test dose practice is mandatory in any patient with a decreased calculated
glomerular filtration rate or other significant risk factors for hematologic
toxicity. Using a test dose also provides an additional safeguard against rare,
idiosyncratic reactions to methotrexate.
Doses are
usually started with lower initial levels to minimize side effects and adjusted
to achieve clinical effectiveness. The usual weekly
dose for psoriasis is 10–15 mg, although doses up to 25 mg per week are not
uncommonly used, except in patients with renal insufficiency. Ordinarily, the dose should not exceed 25 mg weekly
and, in view of the reduced bioavailability of oral MTX at higher doses,
consideration should be given to switching to a subcutaneous route of
administration if response remains poor. Reported schedules have
been to start at lower dosages (e.g., 7.5 mg/week) and gradually increased,
whereas others recommend starting at the anticipated target dosage (e.g., 15
mg/week).
The
dose may be gradually increased by 2.5 to 5 mg every 2–4 weeks until
satisfactory results are obtained with minimal toxicity. Once disease control
has been attained for at least 1–2 months, the MTX can be tapered by 2.5 mg
every 1–2 weeks to the lowest dose that still maintains disease control. It can take 4 to 8
weeks to see a response to changes in methotrexate dose. Some patients can be
gradually weaned off therapy and restarted if the disease flares. The goal is
to both decrease the total cumulative dose and improve tolerability.
MONITORING GUIDELINES FOR METHOTREXATE
Drug Initial screening Follow-up monitoring Special considerations
Methotrexate |
CBC
with PLT count CMP(BUN,
creatinine, and LFTs) Pregnancy
test for women of childbearing potential Serologic
tests for hepatitis A, B, C HIV
testing if indicated Liver
biopsy is only indicated in high risk patients |
1.
CBC with PLT and AST/ALT after initial
2.5–5 mg test dose 2.
3.
CBC with PLT and AST/ALT every week for 2–4
weeks and after each dose escalation, then monthly for 2–4 months, and then
every 3–4 months, if stable 4.
5.
BUN and serum creatinine every 6–12 months 6.
7.
Serum markers and non-invasive imaging
studies used to assess hepatic fibrosis |
1.
Patients being considered for methotrexate therapy are
divided into two groups based on their risk factors for liver injury. Per recent psoriasis treatment guidelines (2009), for
patients without risk factors for hepatic fibrosis, liver biopsy is not
indicated at baseline. For high-risk patients, liver biopsies are done
baseline and then after every 1 g, and in those with grade IIIA liver biopsy
changes, every 6 months or as recommended by a hepatologist 2.
3.
Consider creatinine clearance, especially
in elderly patients |
CMP: Comprehensive metabolic panel
Leukocyte
and platelet counts are depressed maximally approximately 7 to 10 days after
treatment. A drop in these counts below normal levels necessitates reducing or
stopping therapy. Liver function tests (LFTs) are obtained at least 1 week
after the last dose of the drug. MTX causes transient elevations in LFTs for 1
to 3 days after its administration, so a false-positive elevation might be seen
if the patient is tested too soon.
Side Effects
Short-term
side effects include nausea, anorexia, fatigue, oral ulcerations and
stomatitis, mild leukopenia, thrombocytopenia, and macrocytic anemia. These are
dose-related and rapidly reversible and related to renal and hematologic
function. Switching among triple dosing, weekly oral dosing, and intramuscular
dosing may decrease these reactions.
The most important major side effects of MTX include pancytopenia,
and hepatotoxicity. Pancytopenia typically develops early, compared to hepatic
fibrosis and cirrhosis, which take years to develop. There is considerable
debate regarding the risk of hepatotoxicity in psoriasis patients on long-term
MTX, and recent studies have shown that methotrexate-associated fibrosis and
cirrhosis may be less than initially reported. Compounding risk factors for
developing hepatotoxicity from MTX are well established. Before prescribing
MTX, references discussing this issue in detail should be reviewed and
consultation with a hepatologist considered. For those without risk factors, consider
liver biopsy in patients with cumulative doses of more than 3.5-4 gm of
methotrexate and repeated liver biopsies after each subsequent 1.5-gm dosage,
based on LFT results, risk factors (e.g., diabetes and obesity) or in
consultation with a hepatologist.
RISK FACTORS FOR DEVELOPING
HEPATIC TOXICITY FROM METHOTREXATE |
·
Persistent abnormal liver function tests ·
History of chronic liver disease including chronic hepatitis B or C viral infection ·
Previous or concurrent excessive alcohol
intake ·
Family history of inheritable liver disease ·
Diabetes mellitus ·
Obesity ·
Hyperlipidemia ·
History of significant exposure to
hepatotoxic drugs or chemicals ·
Lack of folate supplementation |
While the gold standard for
detection of hepatic fibrosis remains liver biopsy, this invasive procedure
harbors risks including subcapsular hemorrhage. A variety of serum markers and
non-invasive imaging studies have been developed to try to detect hepatic fibrosis.
Elevated levels of procollagen III aminopeptide (PIIINP) have been reported to
be a serologic marker of fibrosis, but this assay does not specify which organ
has fibrosis and it can be unreliable in patients with psoriatic arthritis or
other inflammatory diseases.
Additional
serologic assays for detecting hepatic fibrosis have been developed including:
(1) FIBROSpect® II – measures circulating
levels of α2-macroglobulin,
tissue inhibitor of metalloproteinase-1 (TIMP-1), and hyaluronic acid; and (2) FibroTest® – measures circulating levels of α2-macroglobulin,
haptoglobin, apolipoprotein A1, γ-glutamyl transpeptidase, and total bilirubin. In
both, the value of each serum marker is incorporated into a composite score.
For the detection of moderate-to-severe fibrosis, these assays possess fairly
good diagnostic accuracy, with a sensitivity of 47–77% and a specificity of
78–90%; however, they have been used primarily to evaluate patients with
chronic hepatitis C viral infection. Imaging studies being investigated
include: (1) ultrasound-based transient elastography (FibroScan®) – a rapid, non-invasive measure of liver stiffness
that is unreliable in obese patients; and (2) magnetic resonance elastography.
Both the tests are not widely utilized and have limitations. In the future, it
is likely that some combination of serologic assays plus imaging studies will
allow selection of those patients on MTX most at risk for hepatic fibrosis in
whom a liver biopsy can then be performed.
Photosensitivity
may occur with MTX and patients should take appropriate sun precautions.
Gastrointestinal intolerance is often abated with concomitant folic acid
therapy. Other MTX-induced side effects include accelerated rheumatoid
(cutaneous) nodulosis and reversible lymphoproliferative disorders.
METHOTREXATE (MTX) –
COUNSELING FOR PATIENTS OF CHILDBEARING POTENTIAL |
Women |
·
Should not become pregnant while receiving
MTX ·
MTX can cause birth defects and abortions ·
Must use highly effective means of
contraception (e.g. OCPs; IUD; condom + spermicide) ·
After discontinuing MTX, wait at least one
ovulatory cycle before attempting to become pregnant |
Men |
·
May lead to low sperm counts ·
Drug is present in semen ·
Must use highly effective means of
contraception (e.g. OCPs, IUD [female partner]; condom + spermicide) ·
After discontinuing MTX, wait 3 months
before attempting to have partner become pregnant |
Use in Pregnancy and Lactation
MTX is an abortifacient and teratogen and should not be used
during pregnancy. MTX can impair fertility by
adversely affecting oogenesis and spermatogenesis, and by causing menstrual
dysfunction: these effects are considered to be reversible on discontinuing
treatment. It also has a mutagenic potential and thus it would seem prudent to
advise both men and women to avoid conception whilst taking MTX and for up to 6
months thereafter;
should pregnancy ensue before this time period, consider genetic counseling.
The
American Academy of Pediatrics considers MTX to be contraindicated in
breastfeeding owing to concerns of immune suppression, growth retardation, and
carcinogenesis.
Mothers receiving methotrexate should not breast-feed.
Drug Interactions
The interaction of MTX
with other drugs can occur via a number of mechanisms:
·
Reduced absorption from
gut: digoxin (absorption reduced by MTX), neomycin
· Displacement from plasma proteins: NSAIDs (aspirin, diclofenac, ibuprofen, indometacin, ketoprofen, meloxicam and naproxen), sulphonamides
·
Added antifolate
effect: nitrous oxide, trimethoprim, sulphonamides, dapsone, phenytoin, pyrimethamine
·
Diminished renal
excretion: ciprofloxacin, NSAIDs, omeprazole, penicillins, probenecid, sulphonamides
·
Increased renal
excretion: acetazolamide
·
Cumulative
toxicity: tetracyclines, acitretin, clozapine, ciclosporin, cisplatin, leflunomide, alcohol
·
Other: theophylline (plasma
levels increased by MTX)
Numerous
medications may interact with methotrexate by a variety of mechanisms that can
result in elevated drug levels, thereby increasing the risk for methotrexate
toxicity.
After
absorption, methotrexate binds to serum albumin, NSAIDs (aspirin, diclofenac, ibuprofen, indometacin, ketoprofen, meloxicam and naproxen), sulfonamides,
phenytoin, and antibiotics including penicillin, minocycline, chloramphenicol,
and trimethoprim may decrease the binding of methotrexate to albumin, leading
to increased serum levels of methotrexate. Several other medications including
colchicine, cyclosporin A (CsA), probenecid, NSAIDs, omeprazole, penicillins,
ciprofloxacin and sulfonamides may lead to decreased renal tubular excretion
leading to decreased renal elimination of methotrexate and increased serum
levels. Trimethoprim, sulfonamides, and dapsone also
inhibit the folate metabolic pathway and markedly increase the risk for
pancytopenia with concomitant use. Acitretin has been used successfully
in combination with methotrexate despite the potential for hepatotoxicity from
both medications. Alcohol may cause synergistic liver
damage in combination with MTX.
Risk Factors for Hematologic Toxicity from
Methotrexate
1.
Renal
insufficiency
2.
Advanced
age
3.
Lack
of folate supplementation
4.
Methotrexate
dosing errors (inadvertent daily dosing)
5.
Drug
interactions: displacement of MTX
from protein‐binding sites
(particularly by NSAIDs or sulphonamides)
6.
Hypoalbuminemia
7.
Greater
than moderate alcohol intake
Monitoring for Hepatotoxicity in Patients with
No Risk Factors for Hepatotoxicity
1.
No
baseline liver biopsy
2.
Monitor
LFT results monthly for the first 6 mo and then every 1-3 mo thereafter
- For
elevations <2-fold upper limit of normal: repeat in 2-4 wk
- For
elevations >2-fold but <3-fold upper limit of normal: closely monitor, repeat in 2-4 wk, and decrease dose as needed
- For
persistent elevations in 5 of 9 AST levels during a 12-mo period or if there is
a decline in the serum albumin level below the normal range with normal
nutritional status, in a patient with well-controlled disease, a liver biopsy
should be performed
3.
Consider
liver biopsy after 3.5-4.0 gm total cumulative dosage
4.
Consider
switching to another agent or discontinuing therapy after 3.5-4.0 gm total
cumulative dosage
Monitoring for Hepatotoxicity in Patients with
Risk Factors for Hepatotoxicity
1.
Consider
the use of a different systemic agent.
2.
Consider
delayed baseline liver biopsy (after 2-6 months of therapy to establish
medication efficacy and tolerability).
3.
Perform
repeated liver biopsies after approximately 1.5 gm of methotrexate.
Folate supplementation
Folate
supplementation reduces hematologic, gastrointestinal, and hepatotoxic side
effects without decreasing the efficacy. Options for folate supplementation
include folic acid 1 mg daily or 5 mg once weekly except for the day of
methotrexate dosing, reduces the frequency of side effects.
Lung toxicity
Methotrexate-induced
lung injury can be sudden and severe. The presenting symptom is usually a new
onset of cough and shortness of breath. It is most often a subacute process, in
which symptoms are commonly present for several weeks before diagnosis. Approximately
50% of the cases are diagnosed within 32 weeks from initiation of MTX
treatment. A patient who recovers from MTX lung injury should not be retreated.
Earlier recognition and drug withdrawal may avoid the serious and sometimes
fatal outcome. The strongest predictors of lung injury are older age, diabetes,
rheumatoid pleuropulmonary involvement, previous use of disease-modifying
antirheumatic drugs, and hypoalbuminemia.
Recall of sunburn
Patients
taking methotrexate with a previous history of sunburn or radiation burn may
experience a flare-up of symptoms in the areas that had been burned. This
reaction is distinct from true photosensitivity.
Therefore patients on MTX should take appropriate sun precautions.
ACITRETIN
Acitretin is an oral retinoid and
one of the safest systemic psoriasis therapies. It binds to retinoic acid receptors
and reduces keratinocyte proliferation and also reduces Th17 cells with a
concomitant increase in regulatory T cells. Acitretin is not immunosuppressant
and therefore can be considered in clinical situations where other systemic
medications are contraindicated, such as active malignancy.
INDICATIONS AND
CONTRAINDICATIONS FOR ACITRETIN |
Indications |
·
Severe
psoriasis that cannot be managed by topical treatments or photo(chemo)therapy ·
Monotherapy
is indicated for erythrodermic or pustular psoriasis ·
Combination
therapy is indicated for chronic plaque psoriasis |
Contraindications |
Absolute ·
Severe
liver dysfunction ·
Severe
kidney dysfunction (elimination reduced) ·
Pregnancy
or lactation ·
Women
of childbearing potential who cannot guarantee adequate contraception during
and up to 3 years following discontinuation of acitretin ·
Hyperlipidemia,
especially hypertriglyceridemia, that cannot be controlled ·
Excessive
alcohol intake ·
Unreliable
patient Relative ·
Concomitant
medications that interfere with retinoid bioavailability or whose metabolism
is altered† ·
Concomitant
hepatotoxic drugs, e.g. methotrexate‡ ·
Poorly
controlled diabetes mellitus ·
History
of pancreatitis ·
Use
of contact lenses ·
Atherosclerosis |
† Requires
dose adjustment and careful monitoring, e.g. phenytoin competes for plasma
protein binding.
‡ Reserved
for treatment-resistant patients.
As monotherapy, acitretin is less effective for chronic plaque psoriasis. In one study, 23% of patients
treated with 50 mg of acitretin daily for 8 weeks achieved ≥75% improvement in
their PASI. Combination treatment with photo (chemo) therapy and/or vitamin D3 analogues results in a substantial improvement
in clinical response. Maximal therapeutic efficacy is reached after 2–3 months.
Acitretin has been shown to be an effective maintenance therapy. As
monotherapy, acitretin is highly effective in erythrodermic and pustular psoriasis.
Its efficacy in nail psoriasis and psoriatic arthritis is only modest.
In patients with chronic plaque psoriasis, 0.5 mg/kg/day is the
initial dosage, which can be increased depending upon the clinical response and
side effects. For erythrodermic psoriasis, the initial dosage is
0.25 mg/kg/day, and in pustular psoriasis, the dose should be maximized, i.e.
up to 1 mg/kg/day. In patients with chronic plaque psoriasis, mild cheilitis
(just perceivable by the patient) is the goal, whereas in patients with pustular
psoriasis, a dose that causes a clinically apparent but tolerable cheilitis is
an endpoint.
Optimum dose range for monotherapy
is 25-50 mg/day.
Lower
doses of acitretin at 10 to 25 mg daily are usually effective when used in
combination with phototherapy. This
results in faster and more complete responses to PUVA and to UVB. Significantly lower ultraviolet doses are
required when retinoids are added to a phototherapy regimen.
ACETRETIN – EVALUATION PRIOR
TO AND DURING TREATMENT |
Pre-acitretin screening |
·
History
to exclude contraindications ·
Complete
blood count ·
Liver
function tests (AST, ALT, γGT, alkaline phosphatase, bilirubin) ·
Serum
triglycerides, cholesterol, HDL ·
Glucose ·
Serum
creatinine ·
Pregnancy
test ·
Consider
spinal X-ray (often initially performed during the first 3 months of therapy
if long-term treatment is anticipated) |
Evaluation during acitretin
treatment |
· Monitor mucocutaneous side effects ·
Serum
triglycerides, cholesterol/HDL, and liver function tests (every month for the
first 2 months then every 2–3 months) · CBC and serum creatinine (elderly patients or
patients with mild to moderate renal dysfunction) every 3 months ·
Monitor
for development of hyperostosis by history (twice yearly) and by X-ray of
spine (e.g. once yearly or every other year in patients receiving long-term
treatment) ·
Pregnancy
test (twice yearly throughout treatment) |
Side effects
Mucocutaneous side effects,
hepatotoxicity, and alterations in serum lipid profile are dose-dependent.
Acitretin is
teratogenic. In the presence of ethanol, acitretin is esterified to etretinate.
Etretinate persists in tissue for years. Therefore acitretin is not prescribed
to women of child-bearing potential who may become pregnant within 3 years. In
doses of 50 mg per day or higher, mucocutaneous side effects are common and
include cheilitis, conjunctivitis, and hair loss, failure to develop normal
nail plates, dry skin, and “sticky skin.” Periungual pyogenic granulomas can
develop. Headache is a possible sign of pseudotumor cerebri. Elevation of serum
lipid level, particularly triglycerides, can be prevented by concomitant
administration of gemfibrozil or atorvastatin. Elevation of liver function
tests can occur.
CYCLOSPORINE
Cyclosporine (CS) at dosages of 2.5
to 5.0 mg/kg/day administered to reliable, carefully selected patients who are
closely monitored for both clinical and laboratory parameters produces fast and
favorable results for severe plaque psoriasis. Cyclosporine is indicated for
the treatment of severe, recalcitrant, plaque psoriasis in adults who are
immunocompetent. Cyclosporine is also effective in treating pustular,
erythrodermic, and nail psoriasis. Efficacy of cyclosporine has been
demonstrated in all variants of psoriasis (including nail psoriasis), but less
so for psoriatic arthropathy. In general, a reduction of the PASI of 60–70% can
be reached within 4 weeks of treatment. During long-term treatment, there are
no signs of tachyphylaxis.
Recommendations for
Cyclosporine
1.
Indication: Adult, immunocompetent patients with severe, recalcitrant psoriasis
1.
Severe
is defined by the FDA as extensive or disabling plaque psoriasis
2.
Recalcitrant
is defined by the FDA as those patients who have failed to respond to at least
one systemic therapy or in patients for whom other systemic therapies are
contraindicated, or cannot be tolerated
3.
Some
guidelines suggest use of cyclosporine in moderate-to-severe psoriasis
4.
Efficacy
observed in erythrodermic psoriasis, generalized pustular psoriasis, and
palmoplantar psoriasis
MONITORING GUIDELINES FOR CYCLOSPORINE
Drug Initial
screening Follow-up
monitoring Special considerations
Cyclosporine |
1.
At
least 2 baseline blood pressure readings 2.
At
least 2 baseline BUN and serum creatinine levels 3.
CMP
(must include BUN, magnesium, potassium and uric acid) 4.
CBC
with PLT count 5.
UA
with microscopic examination 6.
Fasting
lipid profile (TG, cholesterol, HDL) |
1.
CBC
with PLT count at 1 month, then every 2–4 months 2.
CMP
(must include BUN, creatinine, uric acid, potassium, magnesium) every 2 weeks
for 1–2 months, then monthly or every 3 months if stable 3.
UA
with microscopic examination every month if abnormal; otherwise yearly 4.
Fasting
lipid panel every 2–4 weeks for 1–2 months, then every 3 months if stable |
1.
Blood
pressure must be monitored every 2 weeks for first 3 months and then monthly
if stable 2.
Lower
dose by 25–50% if serum creatinine remains elevated (over 2 weeks) by >25%
of baseline 3.
Lower
dose by 25–50%, or discontinue, if at any time serum creatinine is elevated
by ≥50% of baseline 4.
Consider
creatinine clearance if >6 months of therapy 5.
Skin
examination at least yearly and age-appropriate cancer screening 6.
Consider
trough whole drug cyclosporine level if non-compliance, poor absorption or
drug interactions are suspected |
Side effects
Cyclosporine can produce dramatic rapid improvement
of psoriasis, but this must be balanced by the requirement for an appropriate
replacement therapy, given the need to ultimately stop cyclosporine therapy.
The most
serious side effects of cyclosporine use in psoriasis are nephrotoxicity and
hypertension.
An important aspect is assessment
of renal function, and creatinine clearance should be estimated using the
Cockcroft–Gault formula:
In elderly patients and patients
with a history of hypertension, the risks of renal impairment and hypertension
are increased.
Cyclosporine
treatment in psoriatic patients has been reported to increase the frequency of
SCCs, especially in those previously treated with PUVA. The underlying
mechanism is not mutagenesis but a decrease in immunosurveillance of the skin.
In particular, psoriatic patients who have been exposed to high cumulative
doses of UV radiation are at risk for development of cutaneous malignancies.
Although cyclosporine is an immunosuppressive agent, no increase in serious
infections has been reported in patients treated with cyclosporine alone. Other
side effects include: gastrointestinal discomfort, hypertrichosis,
paresthesias, gingival hyperplasia, headache, vertigo, muscle cramps, and
tremor. Metabolic side effects include hyperkalemia, hypomagnesemia, hyperuricemia
(due to decreased clearance of uric acid), and elevated cholesterol and
triglycerides.
Contraindications |
Absolute ·
Impaired
renal function ·
Uncontrolled
hypertension ·
Past
or present malignancy ·
History
of excessive photo (chemo) therapy (>200 PUVA treatments) or concurrent
photo (chemo) therapy ·
Radiotherapy ·
Severe
infections ·
Hypersensitivity
to cyclosporine ·
Unreliable patient Relative ·
Active
infections ·
Concomitant
drugs affecting cyclosporine pharmacokinetics† ·
Primary
or secondary immunodeficiency ·
Concomitant
immunosuppressive therapy ·
History
of arsenic exposure ·
Pregnancy
or lactation ·
Concurrent
methotrexate administration ·
Significant
hepatic disease ·
Hyperuricemia,
hyperkalemia ·
Vaccination
with live vaccines ·
Seizure
disorder ·
Poorly
controlled diabetes mellitus ·
Severe
chronic organ dysfunction ·
Alcohol
and drug abuse ·
Malabsorption |
† Requires
dose adjustment and careful monitoring; cyclosporine is inactivated by the
cytochrome P450 3A isoform.
Dosage
Treatment is
started from 2.5 to 5.0 mg/kg/day in two divided doses as clinically indicated
and then the dosage is adjusted by 0.5 to 1 mg/kg/day every 1 week.
The starting
dose depends on the clinical state. There are two approaches to determine the
starting dosage. The speed of improvement and the success rate are proportional
to the dosage.
Low-dose
approach
Start at 2.5
mg/kg/day, and wait at least 1 month before considering increasing the dosage.
This approach with slow increments in dosage increase is for patients with
stable, generalized psoriasis or for patients in whom the severity lies between
moderate and severe. Increase the dosage by 0.5 to 1.0 mg/kg/day increments
every week, up to a maximum of 5 mg/kg/day if needed.
High-dose
approach
Start at a 5
mg/kg/day dose when rapid improvement is critical. Patients with severe
inflammatory flares, patients with recalcitrant cases that have failed to
respond to other modalities, or distressed patients in a crisis situation are
candidates for the high-dose approach. High doses are usually well tolerated
for short-term use. As soon as there is a response, the dosage is decreased by
0.5 to 1 mg/kg/day, but no more than one step in the decrement of dosage per
week, until the minimum effective maintenance dosage is defined.
Intermittent
short courses
Another
therapeutic strategy to manage moderate-to-severe plaque psoriasis with
cyclosporine is the use of intermittent short courses. It is effective and well
tolerated and reduces the risk of side effects. Cyclosporine is initially given
at a dose of 2.5 mg/kg/day in two divided doses. This dosage could be increased
by increments of 0.5 to 1.0 mg/kg/day each week up to a maximum of 5 mg/kg/day.
Treatment is continued until clearance of psoriasis, defined as 90% or more
reduction in the area affected occurs or for a maximum of 12 weeks.
Cyclosporine is stopped abruptly. On relapse, patients are given another course
of cyclosporine, commencing at the optimum dose from the previous treatment
period. Intermittent short courses for up to 2 years appear safe and well
tolerated.
Weekend
therapy
Initiate
treatment with cyclosporin A (CsA) at 5 mg/kg/day for 12 weeks (induction
treatment). Then treat with CsA 5 mg/kg/day for 2 consecutive days (on
weekends) per week.
TARGETED IMMUNOMODULATORS
(“BIOLOGIC” THERAPIES) FOR PSORIASIS
The immunological basis of psoriasis continues to be refined and the
ability of technology to produce targeted therapies has revolutionized
treatment of psoriasis. Psoriasis is an autoimmune disorder involving
activation of dendritic cells that stimulate aberrant T- cell pathways. Biologic agents are proteins that can be
synthesized using recombinant DNA techniques (genetic engineering). Biologic
agents bind to specific cells and proteins and do not have multiorgan adverse
effects as seen with acitretin, cyclosporine, and methotrexate. The potential
for interaction with other drugs is very low.
Biologic therapies are indicated for patients with
moderate to severe psoriasis and/or psoriatic arthritis. Several guidelines
restrict their use to “high-need patients” in whom all other existing
treatments are contraindicated or have led to insufficient improvement, whereas
some investigators have voiced the opinion that biologic therapies should be an
option for patients who have failed to respond adequately to one classic
systemic treatment; the latter recommendation has to be balanced against the
high cost of these medications.
Their two major targets are T cells and cytokines,
including TNF-α, IL-12/23, and IL-17.
COMMERCIALLY
AVAILABLE BIOLOGIC AGENTS FOR THE TREATMENT OF PSORIASIS |
|||
Biologic agent |
Target |
Molecule |
Approved* |
Etanercept |
TNF-α† |
Human fusion protein |
FDA + EMA |
Infliximab |
TNF-α‡ |
Chimeric antibody |
FDA + EMA |
Adalimumab |
TNF-α‡ |
Human antibody |
FDA + EMA |
Ustekinumab |
p40 subunit of
IL-12/23 |
Human antibody |
FDA + EMA |
Secukinumab |
IL-17A |
Human antibody |
FDA + EMA |
Ixekizumab |
IL-17A |
Humanized antibody |
FDA + EMA |
Brodalumab |
IL-17 receptor |
Human antibody |
FDA + EMA |
Guselkumab |
IL-23 |
Human antibody |
FDA |
EMA, European Medicines Agency; FDA, Food and Drug
Administration; IL, interleukin; TNF, tumor necrosis factor.
Several biologic therapies are currently available worldwide for the treatment of psoriasis and/or psoriatic arthritis: adalimumab, etanercept, infliximab, ustekinumab, secukinumab, ixekizumab, brodalumab and guselkumab. Despite the availability of effective topical and classic systemic medications as well as phototherapy, safety concerns, lack of efficacy, and inconveniences are important restrictions, in particular for long-term use. For such patients, biologic agents can improve their skin disease as well as psoriatic arthritis (especially TNF-α inhibitors), with prevention of permanent destructive changes. Some investigators have proposed that reduction of inflammation due to active psoriasis may impact the development of metabolic syndrome and cardiovascular diseases.
INDICATIONS AND
CONTRAINDICATIONS FOR COMMERCIALLY AVAILABLE TARGETED IMMUNOMODULATORS
(“BIOLOGIC” AGENTS) |
Indications |
General indications |
Patients
with moderate to severe psoriasis, eligible for a systemic treatment Patients
with psoriatic arthritis, particularly those who have failed other
disease-modifying antirheumatic drugs (DMARDs) |
Restricted indication |
Patients
with moderate to severe psoriasis who are not candidates for topical
treatments, photo(chemo)therapy, or classic systemic treatments because of
insufficient efficacy or contraindications† |
Contraindications |
Absolute ·
Significant viral, bacterial or fungal
infection, including active Salmonella or
dimorphic fungal infection ·
Increased risk for developing sepsis ·
Active tuberculosis ·
Allergic reaction to the biologic agent ·
Selective for TNF-α inhibitors: ANA+
(especially if high titer) or autoimmune connective tissue disease, blood
dyscrasias, congestive heart failure (NYHA grade III or IV), or
demyelinization disorders (the latter also if in first-degree relative) Relative ·
Selective for ustekinumab: BCG
vaccination within the past 12 months (mode of action expected to increase
susceptibility to mycobacterial infections) ·
Selective for secukinumab, ixekizumab, and
brodalumab: active Crohn disease (may cause exacerbation of bowel
disease) ·
History of hepatitis B viral (HBV)
infection** ·
History of hepatitis C viral infection
(risk of activation)*** ·
Immunosuppressed patient ·
Pregnancy (TNF-α inhibitors,
ustekinumab and secukinumab are category B; ixekizumab was released after
2015 ·
Breastfeeding ·
Malignancy within the past 5 years (does
not include adequately treated single cutaneous squamous or basal cell
carcinoma) ·
Excessive chronic sun exposure or
photo(chemo)therapy |
† Guttate, pustular, or erythrodermic
psoriasis are not established indications, but there is anecdotal evidence for
efficacy and safety.
** If HBsAg-positive, measure HBV DNA and
treat with antiviral agent (e.g. tenofovir, entecavir, telbivudine) prior to
immunosuppressive therapy. Monitor for reactivation in those with anti-HBs or
-HBc antibodies who are HBsAg-negative
*** Need to monitor liver function tests and
viral load.
With
regard to the percentage of patients achieving PASI 75 improvement, there is
significant overlap between the efficacies of biologic agents (as assessed
after 3 months of therapy) and those of photo(chemo)therapy and classic
systemic medications such as MTX and cyclosporine. In general, TNF-α inhibitors and anti-IL-12/23 antibodies have
substantial efficacy and enable long-term control of psoriasis, as do
secukinumab and ixekizumab recently approved anti-IL-17A antibodies. The latter
have demonstrated greater efficacy than etanercept or ustekinumab in
clinical trials.
US Food and Drug
Administration (FDA) warnings and precautions for targeted immune modulators
with dermatologic indications.
Live vaccines should not be given to patients receiving these medications. BCG,
bacillus Calmette–Guérin; IL, interleukin; LE, lupus erythematosus; TNF, tumor
necrosis factor.
FDA WARNINGS AND PRECAUTIONS
FOR TARGETED IMMUNE MODULATORS WITH DERMATOLOGIC INDICATIONS |
TNF inhibitors: adalimumab,
certolizumab, etanercept, golimumab, infliximab |
Increased
risk of serious infections, including tuberculosis, bacterial sepsis,
systemic fungal infections (e.g. histoplasmosis), and infections due to
opportunistic pathogens Risk
of hepatitis B virus reactivation Malignancies
(especially lymphomas) have been reported in patients receiving these agents,
including children and adolescents For infliximab, lymphomas are seen more often than in the
general population and fatal hepatosplenic T-cell lymphomas have developed in
patients with inflammatory bowel disease who were also receiving azathioprine
or 6-mercaptopurine An
increased risk of non-melanoma skin cancer and melanoma has been observed in
patients with rheumatoid arthritis treated with TNF inhibitors Congestive
heart failure (exacerbation or new onset) has been observed Demyelinating
disease (exacerbation or new onset) has been observed Anaphylaxis
or severe allergic reactions can occur, including serum sickness-like
reactions to infliximab Other
potential adverse events include autoimmune hepatitis, cytopenias, and a
lupus-like syndrome; |
Ustekinumab |
Serious
infections have been observed; may increase the risk of infection and
reactivation of latent infections Patients
genetically deficient in IL-12/IL-23 have an increased risk of severe
infections with mycobacteria and Salmonella BCG
vaccination should not be given in the year prior to initiation or the year
following completion of ustekinumab therapy Could
potentially increase the risk of malignancies Hypersensitivity
reactions (e.g. angioedema, anaphylaxis) can occur Reversible
posterior leukoencephalopathy syndrome has been reported |
IL-17 inhibitors: ixekizumab,
secukinumab, brodalumab |
Serious
infections have been observed; may increase the risk of infection and
reactivation of latent infections Patients
genetically deficient in IL-17 are prone to chronic mucocutaneous candidiasis New
onset and exacerbation of inflammatory bowel disease have occurred Hypersensitivity
reactions (e.g. angioedema, anaphylaxis) can develop For brodalumab: suicidal ideation and behavior, including
complete suicides, have occurred |
Recommended laboratory
evaluation for patients receiving targeted immune modulators for dermatologic
disorders
Laboratory testing |
|
Prior to treatment |
During treatment |
PPD**/interferon-γ release assay† and/or (e.g. if immunosuppression or history of
tuberculosis) chest X-ray CBC
and CMP Hepatitis
B and C virus serologic profiles Consider
HIV testing |
Annual
PPD**/interferon-γ release assay† and/or chest X-ray CBC
and CMP every 3–12 months, or as noted below or clinically indicated |
** ≥5 mm of induration should be considered
as positive.
† e.g. QuantiFERON® TB Gold or T-SPOT®.TB.
CUTANEOUS SIDE EFFECTS OF
TUMOR NECROSIS FACTOR INHIBITORS |
New-onset
psoriasis, especially palmoplantar pustulosis Interstitial
granulomatous dermatitis and other granulomatous eruptions Cutaneous
small vessel vasculitis Eczematous
eruptions Lichenoid
dermatitis Lupus-like
syndrome-associated malar rash or discoid lesions Other
types of cutaneous lupus, e.g. SCLE, chilblain lupus |
New-onset
palmoplantar and inverse psoriasis has also been reported to be by the
interleukin-17 inhibitor sekukinimab. Allergic reactions manifesting with
urticaria and angioedema can also occur with most agents.
Vaccination
Live
vaccines are contraindicated in patients receiving TNF inhibitors. It is
recommended that patients (especially children) be brought up-to-date with all
immunizations prior to initiating treatment with TNF inhibitors.
Consider giving pneumococcal, hepatitis A and
hepatitis B, influenza, and tetanus-diphtheria vaccines before initiation of
immunosuppressive therapy. Once immunosuppressive therapy has begun, patients
must avoid vaccination with live vaccines (including varicella; mumps, measles,
and rubella; oral typhoid; yellow fever) and live-attenuated vaccines
(including intranasal influenza and the herpes-zoster vaccine). Studies show
adequate but attenuated immune responses to killed virus vaccines such as
influenza vaccination and pneumococcal vaccine.
VACCINES THAT CONTAIN LIVE
ATTENUATED VIRUSES OR BACTERIA |
1.
Adenovirus 2.
Bacillus Calmette–Guérin (BCG) 3.
Cholera, oral form in the US 4.
Herpes zoster (shingles) 5.
Influenza (including H1N1), intranasal form 6.
Measles, mumps, and rubella 7.
Rotavirus 8.
Polio, oral form 9.
Typhoid, oral form Vaccinia
(smallpox vaccine) Varicella Yellow
fever |
TNF-α inhibitors for the treatment
of psoriasis
The
introduction of TNFi has revolutionized the management of severe psoriasis.
They are effective and are the most commonly prescribed biologic agents for
psoriasis. Currently, three TNFi are licensed for this indication; adalimumab,
etanercept and infliximab. The indication of psoriasis is same for each agent
being limited to moderate/severe disease when other treatments (methotrexate,
ciclosporin and phototherapy) have been ineffective, or are contraindicated.
Patients
with an inadequate response to one TNF inhibitor may derive benefit from
switching to another. Sequential tumor necrosis factor-α antagonist
treatment can be considered for patients with psoriasis who do not respond, or
lose response, during anti–tumor necrosis factor treatment.
Screening
for latent tuberculosis infection
Patients on
anti–tumor necrosis factor-α therapy are at
risk of developing active tuberculosis. Perform tuberculin skin testing before
initiating therapy and at 6- to 12-month intervals during therapy. However,
there have been cases reported in which psoriasis patients on long-term
etanercept therapy have manifested negative tuberculosis results and positive
interferon-gamma release assay results. This suggests that tuberculin skin
testing might be unreliable during long-term anti–tumor necrosis factor-αtherapy.
QuantiFERON-TB Gold testing (interferon-gamma release assay) may be a more
appropriate primary test in patients with risk factors for false-negative
tuberculin skin test results. Interferon-gamma release assays may be very
expensive.
Mechanism of action
Infliximab, adalimumab,
golimumab, and certolizumab pegol are monoclonal antibodies, while etanercept
is a dimeric fusion protein composed of the extracellular portions of two p75
TNF receptors linked to the Fc portion of IgG1. Although all four monoclonal
antibodies target human TNF, infliximab is chimeric (human–mouse) IgG1,
adalimumab and golimumab are human recombinant IgG1 and certolizumab pegol is a
humanized pegylated Fab fragment.
Clinical
studies have found that Infliximab and adalimumab to be slightly more effective
than etanercept in the treatment of psoriasis. The different effects of these
agents are associated with selectivity in their ability to perturb these
receptor ligand interactions. It is known that Infliximab, adalimumab and
etanercept bind TNF differently; infliximab and adalimumab bind to both soluble
and membrane bound TNF, whereas etanercept bind primarily to soluble TNF.
Binding to membrane bound TNF can induce a dose dependent increase in apoptosis
of T cells.
Mechanism of action of the tumor necrosis factor
(TNF) inhibitors.
All of the TNF inhibitors can bind to soluble
TNF and block its ability to activate TNF receptors. By interacting with
membrane-bound TNF, the IgG1 monoclonal antibodies can also activate
complement-dependent cytotoxicity and induce cellular apoptosis. This capacity
to destroy TNF-producing cells may explain the greater efficacy of these
monoclonal antibodies compared to etanercept in the treatment of granulomatous
conditions as well as the potentially higher risk of infections associated with
their use. In addition to the anti-inflammatory effects of TNF inhibitors,
blocking TNF signaling may have an impact on the neuroendocrine system.
Contraindications
Each TNF
inhibitor is contraindicated in patients with known hypersensitivity to that
particular medication, and infliximab is contraindicated in those with an
allergy to murine proteins. These agents should be avoided in patients with a
significant active infection, malignancy, congestive heart failure (especially
if unstable), or multiple sclerosis.
Recommended Laboratory
evaluation for patients receiving targeted immune modulators for dermatologic
disorders.
Laboratory testing |
|
Prior to treatment |
During treatment |
PPD**/interferon-γ release assay† and/or (e.g. if immunosuppression or history of
tuberculosis) chest X-ray CBC
and CMP Hepatitis
B and C virus serologic profiles Consider
HIV testing |
Annual
PPD**/interferon-γ release assay† and/or chest X-ray CBC
and CMP every 3–12 months, or as noted below or clinically indicated |
** ≥5 mm of induration should be considered
as positive.
† e.g. QuantiFERON® TB Gold or T-SPOT®.TB.
Adalimumab
Indications
Adalimumab
(a fully
human monoclonal antibody with affinity for TNF‐α) is approved for the treatment of moderate to severe plaque-type
psoriasis in adults. Additional approved indications include psoriatic
arthritis, rheumatoid arthritis, ankylosing spondylitis, uveitis, and Crohn
disease in adults as well as JIA and Crohn disease in children ≥2 years and ≥6
years of age, respectively.
Dosages
Adalimumab
is supplied in 10–80 mg prefilled syringes and a 40 mg autoinjector; it is
administered subcutaneously. For the treatment of psoriasis, an initial loading
dose of 80 mg is typically given on day 0, followed by 40 mg on day 8 and then
40 mg every other week. Loss of efficacy
of adalimumab over time related to the production of anti-adalimumab antibodies
can occur and may be prevented by the concurrent administration of low-dose
weekly methotrexate; loss of efficacy has also been associated with the
development of antinuclear antibodies. The concomitant use of adalimumab and
methotrexate is approved for the treatment of psoriatic arthritis, rheumatoid arthritis,
and JIA.
The
onset of treatment response in psoriasis is rapid, being significant at 2 weeks
and maximal between weeks 12 and 16. PASI‐75 and PASI‐90
at 16 weeks are seen in 80% and 50%, respectively, of patients receiving the
licensed dose (adalimumab 80 mg at week 0 then 40 mg every other week beginning
at week 1). Adalimumab has shown greater efficacy after 16 weeks of treatment
than methotrexate (7.5 mg/week initial dose increasing to a maximum of 25
mg/week as tolerated). Adalimumab is effective for chronic plaque psoriasis of
the hands and feet. Adalimumab efficacy was well maintained over more than 3
years of continuous therapy for patients with sustained initial PASI-75
responses (i.e., 75% improvement in the Psoriasis Area and Severity Index
score). Many dermatologist prescribed adalimumab as first-line treatment for
psoriasis.
Side effects
Side effects shared with other TNF inhibitors, including risk of
infection with mycobacteria and fungi. Moderately painful injection site reactions
are noted in up to 15% of patients. These reactions usually resolve
spontaneously within the first 2 months of therapy.
Interactions
The
concomitant administration of other targeted immune modulators with adalimumab
may increase the risk of infection and should be avoided.
Use in pregnancy
Adalimumab is classified as
pregnancy category B. Its safety during lactation is unknown.
Infliximab
It is a chimeric monoclonal antibody that has high
specificity, affinity, and avidity for TNF alfa.
Indication
Infliximab is approved for the treatment of adults with chronic
severe plaque-type psoriasis. Compared with the other TNF inhibitors, its onset
of action tends to be faster and a higher proportion of patients (~75–85%)
achieve a 75% reduction in the PASI score. Infliximab is also approved for the
treatment of adults with psoriatic arthritis, Crohn disease (including
associated fistulas), ulcerative colitis, rheumatoid arthritis and ankylosing
spondylitis, as well as children ≥6 years of age with Crohn disease and
ulcerative colitis. Infliximab also has efficacy in psoriatic
nail disease.
Dosages
In patients with plaque
psoriasis, the
drug is given by slow intravenous infusion over 2 hours and dose is based on
body weight (5-10 mg/kg at weeks 0, 2 and 6 and then every 8 weeks). A response
is often seen after the first infusion, and after an induction phase of three
infusions 80% of patients at 5mg/kg dose and 90% of patients at 10mg/kg dose
achieves PASI‐75.
This rapid and reliable response makes the drug particularly suitable in urgent
circumstances such as erythrodermic psoriasis and generalized pustular
psoriasis. In the maintenance phase, efficacy is maintained to 50 weeks. Neutralizing anti-chimeric antibodies have been
reported in a high proportion of individuals treated with infliximab, and
correlate with lower serum drug levels, loss of efficacy and the development of
infusion reactions; the concurrent administration of
low-dose weekly methotrexate may help to prevent the formation of ADA.
Specific side effects
Infusion-related reactions and
serum sickness are the most commonly reported side effect of infliximab. Infusion-related reactions occur in 15% of patients treated.
Most are mild, consisting of pruritus or urticaria. Some patients will have
moderate reactions consisting of chest pain, hypertension, and shortness of
breath and only rarely will severe reactions with hypotension and anaphylaxis
occur. Infusion reaction risk has been linked to the presence of human
anti-chimeric antibodies. Risk of reactions
can usually be managed by slowing the rate of infusion or stopping treatment
entirely or concomitant use of methotrexate
Interactions
Infliximab should not be
administered together with other targeted immune modulators due to the
potential for additive immunosuppression.
Use in pregnancy
Infliximab is rated pregnancy
category B. However, because it crosses the placenta and can lead to an
increased risk of infection in the infant, use in pregnancy should be avoided
unless the benefits outweigh the risks. It is not known whether infliximab is
secreted in breast milk, and its administration to nursing mothers is not
recommended.
Etanercept
Etanercept,
a fully human soluble TNF‐α receptor fusion protein that binds and
neutralizes TNF‐α. Response is of slower onset than with
infliximab, becoming evident after 4–8 weeks of treatment.
Indications
Etanercept
has been approved for the treatment of moderate to severe plaque-type psoriasis
in adults since 2004 and in children ages 4–17 years since 2016. Depending on
the regimen (see below), ~30–60% of patients achieve a 75% improvement in their
Psoriasis Area and Severity Index (PASI) score; significant decreases in
fatigue and depression have also been described. Other approved indications for
etanercept include rheumatoid arthritis, psoriatic arthritis and ankylosing
spondylitis in adults as well as juvenile idiopathic arthritis (JIA) in
children ≥2 years of age.
Dosages
The dosing of etanercept differs in psoriasis
than for its other indications. In rheumatoid and psoriatic arthritis,
etanercept are often used in combination with methotrexate. In psoriasis, all
clinical studies have been performed with etanercept as monotherapy. Rebound
does not typically occur when etanercept is discontinued. The approved dose for moderate to severe plaque psoriasis
is 50 mg twice/week given subcutaneously for 3 months followed by 50 mg
once/week. Some physicians choose to initiate therapy
at a dose of 50 mg weekly and only escalate the dose if the patient fails to
respond after 6 to 12 months. The recommended dose for pediatric psoriasis is
0.8 mg/kg (maximum 50 mg) weekly.
Specific side effects
Injection
site reactions are the most common side effect of etanercept. Mean duration of
reactions is 3 to 5 days; these reactions generally occur in the first month
and subsequently decrease. Erythema, pruritus, pain,
and swelling have been described, and “recall” reactions at sites of previous
injections have been reported. These reactions do not progress to anaphylaxis.
The needle cover of the prefilled etanercept syringe contains latex so this
formulation should not be used in latex-sensitive patients. Loss of efficacy
over time may occur. This is possibly related to the development of
antibodies. Pooled clinical trial data
demonstrate that etanercept is a safe drug even when used at higher doses or
extended periods of time.
Interactions
The
concomitant use of etanercept and methotrexate is approved for the treatment of
rheumatoid arthritis and psoriatic arthritis. Due to the potential for
increased immunosuppression, combination of etanercept with other targeted
immune modulators should generally be avoided. Etanercept has been used together
with narrowband UVB without an increase in adverse events and with added
therapeutic benefit.
Use in pregnancy
Etanercept
is pregnancy category B. Studies in pregnant women have not been conducted. It
has been suggested that etanercept and other TNF inhibitors be avoided after
the first trimester. It is not known whether etanercept is secreted in breast
milk; however, in general, this drug is not absorbed via an oral route.
Golimumab
Golimumab
is a human recombinant IgG1 monoclonal antibody with specificity for human TNF.
It is approved for the treatment of psoriatic arthritis, rheumatoid arthritis,
ankylosing spondylitis, and ulcerative colitis; it can be administered along
with methotrexate. It is supplied in a 50 mg prefilled syringe or autoinjector
that is administered subcutaneously monthly for psoriatic arthritis.
Certolizumab
Certolizumab
pegol is a pegylated Fab fragment of a humanized monoclonal antibody with
specificity for human TNF. It is approved for the treatment of psoriatic
arthritis, rheumatoid arthritis, and Crohn disease. It is supplied in a 200 mg
prefilled syringe and administered subcutaneously. For psoriatic arthritis,
400 mg is given at weeks 0, 2 and 4, followed by 200 mg every 2 weeks or 400 mg
monthly for maintenance.
Biologics that target cytokines interleukin-12 and
interleukin-23
Psoriasis
results in part from the activation and migration of T cells into the epidermis
and the release of inflammatory mediators, which leads to hyper proliferation
of skin cells. The cytokines interleukin-12 (IL-12) and IL-23 are produced by
activated antigen-presenting cells and have a role in the differentiation and
proliferation of type 1 T-helper cells. There is evidence of high levels of
IL-12 and IL-23 expression in psoriatic lesions.
Site
of action of ustekinumab (anti-p40) and IL-23 inhibitors (anti- p19)
Ustekinumab and Guselkumab
Ustekinumab is a monoclonal antibody that
targets IL-12 and IL-23, whereas guselkumab is a monoclonal antibody that
target IL-23.
Mechanism of action
Ustekinumab is a human IgG1 monoclonal antibody that targets
interleukin-12 (IL-12) and IL-23. It binds to the p40 subunit, common to both
IL-12 and IL-23, which prevents these cytokines from binding to the cell
surface of T cells, thereby disrupting the inflammatory cascade implicated in
psoriasis.
Guselkumab is also a human monoclonal antibody (IgG1lamda) that
selectively targets the p19 subunit of IL-23. This is a highly effective
biological agent for psoriaisis.
Indications
Ustekinumab and guselkumab are approved for the treatment of adults with moderate to severe plaque-type psoriasis, with ~65–80% and ~80–90% of patients achieving a 75% improvement in their PASI score. In addition to psoriasis, ustekinumab is also approved for the treatment of various aspects of psoriatic arthritis (peripheral and axial disease, dactylitis, enthesitis) and Crohn disease.
For the treatment of moderate-to-severe plaque psoriasis, ustekinumab may be more efficacious than adalimumab and etanercept, but not infliximab. Ustekinumab produces a stable clinical response over time, with no rebound after withdrawal of the drug and a retreatment PASI-75 response of approximately 85%. Although approved for psoriatic arthritis (PsA), in some patients PsA may worsen on ustekinumab. A small percentage of patients develop anti-ustekinumab antibodies and it is not known if these antibodies affect clinical outcomes.
Dosages
For the treatment of psoriasis, ustekinumab is administered as a
subcutaneous injection of 45 mg for patients who weigh 100 kg or less, and 90 mg
for patients who weigh more than 100 kg. An initial dose is administered
subcutaneously at week 0, followed by another dose at week 4, and then a
further dose every 12 weeks. No dose adjustment is needed for patients older
than age 65. It is not recommended for children younger than age 18 because of
a lack of data. At week 12, a PASI-75 response was achieved by 67% of patients
receiving 45 mg of ustekinumab and 75% of patients receiving 90 mg of
ustekinumab. Consider increasing dose to
once every 8 weeks with ustekinumab 90 mg in patients who do not achieve
PASI-75 at 28 weeks.
At week 16, 80% of patients on guselkumab achieve a PASI 75 and
73% a PASI 90.
Contraindications
Ustekinumab and IL-23 inhibitors
are contraindicated in patients with known sensitivity to the agents, and they
should be avoided in patients with serious active infections or malignancies.
Major side effects
Ustekinumab
is associated with an increased incidence of mucocutaneous candidiasis (~5% of
patients) and a potential risk of severe and disseminated infections with
mycobacteria and Salmonella. These particular infections
mirror those seen in patients with genetic deficiencies in the IL-12/IL-23
signaling pathway. Assessment for tuberculosis is required at baseline and
typically repeated annually during therapy with all IL-12/23 or 23 inhibitors.
Patients with untreated latent tuberculosis should receive antituberculous
therapy prior to beginning treatment.
A
possible excess of major adverse cardiovascular events (MACEs) such as
myocardial infarction or stroke was noted in patients who received
anti-IL-12/23 therapy in individual clinical trials, but meta-analyses and a
5-year follow-up study did not find a significant increase in the risk of MACEs
associated with the use of these agents. Although the effects of IL-12/23
inhibitors on vascular inflammation and thrombosis, especially early in the
course of therapy, remain to be determined, cardiovascular risk factors should
be assessed prior to initiating treatment. Injection site reactions can occur
but are less frequent than with the TNF inhibitors. Retiform purpura
progressing to cutaneous necrosis on the leg was reported in a patient treated
with ustekinumab.
Interactions
Live
vaccines should be avoided. The response to vaccines may be muted. Other
targeted immune modulators should not be administered together with ustekinumab
or IL-23 inhibitors. The concomitant administration of other immunosuppressive
agents might increase the risk of infection.
Use in pregnancy
Ustekinumab is pregnancy category
B; guselkumab was approved after June of 2015 and therefore was not labeled
with a pregnancy category. The safety of these drugs during lactation is
unknown.
Recommendations for Ustekinumab
Indication: Moderate-to-severe
plaque psoriasis in adults
Dosing: 45 mg for people who weigh 100 kg or less, and
90 mg for people who weigh more than 100 kg. An initial dose is administered
subcutaneously at week 0, followed by another dose at week 4, and then a
further dose every 12 weeks. No dose adjustment is needed for patients older
than age 65. It is not recommended for children younger than age 18 because of
a lack of data.
Short-term response: At week 12, a
PASI-75 response was achieved by 67% of patients receiving 45 mg of
ustekinumab, 66% of patients receiving 90 mg of ustekinumab.
Long-term response: Maintenance of
PASI-75 was significantly superior with continuous ustekinumab treatment
compared with treatment withdrawal; at week 76, 84% of patients rerandomized to
maintenance treatment achieved a PASI-75 response compared with 19% of patients
rerandomized to placebo.
Increasing dose: Consider
increasing dose to once every 8 weeks with ustekinumab 90 mg in patients who do
not achieve PASI-75 at 28 weeks.
Toxicity
·
The most common adverse reactions (>10%) are
nasopharyngitis and upper respiratory tract infection. Most were considered
mild and did not necessitate drug discontinuation.
·
Contraindications include clinically important
active infection.
·
Reports of
serious adverse events, including serious infections, malignancies, and cardiovascular
events, were low.
Baseline monitoring
1.
PPD and consider chest x-ray
2.
LFT, CBC, and hepatitis profile
Ongoing monitoring
3.
Consider yearly PPD and periodic CBC and LFT
4.
Periodic history and physical examination
Interleukin-17 Inhibitors
Plaque psoriasis arises in part because of activation of autoimmune CD4+ and CD8+ Th17 cells that proliferate and target the epidermis where they interact with keratinocyte (keratin 7 and LL-37) and melanocyte (ADAMTSL, a disintegrin and metalloproteinase with thrombospondin motifs like 5) auto antigens and produce IL-17 and IL-22. Currently, there are three biological agents targeting the IL-17 pathway: secukinumab and ixekizumab, which target IL-17A, and brodalumab, which blocks the IL-17 receptor A (IL-17R). These biological agents are among the most effective for the treatment of moderate to severe plaque psoriasis. These agents are relatively fast-acting with results as fast as 4 weeks.
Site
of action of IL-17 inhibitors
Contraindications
Each IL-17 inhibitor is
contraindicated in patients with a known hypersensitivity to that particular
agent or components of its formulation. These medications should be used with
caution in patients with chronic/recurrent infections or inflammatory bowel
disease, with Crohn disease representing a contraindication for brodalumab.
Administration of an IL-17 inhibitor should be avoided or discontinued in
patients with a serious active infection.
Major side effects
The most
common adverse events in clinical trials of IL-17 inhibitors were
nasopharyngitis, upper respiratory tract infections, and injection-site
reactions. Hypersensitivity reactions have also been reported, including
anaphylaxis, angioedema, and urticaria. Mucocutaneous candidiasis, most often
oral or vulvovaginal, develops in ~5% of patients; this reflects the important
role of IL-17 in defense against Candida. These
infections are typically mild or moderate and resolve with standard treatment.
Neutropenia (<1500 cells/mm3) occurs in ~1–2%
of patients, which is similar to the frequency in patients receiving
etanercept. Assessment for tuberculosis is required at baseline and typically
repeated annually during therapy. Patients with untreated latent tuberculosis
should receive antituberculous therapy prior to beginning treatment.
New onset and exacerbation of
Crohn disease and ulcerative colitis have occurred in patients receiving IL-17
inhibitors. An increased risk of adverse cardiovascular events was not observed
in clinical trials.
Interactions
Live
vaccines should be avoided. To avert a potential increased risk of infection,
other targeted immune modulators should not be coadministered with IL-17
inhibitors.
Use in pregnancy
Secukinumab is pregnancy category
B; the other IL-17 inhibitors were approved after June of 2015 and therefore
were not labeled with a pregnancy category. There are no available data on
IL-17 inhibitor use in pregnant women, although human IgG is known to cross the
placenta. Safety during lactation is unknown.
Secukinumab
Mechanism of action
Secukinumab is a recombinant human IgG1κ monoclonal antibody that binds to IL-17A and blocks its interaction with the IL-17A receptor.
Indications
Secukinumab
is approved for moderate to severe plaque psoriasis in adults. After 12 weeks
of treatment, approximately 77% and 67% of patients treated with the 300 mg and
150 mg doses, respectively, achieve a 75% improvement in their PASI score. This
response is durable with 76% of patients achieving a 90% reduction in PASI at
52 weeks. It is also approved for psoriatic arthritis and ankylosing spondylitis.
Dosages
Secukinumab is administered as a
subcutaneous injection of 300 mg at weeks 0, 1, 2, 3, and 4; thereafter, it is
given every 4 weeks. A dose of 150 mg may be considered for patients weighing
<90 kg. It is available in 150 mg prefilled syringes and autoinjectors.
Ixekizumab
Mechanism of action
Ixekizumab
is a humanized IgG4 monoclonal antibody that binds and inhibits IL-17A,
resulting in neutralization of IL-17A homodimers and IL-17A/F heterodimers. In
its hinge region, there is substitution of proline for serine which prevents
the formation of half-antibodies (half-mers) that can undergo Fab-arm exchange
with endogenous human IgG4.
Indications
Ixekizumab
is approved for the treatment of moderate to severe plaque-type psoriasis and PsA.
After 12 weeks, 90% of patients are capable of achieving a PASI 75, making Ixekizumab
one of the most effective biological agents for psoriasis.
Dosages
Ixekizumab is administered as a
subcutaneous injection, with a loading dose of 160 mg followed by 80 mg every 2
weeks for 12 weeks and then every 4 weeks. It is supplied in 80 mg prefilled
syringes and autoinjectors.
Brodalumab
Mechanism of action
Brodalumab
is a human IgG2κ monoclonal antibody that
selectively binds the IL-17 receptor A, inhibiting its interactions with
IL-17A/F and IL-17E (also known as IL-25).
Indications
Brodalumab
is approved for the treatment of moderate to severe psoriasis in adults who
have failed or become unresponsive to other systemic therapies. Approximately
80–85% and 60–70% of patients treated with the 210 mg and 140 mg doses,
respectively, achieve a 75% reduction in their PASI score. This is a very
effective biological agent for psoriasis, however, there is a concern about suicidal
ideation and completed suicides in clinical trials.
Dosages
Brodalumab is available in
prefilled syringes and administered as a 210 mg subcutaneous injection every 2
weeks.
PHOSPHODIESTERASE
4 INHIBITOR
A novel,
small-molecule inhibitor of phosphodiesterase 4, apremilast works
intracellularly to reduce the production of proinflammatory mediators and
increase those that are anti-inflammatory. When administered orally, apremilast
is 70–75% bioavailable, with peak plasma concentrations observed at ~2.5 hours.
Nearly 70% of the drug is bound to plasma proteins, and it is metabolized by
cytochrome P450 (CYP) enzymes, predominately CYP3A4; this is followed by
glucuronidation and non-CYP-mediated hydrolysis. Apremilast has a terminal
elimination half-life of 6–9 hours and is excreted in the urine and feces.
Mechanism of Action
Apremilast
inhibits phosphodiesterase 4, which is an intracellular enzyme that degrades
cAMP and represents the predominant phosphodiesterase in keratinocytes,
dendritic cells, monocytes, and neutrophils. Increasing intracellular cAMP
levels activates protein kinase A, leading to enhanced expression of several
transcription factors including cAMP-response element binding protein (CREB), while inhibiting others such as
nuclear factor kappa B (NF-κB). By inhibiting
phosphodiesterase 4 and increasing intracellular cAMP levels, apremilast has
multiple downstream effects: it decreases the production of inflammatory
mediators such as TNF-α, IFN-γ, and interleukins (IL)-2, -12, and -23; it increases
the production of anti-inflammatory mediators including IL-10; and it inhibits
natural killer responses.
Dosages
The recommended dosage for
psoriatic arthritis and psoriasis is 30 mg twice daily. In order to reduce
gastrointestinal symptoms, an upward titration of the dose by 10 mg/day is
recommended, starting with an initial dose of 10 mg/day. Apremilast is
available in 10, 20 and 30 mg tablets. For individuals with severe renal
impairment, the recommended maximum daily dose is 30 mg; there is no dosing
adjustment for hepatic impairment.
Major Side Effects
The most common side effects are gastrointestinal,
especially nausea, vomiting and diarrhea, and they are most evident during the first 15
days of administration and gradually resolve over several weeks. Headache and
nasopharyngitis are additional potential side effects. Because depression,
including suicidal ideation, can be observed in up to 1% of patients, patients
with a history of depression should be monitored closely. Loss of ~5–10% of
body weight occurs in ~10% of patients. No laboratory monitoring is
recommended, but serial measurements of weight can be performed.
Indications
Apremilast is FDA-approved for the treatment of adult patients with active psoriatic arthritis and patients with moderate to severe plaque psoriasis who are candidates for phototherapy or systemic therapy. At week 16, only 33.1% of patients receiving apremilast (30 mg BID) achieve a PASI 75. It is currently being investigated for other inflammatory skin diseases including discoid LE, lichen planus, granulomatous dermatoses, and atopic dermatitis.
Contraindications
Apremilast is contraindicated in
patients with known hypersensitivity to the drug or its components. Dosing should
be adjusted for renal failure. Relative contraindications include a history of
depression or suicidal ideation.
Use in Pregnancy and Lactation
Apremilast was formerly pregnancy
category C. However, it has not been evaluated in well-controlled studies involving
pregnant women and should only be used during pregnancy if the potential
benefit justifies the potential risk. It is not known whether apremilast or its
metabolites are present in human milk.
Drug Interactions
Apremilast is a strong cytochrome p450 inducer and should not be used with drugs metabolized by this pathway such as rifampin, phenobarbital, carbamazepine, and phenytoin.
MANAGEMENT OF PSORIASIS AT
SPECIFIC SITES |
|
Specific sites |
Special considerations and practical
treatments |
Scalp |
Remove
scale with topical 10–15% salicylic acid Apply
potent or ultrapotent corticosteroids in a lotion, gel, foam or shampoo
formulation, either alone or in combination with calcipotriene |
Face; groin, axilla, and other body folds |
First-line:
mild topical corticosteroids Second-line:
topical calcineurin inhibitors or tacalcitol/calcitriol in combination
with a mild topical corticosteroid |
Nail |
Differentiate
proximal versus distal nail unit pathology due to nail matrix versus nail bed
pathology, respectively; consider coexisting onychomycosis For distal
nail unit pathology: vitamin D3 analogues
topically (limited evidence) For
proximal pathology: intralesional corticosteroids and/or systemic
antipsoriatic therapies |
How to combine treatments
Enhanced clinical response and a
possible reduction in side effects are the goals of combination therapy.
However, for safety reasons, some combinations are contraindicated.
Beneficial combinations
For
topical treatments, the combination of calcipotriene with super potent topical
corticosteroids has been shown to be more effective than either treatment as
monotherapy, whereas the combination of calcipotriene plus medium-strength
corticosteroids did not have a substantial additive effect compared with each
as monotherapy. However, the irritation caused by calcipotriene can be reduced
by the addition of medium-strength corticosteroids. Improved efficacy has also
been observed with the combination of calcipotriene–cyclosporine and
calcipotriene–acitretin, compared with each as monotherapy. For example, with
low-dose cyclosporine (2 mg/kg/day) plus topical calcipotriene, marked
improvement or clearing was seen in 90% of patients. In a dose-escalating study
of acitretin, the combination of acitretin and calcipotriene was more effective
and permitted a lower dose of both acitretin and calcipotriene as compared to
each as monotherapy. The combination of calcipotriene and PUVA is characterized
by a marked reduction in the cumulative dose of UVA required and enhanced
efficacy, compared with the respective monotherapies. However, evidence for a
beneficial effect of the combination of UVB and calcipotriene is less
convincing. Combining etanercept (25 mg subcutaneously once weekly) and
acitretin has been shown to be as effective as etanercept 25 mg subcutaneously
twice weekly.
Topical corticosteroids are
frequently combined with other antipsoriatic treatments. Although increases in
the duration of subsequent remission periods have been noted, controlled
studies are required to substantiate these claims. Depending upon the anatomic
site, it is advisable to prescribe daily potent or medium-strength
corticosteroids for a maximum of 4–8 weeks. Then intermittent schedules (e.g.
2–3 applications per week) can be considered.
Contraindicated combinations
and combinations with restricted use
The
combination of acitretin and cyclosporine carries the risk of accumulation of
cyclosporine, because cyclosporine is inactivated by the cytochrome P450
system, which is inhibited by acitretin. An increased occurrence of SCCs has
been observed in patients treated with the combination of cyclosporine and
PUVA, either simultaneously or sequentially (PUVA followed by cyclosporine). As
a result, the combination of cyclosporine and PUVA is contraindicated. Although
a combination of methotrexate and PUVA has been reported to be safe, long-term
data are not available.
The
combination of coal tar with PUVA is contraindicated as it may induce
significant phototoxic responses. The combination of cyclosporine and
methotrexate has been viewed as high risk because both are immunosuppressants.
However, this combination has been used with success by rheumatologists, and,
in recalcitrant psoriasis, the combination has proved to be very effective
without major side effects. The combination of methotrexate and acitretin has
been used in those patients in whom all treatments have failed. Although this
combination can be very effective, severe hepatotoxicity has been reported;
therefore, careful monitoring is mandatory.
How to manage childhood psoriasis
Topical calcipotriene is considered as first-line therapy
for mild to moderate juvenile psoriasis, combined with mild- to
moderate-strength topical corticosteroids (if necessary). For
treatment-resistant flexural and/or facial psoriasis, tacrolimus 0.1% ointment can
be added to the treatment regimen. The next step is to recommend treatment with
narrowband UVB, especially in adolescents. Although controversial, the use of
antibiotics can be considered in patients with guttate psoriasis where there is
suspicion of a streptococcal infection. Of the systemic medications,
methotrexate is regarded as the treatment of choice, with retinoids considered
in pustular and erythrodermic psoriasis. Cyclosporine is occasionally utilized
for exceptional cases, and etanercept should be considered as a third-line drug
for therapy-resistant psoriasis.
TREATMENT LADDER
Mild
plaque psoriasis without psoriatic arthritis
First line
·
Coal tar
·
Potent topical corticosteroid
·
Vitamin D analogue
Second line
·
Local NB‐UVB or PUVA
·
Excimer laser
Moderate
to severe plaque psoriasis without psoriatic arthritis
First line
·
NB‐UVB or PUVA
Second line
·
Acitretin
·
Ciclosporin
·
Methotrexate
Third line
·
Adalimumab
·
Etanercept
·
Infliximab
·
Secukinumab
·
Ustekinumab
Moderate to severe plaque psoriasis with psoriatic arthritis
First line
·
Apremilast
·
Methotrexate
Second line
·
Adalimumab
·
Etanercept
·
Infliximab
·
Ustekinumab
Third line
·
Combination therapy
Future Developments and Treatments
To date, a stepwise approach is
the treatment paradigm for psoriasis, starting with a topical agent, then
phototherapy or a classic systemic medication, and ultimately a “biologic”
therapy. It remains to be determined whether the introduction of even more
targeted biologic therapies (e.g. inhibitors of IL-17A, IL-23, and the IL-17
receptor) will influence this paradigm. In the future, there will be additional
targeted biologic therapies (e.g. inhibitors of IL-20, IL-22, GM-CSF), topical
or oral small molecules (e.g. JAK1/2 inhibitors, protein kinase C inhibitors,
p38 kinase inhibitors), and oligonucleotides that target Toll-like receptors 7,
8 and 9 or function as a STAT3 decoy. Hopefully, the future development of
biomarkers that predict individual responses to different therapies will
provide a more cost-effective approach to therapy.
Conclusion
Psoriasis
is a complex disease. Three way interactions between adaptive immunity, innate
immunity and skin barrier defect may best explain the pathophysiology of
psoriasis. That the role of adaptive immunity is crucial to the development of
psoriasis is beyond doubt, as validated by the successful use of biologic
response modifiers. However, there is increasing evidence on the role played by
innate immunity and the skin barrier function in initiating and perpetuating
inflammation in psoriasis. This is likely to open up newer targets for
therapeutic interventions.
French guidelines on the use of systemic treatments for
moderate-to-severe psoriasis in adults, 2019
Treatment algorithm for patients suffering from plaque psoriasis
without comorbidities. CSA, cyclosporin; MTX, methotrexate; NBUVB, Narrowband
UVB; PUVA, psoralen UVA; re-PUVA, retinoid psoralen UVA